Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-r6qrq Total loading time: 0 Render date: 2024-04-27T14:50:16.568Z Has data issue: false hasContentIssue false

Section 4 - Treatment of Male Infertility

Published online by Cambridge University Press:  08 July 2023

Larry I. Lipshultz
Affiliation:
Baylor College of Medicine, Texas
Stuart S. Howards
Affiliation:
University of Virginia
Craig S. Niederberger
Affiliation:
University of Illinois, Chicago
Dolores J. Lamb
Affiliation:
Weill Cornell Medical College, New York
Get access
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2023

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Carlstrom, K, Eriksson, A, Stege, R, Rannevik, G. Relationship between serum testosterone and sex hormone-binding globulin in adult men with intact or absent gonadal function. Int J Androl 1990;13:6773.Google Scholar
Patel, DP, Brant, WO, Myers, JB, et al. Sperm concentration is poorly associated with hypoandrogenism in infertile men. Urology 2015;85:1062–7.Google Scholar
Sussman, EM, Chudnovsky, A, Niederberger, CS. Hormonal evaluation of the infertile male: has it evolved? Urol Clin North Am 2008;35:147–55, vii.CrossRefGoogle ScholarPubMed
Ross, LS, Kandel, GL, Prinz, LM, Auletta, F. Clomiphene treatment of the idiopathic hypofertile male: high-dose, alternate-day therapy. Fertil Steril 1980;33:618–23.Google Scholar
Kathrins, M, Niederberger, C. Diagnosis and treatment of infertility-related male hormonal dysfunction. Nat Rev Urol 2016;13:309–23.Google Scholar
Kim, ED, McCullough, A, Kaminetsky, J. Oral enclomiphene citrate raises testosterone and preserves sperm counts in obese hypogonadal men, unlike topical testosterone: restoration instead of replacement. BJU Int 2016;117:677–85.Google Scholar
Goldstein, SR, Siddhanti, S, Ciaccia, AV, Plouffe, L Jr. A pharmacological review of selective oestrogen receptor modulators. Hum Reprod Update 2000;6:212–24.Google Scholar
Hussein, A, Ozgok, Y, Ross, L, Rao, P, Niederberger, C. Optimization of spermatogenesis-regulating hormones in patients with non-obstructive azoospermia and its impact on sperm retrieval: a multicentre study. BJU Int 2013;111:E110–14.Google Scholar
Helo, S, Mahon, J, Ellen, J, et al. Serum levels of enclomiphene and zuclomiphene in men with hypogonadism on long-term clomiphene citrate treatment. BJU Int 2017;119:171–6.Google Scholar
Krzastek, SC, Sharma, D, Abdullah, N, et al. Long-term safety and efficacy of clomiphene citrate for the treatment of hypogonadism. J Urol 2019;202:1029–35.Google Scholar
Guay, AT, Bansal, S, Heatley, GJ. Effect of raising endogenous testosterone levels in impotent men with secondary hypogonadism: double blind placebo-controlled trial with clomiphene citrate. J Clin Endocrinol Metab 1995;80:3546–52.Google ScholarPubMed
Shabsigh, A, Kang, Y, Shabsign, R, et al. Clomiphene citrate effects on testosterone/estrogen ratio in male hypogonadism. J Sex Med 2005;2:716–21.CrossRefGoogle ScholarPubMed
Da Ros, CT, Averbeck, MA. Twenty-five milligrams of clomiphene citrate presents positive effect on treatment of male testosterone deficiency – a prospective study. Int Braz J Urol 2012;38:512–18.CrossRefGoogle ScholarPubMed
Mazzola, CR, Katz, DJ, Loghmanieh, N, Nelson, CJ, Mulhall, JP. Predicting biochemical response to clomiphene citrate in men with hypogonadism. J Sex Med 2014;11:2302–7.Google Scholar
Roth, LW, Ryan, AR, Meacham, RB. Clomiphene citrate in the management of male infertility. Semin Reprod Med 2013;31:245–50.CrossRefGoogle ScholarPubMed
Patel, DP, Brant, WO, Myers, JB, et al. The safety and efficacy of clomiphene citrate in hypoandrogenic and subfertile men. Int J Impot Res 2015;27:221–4.Google Scholar
Hussein, A, Ozgok, Y, Ross, L, Niederberger, C. Clomiphene administration for cases of nonobstructive azoospermia: a multicenter study. J Androl 2005;26:787–91; discussion 92–3.CrossRefGoogle ScholarPubMed
Reifsnyder, JE, Ramasamy, R, Husseini, J, Schlegel, PN. Role of optimizing testosterone before microdissection testicular sperm extraction in men with nonobstructive azoospermia. J Urol 2012;188:532–6.CrossRefGoogle ScholarPubMed
Chehab, M, Madala, A, Trussell, JC. On-label and off-label drugs used in the treatment of male infertility. Fertil Steril 2015;103:595604.CrossRefGoogle ScholarPubMed
Pavlovich, CP, King, P, Goldstein, M, Schlegel, PN. Evidence of a treatable endocrinopathy in infertile men. J Urol 2001;165:837–41.CrossRefGoogle ScholarPubMed
Helo, S, Ellen, J, Mechlin, C, et al. A randomized prospective double-blind comparison trial of clomiphene citrate and anastrozole in raising testosterone in hypogonadal infertile men. J Sex Med 2015;12:1761–9.Google Scholar
Shoshany, O, Abhyankar, N, Mufarreh, N, Daniel, G, Niederberger, C. Outcomes of anastrozole in oligozoospermic hypoandrogenic subfertile men. Fertil Steril 2017;107:589–94.Google Scholar
Saylam, B, Efesoy, O, Cayan, S. The effect of aromatase inhibitor letrozole on body mass index, serum hormones, and sperm parameters in infertile men. Fertil Steril 2011;95:809–11.Google Scholar
Cavallini, G, Biagiotti, G, Bolzon, E. Multivariate analysis to predict letrozole efficacy in improving sperm count of non-obstructive azoospermic and cryptozoospermic patients: a pilot study. Asian J Androl 2013;15:806–11.Google Scholar
Gregoriou, O, Bakas, P, Grigoriadis, C, Creatsa, M, Hassiakos, D, Creatsas, G. Changes in hormonal profile and seminal parameters with use of aromatase inhibitors in management of infertile men with low testosterone to estradiol ratios. Fertil Steril 2012;98:4851.CrossRefGoogle ScholarPubMed
Alder, NJ, Keihani, S, Stoddard, GJ, Myers, JB, Hotaling, JM. Combination therapy with clomiphene citrate and anastrozole is a safe and effective alternative for hypoandrogenic subfertile men. BJU Int 2018;122:688–94.Google Scholar
Shiraishi, K, Ohmi, C, Shimabukuro, T, Matsuyama, H. Human chorionic gonadotrophin treatment prior to microdissection testicular sperm extraction in non-obstructive azoospermia. Hum Reprod 2012;27:331–9.Google Scholar
Kobori, Y, Suzuki, K, Iwahata, T, et al. Induction of spermatogenesis by rhFSH for azoospermia due to spermatogenic dysfunction with maturation arrest: five case series. Syst Biol Reprod Med 2015;61:168–70.CrossRefGoogle ScholarPubMed
Santi, D, De Vicentis, S, Aifano, P, et al. Use of follicle-stimulating hormone for the male partner of idiopathic infertile couples in Italy: results from a multicentre, observational, clinical practice survey. Andrology 2020;8:637–44.CrossRefGoogle ScholarPubMed
Mao, JF, Liu, ZX, Nie, M, et al. Pulsatile gonadotropin-releasing hormone therapy is associated with earlier spermatogenesis compared to combined gonadotropin therapy in patients with congenital hypogonadotropic hypogonadism. Asian J Androl 2017;19:680–5.Google Scholar
Buchter, D, Behre, HM, Kliesch, S, Nieschlag, E. Pulsatile GnRH or human chorionic gonadotropin/human menopausal gonadotropin as effective treatment for men with hypogonadotropic hypogonadism: a review of 42 cases. Eur J Endocrinol 1998;139:298303.Google Scholar
Bouloux, PM, Nieschlag, E, Burger, HG, et al. Induction of spermatogenesis by recombinant follicle-stimulating hormone (puregon) in hypogonadotropic azoospermic men who failed to respond to human chorionic gonadotropin alone. J Androl 2003;24:604–11.CrossRefGoogle ScholarPubMed
Vicari, E, Mongioi, A, Calogero, AE, et al. Therapy with human chorionic gonadotrophin alone induces spermatogenesis in men with isolated hypogonadotrophic hypogonadism – long-term follow-up. Int J Androl 1992;15:320–9.Google Scholar
Warne, DW, Decosterd, G, Okada, H, Yano, Y, Koide, N, Howles, CM. A combined analysis of data to identify predictive factors for spermatogenesis in men with hypogonadotropic hypogonadism treated with recombinant human follicle-stimulating hormone and human chorionic gonadotropin. Fertil Steril 2009;92:594604.Google Scholar
Rastrelli, G, Corona, G, Mannucci, E, Maggi, M. Factors affecting spermatogenesis upon gonadotropin-replacement therapy: a meta-analytic study. Andrology 2014;2:794808.CrossRefGoogle ScholarPubMed
Finkel, DM, Phillips, JL, Snyder, PJ. Stimulation of spermatogenesis by gonadotropins in men with hypogonadotropic hypogonadism. N Engl J Med 1985;313:651–5.CrossRefGoogle ScholarPubMed
Ramasamy, R, Stahl, PJ, Schlegel, PN. Medical therapy for spermatogenic failure. Asian J Androl 2012;14:5760.Google Scholar
Liu, PY, Baker, HW, Jayadev, V, Zacharin, M, Conway, AJ, Handelsman, DJ. Induction of spermatogenesis and fertility during gonadotropin treatment of gonadotropin-deficient infertile men: predictors of fertility outcome. J Clin Endocrinol Metab 2009;94:801–8.Google Scholar
Webster, J, Piscitelli, G, Polli, A, Ferrari, CI, Ismail, I, Scanlon, MF. A comparison of cabergoline and bromocriptine in the treatment of hyperprolactinemic amenorrhea. Cabergoline Comparative Study Group. N Engl J Med 1994;331:904–9.Google Scholar
Samplaski, MK, Loai, Y, Wong, K, Lo, KC, Grober, ED, Jarvi, KA. Testosterone use in the male infertility population: prescribing patterns and effects on semen and hormonal parameters. Fertil Steril 2014;101:64–9.Google Scholar
Turek, PJ, Williams, RH, Gilbaugh, JH, 3rd, Lipshultz, LI. The reversibility of anabolic steroid-induced azoospermia. J Urol 1995;153:1628–30.Google Scholar
Anderson, RA, Wu, FC. Comparison between testosterone enanthate-induced azoospermia and oligozoospermia in a male contraceptive study. II. Pharmacokinetics and pharmacodynamics of once weekly administration of testosterone enanthate. J Clin Endocrinol Metab 1996;81:896901.Google Scholar
[No authors listed]. Contraceptive efficacy of testosterone-induced azoospermia in normal men. World Health Organization Task Force on methods for the regulation of male fertility. Lancet 1990;336:955–9.Google Scholar
Wenker, EP, Dupree, JM, Langille, GM, et al. The use of HCG-based combination therapy for recovery of spermatogenesis after testosterone use. J Sex Med 2015;12:1334–7.CrossRefGoogle ScholarPubMed
Ko, EY, Siddiqi, K, Brannigan, RE, Sabanegh, ES Jr. Empirical medical therapy for idiopathic male infertility: a survey of the American Urological Association. J Urol 2012;187:973–8.Google Scholar
Chua, ME, Escusa, KG, Luna, S, Tapia, LC, Dofitas, B, Morales, M. Revisiting oestrogen antagonists (clomiphene or tamoxifen) as medical empiric therapy for idiopathic male infertility: a meta-analysis. Andrology 2013;1:749–57.Google Scholar
Willets, AE, Corbo, JM, Brown, JN. Clomiphene for the treatment of male infertility. Reprod Sci 2013;20:739–44.Google Scholar
Vandekerckhove, P, Lilford, R, Vail, A, Hughes, E. Clomiphene or tamoxifen for idiopathic oligo/asthenospermia. Cochrane Database Syst Rev 2000;2:CD000151.Google Scholar
Shuling, L, Sie Kuei, ML, Saffari, SE, et al. Do men with normal testosterone-oestradiol ratios benefit from letrozole for the treatment of male infertility? Reprod Biomed Online 2019;38:3945.Google Scholar
Caroppo, E, Niederberger, C, Vizziello, GM, D’Amato, G. Recombinant human follicle-stimulating hormone as a pretreatment for idiopathic oligoasthenoteratozoospermic patients undergoing intracytoplasmic sperm injection. Fertil Steril 2003;80:1398–403.Google Scholar
Foresta, C, Bettella, A, Garolla, A, Ambrosini, G, Ferlin, A. Treatment of male idiopathic infertility with recombinant human follicle-stimulating hormone: a prospective, controlled, randomized clinical study. Fertil Steril 2005;84:654–61.Google Scholar
Attia, AM, Abou-Setta, AM, Al-Inany, HG. Gonadotrophins for idiopathic male factor subfertility. Cochrane Database Syst Rev 2013;8:CD005071.Google Scholar
Showell, MG, Mackenzie-Proctor, R, Brown, J, Yazdani, A, Stankiewicz, MT, Hart, RJ. Antioxidants for male subfertility. Cochrane Database Syst Rev 2014;12:CD007411.Google Scholar
Intasqui, P, Antoniassi, MP, Camargo, M, et al. Differences in the seminal plasma proteome are associated with oxidative stress levels in men with normal semen parameters. Fertil Steril 2015;104:292301.CrossRefGoogle ScholarPubMed
Aitken, RJ, Clarkson, JS. Cellular basis of defective sperm function and its association with the genesis of reactive oxygen species by human spermatozoa. J Reprod Fertil 1987;81:459–69.Google Scholar
Zini, A, de Lamirande, E, Gagnon, C. Reactive oxygen species in semen of infertile patients: levels of superoxide dismutase- and catalase-like activities in seminal plasma and spermatozoa. Int J Androl 1993;16:183–8.Google Scholar
Aslam, MA, Groothuis, TG, Smits, MA, Woelders, H. Effect of corticosterone and hen body mass on primary sex ratio in laying hen (Gallus gallus), using unincubated eggs. Biol Reprod 2014;90:76.Google Scholar
Omar, MI, Pal, RP, Kelly, BD, et al. Benefits of empiric nutritional and medical therapy for semen parameters and pregnancy and live birth rates in couples with idiopathic infertility: a systematic review and meta-analysis. Eur Urol 2019;75:615–25.Google Scholar
Samplaski, MK, Clemesha, CG. Discrepancies between the internet and academic literature regarding vitamin use for male infertility. Transl Androl Urol 2018;7:S193–7.Google Scholar
Anderson, D, Schmid, TE, Baumgartner, A, Cemeli-Carratala, E, Brinkworth, MH, Wood, JM. Oestrogenic compounds and oxidative stress (in human sperm and lymphocytes in the Comet assay). Mutat Res 2003;544:173–8.Google Scholar
Mendiola, J, Torres-Cantero, AM, Vioque, J, et al. A low intake of antioxidant nutrients is associated with poor semen quality in patients attending fertility clinics. Fertil Steril 2010;93:1128–33.Google Scholar
Eskenazi, B, Kidd, SA, Marks, AR, Sloter, E, Block, G, Wyrobek, AJ. Antioxidant intake is associated with semen quality in healthy men. Hum Reprod 2005;20:1006–12.Google Scholar
Dawson, EB, Harris, WA, Teter, MC, Powell, LC. Effect of ascorbic acid supplementation on the sperm quality of smokers. Fertil Steril 1992;58:1034–9.Google Scholar
Dawson, EB, Harris, WA, Rankin, WE, Charpentier, LA, McGanity, WJ. Effect of ascorbic acid on male fertility. Ann N Y Acad Sci 1987;498:312–23.Google Scholar
Rolf, C, Cooper, TG, Yeung, CH, Nieschlag, E. Antioxidant treatment of patients with asthenozoospermia or moderate oligoasthenozoospermia with high-dose vitamin C and vitamin E: a randomized, placebo-controlled, double-blind study. Hum Reprod 1999;14:1028–33.Google Scholar
Zareba, P, Colaci, DS, Afeiche, M, et al. Semen quality in relation to antioxidant intake in a healthy male population. Fertil Steril 2013;100:1572–9.Google Scholar
Schmid, TE, Eskenazi, B, Marchetti, F, et al. Micronutrients intake is associated with improved sperm DNA quality in older men. Fertil Steril 2012;98:11307 e1.Google Scholar
Akmal, M, Qadri, JQ, Al-Waili, NS, Thangal, S, Haq, A, Saloom, KY. Improvement in human semen quality after oral supplementation of vitamin C. J Med Food 2006;9:440–2.Google Scholar
Lafuente, R, Gonzalez-Comadran, M, Sola, I, et al. Coenzyme Q10 and male infertility: a meta-analysis. J Assist Reprod Genet 2013;30:1147–56.Google Scholar
Nadjarzadeh, A, Shidfar, F, Amirjannati, N, et al. Effect of Coenzyme Q10 supplementation on antioxidant enzymes activity and oxidative stress of seminal plasma: a double-blind randomised clinical trial. Andrologia 2014;46:177–83.Google Scholar
Safarinejad, MR, Safarinejad, S, Shafiei, N, Safarinejad, S. Effects of the reduced form of coenzyme Q10 (ubiquinol) on semen parameters in men with idiopathic infertility: a double-blind, placebo controlled, randomized study. J Urol 2012;188:526–31.Google Scholar
Balercia, G, Buldreghini, E, Vignini, A, et al. Coenzyme Q10 treatment in infertile men with idiopathic asthenozoospermia: a placebo-controlled, double-blind randomized trial. Fertil Steril 2009;91:1785–92.Google Scholar
Blomberg, JM. Vitamin D and male reproduction. Nat Rev Endocrinol 2014;10:175–86.Google Scholar
Kwiecinski, GG, Petrie, GI, DeLuca, HF. Vitamin D is necessary for reproductive functions of the male rat. J Nutr 1989;119:741–4.Google Scholar
Uhland, AM, Kwiecinski, GG, DeLuca, HF. Normalization of serum calcium restores fertility in vitamin D-deficient male rats. J Nutr 1992;122:1338–44.Google Scholar
Blomberg Jensen, M, Bjerrum, PJ, Jessen, TE, et al. Vitamin D is positively associated with sperm motility and increases intracellular calcium in human spermatozoa. Hum Reprod 2011;26:1307–17.Google Scholar
Yang, B, Sun, H, Wan, Y, et al. Associations between testosterone, bone mineral density, vitamin D and semen quality in fertile and infertile Chinese men. Int J Androl 2012;35:783–92.Google Scholar
Blomberg Jensen, M, Lawaetz, JG, Petersen, JH, Juul, A, Jorgensen, N. Effects of vitamin D supplementation on semen quality, reproductive hormones, and live birth rate: a randomized clinical trial. J Clin Endocrinol Metab 2018;103:870–81.Google Scholar
Xu, B, Chia, SE, Tsakok, M, Ong, CN. Trace elements in blood and seminal plasma and their relationship to sperm quality. Reprod Toxicol 1993;7:613–18.Google Scholar
Chia, SE, Ong, CN, Chua, LH, Ho, LM, Tay, SK. Comparison of zinc concentrations in blood and seminal plasma and the various sperm parameters between fertile and infertile men. J Androl 2000;21:53–7.Google Scholar
Mankad, M, Sathawara, NG, Doshi, H, Saiyed, HN, Kumar, S. Seminal plasma zinc concentration and alpha-glucosidase activity with respect to semen quality. Biol Trace Elem Res 2006;110:97106.Google Scholar
Zhao, RP, Xiong, CL. [Zinc content analysis in serum, seminal plasma and spermatozoa of asthenozoospermic and oligoasthenozoospermic patients]. Zhonghua Nan Ke Xue 2005;11:680–2.Google Scholar
Wong, WY, Flik, G, Groenen, PM, et al. The impact of calcium, magnesium, zinc, and copper in blood and seminal plasma on semen parameters in men. Reprod Toxicol 2001;15:131–6.Google Scholar
Bakalczuk, S, Robak-Cholubek, D, Jakiel, G, Krasucki, W. [Level of zinc and magnesium in semen taken from male partners of married infertile couples]. Ginekol Pol 1994;65:6770.Google Scholar
Steiner, AZ, Hansen, KR, Barnhardt, KT. The effect of antioxidants on male factor infertility: the Males, Antioxidants, and Infertility (MOXI) randomized clinical trial. Fertil Steril 2020;113:552–560.Google Scholar
Carpino, A, Siciliano, L, Petroni, MF, De Stefano, C, Aquila, S, Ando, S. Low seminal zinc bound to high molecular weight proteins in asthenozoospermic patients: evidence of increased sperm zinc content in oligoasthenozoospermic patients. Hum Reprod 1998;13:111–14.Google Scholar
Fuse, H, Kazama, T, Ohta, S, Fujiuchi, Y. Relationship between zinc concentrations in seminal plasma and various sperm parameters. Int Urol Nephrol 1999;31:401–8.Google Scholar
Colagar, AH, Marzony, ET, Chaichi, MJ. Zinc levels in seminal plasma are associated with sperm quality in fertile and infertile men. Nutr Res 2009;29:82–8.Google Scholar
Schisterman, EF, Sjaarda, LA, Clemons, T, et al. Effect of folic acid and zinc supplementation in men on semen quality and live birth among couples undergoing inferility treatment: a randomized clinical trial. JAMA 2020;323:3548.Google Scholar
Forges, T, Monnier-Barbarino, P, Alberto, JM, Gueant-Rodriguez, RM, Daval, JL, Gueant, JL. Impact of folate and homocysteine metabolism on human reproductive health. Hum Reprod Update 2007;13:225–38.Google Scholar
Wong, WY, Merkus, HM, Thomas, CM, Menkveld, R, Zielhuis, GA, Steegers-Theunissen, RP. Effects of folic acid and zinc sulfate on male factor subfertility: a double-bind, randomized, placebo-controlled trial. Fertil Steril 2002;77:491–8.Google Scholar
Agarwal, A, Said, TM. Carnitines and male infertility. Reprod Biomed Online 2004;8:376–84.Google Scholar
Vicari, E, Calogero, AE. Effects of treatment with carnitines in infertile patients with prostato-vesiculo-epididymitis. Hum Reprod 2001;16:2338–42.CrossRefGoogle ScholarPubMed
Lenzi, A, Lombardo, F, Gandini, L, Dondero, F. [Metabolism and action of L-carnitine: its possible role in sperm tail function]. Arch Ital Urol Nefrol Androl 1992;64:187–96.Google Scholar
Schoenfeld, C, Amelar, RD, Dubin, L. Stimulation of ejaculated human spermatozoa by caffeine. A preliminary report. Fertil Steril 1973;24:772–5.Google Scholar
Dimitriadis, F, Giannakis, D, Pardalidis, N, et al. Effects of phosphodiesterase-5 inhibitors on sperm parameters and fertilizing capacity. Asian J Androl 2008;10:115–33.Google Scholar
Dimitriadis, F, Tsambalas, S, Tsounapi, P, et al. Effects of phosphodiesterase-5 inhibitors on Leydig cell secretory function in oligoasthenospermic infertile men: a randomized trial. BJU Int 2010;106:1181–5.Google Scholar
Lacham-Kaplan, O, Trounson, A. The effects of the sperm motility activators 2-deoxyadenosine and pentoxifylline used for sperm micro-injection on mouse and human embryo development. Hum Reprod 1993;8:945–52.Google Scholar
Curtis, KM, Savitz, DA, Arbuckle, TE. Effects of cigarette smoking, caffeine consumption, and alcohol intake on fecundability. Am J Epidemiol 1997;146:3241.Google Scholar
Vine, MF, Setzer, RW Jr, Everson, RB, Wyrobek, AJ. Human sperm morphometry and smoking, caffeine, and alcohol consumption. Reprod Toxicol 1997;11:179–84.Google Scholar
Klonoff-Cohen, H, Bleha, J, Lam-Kruglick, P. A prospective study of the effects of female and male caffeine consumption on the reproductive endpoints of IVF and gamete intra-Fallopian transfer. Hum Reprod 2002;17:1746–54.Google Scholar
Minelli, A, Bellezza, I. Methylxanthines and reproduction. Handb Exp Pharmacol 2011:349–72.Google Scholar
Beaven, CM, Hopkins, WG, Hansen, KT, Wood, MR, Cronin, JB, Lowe, TE. Dose effect of caffeine on testosterone and cortisol responses to resistance exercise. Int J Sport Nutr Exerc Metab 2008;18:131–41.Google Scholar
Karmon, AE, Toth, TL, Chiu, YH, et al. Male caffeine and alcohol intake in relation to semen parameters and in vitro fertilization outcomes among fertility patients. Andrology 2017;5:354–61.Google Scholar
Jensen, TK, Swan, SH, Skakkebaek, NE, Rasmussen, S, Jorgensen, N. Caffeine intake and semen quality in a population of 2,554 young Danish men. Am J Epidemiol 2010;171:883–91.Google Scholar
Schmid, TE, Eskenazi, B, Baumgartner, A, et al. The effects of male age on sperm DNA damage in healthy non-smokers. Hum Reprod 2007;22:180–7.Google Scholar
Salas-Huetos, A, Bullo, M, Salas-Salvado, J. Dietary patterns, foods and nutrients in male fertility parameters and fecundability: a systematic review of observational studies. Hum Reprod Update 2017;23:371–89.Google Scholar
Omori, K, Kotera, J. Overview of PDEs and their regulation. Circ Res 2007;100:309–27.Google Scholar
Scott, L, Smith, S. Human sperm motility-enhancing agents have detrimental effects on mouse oocytes and embryos. Fertil Steril 1995;63:166–75.Google Scholar
Aversa, A, Mazzilli, F, Rossi, T, Delfino, M, Isidori, AM, Fabbri, A. Effects of sildenafil (Viagra) administration on seminal parameters and post-ejaculatory refractory time in normal males. Hum Reprod 2000;15:131–4.Google Scholar
Purvis, K, Muirhead, GJ, Harness, JA. The effects of sildenafil on human sperm function in healthy volunteers. Br J Clin Pharmacol 2002;53 Suppl 1:53S60S.Google Scholar
Hellstrom, WJ, Gittelman, M, Jarow, J, et al. An evaluation of semen characteristics in men 45 years of age or older after daily dosing with tadalafil 20mg: results of a multicenter, randomized, double-blind, placebo-controlled, 9-month study. Eur Urol 2008;53:1058–65.Google Scholar
Jarvi, K, Dula, E, Drehobl, M, Pryor, J, Shapiro, J, Seger, M. Daily vardenafil for 6 months has no detrimental effects on semen characteristics or reproductive hormones in men with normal baseline levels. J Urol 2008;179:1060–5.Google Scholar
Rago, R, Salacone, P, Caponecchia, L, Marcucci, I, Fiori, C, Sebastianelli, A. Effect of vardenafil on semen parameters in infertile men: a pilot study evaluating short-term treatment. J Endocrinol Invest 2012;35:897900.Google Scholar
Pourian, MR, Kvist, U, Bjorndahl, L, Oliw, EH. Rapid and slow hydroxylators of seminal E prostaglandins among men in barren unions. Andrologia 1995;27:71–9.Google Scholar
Bartke, A, Kupfer, D, Dalterio, S. Prostaglandins inhibit testosterone secretion by mouse testes in vitro. Steroids 1976;28:81–8.Google Scholar
Abbatiello, ER, Kaminsky, M, Weisbroth, S. The effect of prostaglandins and prostaglandin inhibitors on spermatogenesis. Int J Fertil 1975;20:177–82.Google Scholar
Cohen, MS, Colin, MJ, Golimbu, M, Hotchkiss, RS. The effects of prostaglandins on sperm motility. Fertil Steril 1977;28:7885.Google Scholar
Barkay, J, Harpaz-Kerpel, S, Ben-Ezra, S, Gordon, S, Zuckerman, H. The prostaglandin inhibitor effect of antiinflammatory drugs in the therapy of male infertility. Fertil Steril 1984;42:406–11.Google Scholar
Kristensen, DM, Desdoits-Lethimonier, C, Mackey, AL, et al. Ibuprofen alters human testicular physiology to produce a state of compensated hypogonadism. Proc Natl Acad Sci U S A 2018;115:E715–24.Google Scholar
Stutz, G, Martini, AC, Ruiz, RD, Fiol De Cuneo, M, Munoz, L, Lacuara, JL. Functional activity of mouse sperm was not affected by low doses of aspirin-like drugs. Arch Androl 2000;44:117–28.Google Scholar
Kavoussi, PK, Gilkey, MS, Hunn, C, et al. Ibuprofen does not have an adverse impact on semen parameters. J Assist Reprod Genet 2018;35:2201–4.Google Scholar

References

Aggour, A, Mostafa, H, Maged, W. Endoscopic management of ejaculatory duct obstruction. Int Urol Nephrol 1998;30:481–5.Google Scholar
Modgil, V, Rai, S, Ralph, DJ, Muneer, A. An update on the diagnosis and management of ejaculatory duct obstruction. Nat Rev Urol 2016;13:1320.Google Scholar
Chen, R, Wang, L, Sheng, X, et al. Transurethral seminal vesiculoscopy for recurrent hemospermia: experience from 419 cases. Asian J Androl 2018;20:438–41.Google Scholar
Pryor, JP, Hendry, WF. Ejaculatory duct obstruction in subfertile males: analysis of 87 patients. Fertil Steril 1991;56:725–30.Google Scholar
Tu, XA, Zhuang, JT, Zhao, L, et al. Transurethral bipolar plasma kinetic resection of ejaculatory duct for treatment of ejaculatory duct obstruction. J Xray Sci Technol 2013;21:293302.Google Scholar
Turek, PJ, Magana, JO, Lipshultz, LI. Semen parameters before and after transurethral surgery for ejaculatory duct obstruction. J Urol 1996;155:1291–3.Google Scholar
Li, YF, Liang, PH, Sun, ZY, et al. Imaging diagnosis, transurethral endoscopic observation, and management of 43 cases of persistent and refractory hematospermia. J Androl 2012;33:906–16.CrossRefGoogle ScholarPubMed
Jarow, JP. Transrectal ultrasonography of infertile men. Fertil Steril 1993;60:1035–9.Google Scholar
Avellino, GJ, Lipshultz, LI, Sigman, M, Hwang, K. Transurethral resection of the ejaculatory ducts: etiology of obstruction and surgical treatment options. Fertil Steril 2019;111:427–43.Google Scholar
Jurewicz, M, Gilbert, BR. Imaging and angiography in male factor infertility. Fertil Steril 2016;105:1432–42.Google Scholar
Nguyen, HT, Etzell, J, Turek, PJ. Normal human ejaculatory duct anatomy: a study of cadaveric and surgical specimens. J Urol 1996;155:1639–42.Google Scholar
McMahon, S. An anatomical study by injection technique of the ejaculatory ducts and their relations. J Anat 1938;72(Pt 4):556–74.Google Scholar
McCarthy, JF, Bitter, JS, Klemperer, P. Anatomical and histological study of the verumontanum with especial reference to the ejaculatory ducts. J Urol 1927;17:116.Google Scholar
Kim, ED, Onel, E, Honig, SC, Lipshultz, LI. The prevalence of cystic abnormalities of the prostate involving the ejaculatory ducts as detected by transrectal ultrasound. Int Urol Nephrol 1997;29:647–52.Google Scholar
Kirkali, Z, Yigitbasi, O, Diren, B, Hekimoglu, B, Ersoy, H. Cysts of the prostate, seminal vesicles and diverticulum of the ejaculatory ducts. Eur Urol 1991;20:7780.Google Scholar
Shebel, HM, Farg, HM, Kolokythas, O, El-Diasty, T. Cysts of the lower male genitourinary tract: embryologic and anatomic considerations and differential diagnosis. Radiographics 2013;33:1125–43.Google Scholar
Worischeck, JH, Parra, RO. Transrectal ultrasound in the evaluation of men with low volume azoospermia. J Urol 1993;149(5 Pt 2):1341–4.Google Scholar
Van Poppel, H, Vereecken, R, De Geeter, P, Verduyn, H. Hemospermia owing to utricular cyst: embryological summary and surgical review. J Urol 1983;129:608–9.Google Scholar
Meschede, D, Dworniczak, B, Behre, HM, et al. CFTR gene mutations in men with bilateral ejaculatory-duct obstruction and anomalies of the seminal vesicles. Am J Hum Genet 1997;61:1200–2.Google Scholar
Jarvi, K, Zielenski, J, Wilschanski, M, et al. Cystic fibrosis transmembrane conductance regulator and obstructive azoospermia. Lancet 1995;345:1578.Google Scholar
Littrup, PJ, Lee, F, McLeary, RD, Wu, D, Lee, A, Kumasaka, GH. Transrectal US of the seminal vesicles and ejaculatory ducts: clinical correlation. Radiology 1988;168:625–8.Google Scholar
Meacham, RB, Hellerstein, DK, Lipshultz, LI. Evaluation and treatment of ejaculatory duct obstruction in the infertile male. Fertil Steril 1993;59:393–7.Google Scholar
Weintraub, CM. Transurethral drainage of the seminal tract for obstruction, infection and infertility. Br J Urol 1980;52:220–5.Google Scholar
Fisch, H, Lambert, SM, Goluboff, ET. Management of ejaculatory duct obstruction: etiology, diagnosis, and treatment. World J Urol 2006;24:604–10.Google Scholar
Weintraub, MP, de Mouy, E, Hellstrom, WJG. Newer modalities in the diagnosis and treatment of ejaculatory duct obstruction. J Urol 1993;150:1150–4.Google Scholar
Jarow, JP, Espeland, MA, Lipshultz, LI. Evaluation of the azoospermic patient. J Urol 1989;142:62–5.Google Scholar
Kochakarn, W, Leenanupunth, C, Muangman, V, Ratana-Olarn, K, Viseshsindh, V. Ejaculatory duct obstruction in the infertile male: experience of 7 cases at Ramathibodi Hospital. J Med Assoc Thai 2001;84:1148–52.Google Scholar
Paick, J, Kim, SH, Kim, SW. Ejaculatory duct obstruction in infertile men. BJU Int 2000;85:720–4.Google Scholar
Hellerstein, DK, Meacham, RB, Lipshultz, LI. Transrectal ultrasound and partial ejaculatory duct obstruction in male infertility. Urology 1992;39:449–52.Google Scholar
Nagler, HM, Rotman, M, Zoltan, E, Fisch, H. The natural history of partial ejaculatory duct obstruction. J Urol 2002;167:253–4.Google Scholar
Schroeder-Printzen, I, Ludwig, M, Köhn, F, Weidner, W. Surgical therapy in infertile men with ejaculatory duct obstruction: technique and outcome of a standardized surgical approach. Hum Reprod 2000;15:1364–8.Google Scholar
Jarow, JP. Seminal vesicle aspiration in the management of patients with ejaculatory duct obstruction. J Urol 1994;152:899901Google Scholar
Jarow, JP. Seminal vesicle aspiration of fertile men. J Urol 1996;156:1005–7.Google Scholar
Purohit, RS, Wu, DS, Shinohara, K, Turek, PJ. A prospective comparison of 3 diagnostic methods to evaluate ejaculatory duct obstruction. J Urol 2004;171:232–5; discussion 235–6.Google Scholar
Engin, G, Kadioğlu, A, Orhan, I, Akdöl, S, Rozanes, I. Transrectal US and endorectal MR imaging in partial and complete obstruction of the seminal duct system. A comparative study. Acta Radiol 2000;41:288–95.Google Scholar
Farley, S, Barnes, R. Stenosis of ejaculatory ducts treated by endoscopic resection. J Urol 1973;109:664–6.Google Scholar
Porch, PP. Aspermia owing to obstruction of distal ejaculatory duct and treatment by transurethral resection. J Urol 1978;119:141–2.Google Scholar
Cornel, EB, Dohle, GR, Meuleman, EJ. Transurethral deroofing of midline prostatic cyst for subfertile men. Hum Reprod 1999;14:2297–300.Google Scholar
Dik, P, Lock, TM, Schrier, BP, Zeijlemaker, BY, Boon, TA. Transurethral marsupialization of a medial prostatic cyst in patients with prostatitis-like symptoms. J Urol 1996;155:1301–4.Google Scholar
Netto, NR, Esteves, SC, Neves, PA. Transurethral resection of partially obstructed ejaculatory ducts: seminal parameters and pregnancy outcomes according to the etiology of obstruction. J Urol 1998;159:2048–53.Google Scholar
Goluboff, ET, Kaplan, SA, Fisch, H. Seminal vesicle urinary reflux as a complication of transurethral resection of ejaculatory ducts. J Urol 1995;153:1234–5.Google Scholar
Vazquez-Levin, MH, Dressler, KP, Nagler, HM. Urine contamination of seminal fluid after transurethral resection of the ejaculatory ducts. J Urol 1994;152(6 Part 1):2049–52.Google Scholar
Vicente, J, del Portillo, L, Ma Pomerol, M. Endoscopic surgery in distal obstruction of the ejaculatory ducts. Eur Urol 1983;9:338–40.Google Scholar
Carson, CC. Transurethral resection for ejaculatory duct stenosis and oligospermia. Fertil Steril 1984;41:482–4.Google Scholar
Kadioǧlu, A, Cayan, S, Tefekli, A, Orhan, I, Engin, G, Turek, PJ. Does response to treatment of ejaculatory duct obstruction in infertile men vary with pathology? Fertil Steril 2001;76:138–42.Google Scholar
Ozgök, Y, Tan, MO, Kilciler, M, Tahmaz, L, Kibar, Y. Diagnosis and treatment of ejaculatory duct obstruction in male infertility. Eur Urol 2001;39:24–9.Google Scholar
Fuse, H, Mizuno, I, Iwasaki, M, Akashi, T. Transurethral treatment of ejaculatory duct obstruction in infertile men. Arch Androl 2003;49:429–31.Google Scholar
Tu, XA, Zhao, LY, Zhao, L, et al. Efficacy of transurethral resection of ejaculatory duct for treatment of ejaculatory duct obstruction: report of 60 cases. [Article in Chinese] Beijing Da Xue Xue Bao 2011;43:559–61.Google Scholar
El-Assmy, A, El-Tholoth, H, Abouelkheir, RT, Abou-El-Ghar, ME. Transurethral resection of ejaculatory duct in infertile men: outcome and predictors of success. Int Urol Nephrol 2012;44:1623–30.Google Scholar
Tu, XA, Zhuang, JT, Zhao, L, et al. Transurethral bipolar plasma kinetic resection of ejaculatory duct for treatment of ejaculatory duct obstruction. J Xray Sci Technol 2013;21:293302.Google Scholar
Goldwasser, BZ, Weinerth, JL, Carson 3rd, CC. Ejaculatory duct obstruction: the case for aggressive diagnosis and treatment. J Urol 1985;134:964–6.Google Scholar
Wang, H, Ye, H, Xu, C, et al. Transurethral seminal vesiculoscopy using a 6F vesiculoscope for ejaculatory duct obstruction: initial experience. J Androl 2012;33:637–43.Google Scholar
Tang, SX, Zhou, HL, Ding, YL. Effectiveness of transurethral seminal vesiculoscopy in the treatment of persistent hematospermia, and oligoasthenozoospermia and azoospermia from ejaculatory duct obstruction. [Article in Chinese] Zhonghua Yi Xue Za Zhi 2016;96:2872–5.Google Scholar
Lawler, LP, Cosin, O, Jarow, JP, Kim, HS. Transrectal US-guided seminal vesiculography and ejaculatory duct recanalization and balloon dilation for treatment of chronic pelvic pain. J Vasc Interv Radiol 2006;17:169–73.Google Scholar
Jarow, JP, Zagoria, RJ. Antegrade ejaculatory duct recanalization and dilation. Urology 1995;46:743–6.Google Scholar
Lee, JY, Diaz, RR, Choi, YD, Cho, KS. Hybrid method of transurethral resection of ejaculatory ducts using holmium:yttriumaluminium garnet laser on complete ejaculatory duct obstruction. Yonsei Med J 2013;54:1062–5.Google Scholar
Halpern, EJ, Hirsch, IH. Sonographically guided transurethral laser incision of a müllerian duct cyst for treatment of ejaculatory duct obstruction. Am J Roentgenol 2000;175:777–8.Google Scholar
Barone, MA, Hutchinson, PL, Johnson, CH, Hsia, J, Wheeler, J. Vasectomy in the United States, 2002. J Urol 2006;176:232–6; discussion 236.Google Scholar
Sharma, V, Le, BV, Sheth, KR, et al. Vasectomy demographics and postvasectomy desire for future children: results from a contemporary national survey. Fertil Steril 2013;99:1880–5.Google Scholar
Potts, JM, Pasqualotto, FF, Nelson, D, Thomas, AJ, Agarwal, A. Patient characteristics associated with vasectomy reversal. J Urol 1999;161:1835–9.Google Scholar
Martin, E, Carnett, JB, Levi, JV, Pennington, ME. The surgical treatment of sterility due to obstruction at the epididymis; together with a study of the morphology of human spermatozoa. Univ Pa Med Bull 1902;15:215.Google Scholar
O’Conor, VJ. Anastomosis of the vas deferens after purposeful division for sterility. J Urol 1948;59:229–33.Google Scholar
Derrick, FC, Yarbrough, W, D’Agostino, J. Vasovasostomy: results of questionnaire of members of the American Urological Association. J Urol 1973;110:556–7.Google Scholar
Silber, SJ. Microsurgery in clinical urology. Urology 1975;6:150–3.Google Scholar
Pavlovich, CP, Schlegel, PN. Fertility options after vasectomy: a cost-effectiveness analysis. Fertil Steril 1997;67:133–41.Google Scholar
Belker, AM, Thomas, AJ, Fuchs, EF, Konnak, JW, Sharlip, ID. Results of 1,469 microsurgical vasectomy reversals by the Vasovasostomy Study Group. J Urol 1991;145:505–11.Google Scholar
Chawla, A, O’Brien, J, Lisi, M, Zini, A, Jarvi, K. Should all urologists performing vasectomy reversals be able to perform vasoepididymostomies if required? J Urol 2004;172:1048–50.Google Scholar
Fenig, DM, Kattan, MW, Mills, JN, Gisbert, M, Yu, C, Lipshultz, LI. Nomogram to preoperatively predict the probability of requiring epididymovasostomy during vasectomy reversal. J Urol 2012;187:215–18.Google Scholar
Parekattil, SJ, Kuang, W, Agarwal, A, Thomas, AJ. Model to predict if a vasoepididymostomy will be required for vasectomy reversal. J Urol 2005;173:1681–4.Google Scholar
Boyle, KE, Thomas, AJ, Marmar, JL, Hirshberg, S, Belker, AM, Jarow, JP. Sperm harvesting and cryopreservation during vasectomy reversal is not cost effective. Fertil Steril 2006;85:961–4.Google Scholar
Fischer, MA, Grantmyre, JE. Comparison of modified one- and two-layer microsurgical vasovasostomy. BJU Int 2000;85:1085–8.Google Scholar
Jee, SH, Hong, YK. One-layer vasovasostomy: microsurgical versus loupe-assisted. Fertil Steril 2010;94:2308–11.Google Scholar
Chan, PT. The evolution and refinement of vasoepididymostomy techniques. Asian J Androl 2013;15:4955.Google Scholar
Parekattil, SJ, Gudeloglu, A, Brahmbhatt, J, Wharton, J, Priola, KB. Robotic assisted versus pure microsurgical vasectomy reversal: technique and prospective database control trial. J Reconstr Microsurg 2012;28:435–44.Google Scholar
Kavoussi, P. Validation of robot-assisted vasectomy reversal. Asian J Androl 2015;17:245–7.Google Scholar
Marshall, MT, Doudt, AD, Berger, JH, Auge, BK, Christman, MS, Choe, CH. Robot-assisted vasovasostomy using a single layer anastomosis. J Robot Surg 2017;11:299303.Google Scholar
Anger, JT, Goldstein, M. Intravasal “toothpaste” in men with obstructive azoospermia is derived from vasal epithelium, not sperm. J Urol 2004;172:634–6.Google Scholar
Silber, SJ. Microscopic vasectomy reversal. Fertil Steril 1977;28:1191–202.Google Scholar
Owen, E, Kapila, H. Vasectomy reversal. Review of 475 microsurgical vasovasostomies. Med J Aust 1984;140:398400.Google Scholar
Lee, HY. A 20-year experience with vasovasostomy. J Urol 1986;136:413–15.Google Scholar
Silber, SJ. Pregnancy after vasovasostomy for vasectomy reversal: a study of factors affecting long-term return of fertility in 282 patients followed for 10 years. Hum Reprod 1989;4:318–22.Google Scholar
Kabalin, JN, Kessler, R. Macroscopic vasovasostomy re-examined. Urology 1991;38:135–8.Google Scholar
Fox, M. Vasectomy reversal – microsurgery for best results. Br J Urol 1994;73:449–53.Google Scholar
Scovell, JM, Mata, DA, Ramasamy, R, Herrel, LA, Hsiao, W, Lipshultz, LI. Association between the presence of sperm in the vasal fluid during vasectomy reversal and postoperative patency: a systematic review and meta-analysis. Urology 2015;85:809–13.Google Scholar
Patel, SR, Sigman, M. Comparison of outcomes of vasovasostomy performed in the convoluted and straight vas deferens. J Urol 2008;179:256–9.Google Scholar
Peng, J, Yuan, Y, Zhang, Z, Cui, W, Song, W, Gao, B. Microsurgical vasoepididymostomy is an effective treatment for azoospermic patients with epididymal obstruction and prior failure to achieve pregnancy by sperm retrieval with intracytoplasmic sperm injection. Hum Reprod 2014;29:17.Google Scholar
Fuchs, ME, Anderson, RE, Ostrowski, KA, Brant, WO, Fuchs, EF. Pre-operative risk factors associated with need for vasoepididymostomy at the time of vasectomy reversal. Andrology 2016;4:160–2.Google Scholar
Herrel, LA, Goodman, M, Goldstein, M, Hsiao, W. Outcomes of microsurgical vasovasostomy for vasectomy reversal: a meta-analysis and systematic review. Urology 2015;85:819–25.Google Scholar
Belker, AM, Thomas, AJ, Fuchs, EF, Konnak, JW, Sharlip, ID. Results of 1,469 microsurgical vasectomy reversals by the Vasovasostomy Study Group. J Urol 1991;145:505–11.Google Scholar
Ostrowski, KA, Polackwich, AS, Kent, J, Conlin, MJ, Hedges, JC, Fuchs, EF. Higher outcomes of vasectomy reversal in men with the same female partner as before vasectomy. J Urol 2015;193:245–7.Google Scholar
Chan, PT., Goldstein, M. Superior outcomes of microsurgical vasectomy reversal in men with the same female partners. Fertil Steril 2004;81:1371–4.Google Scholar
Kolettis, PN, Woo, L, Sandlow, JI. Outcomes of vasectomy reversal performed for men with the same female partners. Urology 2003;61:1221–3.Google Scholar
Gerrard, ER, Sandlow, JI, Oster, RA, Burns, JR, Box, LC, Kolettis, PN. Effect of female partner age on pregnancy rates after vasectomy reversal. Fertil Steril 2007;87:1340–4.Google Scholar
Dubin, L, Amelar, RD. Magnified surgery for epididymovasostomy. Urology 1984;23:525–8.Google Scholar
Fogdestam, I, Fall, M, Nilsson, S. Microsurgical epididymovasostomy in the treatment of occlusive azoospermia. Fertil Steril 1986;46:925–9.Google Scholar
Silber, SJ. Results of microsurgical vasoepididymostomy: role of epididymis in sperm maturation. Hum Reprod 1989;4:298303.Google Scholar
Dewire, D, Thomas, A. Microsurgical treatment of male infertility. In: Goldstein, M, ed. Surgery of Male Fertility. Philadelphia, PA: Saunders, 1995; pp. 128–34.Google Scholar
Berger, RE. Triangulation end-to-side vasoepididymostomy. J Urol 1998;159:1951–3.Google Scholar
Marmar, JL. Modified vasoepididymostomy with simultaneous double needle placement, tubulotomy and tubular invagination. J Urol 2000;163:483–6.Google Scholar
Chan, PTK, Brandell, RA, Goldstein, M. Prospective analysis of outcomes after microsurgical intussusception vasoepididymostomy. BJU Int 2005;96:598601.Google Scholar
Zhao, L, Tu, XA, Zhuang, JT, et al. Retrospective analysis of early outcomes after a single-armed suture technique for microsurgical intussusception vasoepididymostomy. Andrology 2015;3:1150–3.Google Scholar
Silverstein, JI, Mellinger, BC. Fibrin glue vasal anastomosis compared to conventional sutured vasovasostomy in the rat. J Urol 1991;145:1288–91.Google Scholar
Shekarriz, BM, Thomas, AJJ, Sabanegh, E, Kononov, A, Levin, HS. Fibrin-glue assisted vasoepididymostomy: a comparison to standard end-to-side microsurgical vasoepididymostomy in the rat model. J Urol 1997;158:1602–5.Google Scholar
Busato, WF, Marquetti, AM, Rocha, LC. Comparison of vasovasostomy with conventional microsurgical suture and fibrin adhesive in rats. Int Braz J Urol 2007;33:829–36.Google Scholar
Weiss, JN, Mellinger, BC. Fertility rates with delayed fibrin glue: vasovasostomy in rats. Fertil Steril 1992;57:908–11.Google Scholar
Ho K-L, V, Witte, MN, Bird, ET, Hakim, S. Fibrin glue assisted 3-suture vasovasostomy. J Urol 2005;174(4 Pt 1):1360–3; discussion 1363.Google Scholar
Ball, RA, Steinberg, J, Wilson, LA, Loughlin, KR. Comparison of vasovasostomy techniques in rats utilizing conventional microsurgical suture, carbon dioxide laser, and fibrin tissue adhesives. Urology 1993;41:479–83.Google Scholar
Vankemmel, O, de la Taille, A, Burnouf, T, Rigot, JM, Duchene, F, Mazeman, E. Evaluation of a fibrin sealant free of bovine-derived components in an experimental vas anastomosis study. Urol Int 2000;65:196–9.Google Scholar
Saunders, MM, Baxter, ZC, Abou-Elella, A, Kunselman, AR, Trussell, JC. BioGlue and Dermabond save time, leak less, and are not mechanically inferior to two-layer and modified one-layer vasovasostomy. Fertil Steril 2009;91:560–5.Google Scholar
Mazzilli, R, Defeudis, G, Olana, S, Zamponi, V, Macera, M, Mazzilli, F. The role of ejaculatory dysfunction on male infertility. Clin Ter 2020;171:e523–7.Google Scholar
Shaban, SF, Seager, SW, Lipshultz, LI. Clinical electroejaculation. Med Instrum 1988;22:7781.Google Scholar
Wein, AJ, Kavoussi, LR, Novick, AC, Partin, AW, Peters, CA (eds). Campbell-Walsh Urology, 10th ed. Saint Louis, MO: Elsevier, 2011.Google Scholar
Sønksen, J, Ohl, DA. Penile vibratory stimulation and electroejaculation in the treatment of ejaculatory dysfunction. Int J Androl 2002;25:324–32.Google Scholar
Halpern, JA, Das, A, Faw, CA, Brannigan, RE. Oncofertility in adult and pediatric populations: options and barriers. Transl Androl Urol. 2020;9(Suppl 2):S227–38.Google Scholar
Cito, G, Picone, R, Fucci, R, et al. Reproductive outcomes in infertile men with spinal cord injury (SCI): a retrospective case-control analysis. Urology 2020;141:82–8.Google Scholar
Chong, W, Ibrahim, E, Aballa, TC, Lynne, CM, Brackett, NL. Comparison of three methods of penile vibratory stimulation for semen retrieval in men with spinal cord injury. Spinal Cord 2017;55:921–5.Google Scholar
Brackett, NL, Ead, DN, Aballa, TC, Ferrell, SM, Lynne, CM. Semen retrieval in men with spinal cord injury is improved by interrupting current delivery during electroejaculation. J Urol 2002;167:201–3.Google Scholar
Restelli, AE, Bertolla, RP, Spaine, DM, Miotto, A Jr, Borrelli, M Jr, Cedenho, AP. Quality and functional aspects of sperm retrieved through assisted ejaculation in men with spinal cord injury. Fertil Steril 2009;91:819–25.Google Scholar
da Silva, BFBS, Borrelli, MMD, Fariello, RMBS, et al. Is sperm cryopreservation an option for fertility preservation in patients with spinal cord injury-induced anejaculation? Fertil Steril 2010;94:564–73.Google Scholar
Schatte, EC, Orejuela, FJ, Lipshultz, LI, Kim, ED, Lamb, DJ. Treatment of infertility due to anejaculation in the male with electroejaculation and intracytoplasmic sperm injection. J Urol 2000;163:1717–20.Google Scholar
Brackett, NL, Lynne, CM, Aballa, TC, Ferrell, SM. Sperm motility from the vas deferens of spinal cord injured men is higher than from the ejaculate. J Urol 2000;164:712–15.Google Scholar
Witt, MA, Grantmyre, JE, Lomas, M, Richard, J, Lipshultz, LI. The effect of semen quality of the electrical current and heat generated during rectal probe electroejaculation. J Urol 1992;147:747–9.Google Scholar

References

Schoor, RA, Elhanbly, S, Niederberger, CS, Ross, LS. The role of testicular biopsy in the modern management of male infertility. J Urol 2002;167:197200.Google Scholar
Jarvi, K, Lo, K, Fischer, A, et al. CUA Guideline: The workup of azoospermic males. Can Urol Assoc J 2010;4:163–7.Google Scholar
Bernie, AM, Mata, DA, Ramasamy, R, Schlegel, PN. Comparison of microdissection testicular sperm extraction, conventional testicular sperm extraction, and testicular sperm aspiration for nonobstructive azoospermia: a systematic review and meta-analysis. Fertil Steril 2015;104:1099–103.Google Scholar
Schlegel, PN, Su, L-M, Li, PS. Pathophysiologic changes after testicular sperm extraction: patterns and avoidance of testicular injury. In: Hamamah, S, Olivennes, F, Mieusset, R, Frydman, R, eds. Male Sterility and Motility Disorders: Etiologic Factors and Treatment. New York, NY: Springer-Verlag, 1999; pp. 243–52.Google Scholar
Khadra, A.A., Abdulhadi, I., Ghunain, S., Kilani, Z. Efficiency of percutaneous testicular sperm aspiration as a mode of sperm collection for intracytoplasmic sperm injection in nonobstructive azoospermia. J Urol. 2003;169:603–605.Google Scholar
Gottschalk-Sabag, S., Glick, T., Bar-On, E., Weiss, D.B. Testicular fine needle aspiration as a diagnostic method. Fertil Steril. 1993;59:1129–1131.Google Scholar
Meng, MV, Cha, I, Ljung, BM, Turek, PJ. Relationship between classic histological pattern and sperm findings on fine needle aspiration map in infertile men. Hum Reprod 2000;15:1973–7.Google Scholar
Ron-El, R, Strassburger, D, Friedler, S, et al. Extended sperm preparation: an alternative to testicular sperm extraction in non-obstructive azoospermia. Hum Reprod 1997;12:1222–6.Google Scholar
Fahmy, I, Mansour, R, Aboulghar, M, et al. Intracytoplasmic sperm injection using surgically retrieved epididymal and testicular spermatozoa in cases of obstructive and non-obstructive azoospermia. Int J Androl 1997;20:3744.Google Scholar
Amer, M, Haggar, SE, Moustafa, T, Abd El-Naser, T, Zohdy, W. Testicular sperm extraction: impact of testicular histology on outcome, number of biopsies to be performed and optimal time for repetition. Hum Reprod 1999;14:3030–4.Google Scholar
Schlegel, PN. Testicular sperm extraction: microdissection improves sperm yield with minimal tissue excision. Hum Reprod 1999;14:131–5.Google Scholar
Okada, H, Dobashi, M, Yamazaki, T, et al. Conventional versus microdissection testicular sperm extraction for nonobstructive azoospermia. J Urol 2002;168:1063–7.Google Scholar
Tsujimura, A, Matsumiya, K, Miyagawa, Y, et al. Conventional multiple or microdissection testicular sperm extraction: a comparative study. Hum Reprod 2002;17:2924–9.Google Scholar
Ramasamy, R, Yagan, N, Schlegel, PN. Structural and functional changes to the testis after conventional versus microdissection testicular sperm extraction. Urology 2005;65:1190–4.Google Scholar
Ramasamy, R, Schlegel, PN. Microdissection testicular sperm extraction: effect of prior biopsy on success of sperm retrieval. J Urol 2007;177:1447–9.Google Scholar
Schlegel, PN, Su, LM. Physiological consequences of testicular sperm extraction. Hum Reprod 1997;12:1688–92.Google Scholar
Amer, M, Ateyah, A, Hany, R, Zohdy, W. Prospective comparative study between microsurgical and conventional testicular sperm extraction in non-obstructive azoospermia: follow-up by serial ultrasound examinations. Hum Reprod 2000;15:653–6.Google Scholar
Everaert, K, De Croo, I, Kerckhaert, W, et al. Long term effects of micro-surgical testicular sperm extraction on androgen status in patients with non obstructive azoospermia. BMC Urol 2006;6:9.Google Scholar
Schlegel, PN, Liotta, D, Hariprashad, J, Veeck, LL. Fresh testicular sperm from men with non-obstructive azoospermia works best for ICSI. Urology 2004;64:1069–71.Google Scholar
Ohlander, S, Hotaling, J, Kirshenbaum, E, Niederberger, C, Eisenberg, ML. Impact of fresh versus cryopreserved testicular sperm upon intracytoplasmic sperm injection pregnancy outcomes in men with azoospermia due to spermatogenic dysfunction: a meta-analysis. Fertil Steril 2014;101:344–9.Google Scholar
Fullerton, G, Hamilton, M, Maheshwari, A. Should non-mosaic Klinefelter syndrome men be labelled as infertile in 2009? Hum Reprod 2010;25:588–97.Google Scholar
Dabaja, AA, Schlegel, PN. Microdissection testicular sperm extraction: an update. Asian J Androl 2013;15:35–9.Google Scholar
Damani, MN, Master, V, Meng, MV, Burgess, C, Turek, P, Oates, RD. Postchemotherapy ejaculatory azoospermia: fatherhood with sperm from testis tissue with intracytoplasmic sperm injection. J Clin Oncol 2002;20:930–6.Google Scholar
Hopps, CV, Mielnik, A, Goldstein, M, Palermo, GD, Rosenwaks, Z, Schlegel, PN. Detection of sperm in men with Y chromosome microdeletions of the AZFa, AZFb and AZFc regions. Hum Reprod 2003;18:1660–5.Google Scholar
Oates, RD. Clinical evaluation of the infertile male with respect to genetic etiologies. Syst Biol Reprod Med 2011;57:72–7.Google Scholar
Choi, DY, Gong, IH, Hwang, JH, Oh, JJ, Jong, JY. Detection of Y chromosome microdeletion is valuable in the treatment of patients with nonobstructive azoospermia and oligoasthenoteratozoospermia: sperm retrieval rate and birth rate. Korean J Urol 2013;54:111–16.Google Scholar
Weedin, JW, Bennett, RC, Fenig, DM, Lamb, DJ, Lipshultz, LI. Early versus late maturation arrest: reproductive outcomes of testicular failure. J Urol 2011;186:621–6.Google Scholar
Sheynkin, YR, Ye, Z, Menendez, S, Liotta, D, Veeck, LL, Schlegel, PN. Controlled comparison of percutaneous and microsurgical sperm retrieval in men with obstructive azoospermia. Hum Reprod 1998;13:3086–9.Google Scholar
Lee, R, Li, PS, Goldstein, M, Tanrikut, C, Schattman, G, Schlegel, PN. A decision analysis of treatments for obstructive azoospermia. Hum Reprod 2008;23:2043–9.Google Scholar

References

Weidel, L, Prins, GS. Cryosurvival of human spermatozoa frozen in eight different buffer systems. J Androl 1987;8:41–7.Google Scholar
Centola, G, Jeyendran, RS. Sperm processing techniques. In: Lipshultz, LI, Howards, SS, Niederberger, CS, eds. Infertility in the Male, 4th ed. Cambridge: Cambridge University Press, 2009; pp. 582–92.Google Scholar
Jeyendran, RS, Caroppo, E, Rouen, A, Anderson, A, Puscheck, E. Selecting the most competent sperm for assisted reproductive technologies. Fertil Steril 2019;111:851–63.Google Scholar
Schlegel, PN. What are the assisted reproductive technologies for male infertility? In: Robaire, B, Chan, P, eds. Handbook of Andrology, 2nd ed. East Dundee, IL: American Society of Andrology, 2008; pp. 626. Available from: www.andrologysociety.com/.Google Scholar
Byrd, W. Processing human semen for insemination comparison of methods. In: Centola, G, Ginsberg, KA, eds. Evaluation and Treatment of the Infertile Male. Cambridge: Cambridge University Press, 1996; pp. 89107.Google Scholar
European Society for Human Reproduction. Revised guidelines for good practice in IVF laboratories. 2015 . Available from: www.eshre.eu/Guidelines-and-Legal/Guidelines/Revised-guidelines-for-good-practice-in-IVF-laboratories-(2015).aspx.Google Scholar
World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen, 5th ed. Geneva: World Health Organization, 2010.Google Scholar
Wald, M, Prins, G. Sperm banking: indications and techniques. In: Lipshultz, LI, Howards, SS, Niederberger, CS, eds. Infertility in the Male, 4th ed. Cambridge: Cambridge University Press, 2009; pp. 593602.Google Scholar
De Los Santos, MJ, Apter, S, Coticchio, G, et al. Revised guidelines for good practice in IVF laboratories (2015). Hum Reprod 2016;31:685–6.Google Scholar
Niederberger, CS. Male infertility. In: Wein, AJ, Kavoussi, LR, Partin, AW, Peters, CA, eds. Campbell-Walsh Urology. Philadelphia, PA: Elsevier, 2016; pp. 556–79.Google Scholar
Jefferys, A, Siassakos, D, Wardle, P. The management of retrograde ejaculation: a systematic review and update. Fertil Steril 2012;97:30612.e6.Google Scholar
Mullin, C, Stelling, J, Schoor, R. Intrauterine insemination from the urologist’s perspective. In: Lipshultz, LI, Howards, SS, Niederberger, CS, eds. Infertility in the Male, 4th ed. Cambridge: Cambridge University Press, 2009; pp. 4939.Google Scholar
Phipps, W. Assisted reproductive technology for male factor infertility. In: Centola, G, Ginsburg, KA, eds. Evaluation and Treatment of the Infertile Male. Cambridge: Cambridge University Press, 1996; pp. 130–55.Google Scholar
Morin, SJ, Scott, RT. Knowledge gaps in male infertility: a reproductive endocrinology and infertility perspective. Transl Androl Urol 2018;7(Suppl 3):S283–91.Google Scholar
Pagani, RL, Serafini, PC, Srougi, M. In-vitro fertilization and micromanipulation for male infertility. In: Lipshultz, LI, Howards, SS, Niederberger, CS, eds. Infertility in the Male, 4th ed. Cambridge: Cambridge University Press, 2009; pp. 500–15.Google Scholar
Moolenaar, LM, Cissen, M, De Bruin, JP, et al. Cost-effectiveness of assisted conception for male subfertility. Reprod Biomed Online 2015;30:659–66.Google Scholar
Van Voorhis, BJ, Barnett, M, Sparks, AET, Syrop, CH, Rosenthal, G, Dawson, J. Effect of the total motile sperm count on the efficacy and cost-effectiveness of intrauterine insemination and in vitro fertilization. Fertil Steril 2001;75:661–8.Google Scholar
Prins, GS, Dolgina, R, Studney, P, Kaplan, B, Ross, L, Niederberger, C. Quality of cryopreserved testicular sperm in patients with obstructive and nonobstructive azoospermia. J Urol 1999;161:1504–8.Google Scholar
Rothmann, S. What is sperm banking? When and how is it (or should it be) used in humans? Animals? In: Robaire, B, Chan, P, eds. Handbook of Andrology, 2nd ed. East Dundee, IL: American Society of Andrology, 2008; pp. 414. Available from: www.andrologysociety.com/.Google Scholar
Gliedt, D, Reed, M. Preparation of gametes for assisted reproductive technologies. In: Andrology and Embryology Review Course Manual. St Louis, MO: American Association of Bioanalysists, 2003; pp. 8.18.58.Google Scholar
Ruiz, A, Jeyendran, RS. Sperm processing procedure for intrauterine insemination. In: Yeyendran, RS, ed. Sperm Collection and Processing Methods. Cambridge: Cambridge University Press, 2003; pp. 123–40.Google Scholar
Centola, G. Sperm preparation for insemination. In: Patton, PE, Battaglia, DE, eds. Office Andrology. Totowa, NJ: Humana Press, 2005; pp. 3952.Google Scholar
Said, TM, Grunewald, S, Paasch, U, et al. Advantage of combining magnetic cell separation with sperm preparation techniques. Reprod Biomed Online 2005;10:740–6.Google Scholar
Malvezzi, H, Sharma, R, Agarwal, A, Abuzenadah, AM, Abu-Elmagd, M. Sperm quality after density gradient centrifugation with three commercially available media: a controlled trial. Reprod Biol Endocrinol 2014;12:17.Google Scholar
Allamaneni, SSR, Agrawal, A, Rama, S, Ranganathan, P, Sharma, RK. Comparative study on density gradients and swim-up preparation techniques utilizing neat and cryopreserved spermatozoa. Asian J Androl 2005;7:8692.Google Scholar
Henkel, RR, Schill, WB. Sperm preparation for ART. Reprod Biol Endocrinol 2003;1:122.Google Scholar
Oshio, S, Kaneko, S, Lizuka, R, Mohri, H. Effects of gradient centrifugation on human sperm. Arch Androl 1987;19:8593.Google Scholar
Enciso, M, Iglesias, M, Galán, I, Sarasa, J, Gosálvez, A, Gosálvez, J. The ability of sperm selection techniques to remove single- or double-strand DNA damage. Asian J Androl 2011;13:764–8.Google Scholar
Zorn, B, Vidmar, G, Meden-Vrtovec, H. Seminal reactive oxygen species as predictors of fertilization, embryo quality and pregnancy rates after conventional in vitro fertilization and intracytoplasmic sperm injection. Int J Androl 2003;26:279–85.Google Scholar
Takeshima, T, Yumura, Y, Kuroda, S, Kawahara, T, Uemura, H, Iwasaki, A. Effect of density gradient centrifugation on reactive oxygen species in human semen. Syst Biol Reprod Med 2017;63:192–8.Google Scholar
Ahelik, A, Mändar, R, Korrovits, P, et al. Systemic oxidative stress could predict assisted reproductive technique outcome. J Assist Reprod Genet 2015;32:699704.Google Scholar
Yumura, Y, Iwasaki, A, Saito, K, Ogawa, T, Hirokawa, M. Effect of reactive oxygen species in semen on the pregnancy of infertile couples. Int J Urol 2009;16:202–7.Google Scholar
Boomsma, CM, Heineman, MJ, Cohlen, BJ, Farquhar, C. Semen preparation techniques for intrauterine insemination. Cochrane Database Syst Rev 2007;4:CD004507.Google Scholar
Jeyendran, RS, Perez-Pelaez, M, Crabo, BG. Concentration of viable spermatozoa for artificial insemination. Fertil Steril 1986;45:132–4Google Scholar
Henkel, RR, Franken, DR, Lombard, CJ, Schill, WB. Selective capacity of glass-wool filtration for the separation of human spermatozoa with condensed chromatin: a possible therapeutic modality for male-factor cases? J Assist Reprod Genet 1994;11:395400.Google Scholar
Sterzik, K, De Santo, M, Uhlich, S, Gagsteiger, F, Strehler, E. Glass wool filtration leads to a higher percentage of spermatozoa with intact acrosomes: an ultrastructural analysis. Hum Reprod 1998;13:2506–11.Google Scholar
Larson, KL, Brannian, JD, Timm, BK, Jost, LK, Evenson, DP. Density gradient centrifugation and glass wool filtration of semen remove spermatozoa with damaged chromatin structure. Hum Reprod 1999;14:2015–19.Google Scholar
Johnson, DE, Confino, E, Jeyendran, RS. Glass wool column filtration versus mini-Percoll gradient for processing poor quality semen samples. Fertil Steril 1996;66:459–62.Google Scholar
Van Der Ven, HH, Jeyendran, RS, Al-hasani, S, et al. Glass wool column filtration of human semen: relation to swim-up procedure and outcome of IVF. Hum Reprod 1988;3:85–8.Google Scholar
Tanphaichitr, N, Randall, M, Fitzgerald, L, Lee, G, Seibel, M, Taymor, M. An increase in in vitro fertilization ability of low-density human sperm capacitated by multiple-tube swim-up. Fertil Steril 1987;48:821–7.Google Scholar
Andolz, P, Bielsa, M, Genesca, A, Benet, J, Egozcue, J. Improvement of sperm quality in abnormal semen samples using a modified swim-up procedure. Hum Reprod 1987;2:99101.Google Scholar
Calamera, J, Brugo, S, Quiros, M, Nicholson, R. Computer assisted measurement in normal and pathological human semen, fresh and post swim-up technique. Andrologia 1989;21:340–5.Google Scholar
Yetkinel, S, Kilicdag, EB, Aytac, PC, Haydardedeoglu, B, Simsek, E, Cok, T. Effects of the microfluidic chip technique in sperm selection for intracytoplasmic sperm injection for unexplained infertility: a prospective, randomized controlled trial. J Assist Reprod Genet 2019;36:403–9.Google Scholar
Bower, L, Serafini, P, Wikland, M, Moyer, D. The use of hyaluronic acid for sperm selection – evaluation of sperm head ultrastructure. Int J Fertil 1989;34:420–4.Google Scholar
Barak, Y, Amit, A, Lessing, J, Paz, G, Homonnai, T, Yogey, L. Improved fertilization rate in an in-vitro fertilization program by egg yolk treated sperm. Fertil Steril 1992;58:197–8.Google Scholar
Holmgren, WJ, Jeyendran, RS, Neff, MR, Perez-Pelaez, M, Zaneveld, LJD. Preincubation of human spermatozoa in test-yolk medium: effect on penetration of zona-free hamster oocytes and correlation with other semen characteristics. J Vitr Fertil Embryo Transf 1989;6:207–12.Google Scholar
Jeyendran, RS, Gunawardana, VK, Barisic, D, Wentz, AC. TEST-yolk media and sperm quality. Hum Reprod Update 1995;1:73–9.Google Scholar
Bielfeld, P, Jeyendran, RS, Holmgren, WJ, Zaneveld, LJD. Effect of egg yolk medium on the acrosome reaction of human spermatozoa. J Androl 1990;11:260–9.Google Scholar
Yang, YS, Rojas, FJ, Stone, SC. Acrosome reaction of human spermatozoa in zona-free hamster egg penetration test. Fertil Steril 1988;50:954–9.Google Scholar
Ericsson, R, Langevin, C, Nishino, M. Isolation of fractions rich in human Y sperm. Nature 1973;246:421.Google Scholar
Janson, J-C. On the history of the development of Sephadex. Chromatographia 1987;23:361–5.Google Scholar
Byrd, W, Drobnis, E, Kutteh, W, Marshburn, P, Carr, B. Intrauterine insemination with frozen donor sperm: a prospective randomized trial comparing three different sperm preparation techniques. Fertil Steril 1994;62:850–6.Google Scholar
Amer, M, Metawae, B, Hosny, H, Raef, A. Beneficial effect of adding pentoxifylline to processed semen samples on ICSI outcome in infertile males with mild and moderate asthenozoospermia: a randomized controlled prospective crossover study. Int J Reprod Biomed 2013;11:939–44.Google Scholar
Negri, P, Grechi, E, Tomasi, A, Fabbri, E, Capuzzo, A. Effectiveness of pentoxifylline in semen preparation for intrauterine insemination. Hum Reprod 1996;11:1236–9.Google Scholar
Imoedemhe, DAG, Sigue, AB, Pacpaco, ELA, Olazo, AB. The effect of caffeine on the ability of spermatozoa to fertilize mature human oocytes. J Assist Reprod Genet 1992;9:155–60.Google Scholar
Moghadam, KK, Nett, R, Robins, JC, et al. The motility of epididymal or testicular spermatozoa does not directly affect IVF/ICSI pregnancy outcomes. J Androl 2005;26:619–23.Google Scholar
Zhang, W, Li, F, Cao, H, Li, C. Protective effects of l-carnitine on astheno- and normozoospermic human semen samples during cryopreservation. Zygote 2016;24:293300.Google Scholar
Yang, C, Xu, L, Cui, Y, Wu, B, Liao, Z. Potent humanin analogue (HNG) protects human sperm from freeze-thaw-induced damage. Cryobiology 2019;88:4753.Google Scholar
Saeednia, S, Shabani Nashtaei, M, Bahadoran, H, Aleyasin, A, Amidi, F. Effect of nerve growth factor on sperm quality in asthenozoospermic men during cryopreservation. Reprod Biol Endocrinol 2016;14:18.Google Scholar
Zhang, X, Lu, X, Li, J, Xia, Q, Gao, J, Wu, B. Mito-Tempo alleviates cryodamage by regulating intracellular oxidative metabolism in spermatozoa from asthenozoospermic patients. Cryobiology 2019;91:1822.Google Scholar
Bartoov, B, Berkovitz, A, Eltes, F, Kogosowski, A, Menezo, Y, Barak, Y. Real-time fine morphology of motile human sperm cells is associated with IVF-ICSI outcome. J Androl 2001;23:18.Google Scholar
Rappa, KL, Rodriguez, HF, Hakkarainen, GC, Anchan, RM, Mutter, GL, Asghar, W. Sperm processing for advanced reproductive technologies: where are we today? Biotechnol Adv 2016;34:578–87.Google Scholar
Bartoov, B, Berkovitz, A, Eltes, F, et al. Pregnancy rates are higher with intracytoplasmic morphologically selected sperm injection than with conventional intracytoplasmic injection. Fertil Steril 2003;80:1413–19.Google Scholar
Teixeira, DM, Barbosa, MAP, Ferriani, RA, et al. Regular (ICSI) versus ultra-high magnification (IMSI) sperm selection for assisted reproduction. Cochrane Database Syst Rev 2013;7:CD010167.Google Scholar
Huszar, G, Jakab, A, Sakkas, D, et al. Fertility testing and ICSI sperm selection by hyaluronic acid binding: clinical and genetic aspects. Reprod Biomed Online 2007;14:650–63.Google Scholar
Parmegiani, L, Cognigni, GE, Bernardi, S, Troilo, E, Ciampaglia, W, Filicori, M. “Physiologic ICSI”: Hyaluronic acid (HA) favors selection of spermatozoa without DNA fragmentation and with normal nucleus, resulting in improvement of embryo quality. Fertil Steril 2010;93:598604.Google Scholar
Miller, D, Pavitt, S, Sharma, V, et al. Physiological, hyaluronan-selected intracytoplasmic sperm injection for infertility treatment (HABSelect): a parallel, two-group, randomised trial. Lancet 2019;393:416–22.Google Scholar
Liu, Y, Feenan, K, Chapple, V, Roberts, P. Intracytoplasmic sperm injection using hyaluronic acid or polyvinylpyrrolidone: a time-lapse sibling oocyte study. Hum Fertil 2017;22:3945.Google Scholar
Smith, GD, Takayama, S. Application of microfluidic technologies to human assisted reproduction. Mol Hum Reprod 2017;23:257–68.Google Scholar
Yetkinel, S, Kilicdag, EB, Aytac, PC, Haydardedeoglu, B, Simsek, E, Cok, T. Effects of the microfluidic chip technique in sperm selection for intracytoplasmic sperm injection for unexplained infertility: a prospective, randomized controlled trial. J Assist Reprod Genet 2019;36:403–9.Google Scholar
Stanger, JD, Vo, L, Yovich, JL, Almahbobi, G. Hypo-osmotic swelling test identifies individual spermatozoa with minimal DNA fragmentation. Reprod Biomed Online 2010;21:474–84.Google Scholar
Pang, MG, You, YA, Park, YJ, Oh, SA, Kim, DS, Kim, YJ. Numerical chromosome abnormalities are associated with sperm tail swelling patterns. Fertil Steril 2010;94:1012–20.Google Scholar
Rouen, A, Carlier, L, Heide, S, et al. Potential selection of genetically balanced spermatozoa based on the hypo-osmotic swelling test in chromosomal rearrangement carriers. Reprod Biomed Online 2017;35:372–8.Google Scholar
Tsai, YL, Liu, J, Garcia, JE, Katz, E, Compton, G, Baramki, TA. Establishment of an optimal hypo-osmotic swelling test by examining single spermatozoa in four different hypo-osmotic solutions. Hum Reprod 1997;12:1111–13.Google Scholar
Jeyendran, RS, Van der Ven, HH, Zaneveld, LJD. The hypoosmotic swelling test: an update. Syst Biol Reprod Med 1992;29:105–16.Google Scholar
Coetzee, K, Kruger, TF, Menkveld, R, Lombard, CJ, Swanson, RJ. Hypoosmotic swelling test in the prediction of male fertility. Syst Biol Reprod Med 1989;23:131–8.Google Scholar
Nordhoff, V. How to select immotile but viable spermatozoa on the day of intracytoplasmic sperm injection? An embryologist’s view. Andrology 2015;3:156–62.Google Scholar
Mulhall, JP, Burgess, CM, Cunningham, D, Carson, R, Harris, D, Oates, RD. Presence of mature sperm in testicular parenchyma of men with nonobstructive azoospermia: prevalence and predictive factors. Urology 1997;49:91–5.Google Scholar
Oates, RD, Mulhall, J, Burgess, C, Cunningham, D, Carson, R. Fertilization and pregnancy using intentionally cryopreserved testicular tissue as the sperm source for intracytoplasmic sperm injection in 10 men with nonobstructive azoospermia. Hum Reprod 1997;12:734–9.Google Scholar
Wald, M, Ross, LS, Prins, GS, Cieslak-Janzen, J, Wolf, G, Niederberger, CS. Analysis of outcomes of cryopreserved surgically retrieved sperm for IVF/ICSI. J Androl 2006;27:60–5.Google Scholar
Habermann, H, Seo, R, Cieslak, J, Niederberger, C, Prins, GS, Ross, L. In vitro fertilization outcomes after intracytoplasmic sperm injection with fresh or frozen-thawed testicular spermatozoa. Fertil Steril 2000;73:955–60.Google Scholar
Prins, GS, Weidel, L. A comparative study of buffer systems as cryoprotectants for human spermatozoa. Fertil Steril 1986;46:147–9.Google Scholar
Sawetawan, C, Bruns, ES, Prins, GS. Improvement of post-thaw sperm motility in poor quality human semen. Fertil Steril 1993;60:706–10.Google Scholar
Fiser, P, Fairfull, RW. The effects of rapid cooling (cold shock) of ram semen, photoperiod, and egg yolk in diluents on the survival of spermatozoa before and after freezing. Cryobiology 1986;23:518–24.Google Scholar
Hotaling, J, Wen, J, Belmonte, J, Niederberger, C, Prins, GS. 2034 polymerized alginic acid bead and sperm injection through micro-manipulation as a vehicle for cryopreservation and recovery of small numbers of human sperm. J Urol 2013;189(Suppl 4S):e835.Google Scholar
Coetzee, K, Ozgur, K, Berkkanoglu, M, Bulut, H, Isikli, A. Reliable single sperm cryopreservation in Cell Sleepers for azoospermia management. Andrologia 2016;48:203–10.Google Scholar
Kathrins, M, Abhyankar, N, Shoshany, O, et al. Post-thaw recovery of rare or very low concentrations of cryopreserved human sperm. Fertil Steril 2017;107:1300–4.Google Scholar
Kramer, RY, Garner, DL, Bruns, ES, Ericsson, SA, Prins, GS. Comparison of motility and flow cytometric assessments of seminal quality in fresh, 24‐hour extended and cryopreserved human spermatozoa. J Androl 1993;14:374–84.Google Scholar

Further Reading

Del Vento, F, Vermeulen, M, de Michele, F, et al. Tissue engineering to improve immature testicular tissue and cell transplantation outcomes: one step closer to fertility restoration for prepubertal boys exposed to gonadotoxic treatments. Int J Mol Sci 2018;19:286.Google Scholar
Halpern, JA, Hill, R, Brannigan, RE. Guideline based approach to male fertility preservation. Urol Oncol 2020;38:31–5.Google Scholar
Oktay, K, Harvey, BE, Partridge, AH, et al. Fertility preservation in patients with cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol 2018;36:19942001.Google Scholar
Onofre, J, Baert, Y, Faes, K, Goossens, E. Cryopreservation of testicular tissue or testicular cell suspensions: a pivotal step in fertility preservation. Hum Reprod Update 2016;22:744–61.Google Scholar

References

Howlader, N, Noone, AM, Krapcho, M, et al.; National Cancer Institute (eds). SEER Cancer Statistics Review, 1975–2016. Available from: seer.cancer.gov/csr/1975_2016/.Google Scholar
Burkhamer, J, Kriebel, D, Clapp, R. The increasing toll of adolescent cancer incidence in the US. PLoS ONE 2017;12:e0172986.Google Scholar
Sanft, T, Denlinger, CS, Armenian, S, et al. NCCN Guidelines Insights: Survivorship, Version 2.2019. J Natl Compr Canc Netw 2019;17:784–94.Google Scholar
Borno, HT, Dixit, N. Tailoring survivorship to diverse populations with genitourinary malignancies. Urol Oncol 2020;38:118–20.Google Scholar
Zebrack, BJ, Block, R, Hayes-Lattin, B, et al. Psychosocial service use and unmet need among recently diagnosed adolescent and young adult cancer patients. Cancer 2013;119:201–14.Google Scholar
Knapp, CA, Quinn, GP, Murphy, D. Assessing the reproductive concerns of children and adolescents with cancer: challenges and potential solutions. J Adolesc Young Adult Oncol 2011;1:31–5.Google Scholar
Wyns, C, Collienne, C, Shenfield, F, et al. Fertility preservation in the male pediatric population: factors influencing the decision of parents and children. Hum Reprod 2015;30:2022–30.Google Scholar
Williams, DH. Sperm banking and the cancer patient. Ther Adv Urol 2010;2:1934.Google Scholar
Williams, DHT, Karpman, E, Sander, JC, Spiess, PE, Pisters, LL, Lipshultz, LI. Pretreatment semen parameters in men with cancer. J Urol 2009;181:736–40.Google Scholar
Coward, RM, Kovac, JR, Smith, RP, Lipshultz, LI. Fertility preservation in young men treated for malignancies: options for precancer treatment. Sex Med Rev 2013;1:123–34.Google Scholar
Polland, A, Berookhim, BM. Fertility concerns in men with genitourinary malignancies: treatment dilemmas, fertility options, and medicolegal considerations. Urol Oncol 2016;34:399406.Google Scholar
Shnorhavorian, M, Harlan, LC, Smith, AW, et al. Fertility preservation knowledge, counseling, and actions among adolescent and young adult patients with cancer: a population-based study. Cancer 2015;121:3499–506.Google Scholar
Lee, SJ, Schover, LR, Partridge, AH, et al. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol 2006;24:2917–31.Google Scholar
Loren, AW, Mangu, PB, Beck, LN, et al. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol 2013;31:2500–10.Google Scholar
Quinn, GP, Vadaparampil, ST, Lee, JH, et al. Physician referral for fertility preservation in oncology patients: a national study of practice behaviors. J Clin Oncol 2009;27:5952–7.Google Scholar
Schover, LR, Brey, K, Lichtin, A, Lipshultz, LI, Jeha, S. Oncologists’ attitudes and practices regarding banking sperm before cancer treatment. J Clin Oncol 2002;20:1890–7.Google Scholar
Lee, SJ. Preservation of fertility in patients with cancer. N Engl J Med 2009;360:2680; author reply 23.Google Scholar
Oktay, K, Harvey, BE, Partridge, AH, et al. Fertility preservation in patients with cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol 2018;36:19942001.Google Scholar
Coccia, PF, Altman, J, Bhatia, S, et al. Adolescent and young adult oncology. Clinical practice guidelines in oncology. J Natl Compr Canc Netw 2012;10:1112–50.Google Scholar
Coccia, PF, Pappo, AS, Altman, J, et al. Adolescent and young adult oncology, version 2.2014. J Natl Compr Canc Netw 2014;12:2132.Google Scholar
Coccia, PF, Pappo, AS, Beaupin, L, et al. Adolescent and Young Adult Oncology, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2018;16:6697.Google Scholar
Peccatori, FA, Azim, HA Jr, Orecchia, R, et al. Cancer, pregnancy and fertility: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2013;24 Suppl 6:vi160–70.Google Scholar
Fallat, ME, Hutter, J. Preservation of fertility in pediatric and adolescent patients with cancer. Pediatrics 2008;121:e1461–9.Google Scholar
Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in cancer patients. Fertil Steril 2005;83:1622–8.Google Scholar
Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in patients facing gonadotoxic therapies: a committee opinion. Fertil Steril 2013;100:1224–31.Google Scholar
Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in patients facing gonadotoxic therapies: an Ethics Committee opinion. Fertil Steril 2018;110:380–6.Google Scholar
Salsman, JM, Yanez, B, Smith, KN, et al. Documentation of fertility preservation discussions for young adults with cancer: examining compliance with treatment guidelines. J Natl Compr Canc Netw 2016;14:301–9.Google Scholar
Burns, KC, Hoefgen, H, Strine, A, Dasgupta, R. Fertility preservation options in pediatric and adolescent patients with cancer. Cancer 2018;124:1867–76.Google Scholar
Meistrich, ML. Effects of chemotherapy and radiotherapy on spermatogenesis in humans. Fertil Steril 2013;100:1180–6.Google Scholar
Kondo, N, Takahashi, A, Ono, K, Ohnishi, T. DNA damage induced by alkylating agents and repair pathways. J Nucleic Acids 2010;2010:543531.Google Scholar
Colvin, M. Alkylating agents and platinum antitumor compounds. In: Kufe, DW, Pollock, RE, Weichselbaum, RR, et al., eds. Holland-Frei Cancer Medicine, 6th ed. Hamilton, ON: BC Decker, 2003. Available from: www.ncbi.nlm.nih.gov/books/NBK12354/.Google Scholar
Chow, EJ, Stratton, KL, Leisenring, WM, et al. Pregnancy after chemotherapy in male and female survivors of childhood cancer treated between 1970 and 1999: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol 2016;17:567–76.Google Scholar
Green, DM, Liu, W, Kutteh, WH, et al. Cumulative alkylating agent exposure and semen parameters in adult survivors of childhood cancer: a report from the St Jude Lifetime Cohort Study. Lancet Oncol 2014;15:1215–23.Google Scholar
Poganitsch-Korhonen, M, Masliukaite, I, Nurmio, M, et al. Decreased spermatogonial quantity in prepubertal boys with leukaemia treated with alkylating agents. Leukemia 2017;31:1460–3.Google Scholar
Meistrich, ML, Wilson, G, Brown, BW, da Cunha, MF, Lipshultz, LI. Impact of cyclophosphamide on long-term reduction in sperm count in men treated with combination chemotherapy for Ewing and soft tissue sarcomas. Cancer 1992;70:2703–12.Google Scholar
Rowley, MJ, Leach, DR, Warner, GA, Heller, CG. Effect of graded doses of ionizing radiation on the human testis. Radiation Res 1974;59:665–78.Google Scholar
Centola, GM, Keller, JW, Henzler, M, Rubin, P. Effect of low-dose testicular irradiation on sperm count and fertility in patients with testicular seminoma. J Androl 1994;15:608–13.Google Scholar
Skinner, R, Mulder, RL, Kremer, LC, et al. Recommendations for gonadotoxicity surveillance in male childhood, adolescent, and young adult cancer survivors: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group in collaboration with the PanCareSurFup Consortium. Lancet Oncol 2017;18:e75–90.Google Scholar
Giwercman, A, von der Maase, H, Berthelsen, JG, Rorth, M, Bertelsen, A, Skakkebaek, NE. Localized irradiation of testes with carcinoma in situ: effects on Leydig cell function and eradication of malignant germ cells in 20 patients. J Clin Endocrinol Metab 1991;73:596603.Google Scholar
Djaladat, H, Burner, E, Parikh, PM, Beroukhim Kay, D, Hays, K. The association between testis cancer and semen abnormalities before orchiectomy: a systematic review. J Adolesc Young Adult Oncol 2014;3:153–9.Google Scholar
Agarwal, A, Allamaneni, SS. Disruption of spermatogenesis by the cancer disease process. J Natl Cancer Inst Monogr 2005;34:912.Google Scholar
Shoshany, O, Shtabholtz, Y, Schreter, E, et al. Predictors of spermatogenesis in radical orchiectomy specimen and potential implications for patients with testicular cancer. Fertil Steril 2016;106:70–4.Google Scholar
Berthelsen, JG. Testicular cancer and fertility. Int J Androl 1987;10:371–80.Google Scholar
van Iersel, L, Li, Z, Srivastava, DK, et al. Hypothalamic–pituitary disorders in childhood cancer survivors: prevalence, risk factors and long-term Health outcomes. J Clin Endocrinol Metab 2019;104:6101–15.Google Scholar
Shalet, SM, Tsatsoulis, A, Whitehead, E, Read, G. Vulnerability of the human Leydig cell to radiation damage is dependent upon age. J Endocrinol 1989;120:161–5.Google Scholar
Mostoufi-Moab, S, Seidel, K, Leisenring, WM, et al. Endocrine abnormalities in aging survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Clin Oncol 2016;34:3240–7.Google Scholar
Izard, MA. Leydig cell function and radiation: a review of the literature. Radiother Oncol 1995;34:18.Google Scholar
Sprauten, M, Brydoy, M, Haugnes, HS, et al. Longitudinal serum testosterone, luteinizing hormone, and follicle-stimulating hormone levels in a population-based sample of long-term testicular cancer survivors. J Clin Oncol 2014;32:571–8.Google Scholar
Walsh, PC. Nerve sparing radical prostatectomy for early stage prostate cancer. Semin Oncol 1988;15:351–8.Google Scholar
Pearce, S, Steinberg, Z, Eggener, S. Critical evaluation of modified templates and current trends in retroperitoneal lymph node dissection. Curr Urol Rep 2013;14:511–17.Google Scholar
Crawshaw, M. Male coping with cancer-fertility issues: putting the ‘social’ into biopsychosocial approaches. Reprod Biomed Online 2013;27:261–70.Google Scholar
Dupree, JM. Insurance coverage of male infertility: what should the standard be? Transl Androl Urol 2018;7:S310–16.Google Scholar
Hanson, BM, Eisenberg, ML, Hotaling, JM. Male infertility: a biomarker of individual and familial cancer risk. Fertil Steril 2018;109:619.Google Scholar
Nagirnaja, L, Aston, KI, Conrad, DF. Genetic intersection of male infertility and cancer. Fertil Steril 2018;109:20–6.Google Scholar
Vogelstein, B, Papadopoulos, N, Velculescu, VE, Zhou, S, Diaz, LA Jr, Kinzler, KW. Cancer genome landscapes. Science 2013;339:1546–58.Google Scholar
Menon, S, Rives, N, Mousset-Simeon, N, et al. Fertility preservation in adolescent males: experience over 22 years at Rouen University Hospital. Hum Reprod 2009;24:3744.Google Scholar
Grover, NS, Deal, AM, Wood, WA, Mersereau, JE. Young men with cancer experience low referral rates for fertility counseling and sperm banking. J Oncol Pract 2016;12:465–71.Google Scholar
Sonnenburg, DW, Brames, MJ, Case-Eads, S, Einhorn, LH. Utilization of sperm banking and barriers to its use in testicular cancer patients. Support Care Cancer 2015;23:2763–8.Google Scholar
Rives, N, Perdrix, A, Hennebicq, S, et al. The semen quality of 1158 men with testicular cancer at the time of cryopreservation: results of the French National CECOS Network. J Androl 2012;33:1394–401.Google Scholar
Schrader, M, Muller, M, Sofikitis, N, Straub, B, Krause, H, Miller, K. “Onco-tese”: testicular sperm extraction in azoospermic cancer patients before chemotherapy-new guidelines? Urology 2003;61:421–5.Google Scholar
Furuhashi, K, Ishikawa, T, Hashimoto, H, et al. Onco-testicular sperm extraction: testicular sperm extraction in azoospermic and very severely oligozoospermic cancer patients. Andrologia 2013;45:107–10.Google Scholar
Suzuki, K, Shin, T, Shimomura, Y, Iwahata, T, Okada, H. Spermatogenesis in tumor-bearing testes in germ cell testicular cancer patients. Hum Reprod 2015;30:2853–8.Google Scholar
Fayomi, AP, Peters, K, Sukhwani, M, et al. Autologous grafting of cryopreserved prepubertal rhesus testis produces sperm and offspring. Science 2019;363:1314–19.Google Scholar
Hotaling, JM, Lopushnyan, NA, Davenport, M, et al. Raw and test-thaw semen parameters after cryopreservation among men with newly diagnosed cancer. Fertil Steril 2013;99:464–9.Google Scholar
Hotaling, JM, Patel, DP, Vendryes, C, et al. Predictors of sperm recovery after cryopreservation in testicular cancer. Asian J Androl 2016;18:35–8.Google Scholar
Ohlander, S, Hotaling, J, Kirshenbaum, E, Niederberger, C, Eisenberg, ML. Impact of fresh versus cryopreserved testicular sperm upon intracytoplasmic sperm injection pregnancy outcomes in men with azoospermia due to spermatogenic dysfunction: a meta-analysis. Fertil Steril 2014;101:344–9.Google Scholar
Bernie, AM, Mata, DA, Ramasamy, R, Schlegel, PN. Comparison of microdissection testicular sperm extraction, conventional testicular sperm extraction, and testicular sperm aspiration for nonobstructive azoospermia: a systematic review and meta-analysis. Fertil Steril 2015;104:1099103.e1–3.Google Scholar
Flannigan, R, Bach, PV, Schlegel, PN. Microdissection testicular sperm extraction. Transl Androl Urol 2017;6:745–52.Google Scholar
Meseguer, M, Garrido, N, Remohi, J, et al. Testicular sperm extraction (TESE) and ICSI in patients with permanent azoospermia after chemotherapy. Hum Reprod 2003;18:1281–5.Google Scholar
Hsiao, W, Stahl, PJ, Osterberg, EC, et al. Successful treatment of postchemotherapy azoospermia with microsurgical testicular sperm extraction: the Weill Cornell experience. J Clin Oncol 2011;29:1607–11.Google Scholar
Shoshany, O, Abhyankar, N, Elyaguov, J, Niederberger, C. Efficacy of treatment with pseudoephedrine in men with retrograde ejaculation. Andrology 2017;5:744–8.Google Scholar
Aust, TR, Brookes, S, Troup, SA, Fraser, WD, Lewis-Jones, DI. Development and in vitro testing of a new method of urine preparation for retrograde ejaculation; the Liverpool solution. Fertil Steril 2008;89:885–91.Google Scholar
Fode, M, Ohl, DA, Sonksen, J. A step-wise approach to sperm retrieval in men with neurogenic anejaculation. Nat Rev Urol 2015;12:607–16.Google Scholar
Hsiao, W, Deveci, S, Mulhall, JP. Outcomes of the management of post-chemotherapy retroperitoneal lymph node dissection-associated anejaculation. BJU Int 2012;110:1196–200.Google Scholar
Mulhall, JP, Trost, LW, Brannigan, RE, et al. Evaluation and management of testosterone deficiency: AUA Guideline. J Urol 2018;200:423–32.Google Scholar
Patel, DP, Chandrapal, JC, Hotaling, JM. Hormone-based treatments in subfertile males. Curr Urol Rep 2016;17:56.Google Scholar
Ko, EY, Siddiqi, K, Brannigan, RE, Sabanegh, ES Jr. Empirical medical therapy for idiopathic male infertility: a survey of the American Urological Association. J Urol 2012;187:973–8.Google Scholar
Hussein, A, Ozgok, Y, Ross, L, Niederberger, C. Clomiphene administration for cases of nonobstructive azoospermia: a multicenter study. J Androl 2005;26:787–91; discussion 92–3.Google Scholar
Hussein, A, Ozgok, Y, Ross, L, Rao, P, Niederberger, C. Optimization of spermatogenesis-regulating hormones in patients with non-obstructive azoospermia and its impact on sperm retrieval: a multicentre study. BJU Int 2013;111:E110–4.Google Scholar
Cavallini, G, Beretta, G, Biagiotti, G. Preliminary study of letrozole use for improving spermatogenesis in non-obstructive azoospermia patients with normal serum FSH. Asian J Androl 2011;13:895–7.Google Scholar
Cavallini, G, Biagiotti, G, Bolzon, E. Multivariate analysis to predict letrozole efficacy in improving sperm count of non-obstructive azoospermic and cryptozoospermic patients: a pilot study. Asian J Androl 2013;15:806–11.Google Scholar
Saylam, B, Efesoy, O, Cayan, S. The effect of aromatase inhibitor letrozole on body mass index, serum hormones, and sperm parameters in infertile men. Fertil Steril 2011;95:809–11.Google Scholar
Farhat, R, Al-zidjali, F, Alzahrani, AS. Outcome of gonadotropin therapy for male infertility due to hypogonadotrophic hypogonadism. Pituitary 2010;13:105–10.Google Scholar
Yang, L, Zhang, SX, Dong, Q, Xiong, ZB, Li, X. Application of hormonal treatment in hypogonadotropic hypogonadism: more than ten years experience. Int Urol Nephrol 2012;44:393–9.Google Scholar
Tremblay, A, Beaud, H, Delbes, G. Transgenerational impact of chemotherapy: would the father exposure impact the health of future progeny?. Gynecol Obstet Fertil Senol 2017;45:609–18.Google Scholar
Patel, B, Meeks, H, Wan, Y, et al. Transgenerational effects of chemotherapy: both male and female children born to women exposed to chemotherapy have fewer children. Cancer Epidemiol 2018;56:15.Google Scholar
Ginsberg, JP, Ogle, SK, Tuchman, LK, et al. Sperm banking for adolescent and young adult cancer patients: sperm quality, patient, and parent perspectives. Pediatr Blood Cancer 2008;50:594–8.Google Scholar
Rashedi, AS, de Roo, SF, Ataman, LM, et al. Survey of fertility preservation options available to patients with cancer around the globe. JCO Glob Oncol 2018;4:116.Google Scholar
Onofre, J, Baert, Y, Faes, K, Goossens, E. Cryopreservation of testicular tissue or testicular cell suspensions: a pivotal step in fertility preservation. Hum Reprod Update 2016;22:744–61.Google Scholar
Jensen, AK, Macklon, KT, Fedder, J, Ernst, E, Humaidan, P, Andersen, CY. 86 successful births and 9 ongoing pregnancies worldwide in women transplanted with frozen-thawed ovarian tissue: focus on birth and perinatal outcome in 40 of these children. J Assist Reprod Genet 2017;34:325–36.Google Scholar
Demeestere, I, Simon, P, Dedeken, L, et al. Live birth after autograft of ovarian tissue cryopreserved during childhood. Hum Reprod 2015;30:2107–9.Google Scholar
Van der Ven, H, Liebenthron, J, Beckmann, M, et al. Ninety-five orthotopic transplantations in 74 women of ovarian tissue after cytotoxic treatment in a fertility preservation network: tissue activity, pregnancy and delivery rates. Hum Reprod 2016;31:2031–41.Google Scholar
Raffel, N, Lotz, L, Hoffmann, I, et al. Repetitive maturation of oocytes from non-stimulated xenografted ovarian tissue from a prepubertal patient indicating the independence of human ovarian tissue. Geburtshilfe Frauenheilkd 2017;77:1304–11.Google Scholar
Goossens, E, Geens, M, De Block, G, Tournaye, H. Spermatogonial survival in long-term human prepubertal xenografts. Fertil Steril 2008;90:2019–22.Google Scholar
Poels, J, Van Langendonckt, A, Many, MC, Wese, FX, Wyns, C. Vitrification preserves proliferation capacity in human spermatogonia. Hum Reprod 2013;28:578–89.Google Scholar
Schlatt, S, Honaramooz, A, Ehmcke, J, et al. Limited survival of adult human testicular tissue as ectopic xenograft. Hum Reprod 2006;21:384–9.Google Scholar
Wyns, C, Curaba, M, Martinez-Madrid, B, Van Langendonckt, A, Francois-Xavier, W, Donnez, J. Spermatogonial survival after cryopreservation and short-term orthotopic immature human cryptorchid testicular tissue grafting to immunodeficient mice. Hum Reprod 2007;22:1603–11.Google Scholar
Wyns, C, Van Langendonckt, A, Wese, FX, Donnez, J, Curaba, M. Long-term spermatogonial survival in cryopreserved and xenografted immature human testicular tissue. Hum Reprod 2008;23:2402–14.Google Scholar
Jahnukainen, K, Ehmcke, J, Nurmio, M, Schlatt, S. Autologous ectopic grafting of cryopreserved testicular tissue preserves the fertility of prepubescent monkeys that receive sterilizing cytotoxic therapy. Cancer Res 2012;72:5174–8.Google Scholar
Poels, J, Abou-Ghannam, G, Herman, S, Van Langendonckt, A, Wese, FX, Wyns, C. In search of better spermatogonial preservation by supplementation of cryopreserved human immature testicular tissue xenografts with N-acetylcysteine and testosterone. Front Surg 2014;1:47.Google Scholar
Van Saen, D, Goossens, E, Haentjens, P, Baert, Y, Tournaye, H. Exogenous administration of recombinant human FSH does not improve germ cell survival in human prepubertal xenografts. Reprod Biomed Online 2013;26:286–98.Google Scholar
Brinster, RL, Zimmermann, JW. Spermatogenesis following male germ-cell transplantation. Proc Natl Acad Sci U S A 1994;91:11298–302.Google Scholar
Radford, J. Restoration of fertility after treatment for cancer. Hormone Res 2003;59 Suppl 1:21–3.Google Scholar
Del Vento, F, Vermeulen, M, de Michele, F, et al. Tissue engineering to improve immature testicular tissue and cell transplantation outcomes: one step closer to fertility restoration for prepubertal boys exposed to gonadotoxic treatments. Int J Mol Sci 2018;19:286.Google Scholar
Shields, CWT, Reyes, CD, Lopez, GP. Microfluidic cell sorting: a review of the advances in the separation of cells from debulking to rare cell isolation. Lab Chip 2015;15:1230–49.Google Scholar
Giudice, MG, de Michele, F, Poels, J, Vermeulen, M, Wyns, C. Update on fertility restoration from prepubertal spermatogonial stem cells: how far are we from clinical practice? Stem Cell Res 2017;21:171–7.Google Scholar
Curaba, M, Poels, J, van Langendonckt, A, Donnez, J, Wyns, C. Can prepubertal human testicular tissue be cryopreserved by vitrification? Fertil Steril 2011;95:2123.e9–12.Google Scholar
Badylak, SF. The extracellular matrix as a biologic scaffold material. Biomaterials 2007;28:3587–93.Google Scholar
Letchford, K, Burt, H. A review of the formation and classification of amphiphilic block copolymer nanoparticulate structures: micelles, nanospheres, nanocapsules and polymersomes. Eur J Pharm Biopharm 2007;65:259–69.Google Scholar
Halpern, JA, Hill, R, Brannigan, RE. Guideline based approach to male fertility preservation. Urol Oncol 2020;38:31–5.Google Scholar
Moody, JA, Ahmed, K, Yap, T, Minhas, S, Shabbir, M. Fertility managment in testicular cancer: the need to establish a standardized and evidence-based patient-centric pathway. BJU Int 2019;123:160–72.Google Scholar

References

Patel, AP, Smith, RP. Vasectomy reversal: a clinical update. Asian J Androl 2016;18:365.Google Scholar
Chao, JH, Page, ST. The current state of male hormonal contraception. Pharmacol Ther 2016;163:109–17.Google Scholar
Behre, HM, Zitzmann, M, Anderson, RA, et al. Efficacy and safety of an injectable combination hormonal contraceptive for men. J Clin Endocrinol Metab 2016;101:4779–88.Google Scholar
Matzuk, MM, McKeown, MR, Filippakopoulos, P, et al. Small-molecule inhibition of BRDT for male contraception. Cell 2012;150:673–84.Google Scholar
Miyata, H, Satouh, Y, Mashiko, D, et al. Sperm calcineurin inhibition prevents mouse fertility with implications for male contraceptive. Science 2015;350:442–5.Google Scholar
Castaneda, J, Matzuk, MM. Toward a rapid and reversible male pill. Science 2015;350:385–6.Google Scholar
Ostrowski, KA, Holt, SK, Haynes, B, Davies, BJ, Fuchs, EF, Walsh, TJ. Evaluation of vasectomy trends in the United States. Urology 2018;118:76–9.Google Scholar
Trussell, J, Lalla, AM, Doan, QV, et al. Cost effectiveness of contraceptives in the United States. Contraception 2009;79:5.Google Scholar
Anderson, JE, Warner, L, Jamieson, DJ, et al. Contraceptive sterilization use among married men in the United States: results from the male sample of the National Survey of Family Growth. Contraception 2010;82:230.Google Scholar
Poddar, AK, Roy, S. Disappearance of spermatozoa from semen after vasectomy. J Popul Res 1976;3:61.Google Scholar
De Los Rios Osorio, J, Castro Alvarez, EA. Analysis of 5000 vasectomies in a family planning centre in Medellin-Colombia. Arch Esp Urol 2003;56:53.Google Scholar
Leslie, TA, Illing, RO, Cranston, DW, et al. The incidence of chronic scrotal pain after vasectomy: a prospective audit. BJU Int 2007;100:1330.Google Scholar
Wolf, JS Jr, Bennett, CJ, Dmochowski, RR, Hollenbeck, BK, Pearle, MS, Schaeffer, AJ; Urologic Surgery Antimicrobial Prophylaxis Best Practice Policy Panel. Best practice policy statement on urologic surgery antimicrobial prophylaxis. J Urol 2008;179:1379–90. Erratum in: J Urol 2008;180:2262–3.Google Scholar
Cooper, TP. Use of EMLA cream with vasectomy. Urology 2002;60:135.Google Scholar
Weiss, RS, Li, PS. No-needle jet anesthetic technique for no-scalpel vasectomy. J Urol 2005;173:1677–80.Google Scholar
Cook, LA, Pun, A, van Vliet, H, et al. Scalpel versus no-scalpel incision for vasectomy. Cochrane Database Syst Rev 2007;2:CD004112.Google Scholar
Shapiro, EI, Silber, SJ. Open-ended vasectomy, sperm granuloma, and postvasectomy orchialgia. Fertil Steril 1979;32:546.Google Scholar
Freund, MJ, Couture, M: The presence of spermatozoa in the semen of vasectomized men. J Androl 1982;3:313.Google Scholar
Alderman, PM. The lurking sperm. JAMA 1988;259:3142.Google Scholar
Page, ST, Amory, JK, Bremner, WJ. Advances in male contraception. Endocr Rev 2008;29:465–93.Google Scholar
Page, ST, Amory, JK. Male hormonal contraceptive – are we there yet? Nat Rev Endocrinol 2018;14:685–6.Google Scholar
Mathew, V, Bantwal, G. Male contraception. Indian J Endocrinol Metab 2012;16:910.Google Scholar
Kogan, P, Wald, M. Male contraception: history and development. Urol Clin North Am 2014;41:145–61.Google Scholar
Kanakis, GA, Goulis, DG. Male contraception: a clinically-oriented review. Hormones (Athens) 2015;14:598614.Google Scholar
Narayanan, R, Coss, CC, Dalton, JT. Development of selective androgen receptor modulators (SARMs). Mol Cell Endocrinol 2018;465:134–42.Google Scholar
Goericke-Pesch, S. Long-term effects of GnRH agonists on fertility and behaviour. Reprod Domest Anim 2017;52:336–47.Google Scholar
Long, JE, Lee, MS, Blithe, DL. Male contraceptive development: update on novel hormonal and nonhormonal methods. Clin Chem 2019;65:153–60.Google Scholar
Piotrowska, K, Wang, C, Swerdloff, RS, Liu, PY. Male hormonal contraception: hope and promise. Lancet Diabetes Endocrinol 2017;5:214–23.Google Scholar
Roth, MY, Page, ST, Bremner, WJ. Male hormonal contraception: looking back and moving forward. Andrology 2016;4:412.Google Scholar
Roth, MY. Male hormonal contraception. Virtual Mentor 2012;14:126–32.Google Scholar
Wang, C, Sitruk-Ware, R, Serfaty, D. It is time for new male contraceptives! Andrology 2016;4:773–5.Google Scholar
Wang, C, Festin, MPR, Swerdloff, RS. Male hormonal contraception: where are we now? Curr Obstet Gynecol Rep 2016;5:3847.Google Scholar
Khourdaji, I, Zillioux, J, Eisenfrats, K, Foley, D, Smith, R. The future of male contraception: a fertile ground. Transl Androl Urol 2018;7:S220–35.Google Scholar
Gava, G, Lantadilla, C, Martelli, V, Fattorini, A, Seracchioli, R, Meriggiola, MC. Hot issues in female and male hormonal contraception. Minerva Ginecol 2016;68:7889.Google Scholar
Chao, J, Page, ST, Anderson, RA. Male contraception. Best Pract Res Clin Obstet Gynaecol 2014;28:845–57.Google Scholar
Amory, JK, Bremner, WJ. Oral testosterone in oil plus dutasteride in men: a pharmacokinetic study. J Clin Endocrinol Metab 2005;90:2610–17.Google Scholar
Dorman, E, Bishai, D. Demand for male contraception. Expert Rev Pharmacoeconomics Outcomes Res 2012;12:605–13.Google Scholar
Mashiko, D, Fujihara, Y, Satouh, Y, Miyata, H, Isotani, A, Ikawa, M. Generation of mutant mice by pronuclear injection of circular plasmid expressing Cas9 and single guided RNA. Sci Rep 2013;3:3355.Google Scholar
Fujihara, Y, Ikawa, M. CRISPR/Cas9-based genome editing in mice by single plasmid injection. Methods Enzymol 2014;546:319–36.Google Scholar
Mashiko, D, Young, SAM, Muto, M, et al. Feasibility for a large scale mouse mutagenesis by injecting CRISPR/Cas plasmid into zygotes. Dev Growth Differ 2014;56:122–9.Google Scholar
Miyata, H, Castaneda, JM, Fujihara, Y, et al. Genome engineering uncovers 54 evolutionarily conserved and testis-enriched genes that are not required for male fertility in mice. Proc Natl Acad Sci U S A 2016;113:7704–10.Google Scholar
Noda, T, Sakurai, N, Nozawa, K, et al. Nine genes abundantly expressed in the epididymis are not essential for male fecundity in mice. Andrology 2019;7:644–53.Google Scholar
Van der Spoel, AC, Jeyakumar, M, Butters, TD, et al. Reversible infertility in male mice after oral administration of alkylated imino sugars: a nonhormonal approach to male contraception. Proc Natl Acad Sci U S A 2002;99:17173–8.Google Scholar
Walden, CM, Butters, TD, Dwek, RA, Platt, FM, van der Spoel, AC. Long-term non-hormonal male contraception in mice using N-butyldeoxynojirimycin. Hum Reprod 2006;21:1309–15.Google Scholar
Amory, JK, Muller, CH, Page, ST, et al. Miglustat has no apparent effect on spermatogenesis in normal men. Hum Reprod 2007;22:702–7.Google Scholar
Robaire, B. Advancing towards a male contraceptive: a novel approach from an unexpected direction. Trends Pharmacol Sci 2003;24:326–8.Google Scholar
Murdoch, FE, Goldberg, E. Male contraception: another holy grail. Bioorganic Med Chem Lett 2014;24:419–24.Google Scholar
Shang, E, Nickerson, HD, Wen, D, Wang, X, Wolgemuth, DJ. The first bromodomain of Brdt, a testis-specific member of the BET sub-family of double-bromodomain-containing proteins, is essential for male germ cell differentiation. Development 2007;134:3507–15.Google Scholar
Meistrich, ML, Hughes, TH, Bruce, WR. Alteration of epididymal sperm transport and maturation in mice by oestrogen and testosterone. Nature 1975;258:145–7.Google Scholar
Gregory, M, Cyr, DG. The blood–epididymis barrier and inflammation. Spermatogenesis 2014;4:e979619.Google Scholar
Breton, S, Nair, AV, Battistone, MA. Epithelial dynamics in the epididymis: role in the maturation, protection, and storage of spermatozoa. Andrology 2019;7:631–43.Google Scholar
Guha, SK, Singh, G, Anand, S, Ansari, S, Kumar, S, Koul, V. Phase I clinical trial of an injectable contraceptive for the male. Contraception 1993;48:367–75.Google Scholar
Ansari, AS, Badar, A, Balasubramanian, K, Lohiya, NK. Contraception with RISUG® and functional reversal through DMSO and NaHCO3 in male rabbits. Asian J Androl 2017;19:389–95.Google Scholar
Zaneveld, LJ, Burns, JW, Beyler, S, Depel, W, Shapiro, S. Development of a potentially reversible vas deferens occlusion device and evaluation in primates. Fertil Steril 1988;49:527–33.Google Scholar
Zhao, SC. Vas deferens occlusion by percutaneous injection of polyurethane elastomer plugs: clinical experience and reversibility. Contraception 1990;41:453–9.Google Scholar
Homonnai, ZT, Shilon, M, Paz, GF. Phenoxybenzamine – an effective male contraceptive pill. Contraception 1984;29:479–91.Google Scholar
Kedia, KR, Persky, L. Effect of phenoxybenzamine (dibenzyline) on sexual function in man. Urology 1981;18:620–2.Google Scholar
Amobi, NIB, Smith, ICH. Differential inhibition in the human vas deferens by phenoxybenzamine: a possible mechanism for its contraceptive action. J Reprod Fertil 1995;103:215–21.Google Scholar
Zdrojewicz, Z, Konieczny, R, Papier, P, Szten, F. Brdt bromodomains inhibitors and other modern means of male contraception. Adv Clin Exp Med 2015;24:705–14.Google Scholar
Ratnasooriya, WD, Wadsworth, RM. Impairment of fertility of male rats with prazosin. Contraception 1990;41:441–7.Google Scholar
Kjærgaard, N, Kjærgaard, B, Lauritsen, JG. Prazosin, an adrenergic blocking agent inadequate as male contraceptive pill. Contraception 1988;37:621–9.Google Scholar
Hellstrom, WJG, Wang, R, Peterson, CA, Varady, JC, Gesundheit, N, Sikka, SC. Effects of alprostadil and prazosin on motility, viability and membrane integrity of human sperm. J Urol 1998;159:1559–62.Google Scholar
Rayburn, ER, Gao, L, Ding, J, Ding, H, Shao, J, Li, H. FDA-approved drugs that are spermatotoxic in animals and the utility of animal testing for human risk prediction. J Assist Reprod Genet 2018;35:191212.Google Scholar
Hellstrom, WJG, Sikka, SC. Effects of alfuzosin and tamsulosin on sperm parameters in healthy men: results of a short-term, randomized, double-blind, placebo-controlled, crossover study. J Androl 2009;30:469–74.Google Scholar
Wang, J, Zhao, Y, Jiang, SB, et al. Assessment of tamsulosin as a potential male contraceptive in healthy volunteers. Urology 2012;80:614–17.Google Scholar
Ventura, S, Pennefather, JN. Sympathetic co-transmission to the cauda epididymis of the rat: characterization of postjunctional adrenoceptors and purinoceptors. Br J Pharmacol 1991;102:540–4.Google Scholar
Sewak, R, Teng, B, Learman, LA, Hennekens, CH. Male contraception: prospects for sound and ultrasound. Med Hypotheses 2017;107:14.Google Scholar
Amouroux, M, Mieusset, R, Desbriere, R, et al. Are men ready to use thermal male contraception? Acceptability in two French populations: new fathers and new providers. PLoS ONE 2018;13:123.Google Scholar
Kandeel, FR, Swerdloff, RS. Role of temperature in regulation of spermatogenesis and the use of heating as a method for contraception. Fertil Steril 1988;49:123.Google Scholar
Robinson, D, Rock, J, Menkin, MF. Control of human spermatogenesis by induced changes of intrascrotal temperature. JAMA 1968;204:290–7.Google Scholar
Dismore, L, Van Wersch, A, Swainston, K. Social constructions of the male contraception pill: when are we going to break the vicious circle? J Health Psychol 2016;21:788–97.Google Scholar
Ross, J, Hardee, K. Use of male methods of contraception worldwide. J Biosoc Sci 2017;49:648–63.Google Scholar
O’Rand, MG, Silva, EJR, Hamil, KG. Non-hormonal male contraception: a review and development of an Eppin based contraceptive. Pharmacol Ther 2016;157:105–11.Google Scholar
O’Rand, MG, Widgren, EE, Sivashanmugam, P, et al. Reversible immunocontraception in male monkeys immunized with Eppin. Science 2004;306:1189–90.Google Scholar
O’Rand, MG, Widgren, EE, Beyler, S, Richardson, RT. Inhibition of human sperm motility by contraceptive anti-eppin antibodies from infertile male monkeys: effect on cyclic adenosine monophosphate. Biol Reprod 2009;80:279–85.Google Scholar
Mitra, A, Richardson, RT, O’Rand, MG. Analysis of recombinant human semenogelin as an inhibitor of human sperm motility. Biol Reprod 2010;82:489–96.Google Scholar
Krishnamurthy, H, Kumar, KM, Joshi, CV, et al. Alterations in sperm characteristics of follicle-stimulating hormone (FSH)-immunized men are similar to those of FSH-deprived infertile male bonnet monkeys. J Androl 2000;21:316–27.Google Scholar
Frayne, J, Hall, L. The potential use of sperm antigens as targets for immunocontraception; past, present and future. J Reprod Immunol 1999;43:133.Google Scholar
Primakoff, P, Woolman-Gamer, L, Tung, KSK, Myles, DG. Reversible contraceptive effect of PH-20 immunization in male guinea pigs. Biol Reprod 1997;56:1142–6.Google Scholar
Goldberg, E, VandeBerg, JL, Mahony, MC, Doncel, GF. Immune response of male baboons to testis-specific LDH-C. Contraception 2001;64:93–8.Google Scholar
Li, H, Ding, X, Guan, H, Xiong, C. Inhibition of human sperm function and mouse fertilization in vitro by an antibody against cation channel of sperm 1: the contraceptive potential of its transmembrane domains and pore region. Fertil Steril 2009;92:1141–6.Google Scholar
Khobarekar, BG, Vernekar, V, Raghavan, V, Kamada, M, Maegawa, M, Bandivdekar, AH. Evaluation of the potential of synthetic peptides of 80 kDa human sperm antigen (80 kDaHSA) for the development of contraceptive vaccine for male. Vaccine 2008;26:3711–18.Google Scholar
Bandivdekar, AH. Development of antifertility vaccine using sperm specific proteins. Indian J Med Res 2014;140:73–7.Google Scholar
Avella, MA, Baibakov, BA, Jimenez-Movilla, M, Sadusky, AB, Dean, J. ZP2 peptide beads select human sperm in vitro, decoy mouse sperm in vivo, and provide reversible contraception. Sci Transl Med 2016;8:112.Google Scholar
Kaur, K, Prabha, V. Immunocontraceptives: new approaches to fertility control. Biomed Res Int 2014;2014:868196.Google Scholar
Gupta, SK, Shrestha, A, Minhas, V. Milestones in contraceptive vaccines development and hurdles in their application. Hum Vaccines Immunother 2014;10:911–25.Google Scholar
Lye, RJ, Sipilä, P, Vernet, P, Wagenfeld, A. Male contraception – a topic with many facets. Mol Cell Endocrinol 2004;216:7582.Google Scholar
Coutinho, EM. Gossypol: a contraceptive for men. Contraception 2002;65:259–63.Google Scholar
Meng, GD, Zhu, JC, Chen, ZW, et al. Follow-up of men in the recovery period immediately after the cessation of gossypol treatment. Contraception 1988;37:119–28.Google Scholar
Mishra, RK, Singh, S, Singh, SK. Natural products in regulation of male fertility. Indian J Med Res 2018;148:S107–14.Google Scholar
Mruk, DD. New perspectives in non-hormonal male contraception. Trends Endocrinol Metab 2008;19:5764.Google Scholar
Prajogo, BE, Guliet, D, Queiroz, FE, et al. Isolation of male antifertility compound in N-butanol fraction of Justica gendarussa Burm. f. leaves. Folia Medica Indones 2009;45:2831.Google Scholar
Qian, SZ. Tripterygium wilfordii, a Chinese herb effective in male fertility regulation. Contraception 1987;36:335–45.Google Scholar
Matlin, SA, Belenguer, A, Stacey, VE, et al. Male antifertility compounds from Tripterygium wilfordii Hook f. Contraception 1993;47:387400.Google Scholar
Mishra, RK, Singh, SK. Effect of aqueous leaf extract of Azadirachta indica on the reproductive organs in male mice. Indian J Exp Biol 2005;43:1093–103.Google Scholar
Mishra, RK, Singh, SK. Reversible antifertility effect of aqueous rhizome extract of Curcuma longa L. in male laboratory mice. Contraception 2009;79:479–87.Google Scholar
Cheng, Y-H, Xia, W, Wong, E, et al. Adjudin – a male contraceptive with other biological activities. Recent Pat Endocr Metab Immune Drug Discov 2015;9:6373.Google Scholar
Mruk, DD, Wong, CH, Silvestrini, B, Cheng, CY. A male contraceptive targeting germ cell adhesion. Nat Med 2006;12:1323–8.Google Scholar
Tash, JS, Attardi, B, Hild, SA, Chakrasali, R, Jakkaraj, SR, Georg, GI. A novel potent indazole carboxylic acid derivative blocks spermatogenesis and ss contraceptive in rats after a single oral dose. Biol Reprod 2008;78:1127–38.Google Scholar
Tash, JS, Chakrasali, R, Jakkaraj, SR, et al. Gamendazole, an orally active indazole carboxylic acid male contraceptive agent, targets HSP90AB1 (HSP90BETA) and EEF1A1 (eEF1A), and stimulates Il1a rranscription in rat Sertoli cells. Biol Reprod 2008;78:1139–52.Google Scholar
Schulze, GE, Clay, RJ, Mezza, LE, Bregman, CL, Buroker, RA, Frantz, JD. BMS-189453, a novel retinoid receptor antagonist is a potent testicular toxin. Toxicol Sci 2001;59:297308.Google Scholar
Chung, SSW, Wang, X, Roberts, SS, Griffey, SM, Reczek, PR, Wolgemuth, DJ. Oral administration of a retinoic acid receptor antagonist reversibly inhibits spermatogenesis in mice. Endocrinology 2011;152:2492–502.Google Scholar
Amory, JK, Muller, CH, Shimshoni, JA, et al. Suppression of spermatogenesis by bisdichloroacetyldiamines is mediated by inhibition of testicular retinoic acid biosynthesis. J Androl 2011;32:111–19.Google Scholar
Nya-Ngatchou, J-J, Amory, JK. New approaches to male non-hormonal contraception. Contraception 2013;87:296–9.Google Scholar
Jimenez, T, Sánchez, G, Wertheimer, E, Blanco, G. Activity of the Na,K-ATPase α4 isoform is important for membrane potential, intracellular Ca2+, and pH to maintain motility in rat spermatozoa. Reproduction 2010;139:835–45.Google Scholar
Sanchez, G, Nguyen, ANT, Timmerberg, B, Tash, JS, Blanco, G. The Na,K-ATPase α4 isoform from humans has distinct enzymatic properties and is important for sperm motility. Mol Hum Reprod 2006;12:565–76.Google Scholar
Yang, YH, Wan, Y, Lou, H, Xue, T, Su, P. Relationship between ouabain and asthenozoospermia. J Huazhong Univ Sci Technol - Med Sci 2014;34:8790.Google Scholar
Karachitos, A, Kmita, H. Voltage-dependent anion channel isoform 3 as a potential male contraceptive drug target. Future Med Chem 2019;11:857–67.Google Scholar
Ahmad, K, Foote, RH. Postthaw survival and fertility of frozen bull spermatozoa treated with antibiotics and detergent. J Dairy Sci 1986;69:535–41.Google Scholar
Kwon, WS, Park, YJ, Mohamed, ESA, Pang, MG. Voltage-dependent anion channels are a key factor of male fertility. Fertil Steril 2013;99:354–61.Google Scholar
Malallah, YA, Zissis, NP. Effect of minocycline on the sperm count and activity in infertile men with high plus cell count in their seminal fluid. J Chemother 1992;4:286–9.Google Scholar
Carlson, AE, Burnett, LA, del Camino, D, et al. Pharmacological targeting of native CatSper channels reveals a required role in maintenance of sperm hyperactivation. PLoS One 2009;4:e6844.Google Scholar
Srivastav, A, Changkija, B, Sharan, K, Nagar, GK, Bansode, FW. Influence of antifertility agents Dutasteride and Nifedipine on CatSper gene level in epididymis during sperm maturation in BALB/c mice. Reproduction 2018;155:347–59.Google Scholar
Enders, G. Clinical approaches to male infertility with a case report of possible nifedipine-induced sperm dysfunction. J Am Board Fam Pract 1997;10:131–6.Google Scholar
Benoff, S, Cooper, GW, Hurley, I, et al. The effect of calcium ion channel blockers on sperm fertilization potential. Fertil Steril 1994;62:606–17.Google Scholar
Roehrborn, CG, Boyle, P, Nickel, JC, Hoefner, K, Andriole, G; ARIA3001 ARIA3002 and ARIA3003 Study Investigators. Efficacy and safety of a dual inhibitor of 5-alpha-reductase types 1 and 2 (dutasteride) in men with benign prostatic hyperplasia. Urology 2002;60:434–41.Google Scholar
Shukla, KK, Kwon, WS, Rahman, MS, Park, YJ, You, YA, Pang, MG. Nutlin-3a decreases male fertility via UQCRC2. PLoS One 2013;8:18.Google Scholar
Jones, AR. The antifertility actions of alpha-chlorohydrin in the male. Life Sci 1978;23:1625–45.Google Scholar
Hild, SA, Attardi, BJ, Reel, JR. The ability of a gonadotropin-releasing hormone antagonist, acyline, to prevent irreversible infertility induced by the indenopyridine, CDB-4022, in adult male rats: the role of testosterone. Biol Reprod 2004;71:348–58.Google Scholar
Hild, SA, Marshall, GR, Attardi, BJ, et al. Development of l-CDB-4022 as a nonsteroidal male oral contraceptive: induction and recovery from severe oligospermia in the adult male cynomolgus monkey (Macaca fascicularis). Endocrinology 2007;148:1784–96.Google Scholar
Koduri, S, Hild, SA, Pessaint, L, Reel, JR, Attardi, BJ. Mechanism of action of l-CDB-4022, a potential nonhormonal male contraceptive, in the seminiferous epithelium of the rat testis. Endocrinology 2008;149:1850–60.Google Scholar
Mok, K-W, Mruk, DD, Lie, PPY, Lui, W-Y, Cheng, CY. Adjudin, a potential male contraceptive, exerts its effects locally in the seminiferous epithelium of mammalian testes. Reproduction 2011;141:571–80.Google Scholar
Juneja, SC. Development of infertility at young adult age in a mouse model of human Sandhoff disease. Reprod Fertil Dev 2002;14:407–12.Google Scholar
Blithe, DL. Pipeline for contraceptive development. Fertil Steril 2016;106:1295–302.Google Scholar
Amory, JK. Male contraception. Fertil Steril 2016;106:1303–9.Google Scholar
Chen, S-R, Batool, A, Wang, Y-Q, et al. The control of male fertility by spermatid-specific factors: searching for contraceptive targets from spermatozoon’s head to tail. Cell Death Dis 2016;7:e2472.Google Scholar
Roth, MY, Amory, JK. Beyond the condom: frontiers in male contraception. Semin Reprod Med 2016;34:183–90.Google Scholar
Archambeault, DR, Matzuk, MM. Disrupting the male germ line to find infertility and contraception targets. Ann Endocrinol (Paris) 2014;75:101–8.Google Scholar
Payne, C, Goldberg, E. Male contraception: past, present and future. Curr Mol Pharmacol 2014;7:175–81.Google Scholar
Sipilä, P, Jalkanen, J, Huhtaniemi, IT, Poutanen, M. Novel epididymal proteins as targets for the development of post-testicular male contraception. Reproduction 2009;137:379–89.Google Scholar
Naz, RK, Rowan, S. Update on male contraception. Curr Opin Obstet Gynecol 2009;21:265–9.Google Scholar
Kopf, GS. Approaches to the identification of new nonhormonal targets for male contraception. Contraception 2008;78:1822.Google Scholar
Johnston, DS, Jelinsky, SA, Zhi, Y, Finger, JN, Kopf, GS, Wright, WW. Identification of testis-specific male contraceptive targets: insights from transcriptional profiling of the cycle of the rat seminiferous epithelium and purified testicular cells. Ann N Y Acad Sci 2007;1120:3646.Google Scholar
Schultz, N, Hamra, FK, Garbers, DL. A multitude of genes expressed solely in meiotic or postmeiotic spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci U S A 2003;100:12201–6.Google Scholar
Weber, RFA, Dohle, GR. Male contraception: mechanical, hormonal and non-hormonal methods. World J Urol 2003;21:338–40.Google Scholar
Amann, RP. The cycle of the seminiferous epithelium in humans: a need to revisit? J Androl 2008;29:469–87.Google Scholar
Greenbaum, MP, Iwamori, T, Buchold, GM, Matzuk, MM. Germ cell intercellular bridges. Cold Spring Harb Perspect Biol 2011;3:118.Google Scholar
Clermont, Y. Kinetics of spermatogenesis in mammals: seminiferous epithelium cycle and spermatogonial renewal. Physiol Rev 1972;52:198236.Google Scholar
de Rooij, DG, Russell, LD. All you wanted to know about spermatogonia but were afraid to ask. J Androl 2000;21:776–98.Google Scholar
Roosen-Runge, E. Comparative aspects of spermatogenesis. Biol Reprod 1969;39:2439.Google Scholar
O’Donnell, L. Mechanisms of spermiogenesis and spermiation and how they are disturbed. Spermatogenesis 2014;4:e979623.Google Scholar
Bittman, EL. Timing in the testis. J Biol Rhythms 2016;31:1236.Google Scholar
Johnson, L, Varner, DD. Effect of daily spermatozoan production but not age on transit time of spermatozoa through the human epididymis1 Biol Reprod 1988;39:812–17.Google Scholar
Moore, HDM. Contribution of epididymal factors to sperm maturation and storage. Andrologia 2009;30:233–9.Google Scholar
Jones, J, Mosher, W, Daniels, K. Current contraceptive use in the united states, 2006–2010, and changes in patterns of use since 1995. Sex Stat Sel Reports from Natl Cent Heal Stat 2013;1980:127–73.Google Scholar
Plana, O. Male contraception: research, new methods, and implications for marginalized populations. Am J Mens Health 2017;11:1182–9.Google Scholar
Serfaty, D. A plea for male contraception. Eur J Contracept Reprod Heal Care 2015;20:75–6.Google Scholar

Further Reading

Fayomi, AP, et al. Autologous grafting of cryopreserved prepubertal rhesus testis produces sperm and offspring. Science 2019;363:1314–19.Google Scholar
Ibtisham, F, et al. Progress and future prospect of in vitro spermatogenesis. Oncotarget 2017;8:66709–27Google Scholar
Sato, T, et al. In vitro spermatogenesis in explanted adult mouse testis tissues. PLoS One 2015;10:e0130171.Google Scholar
Wang, R, et al. Artificial intelligence in reproductive medicine. Reproduction 2019;158:R139–54.Google Scholar

References

Rosenwaks, Z, Pereira, N. The pioneering of intracytoplasmic sperm injection: historical perspectives. Reproduction 2017;154:F71–7.Google Scholar
Punab, M, et al. Causes of male infertility: a 9-year prospective monocentre study on 1737 patients with reduced total sperm counts. Hum Reprod 2017;32:1831.Google Scholar
Schwarze, JE, et al. Is there a reason to perform ICSI in the absence of male factor? Lessons from the Latin American Registry of ART. Hum Reprod Open 2017;2017:hox013.Google Scholar
Schlegel, PN. Testicular sperm extraction: microdissection improves sperm yield with minimal tissue excision. Hum Reprod 1999;14:131–5.Google Scholar
Bernie, AM, et al. Comparison of microdissection testicular sperm extraction, convention testicular sperm extraction, and testicular sperm aspiration for nonobstructive azoospermia: a systematic review and meta analysis. Fertil Steril 2017;104:1099103.e1–3.Google Scholar
Najari, BB. Ultrasound evaluation of seminiferous tubules: a promising prognostic tool for men with nonobstructive azoospermia undergoing microsurgical testicular sperm extraction. Fertil Steril 2020;113:73–4.Google Scholar
Yu, Y, et al. Intraoperative assessment of tubules in predicting microdissection testicular sperm extraction outcome in men with Sertoli cell-only syndrome. J Int Med Res 2019;47:722–9.Google Scholar
Caroppo, E, et al. The seminiferous tubule caliber pattern as evaluated at high magnification during microdissection testicular sperm extraction predicts sperm retrieval in patients with non-obstructive azoospermia. Andrology 2019;7:814.Google Scholar
Ramasamy, R, et al. Identification of spermatogenesis with multiphoton microscopy: an evaluation in a rodent model. J Urol 2011;186:2487–92.Google Scholar
Da Costa, R, et al. Spectral features of nuclear DNA in human sperm assessed by Raman microspectroscopy: effects of UV-irradiation and hydration. PLoS One 2018;2018:e0207786.Google Scholar
Liu, F, et al. Real-time Raman microspectroscopy scanning of the single live sperm bound to human zone pellucida. Fertil Steril 2013;99:6849.e4.Google Scholar
Sanchez, V, et al. Oxidative DNA damage in human sperm can be detected by Raman microspectroscopy. Fertil Steril 2012;98:11249.e1–3.Google Scholar
Kubasek, WL, et al. Raman spectra of the model B-DNA oligomer d(CGCGAATTCGCG)2 and of the DNA in living salmon sperm show that both have very similar B-type conformations. Biochemistry 1986;25:7440–5.Google Scholar
Liu, Y, et al. Raman spectroscopy as an ex vivo noninvasive approach to distinguish complete and incomplete spermatogenesis within human seminiferous tubules. Fertil Steril 2014;102:54–60e2.Google Scholar
Zhang, S, et al. Assessment of the use of contrast enhanced ultrasound in guiding microdissection testicular sperm extraction in nonobstructive azoospermia. BMC Urol 2018;18:48.Google Scholar
Nariyoshi, S, et al. US-determined size of seminiferous tubules predict sperm retrieval by microdissection testicular sperm extraction in men with non-obstructive azoospermia. Fertil Steril 2020;113:97104.e2.Google Scholar
Ibtisham, F, et al. Progress and future prospect of in vitro spermatogenesis. Oncotarget 2017;8:66709–27.Google Scholar
Mulder, CL, et al. Spermatogonial stem cell autotransplantation and germline genomic editing: a future cure for spermatogenic failure and prevention of transmission of genomic diseases. Hum Reprod Update 2017;22:561–73.Google Scholar
Sato, T, et al. In vitro spermatogenesis in explanted adult mouse testis tissues. PLoS One 2015;10:e0130171.Google Scholar
Smith, MA, et al. Declining childhood and adolescent cancer mortality. Cancer 2014;120:2497–506.Google Scholar
Sadri-Ardekani, H, et al. Propagation of human spermatogonial stem cells in vitro. JAMA 2009;302:2127–34.Google Scholar
Klosky, JL, et al. Sperm cryopreservation practices among adolescent cancer patients at risk for infertility. Pediatr Hematol Oncol 2009;26:252–60.Google Scholar
Picton, HM, et al. A European perspective on testicular tissue cryopreservation for fertility preservation in prepubertal and adolescent boys. Hum Reprod 2015;30:2463–75.Google Scholar
Sadri-Ardekani, H, et al. In vitro propagation of human prepubertal spermatogonial stem cells. JAMA 2011;305:2416–18.Google Scholar
Fayomi, AP, et al. Autologous grafting of cryopreserved prepubertal rhesus testis produces sperm and offspring. Science 2019;363:1314–19.Google Scholar
Abbasi, F, et al. Revolutionizing male fertility factor research in mice by using the genome editing tool CRISPR/Cas9. Reprod Med Biol 2018;17:310.Google Scholar
Lu, Y, et al. CRISPR/Cas9-mediated genome editing reveals 30 testis-enriched genes dispensable for male fertility in micedagger. Biol Reprod 2019;101:501–11.Google Scholar
Li, X, et al. Restore natural fertility of Kit(w)/Kit(wv) mouse with nonobstructive azoospermia through gene editing on SSCs mediated by CRISPR-Cas9. Stem Cell Res Ther 2019;10:271.Google Scholar
Chu, KY, et al. Artificial intelligence in reproductive urology. Curr Urol Rep 2019;20:52.Google Scholar
Wang, R, et al. Artificial intelligence in reproductive medicine. Reproduction 2019;158:R139–54.Google Scholar
Manna, C, et al. Artificial intelligence techniques for embryo and oocyte classification. Reprod Biomed Online 2013;26:42–9.Google Scholar
Munne, S, et al. Selection of embryos by morphology is less effective than by a combination of aneuploidy testing and morphology observations. Fertil Steril 2009;91:943–5.Google Scholar
McCallam, C, et al. Deep learning-based selection of human sperm with high DNA integrity. Commun Biol 2019;2:250.Google Scholar
Agarwal, A, Sharma, RK. Automation is the key to standardized semen analysis using the automated SQA-V sperm quality analyzer. Fertil Steril 2007;87:156–62.Google Scholar
Khosravi, P, et al. Deep learning enables robust assessment and selection of human blastocysts after in vitro fertilization. NPJ Digit Med 2019;2:21.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×