dostęp otwarty

Tom 3, Nr 3 (2018)
Wytyczne / stanowisko ekspertów
Opublikowany online: 2018-10-31
Pobierz cytowanie

Diagnostyka i leczenie niepłodności — rekomendacje Polskiego Towarzystwa Medycyny Rozrodu i Embriologii (PTMRiE) oraz Polskiego Towarzystwa Ginekologów i Położników (PTGP)

Krzysztof Łukaszuk, Katarzyna Kozioł, Grzegorz Jakiel, Artur Jakimiuk, Piotr Jędrzejczak, Waldemar Kuczyński, Rafał Kurzawa, Leszek Pawelczyk, Michał Radwan, Robert Spaczyński, Mirosław Wielgoś, Sławomir Wołczyński
Ginekologia i Perinatologia Praktyczna 2018;3(3):112-140.

dostęp otwarty

Tom 3, Nr 3 (2018)
Rekomendacje
Opublikowany online: 2018-10-31

Streszczenie

Brak

Streszczenie

Brak
Pobierz cytowanie
Informacje o artykule
Tytuł

Diagnostyka i leczenie niepłodności — rekomendacje Polskiego Towarzystwa Medycyny Rozrodu i Embriologii (PTMRiE) oraz Polskiego Towarzystwa Ginekologów i Położników (PTGP)

Czasopismo

Ginekologia i Perinatologia Praktyczna

Numer

Tom 3, Nr 3 (2018)

Typ artykułu

Wytyczne / stanowisko ekspertów

Strony

112-140

Opublikowany online

2018-10-31

Wyświetlenia strony

4904

Wyświetlenia/pobrania artykułu

39590

Rekord bibliograficzny

Ginekologia i Perinatologia Praktyczna 2018;3(3):112-140.

Autorzy

Krzysztof Łukaszuk
Katarzyna Kozioł
Grzegorz Jakiel
Artur Jakimiuk
Piotr Jędrzejczak
Waldemar Kuczyński
Rafał Kurzawa
Leszek Pawelczyk
Michał Radwan
Robert Spaczyński
Mirosław Wielgoś
Sławomir Wołczyński

Referencje (342)
  1. Mascarenhas M, Flaxman S, Boerma T, et al. National, Regional, and Global Trends in Infertility Prevalence Since 1990: A Systematic Analysis of 277 Health Surveys. PLoS Medicine. 2012; 9(12): e1001356.
  2. Janicka A, Spaczyński R, Kurzawa R, et al. Assisted reproductive medicine in Poland – Fertility and Sterility Special Interest Group of the Polish Gynaecological Society (SPiN PTG) 2012 report. Polish Gynaecology. 2015; 86(12).
  3. Boivin J, Takefman J, Braverman A. The fertility quality of life (FertiQoL) tool: development and general psychometric properties†. Human Reproduction. 2011; 26(8): 2084–2091.
  4. Births, Marriages, and Deaths. BIRTHS. The Lancet. 1888; 131(3369): 605.
  5. Female age-related fertility decline. Fertility and Sterility. 2014; 101(3): 633–634.
  6. Broekmans FJ, Soules MR, Fauser BC. Ovarian aging: mechanisms and clinical consequences. Endocr Rev. 2009; 30(5): 465–493.
  7. Diagnostic evaluation of the infertile female: a committee opinion. Fertility and Sterility. 2015; 103(6): e44–e50.
  8. Current clinical irrelevance of luteal phase deficiency: a committee opinion. Fertility and Sterility. 2015; 103(4): e27–e32.
  9. Diagnostic evaluation of the infertile male: a committee opinion. Fertility and Sterility. 2015; 103(3): e18–e25.
  10. Smoking and infertility: a committee opinion. Fertility and Sterility. 2012; 98(6): 1400–1406.
  11. Practice Committee of the American Society for Reproductive Medicine in collaboration with the Society for Reproductive Endocrinology and Infertility. Electronic address: ASRM@asrm.org, Practice Committee of the American Society for Reproductive Medicine in collaboration with the Society for Reproductive Endocrinology and Infertility, Practice Committee of American Society for Reproductive Medicine in collaboration with Society for Reproductive Endocrinology and Infertility. Optimizing natural fertility: a committee opinion. Fertil Steril. 2013; 100(3): 631–637.
  12. Hart R. Physiological Aspects of Female Fertility: Role of the Environment, Modern Lifestyle, and Genetics. Physiological Reviews. 2016; 96(3): 873–909.
  13. Souter I, Baltagi LM, Kuleta D, et al. Women, weight, and fertility: the effect of body mass index on the outcome of superovulation/intrauterine insemination cycles. Fertil Steril. 2011; 95(3): 1042–1047.
  14. Wischmann T, Korge K, Scherg H, et al. A 10-year follow-up study of psychosocial factors affecting couples after infertility treatment. Hum Reprod. 2012; 27(11): 3226–3232.
  15. Gameiro S, Boivin J, Dancet E, et al. ESHRE guideline: routine psychosocial care in infertility and medically assisted reproduction-a guide for fertility staff. Hum Reprod. 2015; 30(11): 2476–2485.
  16. Ecochard R, Boehringer H, Rabilloud M, et al. Chronological aspects of ultrasonic, hormonal, and other indirect indices of ovulation. BJOG. 2001; 108(8): 822–829.
  17. ROBERTS J, SPANDORFER S, FASOULIOTIS S, et al. Taking a basal follicle-stimulating hormone history is essential before initiating in vitro fertilization. Fertility and Sterility. 2005; 83(1): 37–41.
  18. Arce JC, Marca ALa, Klein BM, et al. Antimüllerian hormone in gonadotropin releasing-hormone antagonist cycles: prediction of ovarian response and cumulative treatment outcome in good-prognosis patients. Fertility and Sterility. 2013; 99(6): 1644–1653.e1.
  19. Chalazonitis A, Tzovara I, Laspas F, et al. Hysterosalpingography: technique and applications. Curr Probl Diagn Radiol. 2009; 38(5): 199–205.
  20. Dreyer K, Rijswijk Jv, Mijatovic V, et al. Oil-Based or Water-Based Contrast for Hysterosalpingography in Infertile Women. New England Journal of Medicine. 2017; 376(21): 2043–2052.
  21. Spring D, Barkan H, Pruyn S. Potential Therapeutic Effects of Contrast Materials in Hysterosalpingography: A Prospective Randomized Clinical Trial. Radiology. 2000; 214(1): 53–57.
  22. Maheux-Lacroix S, Boutin A, Moore L, et al. Hysterosalpingosonography for diagnosing tubal occlusion in subfertile women: a systematic review with meta-analysis. Human Reproduction. 2014; 29(5): 953–963.
  23. Emanuel MH, Vliet Mv, Weber M, et al. First experiences with hysterosalpingo-foam sonography (HyFoSy) for office tubal patency testing. Human Reproduction. 2011; 27(1): 114–117.
  24. Emanuel MH, Exalto N. Hysterosalpingo-foam sonography (HyFoSy): a new technique to visualize tubal patency. Ultrasound in Obstetrics & Gynecology. 2011; 37(4): 498–499.
  25. Dreyer K, Out R, Hompes P, et al. Hysterosalpingo-foam sonography, a less painful procedure for tubal patency testing during fertility workup compared with (serial) hysterosalpingography: a randomized controlled trial. Fertility and Sterility. 2014; 102(3): 821–825.
  26. Smit J, Kasius J, Eijkemans M, et al. Hysteroscopy before in-vitro fertilisation (inSIGHT): a multicentre, randomised controlled trial. The Lancet. 2016; 387(10038): 2622–2629.
  27. Armstrong S, Showell M, Stewart E, et al. Baseline anatomical assessment of the uterus and ovaries in infertile women: a systematic review of the evidence on which assessment methods are the safest and most effective in terms of improving fertility outcomes. Human Reproduction Update. 2017; 23(5): 533–547.
  28. Berker B, Şükür Y, Aytaç R, et al. Infertility work-up: To what degree does laparoscopy change the management strategy based on hysterosalpingography findings? Journal of Obstetrics and Gynaecology Research. 2015; 41(11): 1785–1790.
  29. Preutthipan S, Linasmita V. A prospective comparative study between hysterosalpingography and hysteroscopy in the detection of intrauterine pathology in patients with infertility. J Obstet Gynaecol Res. 2003; 29(1): 33–37.
  30. Maiti GD, Lele P. Hysterosalpingography (HSG), hysteroscopy and laparoscopic evaluation of female genital tract of patient attending tertiary infertility centre and correlation of various modalities. International Journal of Reproduction, Contraception, Obstetrics and Gynecology. 2018; 7(4): 1597.
  31. Role of tubal surgery in the era of assisted reproductive technology: a committee opinion. Fertility and Sterility. 2015; 103(6): e37–e43.
  32. Kalra GS, Campbell S, Nargund G. Ovarian reserve may be compromised after adnexal surgery: Are we sufficiently fertility- focused in our surgical training? Facts Views Vis Obgyn. 2016; 8(2): 104–108.
  33. Dohle G. Infertility. Oxford Medicine Online. 2017.
  34. Jedrzejczak P, Taszarek-Hauke G, Hauke J, et al. Prediction of spontaneous conception based on semen parameters. Int J Androl. 2008; 31(5): 499–507.
  35. Poore RE, Jarow JP, Jarow JP. Transrectal ultrasonography of infertile men. Fertil Steril. 1993; 60(6): 1035–1039.
  36. Mehta A, Jarow JP, Maples P, et al. Defining the. Journal of Andrology. 2012; 33(5): 917–920.
  37. Talarczyk-Desole J, Berger A, Taszarek-Hauke G, et al. Manual vs. computer-assisted sperm analysis: can CASA replace manual assessment of human semen in clinical practice? Ginekologia Polska. 2017; 88(2): 56–60.
  38. World Health Organization (WHO). SpringerReference. .
  39. Cui D, Han G, Shang Y, et al. Antisperm antibodies in infertile men and their effect on semen parameters: A systematic review and meta-analysis. Clinica Chimica Acta. 2015; 444: 29–36.
  40. Check ML, Check JH, Katsoff D, et al. ICSI as an effective therapy for male factor with antisperm antibodies. Arch Androl. 2000; 45(3): 125–130.
  41. The clinical utility of sperm DNA integrity testing: a guideline. Fertility and Sterility. 2013; 99(3): 673–677.
  42. Oleszczuk K, Giwercman A, Bungum M. Sperm chromatin structure assay in prediction of in vitro fertilization outcome. Andrology. 2016; 4(2): 290–296.
  43. Cissen M, Wely M, Scholten I, et al. Measuring Sperm DNA Fragmentation and Clinical Outcomes of Medically Assisted Reproduction: A Systematic Review and Meta-Analysis. PLOS ONE. 2016; 11(11): e0165125.
  44. McCallum T, Milunsky J, Munarriz R, et al. Unilateral renal agenesis associated with congenital bilateral absence of the vas deferens: phenotypic findings and genetic considerations. Hum Reprod. 2001; 16(2): 282–288.
  45. McLachlan R, O'Bryan M. State of the Art for Genetic Testing of Infertile Men. The Journal of Clinical Endocrinology & Metabolism. 2010; 95(3): 1013–1024.
  46. Casals T, Bassas L, Egozcue S, et al. Heterogeneity for mutations in the CFTR gene and clinical correlations in patients with congenital absence of the vas deferens. Hum Reprod. 2000; 15(7): 1476–1483.
  47. Popli K, Bourke S, Stewart J. Fertility issues in men with cystic fibrosis: survey of knowledge and opinion of patients. Fertility and Sterility. 2009; 91(4): 1297–1298.
  48. Flannigan R, Schlegel P. Genetic diagnostics of male infertility in clinical practice. Best Practice & Research Clinical Obstetrics & Gynaecology. 2017; 44: 26–37.
  49. De Braekeleer M, Dao TN. Cytogenetic studies in male infertility: a review. Hum Reprod. 1991; 6(2): 245–250.
  50. Dohle GR. Genetic risk factors in infertile men with severe oligozoospermia and azoospermia. Human Reproduction. 2002; 17(1): 13–16.
  51. Tempest HG, Martin RH. Cytogenetic risks in chromosomally normal infertile men. Curr Opin Obstet Gynecol. 2009; 21(3): 223–227.
  52. Foresta C, Garolla A, Bartoloni L, et al. Genetic abnormalities among severely oligospermic men who are candidates for intracytoplasmic sperm injection. J Clin Endocrinol Metab. 2005; 90(1): 152–156.
  53. Leiva R, Bouchard T, Boehringer H, et al. Random serum progesterone threshold to confirm ovulation. Steroids. 2015; 101: 125–129.
  54. Updated NICE guidelines on individualised care for T2DM. PharmacoEconomics & Outcomes News. 2015; 743(1): 11–11.
  55. Melmed S, Casanueva FF, Hoffman AR, et al. Endocrine Society. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011; 96(2): 273–288.
  56. Gordon CM, Ackerman KE, Berga SL, et al. Functional Hypothalamic Amenorrhea: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2017; 102(5): 1413–1439.
  57. Alexander E, Pearce E, Brent G, et al. 2017 Guidelines of the American Thyroid Association for the Diagnosis and Management of Thyroid Disease During Pregnancy and the Postpartum. Thyroid. 2017; 27(3): 315–389.
  58. Carmina E, Fruzzetti F, Lobo R. Features of polycystic ovary syndrome (PCOS) in women with functional hypothalamic amenorrhea (FHA) may be reversible with recovery of menstrual function. Gynecological Endocrinology. 2017; 34(4): 301–304.
  59. Balen A, Morley L, Misso M, et al. The management of anovulatory infertility in women with polycystic ovary syndrome: an analysis of the evidence to support the development of global WHO guidance. Human Reproduction Update. 2016; 22(6): 687–708.
  60. Rotterdam ESHRE/ASRM-Sponsored PCOS consensus workshop group. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS). Hum Reprod. 2004; 19(1): 41–47.
  61. ESHRE Guideline: management of women with premature ovarian insufficiency. Human Reproduction. 2016; 31(5): 926–937.
  62. Dubourdieu S, Fréour T, Dessolle L, et al. Prospective, randomized comparison between pulsatile GnRH therapy and combined gonadotropin (FSH+LH) treatment for ovulation induction in women with hypothalamic amenorrhea and underlying polycystic ovary syndrome. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2013; 168(1): 45–48.
  63. Dumont A, Dewailly D, Plouvier P, et al. Does polycystic ovarian morphology influence the response to treatment with pulsatile GnRH in functional hypothalamic amenorrhea? Reproductive Biology and Endocrinology. 2016; 14(1).
  64. Roque M, Tostes ACI, Valle M, et al. Letrozole versus clomiphene citrate in polycystic ovary syndrome: systematic review and meta-analysis. Gynecol Endocrinol. 2015; 31(12): 917–921.
  65. Tatsumi T, Jwa SC, Kuwahara A, et al. No increased risk of major congenital anomalies or adverse pregnancy or neonatal outcomes following letrozole use in assisted reproductive technology. Human Reproduction. 2016.
  66. Eftekhar M, Deghani Firoozabadi R, Khani P, et al. Effect of Laparoscopic Ovarian Drilling on Outcomes of In Vitro Fertilization in Clomiphene-Resistant Women with Polycystic Ovary Syndrome. Int J Fertil Steril. 2016; 10(1): 42–47.
  67. Balen A. Ovulation induction in the management of anovulatory polycystic ovary syndrome. Molecular and Cellular Endocrinology. 2013; 373(1-2): 77–82.
  68. Nahuis MJ, Lohuis EO, Kose N, et al. Long-term follow-up of laparoscopic electrocautery of the ovaries versus ovulation induction with recombinant FSH in clomiphene citrate-resistant women with polycystic ovary syndrome: an economic evaluation. Human Reproduction. 2012; 27(12): 3577–3582.
  69. Smithson DS, Vause TDR, Cheung AP. No. 362-Ovulation Induction in Polycystic Ovary Syndrome. J Obstet Gynaecol Can. 2018; 40(7): 978–987.
  70. Tanbo T, Mellembakken J, Bjercke S, et al. Ovulation induction in polycystic ovary syndrome. Acta Obstet Gynecol Scand. 2018; 97(10): 1162–1167.
  71. Tremellen KP, Savulescu J, Tremellen K, et al. Ovarian reserve screening: a scientific and ethical analysis. Hum Reprod. 2014; 29(12): 2606–2614.
  72. Marca ALa, Sighinolfi G, Radi D, et al. Anti-Mullerian hormone (AMH) as a predictive marker in assisted reproductive technology (ART). Human Reproduction Update. 2009; 16(2): 113–130.
  73. Gillam M, Molitch M, Lombardi G, et al. Advances in the Treatment of Prolactinomas. Endocrine Reviews. 2006; 27(5): 485–534.
  74. Dotto PD, Bonuccelli U. Clinical Pharmacokinetics of Cabergoline. Clinical Pharmacokinetics. 2003; 42(7): 633–645.
  75. Pascal-Vigneron V, Weryha G, Bosc M, et al. [Hyperprolactinemic amenorrhea:treatment with cabergoline versus bromocriptine. Results of a national multicenter randomized double-blind study]. Presse Med. 1995; 24(16): 753–757.
  76. Dunselman G, Vermeulen N, Becker C, et al. ESHRE guideline: management of women with endometriosis. Human Reproduction. 2014; 29(3): 400–412.
  77. Hughes E, Brown J, Collins JJ, et al. Clomiphene citrate for unexplained subfertility in women. Cochrane Database Syst Rev. 2010(1): CD000057.
  78. Tung KH. Effect of Anovulation Factors on Pre- and Postmenopausal Ovarian Cancer Risk: Revisiting the Incessant Ovulation Hypothesis. American Journal of Epidemiology. 2005; 161(4): 321–329.
  79. Kurman RJ, Shih IM. The origin and pathogenesis of epithelial ovarian cancer: a proposed unifying theory. Am J Surg Pathol. 2010; 34(3): 433–443.
  80. Wynne B. American Society for Reproductive Medicine (ASRM). Encyclopedia of Global Health. .
  81. Humaidan P, Quartarolo J, Papanikolaou E. Preventing ovarian hyperstimulation syndrome: guidance for the clinician. Fertility and Sterility. 2010; 94(2): 389–400.
  82. Devroey P, Polyzos NP, Blockeel C. An OHSS-Free Clinic by segmentation of IVF treatment. Hum Reprod. 2011; 26(10): 2593–2597.
  83. Misso ML, Costello MF, Dokras A, et al. International PCOS Network. International evidence-based guideline for the assessment and management of polycistic ovary syndorme. 2018(3).
  84. Brown J, Farquhar C. Endometriosis: an overview of Cochrane Reviews. Cochrane Database Syst Rev. 2014(3): CD009590.
  85. Practice Committee of the American Society for Reproductive Medicine. Endometriosis and infertility: a committee opinion. Fertil Steril. 2012; 98(3): 591–598.
  86. Moen M. Endometriosis, an everlasting challenge. Acta Obstetricia et Gynecologica Scandinavica. 2017; 96(6): 783–786.
  87. Duffy JMN, Arambage K, Correa FJS, et al. Laparoscopic surgery for endometriosis. Cochrane Database Syst Rev. 2014(4): CD011031.
  88. Tummon IS, Asher LJ, Martin JS, et al. Randomized controlled trial of superovulation and insemination for infertility associated with minimal or mild endometriosis. Fertil Steril. 1997; 68(1): 8–12.
  89. HASHIM HA, RAKHAWY MEL, ELAAL IA. Randomized comparison of superovulation with letrozole vs. clomiphene citrate in an IUI program for women with recently surgically treated minimal to mild endometriosis. Acta Obstetricia et Gynecologica Scandinavica. 2012; 91(3): 338–345.
  90. Matorras R, Corcóstegui B, Esteban J, et al. Fertility in women with minimal endometriosis compared with normal women was assessed by means of a donor insemination program in unstimulated cycles. American Journal of Obstetrics and Gynecology. 2010; 203(4): 345.e1–345.e6.
  91. Werbrouck E, Spiessens C, Meuleman C, et al. No difference in cycle pregnancy rate and in cumulative live-birth rate between women with surgically treated minimal to mild endometriosis and women with unexplained infertility after controlled ovarian hyperstimulation and intrauterine insemination. Fertil Steril. 2006; 86(3): 566–571.
  92. Surrey E. Endometriosis-Related Infertility: The Role of the Assisted Reproductive Technologies. BioMed Research International. 2015; 2015: 1–8.
  93. Opøien HK, Fedorcsak P, Omland AK, et al. In vitro fertilization is a successful treatment in endometriosis-associated infertility. Fertil Steril. 2012; 97(4): 912–918.
  94. Opøien HK, Fedorcsak P, Byholm T, et al. Complete surgical removal of minimal and mild endometriosis improves outcome of subsequent IVF/ICSI treatment. Reprod Biomed Online. 2011; 23(3): 389–395.
  95. Hart RJ, Hickey M, Maouris P, et al. Excisional surgery versus ablative surgery for ovarian endometriomata. Cochrane Database Syst Rev. 2005(3): CD004992.
  96. Hamdan M, Dunselman G, Li TC, et al. The impact of endometrioma on IVF/ICSI outcomes: a systematic review and meta-analysis. Hum Reprod Update. 2015; 21(6): 809–825.
  97. Yap C, Furness S, Farquhar C. Pre and post operative medical therapy for endometriosis surgery. Cochrane Database Syst Rev. 2004(3): CD003678.
  98. Yang JH, Chen MJ, Chen CD, et al. Optimal waiting period for subsequent fertility treatment after various hysteroscopic surgeries. Fertility and Sterility. 2013; 99(7): 2092–2096.e3.
  99. Ziegler Dde, Gayet V, Aubriot F, et al. Use of oral contraceptives in women with endometriosis before assisted reproduction treatment improves outcomes. Fertility and Sterility. 2010; 94(7): 2796–2799.
  100. Sallam HN, Garcia-Velasco JA, Dias S, et al. Long-term pituitary down-regulation before in vitro fertilization (IVF) for women with endometriosis. Cochrane Database Syst Rev. 2006(1): CD004635.
  101. Hughes E, Brown J, Collins JJ, et al. Ovulation suppression for endometriosis. Cochrane Database Syst Rev. 2007(3): CD000155.
  102. WHO. The International Classification of Diseases and Related Health Problems 2018. http://www who int/classifications/en.
  103. Gelbaya TA, Potdar N, Jeve YB, et al. Definition and epidemiology of unexplained infertility. Obstet Gynecol Surv. 2014; 69(2): 109–115.
  104. Gleicher N, Barad D. Unexplained infertility: does it really exist? Hum Reprod. 2006; 21(8): 1951–1955.
  105. Siristatidis C, Bhattacharya S. Unexplained infertility: does it really exist? Does it matter? Hum Reprod. 2007; 22(8): 2084–2087.
  106. Fatum M, Laufer N, Simon A. Investigation of the infertile couple: should diagnostic laparoscopy be performed after normal hysterosalpingography in treating infertility suspected to be of unknown origin? Hum Reprod. 2002; 17(1): 1–3.
  107. Penzias A, Bendikson K, Butts S, et al. The role of immunotherapy in in vitro fertilization: a guideline. Fertility and Sterility. 2018; 110(3): 387–400.
  108. Abdelazim IA, Purohit P, Farag RH, et al. Unexplained infertility: prevalence, possible causes and treatment options. A review of the literature. J Obstet Gynecol Investig. 2018; 1: 17–22.
  109. Gnoth C, Godehardt E, Frank-Herrmann P, et al. Definition and prevalence of subfertility and infertility. Human Reproduction. 2005; 20(5): 1144–1147.
  110. Collins JA, Burrows EA, Wilan AR. The prognosis for live birth among untreated infertile couples. Fertil Steril. 1995; 64(1): 22–28.
  111. Goldman M, Thornton K, Ryley D, et al. A randomized clinical trial to determine optimal infertility treatment in older couples: the Forty and Over Treatment Trial (FORT-T). Fertility and Sterility. 2014; 101(6): 1574–1581.e2.
  112. Farquhar CM, Liu E, Armstrong S, et al. Intrauterine insemination with ovarian stimulation versus expectant management for unexplained infertility (TUI): a pragmatic, open-label, randomised, controlled, two-centre trial. Lancet. 2018; 391(10119): 441–450.
  113. Verhulst SM, Cohlen BJ, Hughes E, et al. Intra-uterine insemination for unexplained subfertility. Cochrane Database Syst Rev. 2006(4): CD001838.
  114. Ombelet W, Dhont N, Thijssen A, et al. Semen quality and prediction of IUI success in male subfertility: a systematic review. Reprod Biomed Online. 2014; 28(3): 300–309.
  115. Danhof NA, Wely Mv, Repping S, et al. Follicle stimulating hormone versus clomiphene citrate in intrauterine insemination for unexplained subfertility: a randomized controlled trial. Human Reproduction. 2018; 33(10): 1866–1874.
  116. Tjon-Kon-Fat RI, Tajik P, Zafarmand MH, et al. INeS study group†. Can we identify subfertile couples that benefit from immediate in vitro fertilisation over intrauterine insemination? Eur J Obstet Gynecol Reprod Biol. 2016; 202(5): 36–40.
  117. Pandian Z, Gibreel A, Bhattacharya S, et al. In vitro fertilisation for unexplained subfertility. Cochrane Database Syst Rev. 2012(4): CD003357.
  118. Audebert A, Pouly JL, Bonifacie B, et al. Laparoscopic surgery for distal tubal occlusions: lessons learned from a historical series of 434 cases. Fertil Steril. 2014; 102(4): 1203–1208.
  119. Chua S, Akande V, Mol B. Surgery for tubal infertility. Cochrane Database of Systematic Reviews. 2017.
  120. Fan M, Ma L. Effect of salpingectomy on ovarian response to hyperstimulation during in vitro fertilization: a meta-analysis. Fertility and Sterility. 2016; 106(2): 322–329.e9.
  121. Noventa M, Gizzo S, Saccardi C, et al. Salpingectomy before assisted reproductive technologies: a systematic literature review. Journal of Ovarian Research. 2016; 9(1).
  122. Johnson N, Voorst Sv, Sowter M, et al. Surgical treatment for tubal disease in women due to undergo in vitro fertilisation. Cochrane Database of Systematic Reviews. 2010.
  123. Zhang Y, Sun Y, Guo Y, et al. Salpingectomy and Proximal Tubal Occlusion for Hydrosalpinx Prior to In Vitro Fertilization. Obstetrical & Gynecological Survey. 2015; 70(1): 33–38.
  124. Pritts EA, Parker WH, Olive DL. Fibroids and infertility: an updated systematic review of the evidence. Fertil Steril. 2009; 91(4): 1215–1223.
  125. Carranza-Mamane B, Havelock J, Hemmings R, et al. REPRODUCTIVE ENDOCRINOLOGY AND INFERTILITY COMMITTEE, SPECIAL CONTRIBUTOR. The management of uterine fibroids in women with otherwise unexplained infertility. J Obstet Gynaecol Can. 2015; 37(3): 277–285.
  126. Palomba S, Fornaciari E, Falbo A, et al. Safety and efficacy of the minilaparotomy for myomectomy: a systematic review and meta-analysis of randomized and non-randomized controlled trials. Reproductive BioMedicine Online. 2015; 30(5): 462–481.
  127. Tinelli A, Hurst BS, Mettler L, et al. Ultrasound evaluation of uterine healing after laparoscopic intracapsular myomectomy: an observational study. Hum Reprod. 2012; 27(9): 2664–2670.
  128. Koo YJ, Lee JK, Lee YK, et al. Pregnancy Outcomes and Risk Factors for Uterine Rupture After Laparoscopic Myomectomy: A Single-Center Experience and Literature Review. Journal of Minimally Invasive Gynecology. 2015; 22(6): 1022–1028.
  129. Brölmann H, Tanos V, Grimbizis G, et al. Options on fibroid morcellation: a literature review. Gynecological Surgery. 2015; 12(1): 3–15.
  130. Chan YY, Jayaprakasan K, Tan A, et al. Reproductive outcomes in women with congenital uterine anomalies: a systematic review. Ultrasound Obstet Gynecol. 2011; 38(4): 371–382.
  131. Chan YY, Jayaprakasan K, Zamora J, et al. The prevalence of congenital uterine anomalies in unselected and high-risk populations: a systematic review. Hum Reprod Update. 2011; 17(6): 761–771.
  132. Venetis CA, Papadopoulos SP, Campo R, et al. Clinical implications of congenital uterine anomalies: a meta-analysis of comparative studies. Reprod Biomed Online. 2014; 29(6): 665–683.
  133. Rikken J, Kowalik C, Emanuel M, et al. Septum resection for women of reproductive age with a septate uterus. Cochrane Database of Systematic Reviews. 2017.
  134. Candiani GB, Vercellini P, Fedele L, et al. Repair of the uterine cavity after hysteroscopic septal incision. Fertil Steril. 1990; 54(6): 991–994.
  135. AAGL Advancing Minimally Invasive Gynecology Worldwide. AAGL practice report: practice guidelines for management of intrauterine synechiae. J Minim Invasive Gynecol. 2010; 17(1): 1–7.
  136. Conforti A, Alviggi C, Mollo A, et al. The management of Asherman syndrome: a review of literature. Reproductive Biology and Endocrinology. 2013; 11(1): 118.
  137. Hanstede M, Meij Ev, Goedemans L, et al. Results of centralized Asherman surgery, 2003–2013. Fertility and Sterility. 2015; 104(6): 1561–1568.e1.
  138. Bosteels J, Kasius J, Weyers S, et al. Hysteroscopy for treating subfertility associated with suspected major uterine cavity abnormalities. Cochrane Database Syst Rev. 2013(1): CD009461.
  139. Bosteels J, Weyers S, Mol BWJ, et al. Anti-adhesion barrier gels following operative hysteroscopy for treating female infertility: a systematic review and meta-analysis. Gynecol Surg. 2014; 11: 113–127.
  140. Bosteels J, Weyers S, D'Hooghe T, et al. Anti-adhesion therapy following operative hysteroscopy for treatment of female subfertility. Cochrane Database of Systematic Reviews. 2017.
  141. Sardo AD, Calagna G, Scognamiglio M, et al. Prevention of intrauterine post-surgical adhesions in hysteroscopy. A systematic review. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2016; 203: 182–192.
  142. Fatemi HM, Kasius JC, Timmermans A, et al. Prevalence of unsuspected uterine cavity abnormalities diagnosed by office hysteroscopy prior to in vitro fertilization. Hum Reprod. 2010; 25(8): 1959–1965.
  143. AAGL Practice Report: Practice Guidelines for the Diagnosis and Management of Endometrial Polyps. Journal of Minimally Invasive Gynecology. 2012; 19(1): 3–10.
  144. Pérez-Medina T, Bajo-Arenas J, Salazar F, et al. Endometrial polyps and their implication in the pregnancy rates of patients undergoing intrauterine insemination: a prospective, randomized study. Human Reproduction. 2005; 20(6): 1632–1635.
  145. Afifi K, Anand S, Nallapeta S, et al. Management of endometrial polyps in subfertile women: a systematic review. Eur J Obstet Gynecol Reprod Biol. 2010; 151(2): 117–121.
  146. Yanaihara A, Yorimitsu T, Motoyama H, et al. Location of endometrial polyp and pregnancy rate in infertility patients. Fertil Steril. 2008; 90(1): 180–182.
  147. Benschop L, Farquhar C, Poel Nv, et al. Interventions for women with endometrioma prior to assisted reproductive technology. Cochrane Database of Systematic Reviews. 2010.
  148. Donnez J, Wyns C, Nisolle M. Does ovarian surgery for endometriomas impair the ovarian response to gonadotropin? Fertil Steril. 2001; 76(4): 662–665.
  149. Howles CM, Tanaka T, Matsuda T. Management of male hypogonadotrophic hypogonadism. Endocr J. 2007; 54(2): 177–190.
  150. Dabbous Z, Atkin S. Hyperprolactinaemia in male infertility: Clinical case scenarios. Arab Journal of Urology. 2018; 16(1): 44–52.
  151. Chehab M, Madala A, Trussell JC. On-label and off-label drugs used in the treatment of male infertility. Fertility and Sterility. 2015; 103(3): 595–604.
  152. Pastuszak A, Pathak P, Chandrashekar A, et al. Varicocele management in the era of in vitro fertilization/intracytoplasmic sperm injection. Asian Journal of Andrology. 2016; 18(3): 343.
  153. Kohn T, Kohn J, Pastuszak A. Varicocelectomy before assisted reproductive technology: are outcomes improved? Fertility and Sterility. 2017; 108(3): 385–391.
  154. Showell MG, Mackenzie-Proctor R, Brown J, et al. Antioxidants for male subfertility. Cochrane Database Syst Rev. 2014(12): CD007411.
  155. The management of infertility due to obstructive azoospermia. Fertility and Sterility. 2008; 90(5): S121–S124.
  156. Esteves SC. Clinical management of infertile men with nonobstructive azoospermia. Asian J Androl. 2015; 17(3): 459–470.
  157. Mehta A, Sigman M. Management of the dry ejaculate: a systematic review of aspermia and retrograde ejaculation. Fertility and Sterility. 2015; 104(5): 1074–1081.
  158. Gat I, Maman E, Yerushalmi G, et al. Electroejaculation combined with intracytoplasmic sperm injection in patients with psychogenic anejaculation yields comparable results to patients with spinal cord injuries. Fertil Steril. 2012; 97(5): 1056–1060.
  159. Berookhim B, Schlegel P. Azoospermia due to Spermatogenic Failure. Urologic Clinics of North America. 2014; 41(1): 97–113.
  160. American Urological Association Practice Committee of the American Society for Reproductive Medicine I. Report on Evaluation of the Azoospermic Male. ; 2001.
  161. Jarvi K, Lo K, Fischer A, et al. CUA Guideline: The workup of azoospermic males. Can Urol Assoc J. 2010; 4(3): 163–167.
  162. Esteves SC, Miyaoka R, Agarwal A. An update on the clinical assessment of the infertile male. [corrected]. Clinics (Sao Paulo). 2011; 66(4): 691–700.
  163. Brezina P, Kutteh W, Bailey A, et al. Fertility Preservation in the Age of Assisted Reproductive Technologies. Obstetrics and Gynecology Clinics of North America. 2015; 42(1): 39–54.
  164. Williams D, Karpman E, Sander J, et al. Pretreatment Semen Parameters in Men With Cancer. The Journal of Urology. 2009; 181(2): 736–740.
  165. RP KS. Ustawa z dnia 25 czerwca 2015 r. o leczeniu niepłodności. ; 2015.
  166. Cohlen B, Bijkerk A, Poel SV, et al. IUI: review and systematic assessment of the evidence that supports global recommendations. Human Reproduction Update. 2018; 24(3): 300–319.
  167. ESHRE Capri Workshop Group. Intrauterine insemination. Hum Reprod Update. 2009; 15(3): 265–277.
  168. Bhattacharya S, Harrild K, Mollison J, et al. Clomifene citrate or unstimulated intrauterine insemination compared with expectant management for unexplained infertility: pragmatic randomised controlled trial. BMJ. 2008; 337(aug07 2): a716–a716.
  169. Honda T, Tsutsumi M, Komoda F, et al. Acceptable pregnancy rate of unstimulated intrauterine insemination: a retrospective analysis of 17,830 cycles. Reproductive Medicine and Biology. 2014; 14(1): 27–32.
  170. Liu J, Li TC, Wang J, et al. The impact of ovarian stimulation on the outcome of intrauterine insemination treatment: an analysis of 8893 cycles. BJOG: An International Journal of Obstetrics & Gynaecology. 2016; 123: 70–75.
  171. Custers IM, Steures P, Hompes P, et al. Intrauterine insemination: how many cycles should we perform? Hum Reprod. 2008; 23(4): 885–888.
  172. Hill M, Whitcomb B, Lewis T, et al. Progesterone luteal support after ovulation induction and intrauterine insemination: a systematic review and meta-analysis. Fertility and Sterility. 2013; 100(5): 1373–1380.e6.
  173. Talbot J, Lawrence M. In-vitro fertilization: indications, stimulation and clinical techniques. The subfertility handbook. : 88–108.
  174. Bouwmans CAM, Grijseels EWM, Braat DDM, et al. [Indications for in vitro fertilization: comparison of the Dutch guidelines with the published evidence]. Ned Tijdschr Geneeskd. 2002; 146(49): 2339–2342.
  175. Coughlan C, Ledger B, Ola B. In-vitro fertilization. Obstetrics, Gynaecology & Reproductive Medicine. 2011; 21(11): 303–310.
  176. Elzeiny H, Garrett C, Toledo M, et al. A randomised controlled trial of intra-uterine insemination versus in vitro fertilisation in patients with idiopathic or mild male infertility. Aust N Z J Obstet Gynaecol. 2014; 54(2): 156–161.
  177. Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet. 1978; 2(8085): 366.
  178. Bhattacharya S, Hamilton MP, Shaaban M, et al. Conventional in-vitro fertilisation versus intracytoplasmic sperm injection for the treatment of non-male-factor infertility: a randomised controlled trial. Lancet. 2001; 357(9274): 2075–2079.
  179. Davis OK. Rosenwaks surgery Z %J R endocrinology, technology. In vitro fertilization. 1996; 2: 2320–34.
  180. Trounson AO, Leeton JF, Wood C, et al. Pregnancies in humans by fertilization in vitro and embryo transfer in the controlled ovulatory cycle. Science. 1981; 212(4495): 681–682.
  181. Niederberger C, Pellicer A. Introduction. Fertility and Sterility. 2018; 110(1): 4.
  182. Palermo GD, O’Neill CL, Chow S, et al. Intracytoplasmic sperm injection: state of the art in humans. Reproduction. 2017; 154(6): F93–F110.
  183. Palermo G, Joris H, Devroey P, et al. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet. 1992; 340(8810): 17–18.
  184. Rumste Mv, Evers J, Farquhar C. Intra-cytoplasmic sperm injection versus conventional techniques for oocyte insemination during in vitro fertilisation in couples with non-male subfertility. Cochrane Database of Systematic Reviews. 2003.
  185. Boulet SL, Mehta A, Kissin DM, et al. Trends in use of and reproductive outcomes associated with intracytoplasmic sperm injection. JAMA. 2015; 313(3): 255–263.
  186. Nagy Z, Liu J, Cecile J, et al. Using ejaculated, fresh, and frozen-thawed epididymal and testicular spermatozoa gives rise to comparable results after intracytoplasmic sperm injection. Fertil Steril. 1995; 63(4): 808–815.
  187. Silber SJ, Van Steirteghem AC, Liu J, et al. High fertilization and pregnancy rate after intracytoplasmic sperm injection with spermatozoa obtained from testicle biopsy. Hum Reprod. 1995; 10(1): 148–152.
  188. Krausz C, Chianese C. Genetic testing and counselling for male infertility. Current Opinion in Endocrinology & Diabetes and Obesity. 2014; 21(3): 244–250.
  189. Hotaling J, Carrell DT. Clinical genetic testing for male factor infertility: current applications and future directions. Andrology. 2014; 2(3): 339–350.
  190. Martin RH. Cytogenetic determinants of male fertility. Human Reproduction Update. 2008; 14(4): 379–390.
  191. Krausz C, Quintana-Murci L, Barbaux S, et al. A high frequency of Y chromosome deletions in males with nonidiopathic infertility. J Clin Endocrinol Metab. 1999; 84(10): 3606–3612.
  192. Page DC, Silber S, Brown LG. Men with infertility caused by AZFc deletion can produce sons by intracytoplasmic sperm injection, but are likely to transmit the deletion and infertility. Hum Reprod. 1999; 14(7): 1722–1726.
  193. Baker VL, Luke B, Brown MB, et al. Multivariate analysis of factors affecting probability of pregnancy and live birth with in vitro fertilization: an analysis of the Society for Assisted Reproductive Technology Clinic Outcomes Reporting System. Fertil Steril. 2010; 94(4): 1410–1416.
  194. Sunkara SK, Rittenberg V, Raine-Fenning N, et al. Association between the number of eggs and live birth in IVF treatment: an analysis of 400 135 treatment cycles. Hum Reprod. 2011; 26(7): 1768–1774.
  195. Ingerslev HJ, Højgaard A, Hindkjaer J, et al. A randomized study comparing IVF in the unstimulated cycle with IVF following clomiphene citrate. Hum Reprod. 2001; 16(4): 696–702.
  196. Bancsi LF, Broekmans FJM, Mol BWJ, et al. Performance of basal follicle-stimulating hormone in the prediction of poor ovarian response and failure to become pregnant after in vitro fertilization: a meta-analysis. Fertil Steril. 2003; 79(5): 1091–1100.
  197. Nelson S, Klein B, Arce JC. Comparison of antimüllerian hormone levels and antral follicle count as predictor of ovarian response to controlled ovarian stimulation in good-prognosis patients at individual fertility clinics in two multicenter trials. Fertility and Sterility. 2015; 103(4): 923–930.e1.
  198. Massin N. New stimulation regimens: endogenous and exogenous progesterone use to block the LH surge during ovarian stimulation for IVF. Human Reproduction Update. 2017.
  199. Maheshwari A, Gibreel A, Siristatidis C, et al. Gonadotrophin-releasing hormone agonist protocols for pituitary suppression in assisted reproduction. Cochrane Database of Systematic Reviews. 2011.
  200. Al-Inany H, Youssef M, Aboulghar M, et al. Gonadotrophin-releasing hormone antagonists for assisted reproductive technology. Cochrane Database of Systematic Reviews. 2011.
  201. Orvieto R. Controlled Ovarian Stimulation for In Vitro Fertilisation Cycles. Infertility in Women with Polycystic Ovary Syndrome. 2018: 259–270.
  202. Pouwer A, Farquhar C, Kremer J. Long-acting FSH versus daily FSH for women undergoing assisted reproduction. Cochrane Database of Systematic Reviews. 2015.
  203. Coyne K, Purdy M, O’Leary K, et al. Challenges and Considerations in Optimizing Ovarian Stimulation Protocols in Oncofertility Patients. Frontiers in Public Health. 2014; 2.
  204. Shrestha D, La X, Feng HL. Comparison of different stimulation protocols used in in vitro fertilization: a review. Ann Transl Med. 2015; 3(10): 137.
  205. Lambalk CB, Banga FR, Huirne JA, et al. GnRH antagonist versus long agonist protocols in IVF: a systematic review and meta-analysis accounting for patient type. Human Reproduction Update. 2017; 23(5): 560–579.
  206. Youssef M, Veen FV, Al-Inany H, et al. Gonadotropin-releasing hormone agonist versus HCG for oocyte triggering in antagonist-assisted reproductive technology. Cochrane Database of Systematic Reviews. 2014.
  207. Iliodromiti S, Kelsey T, Wu O, et al. The predictive accuracy of anti-Müllerian hormone for live birth after assisted conception: a systematic review and meta-analysis of the literature. Human Reproduction Update. 2014; 20(4): 560–570.
  208. Marca ALa, Grisendi V, Giulini S, et al. Individualization of the FSH starting dose in IVF/ICSI cycles using the antral follicle count. Journal of Ovarian Research. 2013; 6(1): 11.
  209. Marca ALa, Papaleo E, Grisendi V, et al. Development of a nomogram based on markers of ovarian reserve for the individualisation of the follicle-stimulating hormone starting dose in in vitro fertilisation cycles. BJOG: An International Journal of Obstetrics & Gynaecology. 2012; 119(10): 1171–1179.
  210. Bosch E, Andersen AN, Barri P, et al. Follicular and endocrine dose responses according to anti-Müllerian hormone levels in IVF patients treated with a novel human recombinant FSH (FE 999049). Clinical Endocrinology. 2015; 83(6): 902–912.
  211. Olivennes F, Trew G, Borini A, et al. Randomized, controlled, open-label, non-inferiority study of the CONSORT algorithm for individualized dosing of follitropin alfa. Reproductive BioMedicine Online. 2015; 30(3): 248–257.
  212. Lensen S, Wilkinson J, Leijdekkers J, et al. Individualised gonadotropin dose selection using markers of ovarian reserve for women undergoing in vitro fertilisation plus intracytoplasmic sperm injection (IVF/ICSI). Cochrane Database of Systematic Reviews. 2018.
  213. van Rooij IAJ, Bancsi LF, Broekmans FJM, et al. Women older than 40 years of age and those with elevated follicle-stimulating hormone levels differ in poor response rate and embryo quality in in vitro fertilization. Fertil Steril. 2003; 79(3): 482–488.
  214. Devroey P, Polyzos NP, Blockeel C. An OHSS-Free Clinic by segmentation of IVF treatment. Hum Reprod. 2011; 26(10): 2593–2597.
  215. Griesinger G, Diedrich K, Devroey P, et al. GnRH agonist for triggering final oocyte maturation in the GnRH antagonist ovarian hyperstimulation protocol: a systematic review and meta-analysis. Human Reproduction Update. 2005; 12(2): 159–168.
  216. Terranova P. Luteal Phase Defects ☆. Reference Module in Biomedical Sciences. 2017.
  217. Valdes CT, 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): 15–18.
  218. Schoolcraft WB, Surrey ES, Gardner DK. Embryo transfer: techniques and variables affecting success. Fertil Steril. 2001; 76(5): 863–870.
  219. Gingold J, Lee J, Rodriguez-Purata J, et al. Endometrial pattern, but not endometrial thickness, affects implantation rates in euploid embryo transfers. Fertility and Sterility. 2015; 104(3): 620–628.e5.
  220. Fanchin R, Ayoubi JM, Righini C, et al. Uterine contractility decreases at the time of blastocyst transfers. Hum Reprod. 2001; 16(6): 1115–1119.
  221. Bishop C, Stormshak F. Nongenomic Action of Progesterone Inhibits Oxytocin-Induced Phosphoinositide Hydrolysis and Prostaglandin F2αSecretion in the Ovine Endometrium. Endocrinology. 2006; 147(2): 937–942.
  222. Visnova H, Pierson RA, Mrázek M, et al. Effects of barusiban, a selective oxytocin antagonist, on uterine contractility in the luteal phase after controlled ovarian stimulation. Fertility and Sterility. 2012; 98(3): S183.
  223. Casper RF. It's time to pay attention to the endometrium. Fertil Steril. 2011; 96(3): 519–521.
  224. Paulson RJ. Hormonal induction of endometrial receptivity. Fertil Steril. 2011; 96(3): 530–535.
  225. Garcia-Velasco JA, Acevedo B, Alvarez C, et al. Strategies to manage refractory endometrium: state of the art in 2016. Reprod Biomed Online. 2016; 32(5): 474–489.
  226. Guven S, Kart C, Unsal M, et al. Endometrial injury may increase the clinical pregnancy rate in normoresponders undergoing long agonist protocol ICSI cycles with single embryo transfer. European Journal of Obstetrics & Gynecology and Reproductive Biology. 2014; 173: 58–62.
  227. Janát-Amsbury M, Gupta K, Kablitz C, et al. Drug delivery for in vitro fertilization: Rationale, current strategies and challenges. Advanced Drug Delivery Reviews. 2009; 61(10): 871–882.
  228. Roque M, Valle M, Guimarães F, et al. Freeze-all policy: fresh vs. frozen-thawed embryo transfer. Fertility and Sterility. 2015; 103(5): 1190–1193.
  229. Basile N, Garcia-Velasco J. The state of “freeze-for-all” in human ARTs. Journal of Assisted Reproduction and Genetics. 2016; 33(12): 1543–1550.
  230. Maheshwari A, Bhattacharya S. Elective frozen replacement cycles for all: ready for prime time? Human Reproduction. 2012; 28(1): 6–9.
  231. Magdi Y, El-Damen A, Fathi AM, et al. Revisiting the management of recurrent implantation failure through freeze-all policy. Fertil Steril. 2017; 108(1): 72–77.
  232. Weinerman R, Mainigi M. Why we should transfer frozen instead of fresh embryos: the translational rationale. Fertil Steril. 2014; 102(1): 10–18.
  233. Zhu D, Zhang J, Cao S, et al. Vitrified-warmed blastocyst transfer cycles yield higher pregnancy and implantation rates compared with fresh blastocyst transfer cycles--time for a new embryo transfer strategy? Fertil Steril. 2011; 95(5): 1691–1695.
  234. Shapiro B, Daneshmand S, Garner F, et al. Clinical rationale for cryopreservation of entire embryo cohorts in lieu of fresh transfer. Fertility and Sterility. 2014; 102(1): 3–9.
  235. Sharkey AM, Smith SK. The endometrium as a cause of implantation failure. Best Pract Res Clin Obstet Gynaecol. 2003; 17(2): 289–307.
  236. Casper RF, Yanushpolsky EH. Optimal endometrial preparation for frozen embryo transfer cycles: window of implantation and progesterone support. Fertil Steril. 2016; 105(4): 867–872.
  237. Dain L, Bider D, Levron J, et al. Thin endometrium in donor oocyte recipients: enigma or obstacle for implantation? Fertility and Sterility. 2013; 100(5): 1289–1295.e2.
  238. Mansour RT, Aboulghar MA. Optimizing the embryo transfer technique. Hum Reprod. 2002; 17(5): 1149–1153.
  239. Pregnancy Rates after Transfer. Equine Embryo Transfer. 2015: 129–133.
  240. Penzias A, Bendikson K, Butts S, et al. ASRM standard embryo transfer protocol template: a committee opinion. Fertility and Sterility. 2017; 107(4): 897–900.
  241. Mains L, Voorhis BV. Optimizing the technique of embryo transfer. Fertility and Sterility. 2010; 94(3): 785–790.
  242. FLISSER E, GRIFO J. Is what we clearly see really so obvious? Ultrasonography and transcervical embryo transfer—a review. Fertility and Sterility. 2007; 87(1): 1–5.
  243. Phillips JAS, Martins WP, Nastri CO, et al. Difficult embryo transfers or blood on catheter and assisted reproductive outcomes: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2013; 168(2): 121–128.
  244. Sullivan EA, Wang YA, Hayward I, et al. Single embryo transfer reduces the risk of perinatal mortality, a population study. Hum Reprod. 2012; 27(12): 3609–3615.
  245. Gerris JMR. Single embryo transfer and IVF/ICSI outcome: a balanced appraisal. Hum Reprod Update. 2005; 11(2): 105–121.
  246. Stillman RJ, Richter KS, Jones HW. Refuting a misguided campaign against the goal of single-embryo transfer and singleton birth in assisted reproduction. Hum Reprod. 2013; 28(10): 2599–2607.
  247. Lessey BA. Two pathways of progesterone action in the human endometrium: implications for implantation and contraception. Steroids. 2003; 68(10-13): 809–815.
  248. Galliano D, Bellver J, Díaz-García C, et al. ART and uterine pathology: how relevant is the maternal side for implantation? Human Reproduction Update. 2014; 21(1): 13–38.
  249. Huang Bo, Ren X, Wu Li, et al. Elevated Progesterone Levels on the Day of Oocyte Maturation May Affect Top Quality Embryo IVF Cycles. PLOS ONE. 2016; 11(1): e0145895.
  250. Fatemi H, Vaerenbergh IV. Significance of premature progesterone rise in IVF. Current Opinion in Obstetrics and Gynecology. 2015; 27(3): 242–248.
  251. Bosch E, Labarta E, Crespo J, et al. Circulating progesterone levels and ongoing pregnancy rates in controlled ovarian stimulation cycles for in vitro fertilization: analysis of over 4000 cycles. Hum Reprod. 2010; 25(8): 2092–2100.
  252. Halasz M, Szekeres-Bartho J. The role of progesterone in implantation and trophoblast invasion. Journal of Reproductive Immunology. 2013; 97(1): 43–50.
  253. Shah D, Nagarajan N. Luteal insufficiency in first trimester. Indian J Endocrinol Metab. 2013; 17(1): 44–49.
  254. Hudić I, Fatusić Z. Progesterone - induced blocking factor (PIBF) and Th(1)/Th(2) cytokine in women with threatened spontaneous abortion. J Perinat Med. 2009; 37(4): 338–342.
  255. Thipasary Y, Effendi J, Anwar A. The Role of Natural Progesterone Administration on the Levels of Progesterone-induced Blocking Factor, Interleukin-10 and the Prolongation of Gestational Length in Impending Preterm Delivery. Journal of SAFOG. 2015; 7: 118–125.
  256. Shapiro D, Boostanfar R, Silverberg K, et al. Examining the evidence: progesterone supplementation during fresh and frozen embryo transfer. Reproductive BioMedicine Online. 2014; 29: S1–S14.
  257. Roztocka A. Vaginal bleeding during the first 20 weeks of pregnancyand its impact on adverse perinatal outcome. Arch Perinat Med. 2016; 22: 7–16.
  258. Beckers NGM, Macklon NS, Eijkemans MJ, et al. Nonsupplemented luteal phase characteristics after the administration of recombinant human chorionic gonadotropin, recombinant luteinizing hormone, or gonadotropin-releasing hormone (GnRH) agonist to induce final oocyte maturation in in vitro fertilization patients after ovarian stimulation with recombinant follicle-stimulating hormone and GnRH antagonist cotreatment. J Clin Endocrinol Metab. 2003; 88(9): 4186–4192.
  259. Fatemi HM, Popovic-Todorovic B, Papanikolaou E, et al. An update of luteal phase support in stimulated IVF cycles. Hum Reprod Update. 2007; 13(6): 581–590.
  260. Devoto L, Kohen P, Muñoz A, et al. Human corpus luteum physiology and the luteal-phase dysfunction associated with ovarian stimulation. Reproductive BioMedicine Online. 2009; 18: S19–S24.
  261. Hubayter ZR, Muasher SJ. Luteal supplementation in in vitro fertilization: more questions than answers. Fertil Steril. 2008; 89(4): 749–758.
  262. Fatemi H, Camus M, Kolibianakis E, et al. The luteal phase of recombinant follicle-stimulating hormone/gonadotropin-releasing hormone antagonist in vitro fertilization cycles during supplementation with progesterone or progesterone and estradiol. Fertility and Sterility. 2007; 87(3): 504–508.
  263. Huang Na, Situ B, Chen X, et al. Meta-analysis of estradiol for luteal phase support in in vitro fertilization/intracytoplasmic sperm injection. Fertility and Sterility. 2015; 103(2): 367–373.e5.
  264. E. M, A. L. Luteal Phase Support in ART: An Update. Enhancing Success of Assisted Reproduction. 2012.
  265. Csapo AI, Pulkkinen MO, Wiest WG. Effects of luteectomy and progesterone replacement therapy in early pregnant patients. Am J Obstet Gynecol. 1973; 115(6): 759–765.
  266. Vaisbuch E, Leong M, Shoham Z. Progesterone support in IVF: is evidence-based medicine translated to clinical practice? A worldwide web-based survey. Reprod Biomed Online. 2012; 25(2): 139–145.
  267. Tavaniotou A, Smitz J, Bourgain C, et al. Comparison between different routes of progesterone administration as luteal phase support in infertility treatments. Hum Reprod Update. 2000; 6(2): 139–148.
  268. Palomba S, Santagni S, Sala GLa. Progesterone administration for luteal phase deficiency in human reproduction: an old or new issue? Journal of Ovarian Research. 2015; 8(1).
  269. Beta J, Szekeres-Bartho J, Skyfta E, et al. Maternal Serum Progesterone-Induced Blocking Factor at 11–13 Weeks’ Gestation in Spontaneous Early Preterm Delivery. Fetal Diagnosis and Therapy. 2011; 29(3): 197–200.
  270. Progesterone supplementation during the luteal phase and in early pregnancy in the treatment of infertility: an educational bulletin. Fertility and Sterility. 2008; 89(4): 789–792.
  271. Kaser DJ, Ginsburg ES, Missmer SA, et al. Intramuscular progesterone versus 8% Crinone vaginal gel for luteal phase support for day 3 cryopreserved embryo transfer. Fertil Steril. 2012; 98(6): 1464–1469.
  272. Haddad G, Saguan DA, Maxwell R, et al. Intramuscular route of progesterone administration increases pregnancy rates during non-downregulated frozen embryo transfer cycles. J Assist Reprod Genet. 2007; 24(10): 467–470.
  273. Lawrence A. Luteal phase deficiency: What we now know. OBG Manag. 2003; 15: 41–61.
  274. Abdelazim I, Elezz A, Elsherbiny M. Relation between single serum progesterone assay and viability of the first trimester pregnancy. SpringerPlus. 2012; 1(1): 80.
  275. Kratz B, Rasheed A, Holden J. Luteal phase support for documented failure of placental steroidogenesis: A case report. Case Reports in Women's Health. 2017; 14: 1–3.
  276. Sonntag B, Loebbecke KC, Nofer JR, et al. Serum estradiol and progesterone in the mid-luteal phase predict clinical pregnancy outcome in IVF/ICSI cycles. Gynecol Endocrinol. 2013; 29(7): 700–703.
  277. Ferraretti AP, Devroey P, Magli MC, et al. No need for luteal phase support in IVF cycles after mild stimulation: proof-of-concept study. Reprod Biomed Online. 2017; 34(2): 162–165.
  278. Connell M, Szatkowski J, Terry N, et al. Timing luteal support in assisted reproductive technology: a systematic review. Fertility and Sterility. 2015; 103(4): 939–946.e3.
  279. Costea DM, Gunn LK, Hargreaves C, et al. Delayed luteo-placental shift of progesterone production in IVF pregnancy. Int J Gynaecol Obstet. 2000; 68(2): 123–129.
  280. Pritts EA, Atwood AK. Luteal phase support in infertility treatment: a meta-analysis of the randomized trials. Hum Reprod. 2002; 17(9): 2287–2299.
  281. Benmachiche A, Benbouhedja S, Zoghmar A, et al. Impact of Mid-Luteal Phase GnRH Agonist Administration on Reproductive Outcomes in GnRH Agonist-Triggered Cycles: A Randomized Controlled Trial. Frontiers in Endocrinology. 2017; 8.
  282. Papanikolaou EG, Verpoest W, Fatemi H, et al. A novel method of luteal supplementation with recombinant luteinizing hormone when a gonadotropin-releasing hormone agonist is used instead of human chorionic gonadotropin for ovulation triggering: a randomized prospective proof of concept study. Fertil Steril. 2011; 95(3): 1174–1177.
  283. Kumar A, Begum N, Prasad S, et al. Oral dydrogesterone treatment during early pregnancy to prevent recurrent pregnancy loss and its role in modulation of cytokine production: a double-blind, randomized, parallel, placebo-controlled trial. Fertility and Sterility. 2014; 102(5): 1357–1363.e3.
  284. Tournaye H, Sukhikh G, Kahler E, et al. A Phase III randomized controlled trial comparing the efficacy, safety and tolerability of oral dydrogesterone versus micronized vaginal progesterone for luteal support in in vitro fertilization. Human Reproduction. 2017; 32(5): 1019–1027.
  285. Griesinger G, Blockeel C, Tournaye H. Oral dydrogesterone for luteal phase support in fresh in vitro fertilization cycles: a new standard? Fertil Steril. 2018; 109(5): 756–762.
  286. Barbosa M, Valadares N, Barbosa A, et al. Oral dydrogesterone vs. vaginal progesterone capsules for luteal-phase support in women undergoing embryo transfer: a systematic review and meta-analysis. JBRA Assisted Reproduction. 2018.
  287. Humaidan P, Polyzos NP, Alsbjerg B, et al. GnRHa trigger and individualized luteal phase hCG support according to ovarian response to stimulation: two prospective randomized controlled multi-centre studies in IVF patients. Hum Reprod. 2013; 28(9): 2511–2521.
  288. Casper R. Luteal phase support for frozen embryo transfer cycles: intramuscular or vaginal progesterone? Fertility and Sterility. 2014; 101(3): 627–628.
  289. Khan N, Richter K, Newsome T, et al. Matched-samples comparison of intramuscular versus vaginal progesterone for luteal phase support after in vitro fertilization and embryo transfer. Fertility and Sterility. 2009; 91(6): 2445–2450.
  290. Doblinger J, Cometti B, Trevisan S, et al. Subcutaneous Progesterone Is Effective and Safe for Luteal Phase Support in IVF: An Individual Patient Data Meta-Analysis of the Phase III Trials. PLOS ONE. 2016; 11(3): e0151388.
  291. Linden Mv, Buckingham K, Farquhar C, et al. Luteal phase support for assisted reproduction cycles. Cochrane Database of Systematic Reviews. 2015.
  292. Stern JE, Lieberman ES, Macaluso M, et al. Is cryopreservation of embryos a legitimate surrogate marker of embryo quality in studies of assisted reproductive technology conducted using national databases? Fertil Steril. 2012; 97(4): 890–893.
  293. Wennerholm UB, Söderström-Anttila V, Bergh C, et al. Children born after cryopreservation of embryos or oocytes: a systematic review of outcome data. Hum Reprod. 2009; 24(9): 2158–2172.
  294. Maheshwari A, Pandey S, Shetty A, et al. Obstetric and perinatal outcomes in singleton pregnancies resulting from the transfer of frozen thawed versus fresh embryos generated through in vitro fertilization treatment: a systematic review and meta-analysis. Fertility and Sterility. 2012; 98(2): 368–377.e9.
  295. Maheshwari A, Raja EA, Bhattacharya S. Obstetric and perinatal outcomes after either fresh or thawed frozen embryo transfer: an analysis of 112,432 singleton pregnancies recorded in the Human Fertilisation and Embryology Authority anonymized dataset. Fertil Steril. 2016; 106(7): 1703–1708.
  296. Dunietz GL, Holzman C, Zhang Y, et al. Assisted Reproductive Technology and Newborn Size in Singletons Resulting from Fresh and Cryopreserved Embryos Transfer. PLOS ONE. 2017; 12(1): e0169869.
  297. Shi W, Xue X, Zhang S, et al. Perinatal and neonatal outcomes of 494 babies delivered from 972 vitrified embryo transfers. Fertil Steril. 2012; 97(6): 1338–1342.
  298. Wong K, Wely Mv, Mol F, et al. Fresh versus frozen embryo transfers in assisted reproduction. Cochrane Database of Systematic Reviews. 2017.
  299. Shi Y, Sun Y, Hao C, et al. Transfer of Fresh versus Frozen Embryos in Ovulatory Women. N Engl J Med. 2018; 378(2): 126–136.
  300. Vuong L, Dang V, Ho T, et al. IVF Transfer of Fresh or Frozen Embryos in Women without Polycystic Ovaries. New England Journal of Medicine. 2018; 378(2): 137–147.
  301. Ishihara O, Kuwahara A, Saitoh H. Frozen-thawed blastocyst transfer reduces ectopic pregnancy risk: an analysis of single embryo transfer cycles in Japan. Fertil Steril. 2011; 95(6): 1966–1969.
  302. Shapiro B, Daneshmand S, Leon LDe, et al. Frozen-thawed embryo transfer is associated with a significantly reduced incidence of ectopic pregnancy. Fertility and Sterility. 2012; 98(6): 1490–1494.
  303. Roque M, Lattes K, Serra S, et al. Fresh embryo transfer versus frozen embryo transfer in in vitro fertilization cycles: a systematic review and meta-analysis. Fertil Steril. 2013; 99(1): 156–162.
  304. Riggs R, Mayer J, Dowling-Lacey D, et al. Does storage time influence postthaw survival and pregnancy outcome? An analysis of 11,768 cryopreserved human embryos. Fertil Steril. 2010; 93(1): 109–115.
  305. Recommendations for gamete and embryo donation: a committee opinion. Fertility and Sterility. 2013; 99(1): 47–62.e1.
  306. Disposition of abandoned embryos: a committee opinion. Fertility and Sterility. 2013; 99(7): 1848–1849.
  307. Ethics Committee of the American Society for Reproductive Medicine. Electronic address: ASRM@asrm.org, Ethics Committee of the American Society for Reproductive Medicine. Defining embryo donation: an Ethics Committee opinion. Fertil Steril. 2016; 106(1): 56–58.
  308. Liebaers I, Desmyttere S, Verpoest W, et al. Report on a consecutive series of 581 children born after blastomere biopsy for preimplantation genetic diagnosis. Human Reproduction. 2009; 25(1): 275–282.
  309. Desmyttere S, Rycke MDe, Staessen C, et al. Neonatal follow-up of 995 consecutively born children after embryo biopsy for PGD. Human Reproduction. 2011; 27(1): 288–293.
  310. Middelburg K, Heide Mv, Houtzager B, et al. Mental, psychomotor, neurologic, and behavioral outcomes of 2-year-old children born after preimplantation genetic screening: follow-up of a randomized controlled trial. Fertility and Sterility. 2011; 96(1): 165–169.
  311. Natsuaki MN, Dimler LM. Pregnancy and child developmental outcomes after preimplantation genetic screening: a meta-analytic and systematic review. World J Pediatr. 2018; 14(6): 555–569.
  312. Testori S. Preimplantation genetic screening. ; 2015.
  313. Mastenbroek S, Twisk M, Veen Fv, et al. Preimplantation genetic screening: a systematic review and meta-analysis of RCTs. Human Reproduction Update. 2011; 17(4): 454–466.
  314. Mastenbroek S, Twisk M, Echten-Arends Jv, et al. In Vitro Fertilization with Preimplantation Genetic Screening. New England Journal of Medicine. 2007; 357(1): 9–17.
  315. Goldman KN, Nazem T, Berkeley A, et al. Preimplantation Genetic Diagnosis (PGD) for Monogenic Disorders: the Value of Concurrent Aneuploidy Screening. J Genet Couns. 2016; 25(6): 1327–1337.
  316. Geraedts J, Wert GDe. Preimplantation genetic diagnosis. Clinical Genetics. 2009; 76(4): 315–325.
  317. Genetics NH, Board NH. Clinical Commissioning Policy: Pre-implantation Genetic Diagnosis. ; 2013.
  318. Santos M, Kuijk E, Macklon N. The impact of ovarian stimulation for IVF on the developing embryo. REPRODUCTION. 2010; 139(1): 23–34.
  319. Macklon NS, Geraedts JPM, Fauser BC. Conception to ongoing pregnancy: the 'black box' of early pregnancy loss. Hum Reprod Update. 2002; 8(4): 333–343.
  320. GIANAROLI L. The in vivo and in vitro efficiency and efficacy of PGD for aneuploidy. Molecular and Cellular Endocrinology. 2001; 183: S13–S18.
  321. Rosenwaks Z, Handyside AH. Is preimplantation genetic testing for aneuploidy an essential tool for embryo selection or a costly 'add-on' of no clinical benefit? Fertil Steril. 2018; 110(3): 351–352.
  322. Tur-Kaspa I, Jeelani R. Clinical guidelines for IVF with PGD for HLA matching. Reprod Biomed Online. 2015; 30(2): 115–119.
  323. Devolder K. Preimplantation HLA typing: having children to save our loved ones. J Med Ethics. 2005; 31(10): 582–586.
  324. Pennings G, Schots R, Liebaers I. Ethical considerations on preimplantation genetic diagnosis for HLA typing to match a future child as a donor of haematopoietic stem cells to a sibling. Hum Reprod. 2002; 17(3): 534–538.
  325. Lukaszuk K, Kalwak K, Pukszta S, et al. Preimplantation genetic diagnosis of human leukocyte antigen for X-linked immunoproliferative syndrome caused by SAP mutation. Eur J Obstet Gynecol Reprod Biol. 2014; 182: 252–253.
  326. Barad D, Darmon S, Kushnir V, et al. Impact of preimplantation genetic screening on donor oocyte-recipient cycles in the United States. American Journal of Obstetrics and Gynecology. 2017; 217(5): 576.e1–576.e8.
  327. Beukers F, Heide Mv, Middelburg K, et al. Morphologic abnormalities in 2-year-old children born after in vitro fertilization/intracytoplasmic sperm injection with preimplantation genetic screening: follow-up of a randomized controlled trial. Fertility and Sterility. 2013; 99(2): 408–413.e4.
  328. Dahdouh EM, Balayla J, García-Velasco JA. Comprehensive chromosome screening improves embryo selection: a meta-analysis. Fertil Steril. 2015; 104(6): 1503–1512.
  329. Esfandiari N, Bunnell ME, Casper RF. Human embryo mosaicism: did we drop the ball on chromosomal testing? J Assist Reprod Genet. 2016; 33(11): 1439–1444.
  330. Dahdouh EM, Balayla J, Audibert F, et al. Genetics Committee. Technical Update: Preimplantation Genetic Diagnosis and Screening. J Obstet Gynaecol Can. 2015; 37(5): 451–463.
  331. Verpoest W, Staessen C, Bossuyt PM, et al. Preimplantation genetic testing for aneuploidy by microarray analysis of polar bodies in advanced maternal age: a randomized clinical trial. Hum Reprod. 2018; 33(9): 1767–1776.
  332. Rubio C, Bellver J, Rodrigo L, et al. In vitro fertilization with preimplantation genetic diagnosis for aneuploidies in advanced maternal age: a randomized, controlled study. Fertility and Sterility. 2017; 107(5): 1122–1129.
  333. Murugappan G, Shahine L, Perfetto C, et al. Intent to treat analysis ofin vitrofertilization and preimplantation genetic screening versus expectant management in patients with recurrent pregnancy loss. Human Reproduction. 2016; 31(8): 1668–1674.
  334. Rosenwaks Z, Handyside AH, Fiorentino F, et al. The pros and cons of preimplantation genetic testing for aneuploidy: clinical and laboratory perspectives. Fertil Steril. 2018; 110(3): 353–361.
  335. Gleicher N, Orvieto R. Is the hypothesis of preimplantation genetic screening (PGS) still supportable? A review. Journal of Ovarian Research. 2017; 10(1).
  336. Vaiarelli A, Cimadomo D, Capalbo A, et al. Pre-implantation genetic testing in ART: who will benefit and what is the evidence? J Assist Reprod Genet. 2016; 33(10): 1273–1278.
  337. Gleicher N, Metzger J, Croft G, et al. A single trophectoderm biopsy at blastocyst stage is mathematically unable to determine embryo ploidy accurately enough for clinical use. Reproductive Biology and Endocrinology. 2017; 15(1).
  338. Naprotechnology – Trademark Details n.d. https://trademarks.justia.com/757/57/ naprotechnology-75757126.html.
  339. Stanford JB, Parnell TA, Boyle PC. Outcomes from treatment of infertility with natural procreative technology in an Irish general practice. J Am Board Fam Med. 2008; 21(5): 375–384.
  340. Recurrent miscarriage. Infertility in Practice. 2010: 402–410.
  341. Protokół nr 37/2014 z posiedzenia Rady Przejrzystości w dniu 27 października 2014 roku w siedzibie Agencji Ochrony Technologii Medycznych. ; 2014.
  342. Dolińska B, Psychologii I, Wrocławski U. Naprotechnologia – przekłamanie czy nieporozumienie? Nauka. 2011; 1: 115–35.

Regulamin

Ważne: serwis https://journals.viamedica.pl/ wykorzystuje pliki cookies. Więcej >>

Używamy informacji zapisanych za pomocą plików cookies m.in. w celach statystycznych, dostosowania serwisu do potrzeb użytkownika (np. język interfejsu) i do obsługi logowania użytkowników. W ustawieniach przeglądarki internetowej można zmienić opcje dotyczące cookies. Korzystanie z serwisu bez zmiany ustawień dotyczących cookies oznacza, że będą one zapisane w pamięci komputera. Więcej informacji można znaleźć w naszej Polityce prywatności.

Czym są i do czego służą pliki cookie możesz dowiedzieć się na stronie wszystkoociasteczkach.pl.

Wydawcą serwisu jest VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl