Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-5nwft Total loading time: 0 Render date: 2024-05-20T10:34:52.460Z Has data issue: false hasContentIssue false

Chapter 26 - Male Contraception

from 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

Summary

Contraception, or the purposeful temporary inhibition of one’s own fertility, is a vital additional level of control over family planning. Though many potent and reversible contraceptives are on the market, most of these contraceptives are for women – leaving a disproportionate burden of the responsibility for contraception and family planning on women. Reversible forms of contraception available for men are limited to the use of condoms and withdrawal. The only other form of male contraception that approaches the same efficacy as some available options for women is vasectomy, which is not always reversible and often has lasting effects on fertility [1]. To meet the unmet need of potent and reversible male contraception, research and clinical trials for hormonal male contraceptives have been ongoing for nearly the past 50 years; yet roadblocks, such as unfavorable injections and undesirable side effects, have slowed the emergence of this option on the market [2, 3]. New research is now under way for optimized hormonal contraceptives and a myriad of nonhormonal contraceptive options. Early preclinical tests of nonhormonal contraceptive compounds in animal models and in small clinical studies have successfully demonstrated that pharmacological compounds targeting specific proteins can result in complete and reversible contraceptive efficacy. However, since many compounds used in early studies were not specifically tailored for their use as contraceptives in some cases, or due to the potential for off-target effects identified from preclinical testing, more work is needed to improve these compounds for selectivity and to identify novel protein targets [4–6]. Nonetheless, the plethora of research and clinical advances made in this field in recent years has done much to make a male contraceptive pill closer to reality. In this chapter, we discuss the history and recent advances in novel hormonal and nonhormonal contraceptive development and provide a brief overview of how contraceptive compounds modulate the dynamics of sperm production and transport.

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

Patel, AP, Smith, RP. Vasectomy reversal: a clinical update. Asian J Androl 2016;18:365.Google ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Matzuk, MM, McKeown, MR, Filippakopoulos, P, et al. Small-molecule inhibition of BRDT for male contraception. Cell 2012;150:673–84.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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 ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Roth, MY, Page, ST, Bremner, WJ. Male hormonal contraception: looking back and moving forward. Andrology 2016;4:412.CrossRefGoogle ScholarPubMed
Roth, MY. Male hormonal contraception. Virtual Mentor 2012;14:126–32.Google ScholarPubMed
Wang, C, Sitruk-Ware, R, Serfaty, D. It is time for new male contraceptives! Andrology 2016;4:773–5.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Amory, JK, Bremner, WJ. Oral testosterone in oil plus dutasteride in men: a pharmacokinetic study. J Clin Endocrinol Metab 2005;90:2610–17.CrossRefGoogle 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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
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 ScholarPubMed
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

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
×