You are on page 1of 29

0163-769X/08/$20.00/0 Printed in U.S.A.

Endocrine Reviews 29(4):465 493 Copyright 2008 by The Endocrine Society doi: 10.1210/er.2007-0041

Advances in Male Contraception


Stephanie T. Page, John K. Amory, and William J. Bremner
Center for Research in Reproduction and Contraception, Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98195
Despite significant advances in contraceptive options for women over the last 50 yr, world population continues to grow rapidly. Scientists and activists alike point to the devastating environmental impacts that population pressures have caused, including global warming from the developed world and hunger and disease in less developed areas. Moreover, almost half of all pregnancies are still unwanted or unplanned. Clearly, there is a need for expanded, reversible, contraceptive options. Multicultural surveys demonstrate the willingness of men to participate in contraception and their female partners to trust them to do so. Notwithstanding their paucity of options, male methods including vasectomy and condoms account for almost one third of contraceptive use in the United States and other countries. Recent international clinical research efforts have demonstrated high efficacy rates (90 95%) for hormonally based male contraceptives. Current barriers to expanded use include limited delivery methods and perceived regulatory obstacles, which stymie introduction to the marketplace. However, advances in oral and injectable androgen delivery are cause for optimism that these hurdles may be overcome. Nonhormonal methods, such as compounds that target sperm motility, are attractive in their theoretical promise of specificity for the reproductive tract. Gene and protein array technologies continue to identify potential targets for this approach. Such nonhormonal agents will likely reach clinical trials in the near future. Great strides have been made in understanding male reproductive physiology; the combined efforts of scientists, clinicians, industry and governmental funding agencies could make an effective, reversible, male contraceptive an option for family planning over the next decade. (Endocrine Reviews 29: 465 493, 2008)

I. Introduction II. Acceptability of Male Contraception III. Currently Available Male Contraceptive Methods A. Condoms B. Vasectomy IV. Regulation of Testicular Function and Spermatogenesis A. The hypothalamic-pituitary axis: control of gonadotropin secretion B. Testosterone production and androgen action C. Spermatogenesis D. The epididymis V. Male Hormonal Contraception Methods A. Hormonal contraceptive efficacy B. Testosterone alone C. Testosterone-progestin combinations D. Adding GnRH analogs to hormonal contraceptive regimens E. Oral male hormonal contraceptive development F. Reversibility of male hormonal contraception G. Side effects of male hormonal contraceptives H. Selective androgen receptor modulators for male contraception I. Why doesnt hormonal contraception work for all men? VI. Nonhormonal Methods A. Testicular targets B. Epdidymal targets C. Targeting sperm motility or sperm-egg fusion D. Immunological approaches to contraception VII. Conclusions

I. Introduction

ESPITE CURRENTLY AVAILABLE contraceptives, the worlds population exceeds 6.5 billion and is increasing by 75 million yearly (1), and overpopulation continues to be a significant contributor to environmental degradation and human suffering worldwide. Much of our current global population growth is unintended. It is estimated that half of all conceptions are unplanned and half of the resulting pregnancies are undesired (2). In the United States, approximately half of unintended pregnancies are attributable to a failure to use contraception, and the other half to difficulties with contraceptive use or method failure (3). Moreover, in developing countries contraception use is further limited by restricted access to many available methods, both economically and culturally. The contraceptive shortfall results in over 20% of pregnancies ending in abortion (3, 4). In addition, undesired pregnancies result in unwanted children who suffer disproportionately from poverty and neglect (5, 6). Therefore, there is a great need for better access and education
First Published Online April 24, 2008 Abbreviations: DHT, Dihydrotestosterone; DMPA, depomedroxyprogesterone; GAPD, glyceraldehydes 3-phosphate dehydrogenase; GPR, G protein receptor; hCG, human chorionic gonadotropin; HDL, high-density lipoprotein; INSL3, insulin-like factor 3; MENT, 7-methyl-19-nortestosterone; PSA, prostate-specific antigen; sAC, soluble adenylate cyclase; SARM, selective androgen receptor modulator; SFEC, sperm flagellar energy carrier; sNHE, soluble sodiumhydrogen exchanger; TD, testosterone decanoate; TE, testosterone enanthate; TU, testosterone undecanoate. Endocrine Reviews is published by The Endocrine Society (http:// www.endo-society.org), the foremost professional society serving the endocrine community.

465

466

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

regarding existing contraceptives and more contraceptive options. Women have a wide range of contraceptive choices. Research and family planning organizations have traditionally focused upon female methods of contraception because women bear a disproportionate portion of the health and economic consequences of childbearing and rearing. Consequently, women have many contraceptive choices, ranging from daily oral medications to intrauterine devices implanted every 5 yr to sterilization. In the current era, there are only two forms of male contraception available, condoms and vasectomy, with only the former being reliably reversible. In fact, no new, reversible, male-based methods have become commercially available since the condom was introduced more than 300 yr ago. However, despite few available options, male methods account for approximately 14% of contraception worldwide, with prevalence significantly higher in the more developed world where male-directed methods account for more than 30% of contraception. Funding for the development of male contraceptives has faltered since this topic was last reviewed (7). Although support from some public agencies has continued, albeit at reduced levels, the pharmaceutical industry has largely abandoned this potential market. Three major international pharmaceutical companies have cancelled their programs in male contraception over the last 5 yr, citing corporate changes in direction as the motivating force. However, it is difficult to imagine that other forces have not influenced these decisions, including the perception that the safety standards for regulatory approval of such products would require studies of such numbers and length as to be financially infeasible or lacking in incentive for profit. In addition, despite the environmental and health consequences of the ongoing but largely forgotten population crisis, large nonprofits such as the Bill and Melinda Gates Foundation have focused their resources on other global health concerns. Without the support of these organizations in the long run, progress in the development of effective and available male contraceptives is difficult to imagine. In this article, we will review the mechanism of action and recent advances in male contraception, with particular emphasis on newer regimens that may be nearing introduction to clinical practice. First, however, we will discuss the important question of acceptability, because only a contraceptive that men, and their female partners, will reliably use represents a true advance in the field. We will discuss the physiological challenges of male contraceptive development, hormonal methods of contraception, which likely offer the prospect of reaching the market first, and nonhormonal methods, including immune-based products.

ingness to use a male contraceptive if one were available (9, 10). Additional multinational and multiethnic studies of potential hormonally based male contraceptives demonstrate that most men (11), and their female partners (12), would be willing to use such a method of contraception. In fact, 98% of women in stable relationships would be willing to rely on their male partner to use male contraception (12). Acceptability of male-based contraceptive methods is, however, influenced by economic, cultural, religious, and relationship status. For example, higher income and education status are associated with a great willingness to use male contraception (10). Other studies demonstrate marked cultural differences. For example, men in predominantly Muslim Indonesia were less willing to use a male contraceptive and cited religious beliefs as a major objection (10). Male Australian immigrants were less receptive to the notion of male contraception than those born in Australia (13). Among women with current partners in Hong Kong, only 14% felt that they would rely a male-based method in the future if it were available, whereas in Shanghai 71% of these female respondents would use a male-based method were it available (12). Interestingly, more than 70% of women in both locales thought male contraception was a good idea (12). Clearly, mode of delivery is an important factor in the acceptability of male contraceptive regimens. In large surveys, oral forms of contraceptive delivery are favored by most male respondents (10, 14). Depending on their country of origin, women felt their partners were most likely to use a daily pill or monthly injection (12). Injection-based hormonal regimens, the most clinically studied experimental contraceptive method to date, are acceptable to the majority of participants in male contraceptive studies (15, 16). However, whether the injections are once a month (16) or as far apart as every 12 wk (15), participants find the injection schedule and associated discomfort as the biggest disadvantage to these regimens, and 35 40% cite dissatisfaction with the method as a result (15, 16). A combination of a daily transdermal gel with every 3-month injections was slightly less acceptable to participants, with 45% stating they would use such a regimen if it were commercially available but one third citing that the daily gel interfered with their daily routine, making it less acceptable (17). In summary, most men and their female partners are receptive to the notion, and in fact the practice, of male contraception. Overall, a majority of men feel that the responsibility of contraception falls too much on women (11) and are receptive to male-based methods (10), while 7197% of women believe male hormonal contraception is a good idea (12). The time is ripe, therefore, for introduction of new methods to the marketplace.
III. Currently Available Male Contraceptive Methods A. Condoms

II. Acceptability of Male Contraception

It has been suggested that development of male-based contraceptive methods represents a misdirection of resources (8) because contraception is ultimately in the domain of female reproductive health. However, the appeal of a male contraceptive to both men and women is great. In surveys spanning four continents, a majority of men indicate a will-

Barrier methods have been used to prevent pregnancy since ancient times, but sheath-like condoms, originally made from animal intestines, were developed 300 400 yr ago. Today, barrier methods, including the female as well as male condom, are the only effective means, besides absti-

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 467

nence and lifelong mutual monogamy, of preventing sexually transmitted disease transmission. In particular, although male circumcision has recently been shown to significantly reduce male transmission of HIV (18), the condom remains the most effective way of preventing HIV transmission during sexual contact. Condoms are currently used by approximately 20% of couples using contraception in the United States (19, 20), although the United Nations reports that only 5% of reproductive age women worldwide rely on male condoms for contraception (21). Condoms are associated with few adverse side effects; however, they have a marginal contraceptive efficacy. This is mainly due to improper or inconsistent usage, as demonstrated by comparison of pregnancy rates for typical use (15%) vs. perfect use (2%) for couples using condoms for contraception (22, 23). A significant drawback to condoms is their poor long-term compliance, with more than half of users reporting inconsistent use with every act of intercourse (24). In addition, many men dislike condoms because they feel condoms diminish sexual pleasure (25), again hampering consistent usage. However, even when used correctly, condom breakage and slippage are not infrequent, occurring in up to 2 8% of cases (26 30). Since 1920, most condoms have been made of latex rubber; however, latex condom use has been associated with development of latex allergies in both men and women, which can cause skin irritation and even death (31, 32). As a result of latex allergies, polyurethane condoms have been more recently developed and entered the market in the 1990s. Clinical studies with these condoms have demonstrated a slightly higher rate of condom breakage and slippage when compared with latex condoms (26, 29, 30, 33). In addition, some users have greater difficulty with putting on (33) or unrolling (30) polyurethane condoms compared with latex condoms. Most importantly, however, in general polyurethane and latex condoms have similar efficacy rates (26, 28), with the exception of one study reporting slightly greater efficacy with the latex condom (29). Therefore, despite their increased breakage, polyurethane condoms are an important alternative to latex condoms for couples, particularly those with latex allergies.
B. Vasectomy

a delay in the onset of azoospermia of several months and the lack of complete reversibility. Acute pain, blood loss, and surgical site infections are very rare, particularly with the no-scalpel technique (41). However, whereas postoperative pain usually resolves quickly, 6 15% of men experience some degree of postvasectomy chronic testicular discomfort (43 45). In one study of such men, the majority had relief of their symptoms with reversal of the vasectomy (46). Three to 5% of men with vasectomies eventually request reversal, usually due to remarriage (47, 48). Vasectomy reversal, or vasovasostomy, has the potential to restore fertility; however, rates of pregnancy vary from 30 60% (45). Factors that impact postreversal pregnancy rates include microsurgical technique used and the length of time between the vasectomy and the reversal procedure (45, 49). In 20 30% of men, vasovasostomy is unable to restore patency of the vas if more than 8 yr have elapsed since the original vasectomy (49), and pregnancy rates fall to less than 50%. In fact, 20 40% of all couples remain infertile after vasectomy reversal despite restored patency of the vas (as documented by imaging techniques), possibly due to the presence of antisperm antibodies (50, 51). The type of antibody response may be a key factor. One study found that the presence of IgA antisperm antibodies greatly increased rates of fertility failure after vasovasostomy (52). In summary, for these reasons, vasectomy cannot be considered to be a truly reversible method of male contraception and is most appropriate for men who no longer wish to father children. Vasectomy appears to be safe in regard to overall male health. Earlier reports of associations between vasectomy and cardiovascular disease have proven incorrect (53), and more recent concerns about vasectomy and prostate cancer risk have not been substantiated (35, 43, 45, 54 56). In summary, vasectomy is highly effective and safe. The major drawbacks of vasectomy are a risk of chronic testicular discomfort, the inability of surgery to reliably restore fertility when desired, and economic constraints in developing countries.
IV. Regulation of Testicular Function and Spermatogenesis

Vasectomy is a safe, simple, outpatient surgery performed under local anesthesia in which the ductus deferens is severed and the ends ligated through a small scrotal incision. Approximately 500,000 vasectomies are performed yearly in the United States (34), where approximately 10% of couples using contraception rely on this method (19, 20). Worldwide over 40 million men have undergone the procedure (35), accounting for about 5% of active contraception (21). Vasectomies are highly effective, with a failure rate of less than 1% and a low incidence of complications (36 38). The no scalpel technique was introduced in the early 1990s after development in the Sichuan province in China (39). This technique utilizes a single, midline puncture in the scrotal raphe made with scissors, and in experienced hands it is superior to previous procedures because it minimizes blood loss and risk of infection (40 42). The major drawbacks to vasectomy are

The purpose of this section is to introduce the physiological basis of male fertility, with particular attention to potential contraceptive targets. It is not meant to be a comprehensive review of all factors involved in this process. Developmental aspects of the male reproductive tract have been reviewed recently (57).
A. The hypothalamic-pituitary axis: control of gonadotropin secretion

The hypothalamic-pituitary-testicular axis is a classic endocrine loop, with negative feedback of downstream products playing the pivotal regulatory role in maintaining homeostasis. Testicular production of both hormones and sperm is exquisitely regulated by gonadotropins produced by the pituitary, whereas gonadotropin production is under the direct control of pulsatile GnRH secretion from the hypothalamus (Fig. 1). In turn, steroid and peptide hormones

468

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

FIG. 1. The endocrinology of spermatogenesis and male hormonal contraception. Open arrows denote promotion of spermatogenesis; dashed lines denote inhibition of spermatogenesis and hormone production. A, Diagram of the naturally occurring, normal state. B, Diagram of the impact of hormonal contraceptive interventions on the reproductive axis.

produced in the testes provide inhibitory signals to the pituitary and hypothalamus. It has been known for over 25 yr that both temporal and quantitative changes in GnRH secretion disrupt testicular function (58). GnRH secretion from the hypothalamus is directly related to pulsatile release of LH and FSH in the peripheral circulation (59, 60); thus disrupters of GnRH secretion and action are potential contraceptive targets. Over the last 5 yr there have been rapid advances in understanding of the neuroendocrine control of reproduction upstream of GnRH, and these too represent potential new contraceptive targets that have yet to be tested. Initially, studies into the

genetics of familial hypogonadotropic hypogonadism revealed the critical role of the then orphan G protein receptor GPR54 in gonadotropin secretion (61, 62). Previous studies had demonstrated that the 54-amino acid peptide kisspeptin-54 (also called metastin) is the ligand for GPR54 (63 65). Subsequent investigations have demonstrated that: 1) kisspeptin is expressed in the arcuate, periventricular, and anteroventral periventricular nuclei of the forebrain; 2) GPR54 receptors located on GnRH-secreting neurons in the brain respond to kisspeptin; 3) kisspeptin administration in the brain results in GnRH and gonadotropin secretion in animal models; 4) gonadotropin responses to kisspeptin administration in animals are blocked by administration of a GnRH antagonist; and 5) kisspeptin appears to have direct inhibitory effects on pituitary gonadotropin release through interaction with GPR54 expressed on pituitary gonadotropes (reviewed in Ref. 66). Moreover, the negative feedback of testosterone and estradiol on GnRH secretion is mediated via inhibition of kisspeptin production in the arcuate nucleus of the hypothalamus (67, 68). Most recently, in contrast to acute administration of kisspeptin, which stimulates gonadotropin and testosterone release, chronic administration of kisspeptin lowers serum LH levels in monkeys (69) and leads to testicular atrophy in adult male rats similar to that seen with chronic GnRH infusions (70). Together, these studies directly link kisspeptin-GPR54 to male reproduction and demonstrate the importance of this pathway in upstream regulation of testicular function and sex steroid homeostasis (Fig. 1). Similar to GnRH, the kisspeptin-GPR54 neuroendocrine pathway could be exploited for contraception. Inhibitors of kisspeptin, and/or GPR54 agonists and antagonists, in combination with testosterone, are a theoretically viable contraceptive strategy yet to be tested. Negative feedback to the pituitary and hypothalamus is critical to maintain endocrine homeostasis in the male (Fig. 1). Testosterone is the main testicular steroid, but significant aromatase activity in the testicle and peripheral tissues results in production of estradiol as well. Testosterone clearly inhibits kisspeptin transcription (67) and GnRH and gonadotropin secretion (7174). Some of this inhibition is estrogen independent (7578), but estradiol seems to play an important role in steroid negative feedback in the male, particularly by decreasing LH production (79 83). Aromatase inhibition results in significant increases in LH and FSH secretion in normal men (80); normal levels can be restored with estradiol supplementation in a nonlinear fashion, with higher estradiol levels having increasingly less effect on circulating gonadotropin and testosterone levels (81). Moreover, both aromatase inhibition and medical castration result in similar increases in FSH, despite markedly decreased testosterone levels with medical castration compared with aromatase inhibition; this suggests that, at least acutely, FSH regulation is more dependent on estradiol than testosterone (84, 85). In contrast, dihydrotestosterone (DHT), a nonaromatizable androgen, is a poor inhibitor of gonadotropin production in the male when given at or near physiological doses (83, 86), and 5-reductase blockade with finasteride or dutasteride, which lowers serum DHT concentration by as much as 95%, fails to alter gonadotropin levels (87). However, with chronic administration and supraphysiological dosing, exogenous

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 469

DHT inhibits both LH and FSH secretion despite concomitant suppression of testosterone and estradiol (88, 89), demonstrating that aromatization is not an absolute requirement for negative feedback. Like other sex steroids, progesterone is expressed in the male as well as the female, albeit at lower levels (90). The precise role of progesterone in normal male physiology is unknown, but progesterone receptors have been demonstrated in the male hypothalamus, pituitary, and reproductive tract (9193). Mature male progesterone receptor-deficient mice have normal reproductive phenotype, normal serum LH, and mildly decreased FSH levels compared with wild-type animals (94), results which suggest that the physiological role of progestins in the male hypothalamic-pituitary axis is minimal, at least in the rodent. However, progestins enhance male hormonal contraceptive efficacy when combined with androgens (95), an effect attributed to increased hypothalamic-pituitary suppression of gonadotropin secretion either directly or through the androgen receptor. Brady et al. (96) compared the effects of progesterone and the synthetic progestin, desogestrel, on gonadotropin secretion and GnRH responsiveness in normal men. Although both progestins decreased LH and FSH secretion, only progesterone decreased the LH response to GnRH. These results demonstrate that progesterone affects gonadotropin secretion through progesterone receptors and provide evidence for such inhibition at both the hypothalamic and pituitary levels. However, the effects of exogenous progestins on spermatogenesis are greater than can be accounted for by enhanced suppression of gonadotropins alone (97). Therefore, it has been suggested that progestins may act directly on the testes. There is recent evidence directly supporting this hypothesis. First, both membrane-bound and intracellular progesterone receptors have been demonstrated in the human testes (98). Second, Walton et al. (99) recently demonstrated that the exogenous progestin desogestrel, when combined with testosterone and a GnRH antagonist, mediates a direct effect on testicular gene expression. In this context, progestins influenced expression of genes involved in steroidogenesis, Sertoli cell function, and spermatogenesis. Together these data strongly support an effect of progestins on male reproduction independent of their gonadotropin effects. Exploitation of testicular progestin action is an additional mechanism whereby specific progestins could be formulated as male contraceptives. Finally, the nonsteroidal testicular product inhibin B contributes to hormonal feedback in the male. Inhibin B is a dimeric molecule consisting of and subunits and a member of the TGF- family. Inhibin B is predominantly produced by Sertoli cells (100), and inhibin B levels correlate with Sertoli cell number (101). However recent observations in boys with constitutively active LH-receptor mutations resulting in familial male-limited precocious puberty suggest that Leydig cells may produce some inhibin B (102). Indeed, although inhibin B levels rise acutely with FSH administration (103), chronic FSH and spermatogenic suppression results in only a 25% decline in levels, suggesting FSH-independent inhibin B production (104). Similarly, inhibin B levels are low, but detectable, in rare individuals with FSH and FSH receptor mutations (105107). Negative feedback by

inhibin B on FSH production has been demonstrated in 1) normal men, where inhibin B and FSH exhibit an inverse relationship (108, 109); 2) castrate men with very high FSH levels compared with normal men rendered medically castrate (109); 3) men with testicular disorders (103, 110); and 4) male infertility (111) where low inhibin B levels are associated with increased levels of FSH. Together, these data establish the role for inhibin B as a testicular regulator of FSH, as hypothesized by McCullagh (112) over 75 yr ago. Aside from its role in regulating FSH production and as an indicator of Sertoli cell function, no clear role for inhibin B in testicular physiology or spermatogenesis has been established. Theoretically, exogenous inhibin B in conjunction with androgens might further suppress FSH and be an adjunct in a male hormonal contraceptive regimen (Fig. 1). However, to date recombinant inhibin B has not been available for clinical studies, and recent investigations suggest that incomplete FSH suppression may not be pivotal to persistent spermatogenesis in men undergoing male hormonal contraceptive treatment (see Ref. 97 and Section IV.D).
B. Testosterone production and androgen action

Testosterone is produced by the Leydig cells of the testes, primarily in response to LH stimulation. However, there is increasing evidence that there may be non-LH-dependent production of testosterone, which may be critical in maintaining spermatogenesis under conditions of LH inhibition, such as with the hormonal approach to male contraception. LH-receptor (LHR /) knockout mice have low, but detectable levels of circulating testosterone (113, 114). Despite being infertile, a detailed histological examination of testicular tissue from mature LHR / mice revealed low level spermatogenesis that was completely blocked by treatment with an androgen receptor antagonist (113). These results are consistent with low levels of testosterone observed in rodents after hypophysectomy (115, 116). Regulation of non-gonadotropin-dependent testosterone production in men, if it is similar to that in rodents, may be a critical aspect in the development of a universally effective hormonal method of male contraception because this approach relies on blockade of gonadotropin secretion for efficacy. Under normal physiological conditions in young men, 4 6 mg of testosterone is secreted by the testes daily (117119) with a circadian rhythm; the highest level of secretion is in the early morning, and lower levels are found in the circulation during midafternoon (120 122). This pattern is blunted with age, likely due to impairment at multiple levels of the hypothalamic-pituitary-testicular axis (123126). A small percentage of testosterone (1%) is converted to DHT within the testes and elsewhere by the isoenzymes 5-reductase I and II, resulting in circulating DHT levels in the 13 nmol/liter range in normal men. DHT is thought to be a considerably more potent androgen than testosterone, due to its 10-fold greater affinity for the androgen receptor, but its role in spermatogenesis is not known. Significant reductions in serum DHT appear to have a marginal impact on sperm production in normal men (87). In addition, testosterone can be aromatized to estradiol within the testes. The role of estradiol in spermatogenesis is unclear, but increasing evi-

470

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

dence suggests that it may play a role. Men with aromatase deficiency or mutations in the estrogen receptor have impaired spermatogenesis despite otherwise normal secondary sexual development (127130). Moreover, estrogen receptors are expressed in the testes and perhaps on human male germ cells themselves (131133). Both testosterone and its active metabolites therefore appear to be important in qualitatively and quantitatively normal spermatogenesis. Because testicular Leydig cells are the site of testosterone synthesis and secretion under the control of LH, intratesticular testosterone concentrations are 100-fold those of serum (134 138), but the relative importance of this high-androgen environment for spermatogenesis is not well understood. Experimental suppression of LH and FSH levels profoundly decreases testosterone production (137, 139, 140) and dramatically reduces spermatogenesis in men (141, 142). However, results from LH receptor-deficient mice suggest that even markedly reduced levels of androgen can support spermatogenesis (113, 114), although complete blockade of residual androgen action with androgen receptor antagonist flutamide abolished spermatogenesis (113). In addition, using the very effective combination of testosterone enanthate (TE) cyproterone acetate, higher levels of serum testosterone were associated with a lesser degree of spermatogenic suppression (143). These findings have led to the proposal that high levels of exogenous androgens or residual androgen production after gonadotropin withdrawal may be sufficient to support spermatogenesis and result in contraceptive failure (117, 138, 144, 145). However, two recent studies have called this into question (140, 146). In contrast to a rodent model (147), no significant relationship between sperm concentration and intratesticular androgen concentrations was observed in men after prolonged gonadotropin withdrawal in these small studies. Moreover, although it has been suggested that the active metabolite of testosterone, DHT, a more potent androgen, may play a pivotal role in maintaining spermatogenesis during male hormonal contraceptive treatment (117, 138), intratesticular DHT levels did not explain persistent spermatogenesis in these studies. These results are consistent with data in humans where the addition of a 5-reductase inhibitor to a male hormonal contraceptive regimen was not shown to increase the effectiveness of spermatogenic suppression (148, 149). Further investigation into the precise regulation and role of intratesticular androgens in supporting spermatogenesis in men is clearly needed, because targeted disruption of residual androgen production and/or action might be an adjunct to increase effectiveness of current hormonal methods. Classically, androgen action results from ligand binding to the androgen receptor and translocation of the bound, dimerized receptor-ligand complex to the nucleus where it directs androgen-regulated gene transcription via interaction with androgen-response-elements (150). Androgen receptor is expressed in multiple cell types within the testes including Sertoli, Leydig, peritubular myoid, and vascular smooth muscle cells, and on germ cells during the early stages of spermatogenesis (151). Mice completely devoid of androgen receptor throughout development have feminized genitalia, cryptorchid testes, and azoospermia with arrested spermatogenesis at the pachytene spermatocyte stage (152). Due to

the role of androgen receptor signaling in the development of both the external and internal male phenotype, tissuespecific knockouts were required to further delineate the role of androgen receptor in supporting spermatogenesis in specific testicular cell types. Normal fertility is preserved in mice lacking androgen receptor in either germ cells (153) or peritubular myoid cells (154), although in the latter case reduced germ cell numbers, particularly within the epididymis, were noted. In contrast, both Leydig cell and Sertoli cell-specific androgen receptor knockouts are infertile (153, 155157) and have small testes. Interestingly, the level of serum testosterone found in various Sertoli-cell only knockouts varies from low to supraphysiological for unclear reasons (155157). These differences aside, these studies emphasize the critical importance of paracrine actions of androgens within the testes, but the precise mechanism whereby androgen receptor signaling in Leydig and Sertoli cells supports germ cell maturation is not yet clear. Three recent studies examining the transcriptional targets of androgen receptor using Sertolicell only androgen receptor knockouts suggest potential targets, but further functional analyses of these will be required (158 160). In theory, these models suggest that androgen receptor disrupters that could be targeted to specific cell types such as the Sertoli cell might make excellent contraceptives but such compounds have yet to be developed. Data from animals with disrupted androgen receptor and mice treated with androgen receptor blockade with the antagonist flutamide (161) demonstrate that signaling through the androgen receptor is required for spermatogenesis. Modulators of androgen action include tissue-specific expression of coactivators and corepressors as well as genomic variation in the androgen receptor itself. Molecules that bind the androgen receptor yet prohibit or enhance binding of tissuespecific coactivators or corepressors could act as selective androgen receptor modulators (SARMs). Recently, a second, nongenotropic sex steroid receptor-mediated signaling pathway has been proposed that may allow for a second, tissuespecific mechanism for androgen receptor signaling (162 164). Activators of nongenomic estrogen-like signaling, or ANGELS (164, 165), are thought to act predominantly through the MAPK kinase signaling cascade, although their exact mechanism of action and degree of tissue selectivity remain controversial (166). Either SARMs or ANGELS could be employed as part of a hormonal approach to male contraception, allowing for tissue-specific androgen replacement in the setting of reduced gonadotropin and endogenous testosterone production. However, such approaches have yet to be tested in human trials. In addition, naturally occurring variations in the androgen receptor structure could be exploited in the design of such designer androgens. For example, exon 1 of the androgen receptor contains a trinucleotide CAG repeat of variable number, encoding a polymorphic polyglutamine tract and a downstream GGN repeat encoding a polyglycine region (167). The number of CAG repeats is inversely associated with the transcriptional response to androgens in vitro (168, 169) and in vivo (170 172). Impairments in spermatogenesis have been associated with increased CAG repeat length (173, 174), although this is not a consistent finding (175, 176). Expanded GGN repeats are also thought to decrease andro-

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 471

gen receptor signaling, but increased repeats were not associated with impaired spermatogenesis (177) unless found in conjunction with longer CAG repeats (178). The crystal structure of the androgen receptor with expanded exon 1 repeats has not been published to date, so the exact impact of these changes on the ligand binding domain is not known. However, given the subtle androgen effects of CAG and GGN repeats observed clinically, exploitation of naturally occurring variations in androgen receptor is unlikely to prove an effective strategy for contraceptive drug design. Over the last few years, a number of testosterone formulations have become available, including long-acting depot injections, implants, transdermal gels, transdermal patches, and buccal formulations (Table 1). Each of these has its own particular pharmacokinetics (179). Using a hormonal approach to contraception, testosterone is administered in part to block gonadotropin secretion but also to maintain nongonadal androgen-dependent functions such as sexual drive and muscle mass. The importance of mimicking normal physiological testosterone secretion patterns in maintaining both nongonadal androgenicity and contraceptive efficacy using a hormonal approach is not known (180), because head to head trials comparing different forms of testosterone delivery are lacking. Long-acting injections, which are currently being tested as part of a large contraceptive efficacy trial, are associated with peaks and troughs, even after multiple injections (181). Although the transdermal preparations have the most physiological pharmacokinetic profiles, patches have been universally disappointing in contraceptive trials (182184), probably due to low levels of serum testosterone. The only large study using transdermal testosterone gel to date, although highly effective, achieved testosterone levels in the slightly supraphysiological range (185). Significant supraphysiological dosing of testosterone

can be associated with side effects including high-density lipoprotein (HDL) suppression, acne, and increased hemoglobin concentrations (186, 187) that would not be desirable in a regimen designed for long-term use, although these are not consistent findings (188). Therefore, the goal of most hormonal regimens under study is to maintain serum testosterone levels within the normal range, delivering approximately 5 mg/d of testosterone.
C. Spermatogenesis

Male contraceptive strategies may generally be divided into approaches that disrupt spermatogenesis, which are largely hormonally based, and those that disrupt sperm-egg interactions by disrupting either sperm motility or processes involved in fertilization. These latter approaches are nonhormonal. Although this is not meant to be a comprehensive review of our current understanding of spermatogenesis, we provide a brief overview of sperm maturation here to put these strategies into context. In particular, there have been substantial developments in delineating the markers and potential of germ cell stem cells recently, and we refer the reader to recent reviews of this field (189, 190). During spermatogenesis, spermatogonia, through multiple mitotic and meiotic divisions, develop into mature spermatozoa. In brief, this process may be divided into four stages (191). First, type A spermatogonia undergo mitosis, resulting in renewal of germline stem cells as well as type B spermatogonia that continue to undergo differentiation. Second, type B spermatogonia undergo meiosis, generating haploid round spermatids. Next, spermiogenesis occurs, wherein round spermatids mature to motile spermatozoa without undergoing further cell division. Spermiogenesis includes formation of the acrosome, nuclear condensation,

TABLE 1. Androgen formulations available for possible incorporation into a male hormonal contraceptive regimen
Generic name Route and interval of administration Published use in a contraceptive regimen (Ref.) Potential concerns as part of a contraceptive regimen

Testosterone undecanoate 17-Methyltestosterone Testosterone enanthate Testosterone decanoate Testosterone undecanoate Testosterone implants Testosterone patch Testosterone gel Dihydrotestosterone gel Testosterone buccal system MENT implant Testosterone 5-reductase inhibitor

Oral, twice daily Oral, daily Intramuscular, 12 wk Intramuscular, 4 6 wk Intramuscular, 8 12 wk Subcutaneous, 4 months Transdermal nonscrotal, daily Transdermal, daily Transdermal, daily Buccal, daily Subcutaneous, 6 months Injection 12 wk, plus daily oral pill

Yes (228, 229, 241, 381) No Yes (141, 142, 143, 238 240, 242, 243, 250, 251, 382) Yes (246) Yes (217, 230, 231, 254 256, 384) Yes (216, 218, 245, 247, 385) Yes (182184) Yes (185, 381) Yes (381, 383) No Yes (215, 298) Yes (148, 149)

Twice daily dosing, short and variable duration Liver toxicity Injections can be painful, high peak levels Injections can be painful, high peak levels Injections can be painful, nonphysiological pharmacokinetics Surgical placement, occasional painful expulsions Poor efficacy, high frequency of skin irritation Possibility of partner transfer, daily application needed Poor efficacy, not aromatized Untested, very limited clinical use Surgical placement, poor sperm suppression, concern regarding bone effects 5-Reductase inhibitor gave no additional contraceptive benefit

472

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

and extensive cellular reorganization including sperm tail development. Finally, interactions with the Sertoli cell mediate the process of spermiation, wherein cytoplasmic material from the spermatid is removed and the mature sperm is released into the lumen of the seminiferous tubule. This entire process is under strict endocrine regulation, and there is potential for contraceptives to target all or part of this process. Although the role of FSH in human spermatogenesis has been debated, it is clear that both FSH and LH are required for quantitatively and qualitatively normal spermatogenesis in men. This has been demonstrated in a series of human studies wherein gonadotropin production and spermatogenesis were suppressed in men using exogenous testosterone and either LH [in the form of human chorionic gonadotropin (hCG)] or FSH were given back in a selective fashion (192 195). Although either gonadotropin alone can reinitiate spermatogenesis, both LH and FSH are required to achieve normal semen parameters. Matthiesson et al. (196) recently expanded upon this work with detailed examination of the relative stages of spermatogenesis affected by selective gonadotropin supplementation using exogenous testosterone plus depomedroxyprogesterone (DMPA) to suppress endogenous testosterone and spermatogenesis. Data from both rodents and monkeys suggested that FSH is more important than LH for spermatogonial maturation, whereas LH and/or intratesticular androgens are needed for normal completion of meiosis and spermiogenesis/spermiation (197200). Surprisingly, after 6 wk of selective LH or FSH withdrawal, only pachytene sperm differed among men lacking FSH (treated with testosterone DMPA hCG) compared with untreated, control individuals; men lacking LH showed no differences compared with untreated men. Moreover, this was despite a 100-fold reduction in intratesticular testosterone in men treated with testosterone DMPA FSH compared with control and hCG-treated men (196). Although the interpretation of these results is limited by the length of treatment and small numbers of individuals studied, they suggest that either FSH or LH/intratesticular androgens are sufficient to support all stages of spermatogenesis, including spermatogonial maturation, meiosis, spermiogenesis, and spermiation. These data emphasize the need for hormonally based male contraceptive strategies to maximally suppress both LH and FSH to optimize spermatogenic suppression.
D. The epididymis

cell. Therefore, the molecules in these Sertoli-germ cell complexes are attractive contraceptive targets if testes specificity could be demonstrated and appropriate agents developed (203). Within the epididymis, sperm undergo further maturation steps allowing for maximal motility and fertilization capacity, as exemplified by the improved fertility rates when recovered spermatozoa are injected intracytoplasmically into the egg or when spermatozoa from further down the cauda are used for in vitro fertilization (204). There have been a number of detailed analyses of the transcriptional profiles of the rodent epididymis, including detailed segmental analyses (205207). It is possible that in the future such analyses will provide novel contraceptive targets, but at this point these data remain largely descriptive and functional analyses of these novel transcripts are required. Similar analyses using proteomics have begun, again perhaps providing novel, organ-specific targets for intervention (208). Suzuki et al. (209) have recently identified an epididymis-specific promoter from the Lipocalin 5 that may be of use in contrasting animal models to assess these targets. Finally, as they travel through the epididymis, maturing sperm encounter an increasingly hyperosmolar environment, yet upon arrival in the female tract these sperm must adopt to the local, hyposmolar environment to remain motile and competent. This requires significant, regulated volume decreases via ion channels acquired during epididymal maturation that might be targets for contraception in either the male or female (210, 211).
V. Male Hormonal Contraceptive Methods

Once spermatogenesis is complete, sperm are released from the Sertoli cells into the lumen of the seminiferous tubules and move through the epididymis before ejaculation. These processes involve additional, as yet poorly characterized events that might provide further targets for contraception. Both FSH and testosterone are thought to play a role in spermiation (201), the release of the spermatid into the tubule lumen, perhaps through regulation of proteins involved in germ-Sertoli cell adherence (197). In rodents, this multiprotein junctional complex appears to involve N-cadherin and a number of other integrins and kinases and is sensitive to phosphorylation (202). Failure of spermiation results in sperm retention and phagocytosis by the Sertoli

It has been known for almost 70 yr that the chronic administration of testosterone to a man suppresses sperm production (212). Because the administration of testosterone alone fails to completely suppress sperm production in some men, progestogens or GnRH antagonists have been combined with testosterone to further suppress pituitary gonadotropins and improve its contraceptive efficacy. These hormonal contraceptive regimens inhibit sperm production and do not incapacitate existing sperm. Because spermatogenesis takes place over 72 d, there is a 2- to 3-month delay in the onset of full contraceptive effect associated with the use of male hormonal contraceptives, a delay that is similar to that seen with vasectomy, but longer than the period of time required for female oral contraceptives to be effective. In addition, there are ethnic differences in the response of sperm concentrations to male contraceptive regimens. For example, study volunteers in Asia exhibit rates of azoospermia in the 90 100% range on testosterone-alone regimens, whereas Caucasian men have rates of azoospermia closer to 60% with the same treatment (142, 213214, 216). The explanation for this difference is unknown, although it is unlikely to be due to different rates of metabolic clearance of testosterone (122), and it complicates extrapolation of rates of suppression of sperm concentrations between populations.
A. Hormonal contraceptive efficacy

The best test of a contraceptive is to study the method in couples who use it as a sole means of contraception. Al-

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 473

though the efficacy of condoms and vasectomy is well studied, the only experimental male contraceptive methods to date to undergo efficacy testing are hormonal regimens. Four such male hormonal contraceptive efficacy trials have been published (141, 142, 217, 218), and a fifth multicenter study was recently completed. The first of these tested only the fertility of men who became azoospermic with exogenous testosterone administration (141) and demonstrated that the contraceptive efficacy of testosterone-induced azoospermia is almost perfect. However, this study cannot be used to determine the overall efficacy of testosterone alone as a contraceptive because men who did not achieve azoospermia were not allowed to enter the efficacy phase. The three more recent trials have examined the outcomes of all subjects enrolled and allowed men who suppressed below a threshold sperm concentration (1 to 5 million sperm/ml) to use the method as a sole means of birth control. Analysis of these studies reveals that male hormonal contraception has an overall efficacy of approximately 95%, even when men who do not completely suppress sperm production are included (Table 2). This overall figure is only slightly inferior to the efficacy of female hormonal contraceptives. In these studies, a sperm concentration at or below 1 million sperm/ml of ejaculatesevere oligospermiais associated with a risk of pregnancy of approximately 1% (142). Therefore, uniform suppression of all subjects to a sperm concentration no greater than 1 million sperm/ml is thought to be a reasonable short-term goal for male hormonal contraceptive development (219). Aside from those studies mentioned above, male hormonal contraceptive studies examine the impact of exogenous hormones on sperm production, with effectiveness measured as rates of azoospermia (0 sperm/ ml), severe oligospermia (1 million sperm/ml), or oligospermia (3 million sperm/ml) among users.
B. Testosterone alone

1. Testosterone enanthate (TE). As mentioned above, the World Health Organization (WHO) conducted two seminal, multicenter, male hormonal contraceptive efficacy studies. These trials used the injectable testosterone ester, TE, as a single agent for male contraception. The first study enrolled 271 Asian and Caucasian men who were administered 200 mg TE by weekly injection for 6 months (141). Sixty-five percent of the men became azoospermic using this regimen after a mean of 4 months, and 75% of the remaining men became oligoTABLE 2. Male hormonal contraceptive efficacy trials
Study, year published (Ref.) Length of efficacy phase No. of azoospermic men (%) No. of oligo-spermic men (%)

spermic (3 million sperm/ml). The fertility of 119 of the azoospermic men was then evaluated in a 12-month efficacy phase. In these couples, who used TE as their sole means of contraception during this year, only one pregnancy occurred, corresponding to a pregnancy rate of 0.8 pregnancies per 100 person-years and an efficacy rate of over 99%. The second WHO study tested the fertility of men who became either azoospermic or oligospermic (less than 35 million sperm/ml) on 200 mg of TE weekly (142). The study enrolled 399 men, all but eight (2%) of whom became azoospermic or oligospermic and met criteria for entry into the efficacy phase of the study. In this phase, there were no pregnancies fathered by the men who became azoospermic. In men whose sperm concentration was suppressed to less than 35 million/ml, fertility was reduced to 8.1 pregnancies per 100 person-years. The fertility rate for all men who suppressed enough to enter the efficacy phase was 1.4 per 100 person-years, and the overall failure rate (including the eight men who failed to suppress to less than 5 million sperm/ml) was 5.3%, for an overall contraceptive efficacy of 94.7% (Table 2). These two seminal studies demonstrated that a regimen of weekly TE injections is a reversible and effective contraceptive in the large majority of men and is safe over the 18 months of treatment. The main drawbacks to this approach are the delay in onset of full contraceptive action of 3 4 months, the fact that 2% of men fail to suppress below 3 million sperm/ml and therefore remain potentially fertile, and the potential safety concerns associated with long-term, supraphysiological levels of serum testosterone, the longterm consequences of which are not known. Side effects from weekly injections of 200 mg of TE in healthy men include weight gain, a reversible 25% reduction in testicular volume, a 6% increase in hemoglobin, and a 10 15% decrease in serum HDL cholesterol (142, 187, 220). Importantly, these effects appear to be dose-related (186). There was no evidence that exogenous testosterone administration, despite increasing estradiol levels, increases the risk of blood clotting, which could increase the risk of heart attack or stroke. Indeed, one study suggests that the administration of exogenous testosterone in a male contraceptive regimen may exhibit antithrombotic and antiinflammatory effects (221, 222). Quality of life, sexual function, and well-being are maintained or slightly improved with TE-based male contraceptive regimens (220, 223). Overall, the TE regimen was

No. of couples

No. of failures to suppressa (%)

No. of pregnancies (%)

Total no. of failuresb (%)

Overall contraceptive efficacy (95% CI)

WHO, 1996 (142) Gu et al., 2003 (217) Turner et al., 2003 (218) Total

357 305 53 715

12months 6months 12months

268 (75) 130 (43) 49 (93) 447 (62)

81 (23) 166 (54) 2 (3.6) 249 (35)

8 (2.2) 9 (2.9) 2 (3.6) 19 (2.6)

11 (3.1) 1 (0.3) 0 (0.0) 12 (1.7)

19 (5.3) 10 (3.3) 2 (3.6) 31 (4.4)

94.7(9297) 96.7(9598) 96.4(91100) 95.7(94 97)

CI, Confidence interval. a To a sperm count less than 35 million, depending on study. b Defined as pregnancy and/or failure to suppress to enrollment in the efficacy phase.

474

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

rated better than expected by a majority of men in these studies (224). However, the use of weekly im injections in this regimen led to discontinuation by a small number of men. Although useful as a proof of principle, these landmark studies demonstrated that use of weekly TE alone for contraception is limited by side effects, a 2% failure to adequately suppress spermatogenesis, and the impracticality of weekly injections, and suggested that additional agents would be necessary to improve the hormonal approach to male hormonal contraception. 2. Testosterone undecanoate (TU). TU is a long-chain ester that can be administered orally or by injection (225, 226). In testosterone-deficient men, a single injection maintains serum testosterone levels in the normal range for 6 12 wk (226, 227). Oral administration requires dosing two or three times daily, making it less practical (225), and explaining perhaps the somewhat disappointing results of the only (pilot) male hormonal contraceptive study using oral TU (228). Three small trials using TU injections in normal men for male contraception were initially conducted. In the first study, conducted in China, volunteers received monthly doses of 500 or 1000 mg TU. Eleven of the 12 men in the 500-mg group and all 12 in the 1000-mg group became azoospermic, with the 1000-mg group achieving azoospermia more quickly (229). In the two other pilot studies, conducted in Germany, TU was used alone or in combination with a progestin (see Section V.C) (230, 231). In the arms of these studies in which subjects received only TU, eight of 14 men who were administered 1000 mg TU every 6 wk achieved azoospermia, with four of the remaining six suppressing their sperm concentrations below 3 million sperm/ml. Although the regimens and formulations of TU were slightly different than that used in China, it appears that, as was the case with TE, it remains more difficult to suppress sperm concentrations in nonAsian populations even with long-acting androgens such as TU. Subsequently, a large Chinese efficacy trial studied 308 men treated with monthly injections of 500 mg TU after a 1000-mg loading dose (217). Forty-three percent of men suppressed to azoospermia on this regimen, but only nine (3%) of the men failed to suppress to sperm concentrations below 3 million sperm/ml, the criteria for entering the efficacy phase of the study. During the subsequent 12-month contraceptive efficacy phase, 296 couples used the TU injections as a sole means of contraception. Of these, six men had partial return of sperm production, and there was one pregnancy attributed to the reappearance of sperm while on TU therapy. Overall, this regimen has a 96.7% contraceptive efficacy in Chinese men (Table 2). As with TE, injections of TU at this dosing interval resulted in supraphysiological levels of serum testosterone in some men, and nadir testosterone levels were 31% greater than baseline testosterone levels. Similar to TE, injectable TU was associated with weight gain, a 9% increase in hemoglobin, and a 14% decrease in HDL. However, there were no serious adverse events during treatment, and these changes were reversible. Moreover, most participants found the regimen acceptable (16). A phase III trial of contraceptive efficacy with TU in tea seed oil involving 1000

Chinese couples is currently nearing completion, which will add much to the field. 3. Transdermal testosterone. Scrotal transdermal testosterone patches were introduced in the late 1980s (232). Although testosterone patches are useful for the treatment of male hypogonadism, they have not been effective for male contraception, presumably because the serum testosterone levels are insufficient to completely suppress gonadotropin secretion from the pituitary, even with the addition of a progestin. For example, one study combining a testosterone patch with the progestin levonorgestrel resulted in azoospermia in only two of 11 men and counts below 3 million sperm/ml in three others (182). In a second study, testosterone patches were combined with oral levonorgestrel, but less than 50% of subjects achieved azoospermia (183). Skin reactions to the patch are also a problem. In a third testosterone-patch study, which combined testosterone patches with the progestin desogestrel, only 41% of the subjects achieved azoospermia, and 24% of participants withdrew from the study because of skin irritation attributable to the patches (184). Recently, transdermal testosterone gels have been introduced into the marketplace for the treatment of male hypogonadism (233, 234). We recently reported that, in contrast to the testosterone patch, the combination of testosterone gel plus a long-acting injectable progestin, DMPA, resulted in levels of azo- and oligospermia comparable to previous studies with im testosterone (185). These gels hold promise for contraceptive development because they achieve higher serum testosterone levels and are associated with less irritation of the skin than testosterone patches (233). It is possible that in the future, these gels could be combined with a progestin gel as an all-gel approach to male contraception.
C. Testosterone-progestin combinations

Because progestins inhibit the secretion of gonadotropins from the pituitary, several progestins have been combined with testosterone to improve its contraceptive effect by augmenting suppression of gonadotropins. In addition, it has been hypothesized that progestins might also have a direct inhibitory effect on spermatogenesis, although a recent study in nonhuman primates failed to demonstrate such an effect for the progestin norethisterone (235). Testosterone/progestin regimens were first tested for male hormonal contraception in the 1970s (236). One early case report suggested that the contraceptive efficacy of the early combinations was not ideal, with nine of 100 couples conceiving while on treatment, despite instructions to simultaneously use other methods of contraception and sperm concentrations under 10 million sperm/ml (237). Over the following 35 yr, efficacy of testosterone-progestin-based regimens has greatly improved. However, the use of progestins in men can be associated with side effects such as weight gain, additive suppression of serum HDL cholesterol, and perhaps increases in proinflammatory cytokines associated with increased cardiovascular risk (222); thus current protocols aim to minimize these adverse effects (see Section V.G). Early studies of progestins focused on demonstrating im-

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 475

proved sperm suppression compared with testosterone alone, while subsequent studies have aimed at minimizing the doses involved or optimizing the delivery strategy. A randomized, controlled trial of 0.5 mg of oral levonorgestrel with 100 mg of weekly TE showed that the combination was superior to TE alone in achieving azoospermia (67 vs. 33%) by 6 months in Caucasian men (238). Furthermore, the proportion of subjects achieving a sperm concentration of less than 3 million/ml was 94% in the combination group compared with 61% in the TE-alone group. However, the combination regimen resulted in greater weight gain and further decreases in HDL cholesterol when compared with the TEalone group, side effects which have been minimized without compromising effectiveness by lowering the dose of levonorgestrel in subsequent studies (239, 240). Recently, the combination of a long-acting levonorgestrel implant and 1000 mg of TU every 8 wk resulted in azoospermia in 90% of Chinese men, with all subjects suppressing to concentrations fewer than 3 million sperm/ml (241). Another oral progestin, desogestrel, has been tested in male contraceptive regimens with injectable TE (242, 243) or testosterone pellets (400 mg implanted every 12 wk) (244, 245) with overall rates of azoospermia in the 80 90% range in mostly Caucasian populations. The active metabolite of desogestrel is etonogestrel. Oral etonogestrel has been studied in combination with im injections at 4- or 6-wk intervals of the testosterone ester, testosterone decanoate (TD). In a study of 112 subjects, 111 achieved sperm concentrations of less than 1 million sperm/ml (246). Suppression was more rapid in the group receiving TD injections every 4 wk, and side effects were minor. Based upon these results, enthusiasm for the combination of injectable testosterone and a progestin led to two studies combining etonogestrel implants with testosterone. In the first, one or two implanted etonogestrel rods were combined with testosterone pellets and resulted in azoospermia in nine of 14 men in the higher dose etonogestrel group, with 13 of 14 men having sperm counts less than 0.1 million sperm/ml after 24 wk (247). Importantly, there were no significant reductions in HDL cholesterol when the progestin was administered via implant, implying that nonoral administration may be preferable for long-term administration of these compounds. The results from this pilot study were promising enough for a large pharmaceutical company to sponsor a multicenter phase II study of the combination of etonogestrel implants with TD given by im injection every 4 6 wk (248). This combination achieved rates of azoospermia in the 80 90% range, with overall high acceptability and a low incidence of side effects. A follow-up trial of etonogestrel implants combined with TU (which exhibits a half-life superior to that of TD) and involving more than 300 subjects has recently been completed in Europe; these results are not yet published. Success from this trial could in theory result in the first marketed male hormonal contraceptive, although the industry sponsors of this trial (Organon Biosciences and Schering Corporation) may be discontinuing their programs in male contraceptive development. Some progestins such as cyproterone acetate have antiandrogenic properties (249). The antiandrogenic effect of cyproterone may allow it to interfere with testosterone-mediated

spermatogenesis in the testis in addition to its progestogenic inhibition of FSH and LH secretion from the pituitary. In one small trial combining oral cyproterone acetate with 100 mg TE weekly, all men receiving the combination became azoospermic, whereas only three of five in the TE-alone group attained azoospermia (250). In addition, the time required to achieve azoospermia was markedly less in the cyproterone TE groups compared with the testosteronealone group (49 vs. 98 d). This group went on to evaluate lower doses of cyproterone acetate (251), which was also effective, and a completely oral male contraceptive regimen combining cyproterone with twice daily oral TU (228). In the study of the oral regimen, four of eight men suppressed their sperm concentrations below 1 million sperm/ml, with the four remaining subjects suppressing to 1.2 6 million sperm/ml (228). Cyproterone is not currently available in some countries (including the United States). Additionally, a similar antiandrogenic progestogen, dienogest, may be a promising progestin for future clinical trials in male contraception (252). The long-acting injectable progestogen, norethisterone enanthate, has also been tested in male hormonal contraceptive trials (253). In one study, 13 of 14 men who received the combination of 1000 mg TU and 200 mg norethisterone every 6 wk achieved azoospermia after 32 wk of treatment, with HDL suppression and mild weight gain in line with prior studies of testosterone/progestin combinations (231). It has subsequently been demonstrated that high rates of azoospermia (90%) can be maintained when TU and norethisterone enanthate are administered at 8-wk intervals, but not when they are dosed every 12 wk (254). Furthermore, although there is some accumulation of serum testosterone with every 8-wk dosing, the pharmacokinetics of TU are not altered by coadministration of norethisterone (181). This approach, employing an every 8-wk injection schedule, is a significant improvement over the requirement for weekly injections of TE in most prior studies. Alternatively, norethisterone enanthate can also be administered orally in combination with injectable TU (255). Similar to norethisterone enanthate, medroxyprogesterone acetate can be delivered by depot injection (DMPA). Doses identical to those widely used by women for contraception as Depo-Provera (300 mg every 3 months) have been tested in three recent male contraceptive studies. The first was an Australian study combining DMPA with testosterone pellets administered by implantation every 4 6 months (218). In this study, 53 of 55 men achieved sperm concentrations of less than 1 million sperm/ml and entered a 12month efficacy phase in which no pregnancies were observed (Table 2). This important trial was the first testosterone/ progestin efficacy study and confirms that the high rate of pregnancy prevention observed in the original testosteroneonly WHO studies extends to testosterone/progestin combinations. Weight gain and HDL suppression were also very limited in this study. Additionally, a trial of TU at a dose of 1000 mg every 8 wk with DMPA was conducted in China and demonstrated uniform azoospermia in subjects receiving the combination, but not in those receiving TU alone (256). Finally, DMPA injected every 3 months combined with daily transdermal testosterone gel effectively suppressed 19 of 21

476

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

participants to sperm concentrations less than 1 million/ml after 6 months of administration (185). This combination of testosterone plus DMPA is therefore an attractive candidate for larger efficacy studies. In summary, the contraceptive effect of most of the progestins is fairly similar, although smaller doses may be more efficacious for some progestins compared with others and their side effect profiles likely vary (see Section V.G). Combinations of im TU or testosterone pellets and levonorgestrel, etonogestrel, norethindrone, or medroxyprogesterone all result in azoospermia rates of almost 90%. As a result, many researchers now feel that one of these combinations is the most likely to result in a clinically useful contraceptive method. However, larger-scale phase III studies of efficacy at pregnancy prevention will need to be carried out with these regimens before they can be marketed. Suggested guidelines for such a study have been recently outlined (219), and a focus on both pre- and postmarketing side effect profiles is warranted, given the importance of minimizing these in delivery of a long-term medication to a healthy population.

D. Adding GnRH analogs to hormonal contraceptive regimens

GnRH agonists (such as leuprolide) are not effective when used with testosterone for male hormonal contraception (257), perhaps because these agonists tend to allow the continued production of FSH even after chronic administration (257). In contrast to agonists, GnRH antagonists can suppress FSH and LH production within hours of administration and inhibit gonadotropin secretion more completely than agonists. Several trials have investigated the effects of both shortand longer-acting GnRH antagonists as adjuncts in male hormonal contraceptive treatment. Initial very small trials investigated the GnRH antagonist Nal-Glu, administered by daily sc injection, in combination with testosterone to maintain peripheral androgenicity. In the first two trials, 14 of 16 subjects achieved azoospermia by 6 10 wk of treatment (258, 259). But a third small, randomized trial demonstrated no difference in azoospermia with TE plus Nal-Glu compared with TE alone, at least at the doses given (260). Moreover, daily sc injections required for the use of Nal-Glu were not considered to be a feasible part of a long-term contraceptive regimen. It was suggested, therefore, that GnRH antagonists might have a useful role in the induction of azoospermia with maintenance by a combination of hormonal agents. This was demonstrated with Nal-Glu, which induced azoospermia when administered with TE for 12 wk, which was then maintained by TE alone for 20 additional weeks (261). In contrast, the GnRH antagonist cetrorelix, also given by daily injection, combined with the androgen 19-nortestosterone resulted in azoospermia by 12 wk in all six subjects tested; however, azoospermia could not be maintained after the cetrorelix was discontinued (262). It is possible that failure to maintain azoospermia in the second study stemmed from the use of 19-nortestosterone that cannot be aromatized to estrogen, because estrogen may play a role in the feedback suppression of gonadotropin secretion at the pituitary. Indeed, increased

gonadotropin levels were seen associated with spermatogenesis after cessation of cetrorelix in this study (262). Longer-acting GnRH antagonists have recently been developed. A depot formulation of the GnRH antagonist abarelix results in effective gonadotropin suppression for periods up to 1 month (263), but case reports of allergic reactions to this agent may limit its clinical utility. The GnRH antagonist acyline can markedly suppress gonadotropins for up to 2 wk after a single sc administration without significant side effects (264). In a small trial, 12 wk of acyline combined with DMPA and testosterone gel did not demonstrably augment sperm suppression, or the rapidity of sperm suppression, compared with testosterone and DMPA alone (185). These results were surprising because preliminary data demonstrated that administration of acyline in combination with testosterone injections resulted in more rapid gonadotropin suppression than testosterone alone or testosterone plus a progestin (Fig. 2, A. D. Coviello and W. J. Bremner, unpublished data). Additional GnRH antagonists are in development, including orally administered peptide and nonpeptide antagonists (265, 266) and a long-acting (2 yr) GnRH agonist implant that has recently been introduced to the market for treatment of prostate cancer (267). These remain interesting prospects for future contraceptive trials because in theory they could be combined with similarly delivered testosterone formulations without the need for a progestin, perhaps minimizing nonreproductive side effects. In conclusion, while theoretically appealing, GnRH antagonists

FIG. 2. Rate and extent of gonadotropin suppression with various hormonal interventions. Normal healthy men ages 18 55 given TE alone (100 mg im every week), TE plus oral levonorgestrel (LNG, 125 mg oral, daily), TE acyline (300 g/kg sc on day 0 only), or TE LNG acyline (n 7 per group). Error bars represent SEM. Rates of suppression were compared using time-to-event analysis with the event defined as suppression of LH or FSH to 1 IU/liter. Suppression of both LH and FSH was accelerated in both groups receiving acyline (P 0.05) compared with the other two groups. [Data printed with permission of A. D. Coviello and W. J. Bremner.]

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 477

have been relatively understudied compared with testosterone-progestin combinations; however, improved methods of delivery may allow for their incorporation into future contraceptive regimens.
E. Oral male hormonal contraceptive development

Although implant and injection-based hormonal contraceptive regimens have been well studied, many men would prefer an oral formulation for contraception (10). The only orally available testosterone replacement products currently approved for use in the United States are alkylated testosterone derivatives such as methyltestosterone, which can cause liver injury and have been associated with hepatomas (268) and thus are not safe for contraceptive use. The testosterone ester TU is an oral testosterone available in many countries and is not associated with the liver injury seen with methyltestosterone. However, the bioavailability of TU is significantly dependent upon the simultaneous ingestion of a fatty meal (269), and it must be dosed two or three times per day to achieve normal serum testosterone levels, thus making it a poor choice for contraceptive purposes. Oral administration of crystalline testosterone does not greatly increase serum testosterone levels due to extensive hepatic first-pass metabolism and low bioavailability (225, 270 272). However, we recently demonstrated that the addition of a 5-reductase inhibitor to orally administered testosterone in an oil emulsion improved the bioavailability of testosterone compared with oral testosterone alone (273, 274). In these studies, serum testosterone levels within the normal range were achieved for 10 h in normal men with reversible, medically induced hypogonadism when a single dose of testosterone was administered orally in oil together with a 5-reductase inhibitor. Although longer-term studies will be required to demonstrate its safety and efficacy, this form of oral testosterone delivery, in combination with an oral progestin and/or GnRH antagonist raises the possibility of a male pill in the future. An additional orally available androgen that holds promise for hormonal contraception has been reported recently. Attardi et al. (275) demonstrated that the 19-nortestosterone derivative dimethandrolone is active in vivo in a rodent model, causing suppression of LH in castrate rats. Moreover, dimethandrolone has both androgenic and progestational activity, at least in vitro, which might increase its potency as a single agent for contraception (275). Such a strategy remains to be tested in men. Similarly, some steroidal and nonsteroidal SARMs, which have some tissue selectivity for androgen action (see Section V.H) (276), are orally active and represent potentially attractive agents for long-term oral use alone or combined with a progestin.
F. Reversibility of male hormonal contraception

was examined. Recovery was defined as attaining a sperm concentration of more than 20 million sperm/ml, consistent with WHO guidelines for normal semen analyses and normal fertility (278). Importantly, in no case did a subject fail to recover. Median time to recovery was 3.4 months. Increased length of recovery time was associated with longer treatment periods and the use of longer-acting testosterone compounds. Prolonged recovery was not, however, associated with inclusion of a progestin in the regimen. Although the additional recovery data provided by two recently completed multicenter trials will be important to increase the absolute numbers of individuals examined, this analysis provides significant reassurance that the hormonal contraceptive approach is truly reversible in men.
G. Side effects of male hormonal contraceptives

A contraceptive for men must be not only efficacious but also fully reversible. Liu et al. (277) recently published a comprehensive analysis of spermatogenic recovery after experimental male hormonal contraceptive treatment including androgens or an androgen plus a progestin for a minimum of 3 months. Data from 1549 men enrolled in 30 studies

Male hormonal contraceptive regimens can have small but significant effects on a number of nonreproductive parameters, and these need to be examined carefully because such regimens are designed for long-term use in large numbers of men. The extent to which these nonreproductive systems are affected appears to be mediated, at least in part, by differences in the progestin component of the regimen and perhaps by the mode of hormonal delivery (injection vs. oral vs. transdermal). Direct comparisons between regimens have, however, rarely been performed, and it is difficult to extrapolate the relative differences between studies performed at various centers. Since efficacy has been established, at least in small studies, dose-reduction and delivery systems designed to minimize the extragonadal effects of male hormonal contraceptive regimens will be an important aspect of drug development over the next decade. It is possible that, analogous to female oral contraceptives that reduce the incidence of some hormone-dependent cancers (279), male hormonal contraceptives could have health benefits for men. In healthy young men, exogenous androgens increase lean body mass and decrease fat mass in a dose-dependent manner (280), whereas androgen replacement in hypogonadal men results in similar improvements in body composition in addition to increasing bone mineral density (281, 282). It is possible that such benefits could occur in men using hormonal contraception over the long term, although these changes may be modulated by the necessary addition of a progestin (283) or other agent. Increases in lean body mass and decreases in fat mass might be beneficial in reducing the incidence of the metabolic syndrome or cardiovascular disease or in maintaining physical function as men age. Moreover, increases in bone mineral density could decrease fracture risk. Long-term trials and data collection will be required to ascertain whether such benefits are conferred when exogenous androgens are given to healthy men. Little is known about the cardiovascular effects of exogenous androgen-based contraceptive regimens, but weight gain and HDL suppression have been consistent findings. However, total cholesterol and the proatherogenic lipid particle low-density lipoprotein are also suppressed by androgens. High doses of testosterone can cause some of these effects, as in the second WHO efficacy trial of high-dosage im TE (200 mg im/wk), where HDL levels declined by 13% and

478

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

weight increased by 4 kg (142). More physiological dosages of testosterone alone cause significantly less HDL suppression and weight gain but are not as effective at suppressing sperm production and thus have limited clinical utility as a contraceptive (183, 238). The mechanism through which androgens alter serum lipids is not fully understood, but likely is a combination of changes in body fat distribution and stimulation of hepatic lipase activity resulting in decreased lipid particle size and increased lipid density (283). Whereas combining physiological doses of testosterone with a progestin dramatically improves sperm suppression, HDL and weight are also adversely affected (183, 238). At least in the case of some regimens, HDL and weight effects have been clearly shown to be related to the progestin dose (238 240, 242). Together, these data suggest that the progestin component of androgen-progestin-based male hormonal contraceptive regimens may negatively affect metabolic parameters in men. Very little is known about the effects of progestins on cardiovascular risk factors in men. In a small study of men (283) and a large study of women (284), progestins were associated with increased total body and abdominal fat and insulin resistance. The route of administration of the progestin may modulate these metabolic effects, because oral administration is associated with first-pass metabolism in the liver, a critical site for cholesterol metabolism and glucose homeostasis. For example, testosterone combined with oral northethisterone and testosterone plus im northethisterone were equally effective in inducing uniform severe oligospermia, but testosterone plus oral northethisterone induced significantly greater HDL suppression (17%) than testosterone plus im northethisterone (10 12%) (255). Similarly, oral desogestrel significantly suppresses HDL (by 20%), but in a separate trial its active metabolite etonogestrel, delivered by implant, did not cause significant HDL suppression (although HDL levels decline by 722% in dosages that effectively induce severe oligospermia) (242, 245247, 285). Inflammation has been implicated in the development of cardiovascular disease, and a number of inflammatory markers including IL-6 and its product, C-reactive protein, are linked to the development of heart disease. In women, exogenous progestins can increase inflammatory markers (284), but there are little data on the inflammatory effects of progestins in men. In a recent study, norethisterone enanthate administered im in combination with testosterone was associated with an increase in proinflammatory IL-6, whereas testosterone alone resulted in decreased IL-6 (222). Perhaps the most worrisome potential drawback to androgen administration in men is inducing or stimulating prostate diseases including benign prostatic hypertrophy or prostate cancer, both major causes of morbidity and mortality in men. The relationship between circulating androgens and the development of prostate disease in men is controversial. Some epidemiological data correlate high serum testosterone levels with prostate cancer risk (286), although these studies have been conflicting (reviewed in Ref. 287). In contrast, other studies have found a relationship between low serum testosterone levels and occult, aggressive prostate cancer (288, 289). Prostate assessments in previous trials of male hormonal

contraception and of androgen therapy in testosterone-deficient men, while reassuring, have been limited to measurement of serum prostate-specific antigen (PSA), and in some cases, prostate volume (217, 218, 256, 285). Limited data in young men acutely administered im TE at doses as high as 500 mg weekly (five times the physiological dose) for 20 wk did not demonstrate significant elevations in PSA (290), but no other measures of prostate function or morphology were assessed. Given the high prevalence of prostate disease in the developed world, even small changes in disease prevalence associated with exogenous androgen administration in young men could be associated with significant harm. Thus, understanding the long-term effects of exogenous but physiologically dosed androgens on prostate health is a key component to hormonal contraceptive development. Moreover, novel combinations such as a GnRH antagonist plus lowdose androgens with or without a 5-reductase inhibitor might minimize prostate stimulation and deserve further clinical evaluation.
H. Selective androgen receptor modulators for male contraception

There has been recent interest in the development of SARMs that provide tissue-selective androgen action (276). Although development is targeted to hypogonadal men, prostate-sparing androgens that inhibit gonadotropin secretion and spermatogenesis while supporting sexual function and maintaining lean body and bone mass would be attractive for long-term contraceptive use. One approach that might achieve these goals is combining testosterone with a 5-reductase inhibitor. 5-Reductase type II is highly expressed in the prostate and is responsible for the conversion of testosterone to the more potent androgen DHT, resulting in tissue-specific amplification of androgen signaling. In older, hypogonadal men, the combination of TE plus finasteride, a specific inhibitor of 5-reductase type II, maintains the beneficial effects of testosterone on bone mineral density (291), strength, physical performance, lipids, and body composition (292) without precipitating the increases in prostate size or PSA noted with administration of TE alone (291). Moreover, results from the Prostate Cancer Prevention Trial (293) suggest that 5-reductase inhibition may have longterm benefits for men by reducing prostate cancer incidence. In addition, it is possible that the addition of a potent 5reductase inhibitor to testosterone might improve contraceptive efficacy by inhibition of 5-reductase activity in the testes because persistent intratesticular DHT levels have been suggested to contribute to the failure to reach azoospermia in some male hormonal contraceptive studies (99, 117, 138). However, contraceptive trials have yet to demonstrate that the addition of a 5-reductase inhibitor to testosterone plus a progestin further blocks spermatogenesis (140, 148). The compound 7-methyl-19-nortestosterone (MENT) is the first steroidal SARM to be tested as part of a male hormonal contraceptive regimen. The 7-methylation of this compound prevents 5-reduction, effectively minimizing the prostatic activity of MENT (294) and perhaps other androgenic side effects attributed to DHT such as male pattern baldness. Such noncontraceptive benefits could significantly

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 479

improve the appeal of a male hormonal contraceptive to men. In a study in castrated monkeys, MENT showed a 10-fold greater potency (vs. testosterone) for LH inhibition, but only 2-fold greater potency at stimulating prostate growth (295). In men, MENT has a short half-life, probably due to the fact that MENT does not appear to bind to SHBG (296). Therefore, MENT implants have been synthesized to allow for sustained delivery. These implants suppress gonadotropins and androgens by 80 90% in men (297); as a contraceptive, MENT as a single agent resulted in azoospermia in 73% of those individuals who received four implants (298). Recently, MENT implants have been combined with a progestin for contraception (299). Disappointingly, effective sperm suppression was not maintained beyond 12 wk with combined, implantable MENT plus etonogestrel, likely due to the low levels of MENT in the circulation. Future studies with more sustained delivery of MENT or perhaps other selective steroidal SARMS, combined with a progestin or GnRH antagonist, are still a promising approach to contraceptive development. Like MENT, some anabolic steroids have been recently demonstrated in vitro and in animal models to have some degree of tissue selectivity. These studies come predominantly from efforts aimed at understanding the physiological effects of substances that merit banning in professional sport. Some of these have been shown to have tissue-selective androgen action that may make them candidates both for androgen replacement therapy and as part of a contraceptive regimen. For example, desoxymethyltestosterone appears to be significantly more potent for levator ani muscle than at the prostate (300) in a castrate rat model. Similarly, tetrahydrogestinone is less potent than DHT on the rat prostate at doses capable of maintaining muscle mass (301). Both tetrahydrogestinone (302) and dimethandrolone (275) appear to have significant interaction with both the androgen and progestin receptors. Such properties might provide the benefits of increased gonadotropin and spermatogenic suppression, which characterize combined androgen-progestin regimens. Some of these steroids, particularly dimethandrolone that can be administered orally, warrant further testing in humans as part of a contraceptive strategy. Recently, a number of nonsteroidal small molecule SARMs, which may be compatible with oral administration, have been described (276, 303305). In order for these compounds to act as hormonal contraceptives, they must be potent at the pituitary, muscle, and bone while sparing the prostate. One such compound suppressed spermatogenesis in rats by 75% after 10 wk of treatment (306), and another has been shown to maintain bone density and muscle mass while suppressing LH in castrate rats (307). Some of these compounds are currently undergoing clinical evaluation in humans. Nonsteroidal SARMs are particularly appealing because they might allow for oral administration of the androgen component of future male contraceptive regimens and could, in theory, minimize any adverse effects on the prostate. However, their development as contraceptive agents will require a significant effort on the part of the pharmaceutical industry.

I. Why doesnt hormonal contraception work for all men?

A major mystery in the field of male hormonal contraceptive research is why some men fail to sufficiently suppress their sperm counts despite inhibition of serum gonadotropin secretion to extremely low levels. There are no apparent differences in the gonadotropin levels among men who suppress to azoospermia and those who do not; thus the degree of gonadotropin suppression itself is unlikely to be the explanation for these differences (97, 308, 309). As discussed above, an alternative hypothesis has been that persistent intratesticular DHT levels may support ongoing spermatogenesis in this setting (99, 117, 138); however clinical trials do not support an additional contraceptive benefit with the addition of 5-reductase inhibitors to contraceptive regimens (140, 148). Similarly, residual intratesticular testosterone production in some men despite maximal gonadotropin suppression might support spermatogenesis in those men deemed treatment failures. Indeed, work in the LH-receptor knockout mouse supports the concept of basal, non-LH dependent testosterone production capable of supporting low levels of spermatogenesis (113, 310). However, in two small studies we found no relationship between residual intratesticular testosterone levels and sperm production in men undergoing hormonal contraceptive treatment (134, 146). Larger studies are required to confirm these results, but it is possible that, in contrast to studies in rodents, persistent intratesticular androgen levels after hormonal contraceptive treatment do not explain treatment failures and alternative explanations should be pursued. Clinically, it would be of great use if one could predict treatment failures based upon serum markers, either before or during treatment. Although examined in almost every male contraceptive study, baseline sex steroids, gonadotropins, and sperm concentrations do not appear to be predictive of those men who will achieve adequate sperm suppression with hormonal contraceptive treatment. Insulin-like factor 3 (INSL3) is a recently described protein secreted by the testes and found in abundance in the serum (311). INSL3 secretion is regulated, at least in part, by LH, and may be a marker of Leydig cell function (311, 312). Although the physiological role of INSL3 is not completely understood, some data suggest that INSL3 may play a role in germ cell survival (313). Male hormonal contraceptive treatment results in a significant decrease in circulating INSL3 (311, 312). In a retrospective analysis, we recently showed that higher end-oftreatment INSL3 concentrations after male hormonal contraceptive treatment correlated with persistent spermatogenesis and failure to achieve azoospermia (314). Although these results require further confirmation in a prospective fashion, this is the first demonstration of a serum marker that may be associated with persistent spermatogenesis. Exploring a possible functional role for INSL3 in spermatogenesis will be the subject of future studies in this important area. Genetic polymorphisms may explain the difference between men who suppress to azoospermia and those who do not. No such polymorphisms have been identified to date, although three have been examined. Androgen receptor action is influenced, in part, by two trinucleotide repeats lo-

480

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

cated in exon 1. Increased numbers of CAG repeats are associated with decreased androgen activity, whereas the effects of GGN repeats are less clear. One study found that a CAG repeat number of greater than 22 was associated with an increased chance of azoospermia in the setting of incompletely suppressed gonadotropins after hormonal contraceptive treatment (315). However, a second study found that the number of neither CAG nor GGN repeats in the androgen receptor appeared to influence individual responsiveness to hormonal suppression of spermatogenesis (316). These authors went on to examine polymorphisms in the CYP3A4 gene, which encodes an important enzyme in the metabolism of testosterone, and again found no relationship between a known polymorphism in CYP3A4 and sperm suppression (316). Clearly, further investigation will be needed to understand the innate differences in the intratesticular environment that allow some men to continue to produce sperm in the extremely low gonadotropin environment of a male contraceptive regimen. These efforts will likely require a coordinated effort among research centers to yield significant results, due to the relatively small, but critical, numbers of men who fall into the category of hormonal contraceptive treatment failures.
VI. Nonhormonal Methods

Nonhormonal approaches to male contraception can target either sperm production (testicular targets) or sperm motility. These are attractive targets because, theoretically, agents targeted to these processes might be very specific, thus lacking the systemic side effects that plague hormonal methods. In addition, it is possible that such agents could have a much more rapid onset of action. However, development in this area has been slow, and only one of these approaches has reached phase II trials in men (gossypol). Both reversibility and efficacy in animal models continues to be an issue for these approaches, but with advances in genetic and proteomic array approaches, new potential targets will continue to emerge.
A. Testicular targets

Two plant extracts, gossypol, a phenolic compound derived from the cotton plant, and triptolide, derived from the Chinese herbal medicine Trypterigium wilfordii, were promising nonhormonal agents evaluated in the 1980s and 1990s in China. Unfortunately, both agents were found to result in significant, serious side effects and in some cases irreversible infertility, and their development as contraceptives has been abandoned. Gossypol affects both spermatogenesis and sperm motility. Of more than 8000 Chinese men treated with oral gossypol, more than 90% achieved sperm concentrations of less than 1 million/ml (317, 318); however, in 20% of cases this effect was not reversible (319). In addition, hypokalemia was a frequent side effect, in some cases resulting in periodic paralysis (320). A subsequent study suggested that these adverse effects of gossypol may be dose-dependent, but reversible infertility still resulted when lower doses were used (321). However, these side effects caused a reevaluation of the preclinical criteria for contraceptive development (320).

T. wilfordii, an herbal medicine with immunosuppressive properties, impairs sperm motility and density when given to rats and men (322). Purification of the active component of T. wilfordii, triptolide, followed (323). Unfortunately, administration of triptolide to rodents resulted in irreversible infertility (324, 325). Moreover, the immunosuppressive effects of triptolide would likely have prohibited its development as a contraceptive. Mild elevations in scrotal temperature, just above that of the body core, can cause germ cell apoptosis (326, 327). Rodent germ cells appear to be particularly susceptible to testicular heat (328, 329). Although exogenous heat alone is not an effective, long-term contraceptive strategy in men (330), it is possible that heat may decrease the time to low sperm concentration when used in combination with hormonal or nonhormonal contraceptive methods (331). In a recent study, heat using a scrotal water bath at 43 C (30 min/d for 6 consecutive days) in combination with exogenous testosterone decreased sperm count and motility and increased germ cell apoptosis during the first 12 wk of treatment compared with testosterone alone (331). However, in contrast to testosterone plus a progestin, most men using heat plus testosterone failed to achieve sperm concentrations below 1 million/ml after 18 wk of treatment. Therefore, heat plus hormone-based regimens are unlikely to be clinically useful at this point. An alternative strategy to nonhormonal contraception is to target nongerm cells critical for spermatogenesis. In theory, this could be either the Leydig or Sertoli cell, however interruption with Leydig cell function would likely result in systemic hypogonadism requiring the addition of exogenous testosterone. Indenopyridines, serendipitously found to have antifertility effects during toxicology screening for new antihistamines, appear to target both Sertoli cells and germ cells (332334). One such indenopyridine, CDB-4022, when used in combination with a GnRH antagonist, induced reversible infertility in male rats (335). Recently, the l-enantiomer of CDB-4022 was tested in adult cynomolgus monkeys (332). Seven days of oral treatment with l-CDB-4022 resulted in severe oligospermia in all four monkeys treated 17 d after initial administration and remained as such for 6 wk total. Reversibility was demonstrated by wk 16, at which time sperm counts were equivalent to vehicle-treated animals. Gonadotropin and sex steroid concentrations were unaffected, and there were no overt toxicities associated with drug administration. Although l-CDB-4022 clearly affected developing spermatocytes, its precise mechanism of action in the monkey is unclear (332). Given that some studies have demonstrated irreversible testicular effects of CDB-4022 administration in rodents, longer-term studies of l-CDB-4022 in nonhuman primates will likely be necessary before testing in humans. However, these are promising preclinical data for a potential oral, nonhormonal male contraceptive. During their development in the seminiferous tubules, spermatids adhere tightly to Sertoli cells via anchoring proteins in junctions known as apical ectoplasmic specializations (apical ES). Oral administration of 1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide (adjudin) to rats has been shown to disrupt the interaction of spermatid-Sertoli cells by interacting with specific proteins in the apical ES and

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 481

to result in reversible infertility (336 338). However, in some animals, nonreproductive side effects including liver inflammation and muscle atrophy were noted with adjudin administration. These side effects appear to be overcome by directing adjudin delivery to the testes (339). Mruk et al. (339) developed a mutated FSH molecule, lacking three critical glycosylation sites, and conjugated it to adjudin, allowing for significantly less overall drug exposure for the animals and with consistent achievement of reversible infertility. Moreover, endogenous FSH activity appeared to be maintained. Not only adjudin, but also this novel approach to directing agents to the testes is an exciting new area in contraceptive development; however, caution will be required due to the possibility of developing anti-FSH autoantibodies that might compromise reversibility with long-term use.
B. Epididymal targets

During transit through the epididymis, before ejaculation, spermatozoa undergo maturation; thus the epididymis provides another possibly unique contraceptive target. Murine genetic knockout models have provided a number of putative epididymal targets; however, none have reached clinical testing in humans or nonhuman primates. Epididymal-specific proteins could provide points for pharmaceutical intervention. For example, the G-coupled protein receptor HE6 appears to have epididymal duct-specific expression, and HE6-deficient mice are infertile but otherwise phenotypically normal (340, 341). HE6 appears to be critical to fluid resorption in the efferent epididymal ductules, but its precise mechanism of action is not well known. Nevertheless, G-coupled protein receptors can be excellent targets for drug development. Similarly, CRISP-1, a member of the cystiene-rich secretory protein family, appears to have epididymal-specific expression. CRISP-1 appears to be secreted into the lumen of the epididymis where it is postulated to interact with spermatozoa, preventing the initiation of capacitation during sperm transit and storage (342, 343). Pursuit of these, and other, epididymal proteins as contraceptive targets, however, is still a number of years from clinical use because not only do appropriate inhibitors need to be identified, but their specificity and concentration in the reproductive tract will need to be confirmed.
C. Targeting sperm motility or sperm-egg fusion

Recently, several investigators have focused on inhibition of sperm motility as a promising target for male contraceptive development. Ascent of sperm in the female reproductive tract is dependent on initiation of sperm flagellar beating during ejaculation. Sperm progress between 1 and 3 mm/ min propelled by their flagella, which whip three-dimensionally in an elliptical cone (344). Before fertilization, sperm dramatically increase the frequency and force of their flagellar beating, a process termed hyperactivation, to reach and penetrate the ovum. Blocking sperm motility, hyperactivation, or both has great appeal as a male contraceptive. A drug targeting motility or hyperactivation may not need to cross the blood-testes barrier as long as the drug partitioned into seminal fluid and was ejaculated with the sperm. In addition,

such drugs might have a very rapid onset of action, perhaps allowing for administration immediately before intercourse. Unfortunately, agents that interfere with sperm motility may not always be effective across species. Glycosphingolipids are abundant in sperm, and mice deficient in enzymes involved in glycosphingolipid biosynthesis and metabolism have significantly compromised fertility (345347). Gaucher disease is a rare autosomal recessive glycogen storage disease caused by a deficiency in glycosphingolipid metabolism that can be treated, in part, by an inhibitor of glycosphingolipid synthesis, miglustat (N-butyldeoxynojirimycin) (348 350). Administration of miglustat to normal mice resulted in reversible infertility by markedly affecting sperm motility and acrosome morphology (351). However, we recently demonstrated that, in contrast to the initial findings in mice, administration of daily, oral miglustat to healthy men for 6 wk, at doses similar to that used to treat Gaucher disease, had no effect on sperm motility, morphology, or the acrosome reaction (352). In addition, the initial findings in mice were not reproducible among different strains (353), suggesting that genetic variance plays a key role in spermatic sensitivity to miglustat. One approach to blocking sperm motilityand sperm hyperactivation in particular has focused on a group of four novel sperm-specific transmembrane proteins termed CatSpers (354). These transmembrane proteins assemble, likely as a tetramer in conjunction with the transmembrane protein CatSperbeta (355), and allow for calcium entry into the sperm tail through a bicarbonate-activated, voltage-sensitive channel. The rise in intracellular calcium mediated by the CatSpers is directly responsible for the increase in flagellar beat frequency that characterizes sperm hyperactivation. All four CatSpers are critical for fertility, and mice rendered deficient in even a single CatSper are infertile (354, 356) but otherwise phenotypically normal. Thus, CatSpers are promising targets for male contraceptive development because they are highly specific for sperm. However, antagonists for these channels have not yet been reported in the literature and thus are a long way from clinical use. Potassium chloride cotransporters and ion-specific channels are also thought to play an important role in sperm motility. To maintain their motility, ejaculated sperm undergo regulated volume decrease wherein ions and organic osmolytes are effluxed from the sperm in a regulated fashion to prevent cytoplasmic swelling after exposure to the relatively hyposmolar female reproductive tract (211). The nonselective ion channel blocker quinine maximally inhibits regulated volume decrease (357, 358), but sperm-specific potassium and chloride channels, and effective inhibitors, for human sperm have yet to be identified and may not be equivalent to those in the mouse (357). Additional potential targets for inhibiting sperm motility are the proteins of the principal piece of the flagellum. A recent study used mass spectrometry proteomic analyses of isolated human fibrous sheaths to identify several proteins that colocalize to the fibrous sheath of the flagellum (359). Most of the isolated proteins were known glycolytic enzymes, but the authors also identified a unique adenine nucleotide translocase. This protein, called sperm flagellar energy carrier [SFEC, also termed ADP-ATP carrier protein 4

482

Endocrine Reviews, June 2008, 29(4):465 493

Page et al. Advances in Male Contraception

(AAC4)], appears to participate in the transport of ATP within the principal piece. The authors hypothesize that once SFEC has moved ATP to the flagellum core, dynein ATPases allow the microtubules of the flagellum to slide across one another and generate flagellar movement. Structural modeling suggests that SFEC possesses a unique protein motif at the entrance to the nucleotide-binding pocket that might be an attractive drug target. Similarly, the sperm-specific glyceraldehydes 3-phosphate dehydrogenase-S (GAPDS) is bound to the flagellar cytoskeleton and plays a critical role in sperm glycolysis in the mouse (360). GAPDS-deficient male mice are completely infertile, despite normal sperm counts, due to severely impaired motility (360). The human homolog of GAPDS, GAPD2, also appears to be sperm-specific (361). If this is the case, if enzyme-specific inhibitors can be developed that do not interfere with extraspermatic sites of glycosylation, GAPD2 may be an intriguing contraceptive target. Two final intriguing targets for male contraceptive motility research are the unique soluble adenylate cyclase (sAC) and sodium-hydrogen exchanger (sNHE) found in sperm. Increased intracellular cAMP is essential for preparing sperm for fertilization, a process known as capacitation. sAC lacks a transmembrane anchor and produces cAMP in the cytoplasm of the sperm. sAC activity is potentiated by bicarbonate and calcium, known triggers of sperm hyperactivation (362). Therefore, an inhibitor of sAC could utilize two mechanisms to impair fertility: 1) inhibiting sperm motility and hyperactivation; and 2) impairing capacitation. Mice lacking sAC are infertile, and their sperm are immotile due to defects in cAMP production (363, 364). Furthermore, recent work with KH7, a specific inhibitor of sAC, demonstrated that sAC inhibition markedly impairs the ability of mouse sperm to undergo capacitation, achieve a hyperactivated state, and ultimately, fertilize oocytes in vitro (365). Analogous to sAC-null mice, male mice deficient in sNHE are infertile due to immotile sperm (366), and this defect can be overcome in vitro with the addition of cAMP. Wang et al. (367) recently demonstrated that sNHE is required for the full expression of sAC at the protein level and that these two proteins colocalize and form a complex critical for sperm motility. Future work to develop specific inhibitors of sAC and/or sNHE is an intriguing prospect for male contraception. A sperm-specific membrane protein with structural homology to Ig, Izumo, has been recently identified. Data suggest that Izumo is required for sperm-egg fusion (368). Male knockout mice with homozygous Izumo deficiency appear phenotypically normal, with normal sperm numbers, morphology, and motility, yet are infertile. Sperm from Izumodeficient animals could penetrate the zona pellucida of eggs in vitro but could not complete fertilization. However, when intracytoplasmic sperm injection was used to inject Izumodeficient sperm into normal eggs in vitro, the defect was overcome. Its apparent specificity for this critical prefertilization step makes Izumo an attractive target of contraceptive development; however, pharmacological inhibitors of Izumo have yet to be identified. Finally, over the last two decades researchers in India have been developing an injectable intra-vas compound that has

been tested in small phase I and II trials (369, 370). RISUG is a polymer of styrene maleic anhydride dissolved in the vehicle dimethylsulfoxide that is injected into the lumen of the vas deferens using a no-scalpel technique. In the phase II study of 12 men, sustained azoospermia was achieved between 5 and 106 d after the procedure, with the exception of one individual who did not reach azoospermia until d 243, although 100% of his remaining sperm from d 39 onward were nonmotile (370). No pregnancies in the 12 couples studied were reported over the following 12-month period, wherein after the initial 2 months of condom use couples relied only upon the RISUG for contraception. Unfortunately, although RISUG denotes reversible inhibition of sperm under guidance, to date both preclinical and clinical studies have failed to demonstrate reversibility, and the mechanism whereby this might be achieved is not clear. More recent reports from this group have suggested that exposure to the styrene maleic anhydride-dimethylsulfoxide combination within the vas deferens results in irreversible alterations of the sperm membrane via changing the electrostatic and pH environments (371373). Resulting immediate changes in sperm motility and function, if demonstrated clinically, might give such a method an advantage over vasectomy by shortening the postprocedure time to contraceptive efficacy. A phase III trial of this method is apparently under way and hopefully will include data on reversibility. Until such data are published, RISUG should only be considered an experimental alternative to vasectomy.
D. Immunological approaches to contraception

Employing the immune system as a contraceptive by targeting sperm- or egg-specific proteins, or even gonadotropins, has some natural parallel because antisperm antibodies can play a role in infertility. Indeed, sperm antigens have been suggested as ideal targets for contraceptive vaccine development (recently reviewed in Ref. 374). In light of immunological memory, it is difficult conceptually to think of a vaccine approach as a reversible contraceptive method, but a recent study in nonhuman primates suggested that this might be possible. ORand et al. (375) reported that immunization of nine male monkeys with recombinant human eppin resulted in reversible contraception in five of nine animals. Eppin is a testes-epididymal-specific protease inhibitor found on spermatozoa thought to play a role in sperm-semenogelin interaction in the coagulum of human ejaculate (376). Although this represents a novel success in the field of immunocontraception in the male, a number of concerns for further clinical development of this approach were raised in this pilot study. First, booster immunizations were required every 3 wk throughout the treatment to maintain high titer anti-Eppin antibodies, a requirement for contraception. Second, for unknown reasons, two of nine animals failed to mount a significant enough immune response to warrant fertility testing. Finally, two of nine animals failed to sire an offspring after cessation of injections, even after 450 d of observation. It is likely that any vaccine approach to contraception might have similarly inconsistent responses among individuals and that reversibility may also be unpredictable, given the variability in immunogenicity of such a vaccine among individuals. Although controversial, vaccine approaches to contraception

Page et al. Advances in Male Contraception

Endocrine Reviews, June 2008, 29(4):465 493 483

aimed at gonadotropins or GnRH have reached phase I clinical trials in women (377), and a similar approach could in theory also be used in men. Again, variability in the immune response leading to irreversibility and triggering autoimmune disease and perhaps anaphylactic shock has concerned critics in this area (378). In men, GnRH and FSH have been suggested as possible targets for immunocontraception (379), although blockade of the latter would require exogenous testosterone administration for maintenance of androgenicity. Included among most recent work are vaccines aimed at GnRH-1 DNA (380), but there has been very little progress in male contraceptive vaccine development in the last 10 yr, with most work in this area focusing on medical alternatives to castration (permanent sterility) for veterinary use.

introduction, they offer the potential for sperm-specific interventions and thus great promise as reversible contraceptives with minimal side effects suitable for long-term use. Population control remains one of humankinds greatest challenges in the 21st century. The population burden continues to tax our environment and contribute to morbidity and mortality globally. It is the hope of researchers in the field of male contraceptive development that approaches to blocking sperm production and function will bring the dream of the male pill to fruition and significantly impact the health and future of the planet.
Acknowledgments
We thank Dr. Andrea Coviello for permission to include unpublished data (Fig. 2). Received November 26, 2007. Accepted February 26, 2008. Address all correspondence and requests for reprints to: Stephanie T. Page, University of Washington, Box 356138, 1959 NE Pacific Street, Seattle, Washington 98195. E-mail: page@u.washington.edu This work was supported by the National Institute of Child Health and Human Development, a division of the National Institute of Health (NIH), through cooperative agreements U54-HD-12629 and U54 HD42454 as part of the specialized Cooperative Centers Program in Reproductive Research and the Cooperative Contraceptive Research Centers Program (to W.J.B., S.T.P., and J.K.A). In addition, S.T.P. is supported in part by the National Institute of Aging, a Division of the NIH, by Grant K23 AG027238. J.K.A. is supported, in part, by the National Institute of Child Health and Human Development, a Division of the NIH, through Grant K23 HD45386. Disclosures: S.T.P. and J.K.A. have received a lecture fee from Solvay Pharmaceuticals. J.K.A. has been a consultant for the Takeda Pharmaceutical Company. W.J.B. has nothing to disclose.

VII. Conclusions

From a global standpoint, there is clearly a desire and need for more contraceptive options. Couples desire more choices for fertility control, and unplanned pregnancies continue to occur at alarming rates. The hormonal approach to male contraception has made significant progress in the last decade in terms of clinical development. The efficacy of this approach appears to surpass condoms, and these methods seem to appeal to men and their partners. The long experience with female contraceptives sets the bar very high in terms of minimizing side effects for male hormonal contraceptives, thus continued refinements of male hormonal contraceptive regimens to this end are under way. Because the direct health benefits of pregnancy prevention for men are less tangible, male contraceptive development faces additional challenges because agents used for this purpose might be used by healthy individuals over many years. Therefore, acceptance of side effects, or even potential side effects, is very low. New forms of androgen delivery have been introduced over the last 510 yr, including transdermal, oral, and long-acting injectable androgens. These could allow for a very acceptable delivery system for male hormonal contraceptive users. Moreover, the hormonal approach to male fertility control has the potential to further benefit men by contributing to chemoprevention of other diseases, as the female oral contraceptive has. Unfortunately, over the last 2 yr, the major pharmaceutical sponsors of male hormonal contraceptive research have withdrawn their support in this area of product development, making it difficult to complete the final phases of clinical development. Government and not-for-profit sponsors will be needed in this environment to devote the necessary resources for longterm efficacy studies. The potential rewards of preventing unwanted pregnancies and providing more contraceptive choices for couples will hopefully attract visionary leaders to support this mission. Array technology at both the RNA and protein levels is likely to provide an explosion in sperm- and fertilization-specific contraceptive targets in the coming years. Genetic engineering using murine models has already revealed some intriguing nonhormonal targets. Caution must be taken in translating preclinical results, and these methods will require extensive nonhuman primate and human testing. However, although such approaches are a number of years away from clinical

References
1. Population Division of the Department of Economic and Social Affairs of the United Nations Secretariat 2007 World population prospects: the 2006 revision and world urbanization prospects: the 2005 revision. http://esa.un.org/unpp 2. Henshaw SK 1998 Unintended pregnancy in the United States. Fam Plann Perspect 30:24 29, 46 3. Finer LB, Henshaw SK 2003 Abortion incidence and services in the United States in 2000. Perspect Sex Reprod Health 35:6 15 4. Jones RK, Darroch JE, Henshaw SK 2002 Contraceptive use among U.S. women having abortions in 2000 2001. Perspect Sex Reprod Health 34:294 303 5. Besculides M, Laraque F 2004 Unintended pregnancy among the urban poor. J Urban Health 81:340 348 6. Goto A, Yasumura S, Yabe J, Reich MR 2006 Addressing Japans fertility decline: influences of unintended pregnancy on child rearing. Reprod Health Matters 14:191200 7. Anderson RA, Baird DT 2002 Male contraception. Endocr Rev 23:735762 8. Potts M 1996 The myth of a male pill. Nat Med 2:398 399 9. Heinemann K, Saad F, Wiesemes M, Heinemann LA 2005 Expectations toward a novel male fertility control method and potential user types: results of a multinational survey. J Androl 26: 155162 10. Heinemann K, Saad F, Wiesemes M, White S, Heinemann L 2005 Attitudes toward male fertility control: results of a multinational survey on four continents. Hum Reprod 20:549 556 11. Martin CW, Anderson RA, Cheng L, Ho PC, van der Spuy Z, Smith KB, Glasier AF, Everington D, Baird DT 2000 Potential impact of hormonal male contraception: cross-cultural implications for development of novel preparations. Hum Reprod 15:637 645 12. Glasier AF, Anakwe R, Everington D, Martin CW, van der Spuy

484

Endocrine Reviews, June 2008, 29(4):465 493 Z, Cheng L, Ho PC, Anderson RA 2000 Would women trust their partners to use a male pill? Hum Reprod 15:646 649 Weston GC, Schlipalius ML, Vollenhoven BJ 2002 Migrant fathers and their attitudes to potential male hormonal contraceptives. Contraception 66:351355 Weston GC, Schlipalius ML, Bhuinneain MN, Vollenhoven BJ 2002 Will Australian men use male hormonal contraception? A survey of a postpartum population. Med J Aust 176:208 210 Meriggiola MC, Cerpolini S, Bremner WJ, Mbizvo MT, Vogelsong KM, Martorana G, Pelusi G 2006 Acceptability of an injectable male contraceptive regimen of norethisterone enanthate and testosterone undecanoate for men. Hum Reprod 21:20332040 Zhang L, Shah IH, Liu Y, Vogelsong KM, Zhang L 2006 The acceptability of an injectable, once-a-month male contraceptive in China. Contraception 73:548 553 Amory JK, Page ST, Anawalt BD, Matsumoto AM, Bremner WJ 2007 Acceptability of a combination testosterone gel and depomedroxyprogesterone acetate male contraceptive regimen. Contraception 75:218 223 Gray RH, Kigozi G, Serwadda D, Makumbi F, Watya S, Nalugoda F, Kiwanuka N, Moulton LH, Chaudhary MA, Chen MZ, Sewankambo NK, Wabwire-Mangen F, Bacon MC, Williams CF, Opendi P, Reynolds SJ, Laeyendecker O, Quinn TC, Wawer MJ 2007 Male circumcision for HIV prevention in men in Rakai, Uganda: a randomised trial. Lancet 369:657 666 Mosher WD, Martinez GM, Chandra A, Abma JC, Willson SJ 2004 Use of contraception and use of family planning services in the United States: 19822002. Adv Data 350:136 Piccinino LJ, Mosher WD 1998 Trends in contraceptive use in the United States: 19821995. Fam Plann Perspect 30:4 10, 46 United Nations Department of Economic and Social Affairs, Population Division 2005 World contraceptive use 2005. New York; http://www.unpopulation.org Fu H, Darroch JE, Haas T, Ranjit N 1999 Contraceptive failure rates: new estimates from the 1995 National Survey of Family Growth. Fam Plann Perspect 31:56 63 Trussell J 2004 Contraceptive failure in the United States. Contraception 70:89 96 Mosher WD, Pratt WF 1993 AIDS-related behavior among women 15 44 years of age: United States, 1988 and 1990. Adv Data 239:115 Grady WR, Klepinger DH, Billy JO, Tanfer K 1993 Condom characteristics: the perceptions and preferences of men in the United States. Fam Plann Perspect 25:6773 Gallo MF, Grimes DA, Schulz KF 2003 Nonlatex vs. latex male condoms for contraception: a systematic review of randomized controlled trials. Contraception 68:319 326 Macaluso M, Kelaghan J, Artz L, Austin H, Fleenor M, Hook 3rd EW, Valappil T 1999 Mechanical failure of the latex condom in a cohort of women at high STD risk. Sex Transm Dis 26:450 458 Potter WD, de Villemeur M 2003 Clinical breakage, slippage and acceptability of a new commercial polyurethane condom: a randomized, controlled study. Contraception 68:39 45 Steiner MJ, Dominik R, Rountree RW, Nanda K, Dorflinger LJ 2003 Contraceptive effectiveness of a polyurethane condom and a latex condom: a randomized controlled trial. Obstet Gynecol 101: 539 547 Walsh TL, Frezieres RG, Peacock K, Nelson AL, Clark VA, Bernstein L 2003 Evaluation of the efficacy of a nonlatex condom: results from a randomized, controlled clinical trial. Perspect Sex Reprod Health 35:79 86 Levy DA, Khouader S, Leynadier F 1998 Allergy to latex condoms. Allergy 53:11071108 Turjanmaa K 1994 Allergy to natural rubber latex: a growing problem. Ann Med 26:297300 Frezieres RG, Walsh TL, Nelson AL, Clark VA, Coulson AH 1999 Evaluation of the efficacy of a polyurethane condom: results from a randomized, controlled clinical trial. Fam Plann Perspect 31: 81 87 Magnani RJ, Haws JM, Morgan GT, Gargiullo PM, Pollack AE, Koonin LM 1999 Vasectomy in the United States, 1991 and 1995. Am J Public Health 89:9294 Peterson HB, Curtis KM 2005 Clinical practice. Long-acting methods of contraception. N Engl J Med 353:2169 2175

Page et al. Advances in Male Contraception 36. Jamieson DJ, Costello C, Trussell J, Hillis SD, Marchbanks PA, Peterson HB 2004 The risk of pregnancy after vasectomy. Obstet Gynecol 103:848 850 37. Philp T, Guillebaud J, Budd D 1984 Complications of vasectomy: review of 16,000 patients. Br J Urol 56:745748 38. Schmidt SS 1995 Vasectomy by section, luminal fulguration and fascial interposition: results from 6248 cases. Br J Urol 76:373374; discussion 375 39. Li SQ, Goldstein M, Zhu J, Huber D 1991 The no-scalpel vasectomy. J Urol 145:341344 40. Dassow P, Bennett JM 2006 Vasectomy: an update. Am Fam Physician 74:2069 2074 41. Kumar V, Kaza RM, Singh I, Singhal S, Kumaran V 1999 An evaluation of the no-scalpel vasectomy technique. BJU Int 83:283 284 42. Nirapathpongporn A, Huber DH, Krieger JN 1990 No-scalpel vasectomy at the Kings birthday vasectomy festival. Lancet 335: 894 895 43. Awsare NS, Krishnan J, Boustead GB, Hanbury DC, McNicholas TA 2005 Complications of vasectomy. Ann R Coll Surg Engl 87: 406 410 44. Morris C, Mishra K, Kirkman RJ 2002 A study to assess the prevalence of chronic testicular pain in post-vasectomy men compared to non-vasectomised men. J Fam Plann Reprod Health Care 28:142144 45. Schwingl PJ, Guess HA 2000 Safety and effectiveness of vasectomy. Fertil Steril 73:923936 46. Myers SA, Mershon CE, Fuchs EF 1997 Vasectomy reversal for treatment of the post-vasectomy pain syndrome. J Urol 157:518 520 47. Jequier AM 1998 Vasectomy related infertility: a major and costly medical problem. Hum Reprod 13:17571759 48. Potts JM, Pasqualotto FF, Nelson D, Thomas Jr AJ, Agarwal A 1999 Patient characteristics associated with vasectomy reversal. J Urol 161:18351839 49. Belker AM, Thomas Jr AJ, Fuchs EF, Konnak JW, Sharlip ID 1991 Results of 1,469 microsurgical vasectomy reversals by the Vasovasostomy Study Group. J Urol 145:505511 50. Heidenreich A, Bonfig R, Wilbert DM, Strohmaier WL, Engelmann UH 1994 Risk factors for antisperm antibodies in infertile men. Am J Reprod Immunol 31:69 76 51. Kay DJ, Clifton V, Taylor JS, Boettcher B 1993 Anti-sperm antibodies and semen profiles in re-anastomosed men. Reprod Fertil Dev 5:135139 52. Meinertz H, Linnet L, Fogh-Andersen P, Hjort T 1990 Antisperm antibodies and fertility after vasovasostomy: a follow-up study of 216 men. Fertil Steril 54:315321 53. Giovannucci E, Tosteson TD, Speizer FE, Vessey MP, Colditz GA 1992 A long-term study of mortality in men who have undergone vasectomy. N Engl J Med 326:13921398 54. Giovannucci E, Ascherio A, Rimm EB, Colditz GA, Stampfer MJ, Willett WC 1993 A prospective cohort study of vasectomy and prostate cancer in US men. JAMA 269:873 877 55. Bernal-Delgado E, Latour-Perez J, Pradas-Arnal F, Gomez-Lopez LI 1998 The association between vasectomy and prostate cancer: a systematic review of the literature. Fertil Steril 70:191200 56. Cox B, Sneyd MJ, Paul C, Delahunt B, Skegg DC 2002 Vasectomy and risk of prostate cancer. JAMA 287:3110 3115 57. Harley VR, Clarkson MJ, Argentaro A 2003 The molecular action and regulation of the testis-determining factors, SRY (sex-determining region on the Y chromosome) and SOX9 [SRY-related highmobility group (HMG) box 9]. Endocr Rev 24:466 487 58. Belchetz PE, Plant TM, Nakai Y, Keogh EJ, Knobil E 1978 Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin-releasing hormone. Science 202:631 633 59. Santen RJ, Bardin CW 1973 Episodic luteinizing hormone secretion in man. Pulse analysis, clinical interpretation, physiologic mechanisms. J Clin Invest 52:26172628 60. Veldhuis JD, King JC, Urban RJ, Rogol AD, Evans WS, Kolp LA, Johnson ML 1987 Operating characteristics of the male hypothalamo-pituitary-gonadal axis: pulsatile release of testosterone and follicle-stimulating hormone and their temporal coupling with luteinizing hormone. J Clin Endocrinol Metab 65:929 941

13. 14. 15.

16. 17.

18.

19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29.

30.

31. 32. 33.

34. 35.

Page et al. Advances in Male Contraception 61. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E 2003 Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci USA 100:1097210976 62. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno Jr JS, Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB, Slaugenhaupt SA, Gusella JF, ORahilly S, Carlton MB, Crowley Jr WF, Aparicio SA, Colledge WH 2003 The GPR54 gene as a regulator of puberty. N Engl J Med 349:1614 1627 63. Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E, Brezillon S, Tyldesley R, Suarez-Huerta N, Vandeput F, Blanpain C, Schiffmann SN, Vassart G, Parmentier M 2001 The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem 276:3463134636 64. Muir AI, Chamberlain L, Elshourbagy NA, Michalovich D, Moore DJ, Calamari A, Szekeres PG, Sarau HM, Chambers JK, Murdock P, Steplewski K, Shabon U, Miller JE, Middleton SE, Darker JG, Larminie CG, Wilson S, Bergsma DJ, Emson P, Faull R, Philpott KL, Harrison DC 2001 AXOR12, a novel human G protein-coupled receptor, activated by the peptide KiSS-1. J Biol Chem 276:28969 28975 65. Ohtaki N, Yamaguchi A, Goi T, Fukaya T, Takeuchi K, Katayama K, Hirose K, Urano T 2001 Somatic alterations of the DPC4 and Madr2 genes in colorectal cancers and relationship to metastasis. Int J Oncol 18:265270 66. Dungan HM, Clifton DK, Steiner RA 2006 Minireview: kisspeptin neurons as central processors in the regulation of gonadotropinreleasing hormone secretion. Endocrinology 147:1154 1158 67. Navarro VM, Castellano JM, Fernandez-Fernandez R, Barreiro ML, Roa J, Sanchez-Criado JE, Aguilar E, Dieguez C, Pinilla L, Tena-Sempere M 2004 Developmental and hormonally regulated messenger ribonucleic acid expression of KiSS-1 and its putative receptor, GPR54, in rat hypothalamus and potent luteinizing hormone-releasing activity of KiSS-1 peptide. Endocrinology 145:4565 4574 68. Smith JT, Cunningham MJ, Rissman EF, Clifton DK, Steiner RA 2005 Regulation of Kiss1 gene expression in the brain of the female mouse. Endocrinology 146:3686 3692 69. Seminara SB, Dipietro MJ, Ramaswamy S, Crowley Jr WF, Plant TM 2006 Continuous human metastin 4554 infusion desensitizes G protein-coupled receptor 54-induced gonadotropin-releasing hormone release monitored indirectly in the juvenile male Rhesus monkey (Macaca mulatta): a finding with therapeutic implications. Endocrinology 147:21222126 70. Thompson EL, Murphy KG, Patterson M, Bewick GA, Stamp GW, Curtis AE, Cooke JH, Jethwa PH, Todd JF, Ghatei MA, Bloom SR 2006 Chronic subcutaneous administration of kisspeptin-54 causes testicular degeneration in adult male rats. Am J Physiol Endocrinol Metab 291:E1074 E1082 71. Matsumoto AM, Bremner WJ 1984 Modulation of pulsatile gonadotropin secretion by testosterone in man. J Clin Endocrinol Metab 58:609 614 72. Santen RJ 1975 Is aromatization of testosterone to estradiol required for inhibition of luteinizing hormone secretion in men? J Clin Invest 56:15551563 73. Urban RJ, Davis MR, Rogol AD, Johnson ML, Veldhuis JD 1988 Acute androgen receptor blockade increases luteinizing hormone secretory activity in men. J Clin Endocrinol Metab 67:1149 1155 74. Winters SJ, Sherins RJ, Loriaux DL 1979 Studies on the role of sex steroids in the feedback control of gonadotropin concentrations in men. III. Androgen resistance in primary gonadal failure. J Clin Endocrinol Metab 48:553558 75. Kerrigan JR, Veldhuis JD, Rogol AD 1994 Androgen-receptor blockade enhances pulsatile luteinizing hormone production in late pubertal males: evidence for a hypothalamic site of physiologic androgen feedback action. Pediatr Res 35:102106 76. Knuth UA, Hano R, Nieschlag E 1984 Effect of flutamide or cyproterone acetate on pituitary and testicular hormones in normal men. J Clin Endocrinol Metab 59:963969 77. Veldhuis JD, Urban RJ, Dufau ML 1992 Evidence that androgen negative feedback regulates hypothalamic gonadotropin-releasing

Endocrine Reviews, June 2008, 29(4):465 493 485 hormone impulse strength and the burst-like secretion of biologically active luteinizing hormone in men. J Clin Endocrinol Metab 74:12271235 Vermeulen A, Deslypere JP 1985 Long-term transdermal dihydrotestosterone therapy: effects on pituitary gonadal axis and plasma lipoproteins. Maturitas 7:281287 Finkelstein JS, ODea LS, Whitcomb RW, Crowley Jr WF 1991 Sex steroid control of gonadotropin secretion in the human male. II. Effects of estradiol administration in normal and gonadotropinreleasing hormone-deficient men. J Clin Endocrinol Metab 73:621 628 Hayes FJ, Seminara SB, Decruz S, Boepple PA, Crowley Jr WF 2000 Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. J Clin Endocrinol Metab 85:3027 3035 Raven G, de Jong FH, Kaufman JM, de Ronde W 2006 In men, peripheral estradiol levels directly reflect the action of estrogens at the hypothalamo-pituitary level to inhibit gonadotropin secretion. J Clin Endocrinol Metab 91:3324 3328 Rochira V, Zirilli L, Genazzani AD, Balestrieri A, Aranda C, Fabre B, Antunez P, Diazzi C, Carani C, Maffei L 2006 Hypothalamicpituitary-gonadal axis in two men with aromatase deficiency: evidence that circulating estrogens are required at the hypothalamic level for the integrity of gonadotropin negative feedback. Eur J Endocrinol 155:513522 Bagatell CJ, Dahl KD, Bremner WJ 1994 The direct pituitary effect of testosterone to inhibit gonadotropin secretion in men is partially mediated by aromatization to estradiol. J Androl 15:1521 Hayes FJ, DeCruz S, Seminara SB, Boepple PA, Crowley Jr WF 2001 Differential regulation of gonadotropin secretion by testosterone in the human male: absence of a negative feedback effect of testosterone on follicle-stimulating hormone secretion. J Clin Endocrinol Metab 86:5358 Schnorr JA, Bray MJ, Veldhuis JD 2001 Aromatization mediates testosterones short-term feedback restraint of 24-hour endogenously driven and acute exogenous gonadotropin-releasing hormone-stimulated luteinizing hormone and follicle-stimulating hormone secretion in young men. J Clin Endocrinol Metab 86:2600 2606 Wang C, Iranmanesh A, Berman N, McDonald V, Steiner B, Ziel F, Faulkner SM, Dudley RE, Veldhuis JD, Swerdloff RS 1998 Comparative pharmacokinetics of three doses of percutaneous dihydrotestosterone gel in healthy elderly mena clinical research center study. J Clin Endocrinol Metab 83:2749 2757 Amory JK, Wang C, Swerdloff RS, Anawalt BD, Matsumoto AM, Bremner WJ, Walker SE, Haberer LJ, Clark RV 2007 The effect of 5-reductase inhibition with dutasteride and finasteride on semen parameters and serum hormones in healthy men. J Clin Endocrinol Metab 92:1659 1665 Kunelius P, Lukkarinen O, Hannuksela ML, Itkonen O, Tapanainen JS 2002 The effects of transdermal dihydrotestosterone in the aging male: a prospective, randomized, double-blind study. J Clin Endocrinol Metab 87:14671472 Ly LP, Jimenez M, Zhuang TN, Celermajer DS, Conway AJ, Handelsman DJ 2001 A double-blind, placebo-controlled, randomized clinical trial of transdermal dihydrotestosterone gel on muscular strength, mobility, and quality of life in older men with partial androgen deficiency. J Clin Endocrinol Metab 86:4078 4088 Belanger A, Candas B, Dupont A, Cusan L, Diamond P, Gomez JL, Labrie F 1994 Changes in serum concentrations of conjugated and unconjugated steroids in 40- to 80-year-old men. J Clin Endocrinol Metab 79:1086 1090 Luetjens CM, Didolkar A, Kliesch S, Paulus W, Jeibmann A, Bocker W, Nieschlag E, Simoni M 2006 Tissue expression of the nuclear progesterone receptor in male non-human primates and men. J Endocrinol 189:529 539 Heikinheimo O, Mahony MC, Gordon K, Hsiu JG, Hodgen GD, Gibbons WE 1995 Estrogen and progesterone receptor mRNA are expressed in distinct pattern in male primate reproductive organs. J Assist Reprod Genet 12:198 204 Bethea CL, Fahrenbach WH, Sprangers SA, Freesh F 1992 Immunocytochemical localization of progestin receptors in monkey hy-

78. 79.

80.

81.

82.

83. 84.

85.

86.

87.

88.

89.

90.

91.

92.

93.

486

Endocrine Reviews, June 2008, 29(4):465 493 pothalamus: effect of estrogen and progestin. Endocrinology 130: 895905 Schneider JS, Burgess C, Sleiter NC, DonCarlos LL, Lydon JP, OMalley B, Levine JE 2005 Enhanced sexual behaviors and androgen receptor immunoreactivity in the male progesterone receptor knockout mouse. Endocrinology 146:4340 4348 Meriggiola MC, Farley TM, Mbizvo MT 2003 A review of androgen-progestin regimens for male contraception. J Androl 24:466 483 Brady BM, Anderson RA, Kinniburgh D, Baird DT 2003 Demonstration of progesterone receptor-mediated gonadotrophin suppression in the human male. Clin Endocrinol (Oxf) 58:506 512 McLachlan RI, Robertson DM, Pruysers E, Ugoni A, Matsumoto AM, Anawalt BD, Bremner WJ, Meriggiola C 2004 Relationship between serum gonadotropins and spermatogenic suppression in men undergoing steroidal contraceptive treatment. J Clin Endocrinol Metab 89:142149 Shah C, Modi D, Sachdeva G, Gadkar S, Puri C 2005 Coexistence of intracellular and membrane-bound progesterone receptors in human testis. J Clin Endocrinol Metab 90:474 483 Walton MJ, Bayne RA, Wallace I, Baird DT, Anderson RA 2006 Direct effect of progestogen on gene expression in the testis during gonadotropin withdrawal and early suppression of spermatogenesis. J Clin Endocrinol Metab 91:2526 2533 OConnor AE, De Kretser DM 2004 Inhibins in normal male physiology. Semin Reprod Med 22:177185 Ramaswamy S, Marshall GR, McNeilly AS, Plant TM 1999 Evidence that in a physiological setting Sertoli cell number is the major determinant of circulating concentrations of inhibin B in the adult male rhesus monkey (Macaca mulatta). J Androl 20:430 434 Soriano-Guillen L, Mitchell V, Carel JC, Barbet P, Roger M, Lahlou N 2006 Activating mutations in the luteinizing hormone receptor gene: a human model of non-follicle-stimulating hormone-dependent inhibin production and germ cell maturation. J Clin Endocrinol Metab 91:30413047 Anawalt BD, Bebb RA, Matsumoto AM, Groome NP, Illingworth PJ, McNeilly AS, Bremner WJ 1996 Serum inhibin B levels reflect Sertoli cell function in normal men and men with testicular dysfunction. J Clin Endocrinol Metab 81:33413345 Matthiesson KL, Robertson DM, Burger HG, McLachlan RI 2003 Response of serum inhibin B and pro-C levels to gonadotrophic stimulation in normal men before and after steroidal contraceptive treatment. Hum Reprod 18:734 743 Phillip M, Arbelle JE, Segev Y, Parvari R 1998 Male hypogonadism due to a mutation in the gene for the -subunit of folliclestimulating hormone. N Engl J Med 338:1729 1732 Tapanainen JS, Aittomaki K, Min J, Vaskivuo T, Huhtaniemi IT 1997 Men homozygous for an inactivating mutation of the folliclestimulating hormone (FSH) receptor gene present variable suppression of spermatogenesis and fertility. Nat Genet 15:205206 Vaskivuo TE, Aittomaki K, Anttonen M, Ruokonen A, Herva R, Osawa Y, Heikinheimo M, Huhtaniemi I, Tapanainen JS 2002 Effects of follicle-stimulating hormone (FSH) and human chorionic gonadotropin in individuals with an inactivating mutation of the FSH receptor. Fertil Steril 78:108 113 Anderson RA, Wallace EM, Groome NP, Bellis AJ, Wu FC 1997 Physiological relationships between inhibin B, follicle stimulating hormone secretion and spermatogenesis in normal men and response to gonadotrophin suppression by exogenous testosterone. Hum Reprod 12:746 751 Hayes FJ, Pitteloud N, DeCruz S, Crowley Jr WF, Boepple PA 2001 Importance of inhibin B in the regulation of FSH secretion in the human male. J Clin Endocrinol Metab 86:55415546 Illingworth PJ, Groome NP, Byrd W, Rainey WE, McNeilly AS, Mather JP, Bremner WJ 1996 Inhibin-B: a likely candidate for the physiologically important form of inhibin in men. J Clin Endocrinol Metab 81:13211325 Kumanov P, Nandipati K, Tomova A, Agarwal A 2006 Inhibin B is a better marker of spermatogenesis than other hormones in the evaluation of male factor infertility. Fertil Steril 86:332338 McCullagh D 1932 Dual endocrine activity of the testes. Science 76:19 20 Zhang FP, Pakarainen T, Poutanen M, Toppari J, Huhtaniemi I

Page et al. Advances in Male Contraception 2003 The low gonadotropin-independent constitutive production of testicular testosterone is sufficient to maintain spermatogenesis. Proc Natl Acad Sci USA 100:1369213697 Zhang FP, Pakarainen T, Zhu F, Poutanen M, Huhtaniemi I 2004 Molecular characterization of postnatal development of testicular steroidogenesis in luteinizing hormone receptor knockout mice. Endocrinology 145:14531463 Turner TT, Ewing LL, Jones CE, Howards SS, Zegeye B 1985 Androgens in various fluid compartments of the rat testis and epididymis after hypophysectomy and gonadotropin supplementation. J Androl 6:353358 Turner TT, Ewing LL, Jones CE, Howards SS, Zegeye B 1985 Androgens in male rat reproductive tract fluids: hypophysectomy and steroid replacement. Am J Physiol 248:E274 E280 Anderson RA, Wallace AM, Wu FC 1996 Comparison between testosterone enanthate-induced azoospermia and oligozoospermia in a male contraceptive study. III. Higher 5 -reductase activity in oligozoospermic men administered supraphysiological doses of testosterone. J Clin Endocrinol Metab 81:902908 Horton R 1978 Sex steroid production and secretion in the male. Andrologia 10:183194 Vierhapper H, Nowotny P, Waldhausl W 1997 Determination of testosterone production rates in men and women using stable isotope/dilution and mass spectrometry. J Clin Endocrinol Metab 82:14921496 Leymarie P, Roger M, Castanier M, Scholler R 1974 Circadian variations of plasma testosterone and estrogens in normal men. A study by frequent sampling. J Steroid Biochem 5:167171 Piro C, Fraioli F, Sciarra F, Conti C 1973 Circadian rhythm of plasma testosterone, cortisol and gonadotropins in normal male subjects. J Steroid Biochem 4:321329 Wang C, Catlin DH, Starcevic B, Leung A, DiStefano E, Lucas G, Hull L, Swerdloff RS 2004 Testosterone metabolic clearance and production rates determined by stable isotope dilution/tandem mass spectrometry in normal men: influence of ethnicity and age. J Clin Endocrinol Metab 89:2936 2941 Bremner WJ, Vitiello MV, Prinz PN 1983 Loss of circadian rhythmicity in blood testosterone levels with aging in normal men. J Clin Endocrinol Metab 56:1278 1281 Liu PY, Takahashi PY, Roebuck PD, Iranmanesh A, Veldhuis JD 2005 Age-specific changes in the regulation of LH-dependent testosterone secretion: assessing responsiveness to varying endogenous gonadotropin output in normal men. Am J Physiol Regul Integr Comp Physiol 289:R721R728 Tenover JS, Matsumoto AM, Clifton DK, Bremner WJ 1988 Agerelated alterations in the circadian rhythms of pulsatile luteinizing hormone and testosterone secretion in healthy men. J Gerontol 43:M163M169 Veldhuis JD, Veldhuis NJ, Keenan DM, Iranmanesh A 2005 Age diminishes the testicular steroidogenic response to repeated intravenous pulses of recombinant human LH during acute GnRHreceptor blockade in healthy men. Am J Physiol Endocrinol Metab 288:E775E781 Carani C, Qin K, Simoni M, Faustini-Fustini M, Serpente S, Boyd J, Korach KS, Simpson ER 1997 Effect of testosterone and estradiol in a man with aromatase deficiency. N Engl J Med 337:9195 Maffei L, Murata Y, Rochira V, Tubert G, Aranda C, Vazquez M, Clyne CD, Davis S, Simpson ER, Carani C 2004 Dysmetabolic syndrome in a man with a novel mutation of the aromatase gene: effects of testosterone, alendronate, and estradiol treatment. J Clin Endocrinol Metab 89:6170 Aschim EL, Giwercman A, Stahl O, Eberhard J, Cwikiel M, Nordenskjold A, Haugen TB, Grotmol T, Giwercman YL 2005 The RsaI polymorphism in the estrogen receptor- gene is associated with male infertility. J Clin Endocrinol Metab 90:53435348 Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS 1994 Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med 331:1056 1061 Aschim EL, Saether T, Wiger R, Grotmol T, Haugen TB 2004 Differential distribution of splice variants of estrogen receptor in human testicular cells suggests specific functions in spermatogenesis. J Steroid Biochem Mol Biol 92:97106

94.

114.

95. 96. 97.

115.

116. 117.

98. 99.

118. 119.

100. 101.

120. 121. 122.

102.

103.

123. 124.

104.

105. 106.

125.

126.

107.

127. 128.

108.

109. 110.

129.

130.

111. 112. 113.

131.

Page et al. Advances in Male Contraception 132. Lambard S, Galeraud-Denis I, Saunders PT, Carreau S 2004 Human immature germ cells and ejaculated spermatozoa contain aromatase and oestrogen receptors. J Mol Endocrinol 32:279 289 133. Shapiro E, Huang H, Masch RJ, McFadden DE, Wu XR, Ostrer H 2005 Immunolocalization of androgen receptor and estrogen receptors and in human fetal testis and epididymis. J Urol 174: 16951698; discussion 1698 134. Coviello AD, Bremner WJ, Matsumoto AM, Herbst KL, Amory JK, Anawalt BD, Yan X, Brown TR, Wright WW, Zirkin BR, Jarow JP 2004 Intratesticular testosterone concentrations comparable with serum levels are not sufficient to maintain normal sperm production in men receiving a hormonal contraceptive regimen. J Androl 25:931938 135. Coviello AD, Matsumoto AM, Bremner WJ, Herbst KL, Amory JK, Anawalt BD, Sutton PR, Wright WW, Brown TR, Yan X, Zirkin BR, Jarow JP 2005 Low-dose human chorionic gonadotropin maintains intratesticular testosterone in normal men with testosterone-induced gonadotropin suppression. J Clin Endocrinol Metab 90:25952602 136. Heller CG, Rowley MJ, Heller GV 1969 Clomiphene citrate: a correlation of its effect on sperm concentration and morphology, total gonadotropins, ICSH, estrogen and testosterone excretion, and testicular cytology in normal men. J Clin Endocrinol Metab 29:638 649 137. Huhtaniemi I, Nikula H, Rannikko S 1987 Pituitary-testicular function of prostatic cancer patients during treatment with a gonadotropin-releasing hormone agonist analog. I. Circulating hormone levels. J Androl 8:355362 138. McLachlan RI, ODonnell L, Stanton PG, Balourdos G, Frydenberg M, de Kretser DM, Robertson DM 2002 Effects of testosterone plus medroxyprogesterone acetate on semen quality, reproductive hormones, and germ cell populations in normal young men. J Clin Endocrinol Metab 87:546 556 139. Morse HC, Horike N, Rowley MJ, Heller CG 1973 Testosterone concentrations in testes of normal men: effects of testosterone propionate administration. J Clin Endocrinol Metab 37:882 886 140. Matthiesson KL, Stanton PG, ODonnell L, Meachem SJ, Amory JK, Berger R, Bremner WJ, McLachlan RI 2005 Effects of testosterone and levonorgestrel combined with a 5-reductase inhibitor or gonadotropin-releasing hormone antagonist on spermatogenesis and intratesticular steroid levels in normal men. J Clin Endocrinol Metab 90:56475655 141. 1990 Contraceptive efficacy of testosterone-induced azoospermia in normal men. World Health Organization Task Force on methods for the regulation of male fertility. 142. World Health Organization 1996 Contraceptive efficacy of testosterone-induced azoospermia and oligospermia in normal men. Fertil Steril 65:821 829 143. Meriggiola MC, Costantino A, Bremner WJ, Morselli-Labate AM 2002 Higher testosterone dose impairs sperm suppression induced by a combined androgen-progestin regimen. J Androl 23:684 690 144. Amory JK, Page ST, Bremner WJ 2006 Recent advances in male hormonal contraception. Nat Clin Pract Endocrinol Metab 2:32 41 145. Jarow JP, Zirkin BR 2005 The androgen microenvironment of the human testis and hormonal control of spermatogenesis. Ann NY Acad Sci 1061:208 220 146. Page ST, Kalhorn TF, Bremner WJ, Anawalt BD, Matsumoto AM, Amory JK 2007 Intratesticular androgens and spermatogenesis during severe gonadotropin suppression induced by male hormonal contraceptive treatment. J Androl 28:734 41 147. Zirkin BR, Santulli R, Awoniyi CA, Ewing LL 1989 Maintenance of advanced spermatogenic cells in the adult rat testis: quantitative relationship to testosterone concentration within the testis. Endocrinology 124:30433049 148. Kinniburgh D, Anderson RA, Baird DT 2001 Suppression of spermatogenesis with desogestrel and testosterone pellets is not enhanced by addition of finasteride. J Androl 22:88 95 149. McLachlan RI, McDonald J, Rushford D, Robertson DM, Garrett C, Baker HW 2000 Efficacy and acceptability of testosterone implants, alone or in combination with a 5-reductase inhibitor, for male hormonal contraception. Contraception 62:7378 150. Heinlein CA, Chang C 2004 Androgen receptor in prostate cancer. Endocr Rev 25:276 308

Endocrine Reviews, June 2008, 29(4):465 493 487 151. Collins LL, Lee HJ, Chen YT, Chang M, Hsu HY, Yeh S, Chang C 2003 The androgen receptor in spermatogenesis. Cytogenet Genome Res 103:299 301 152. Yeh S, Tsai MY, Xu Q, Mu XM, Lardy H, Huang KE, Lin H, Yeh SD, Altuwaijri S, Zhou X, Xing L, Boyce BF, Hung MC, Zhang S, Gan L, Chang C 2002 Generation and characterization of androgen receptor knockout (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci USA 99:13498 13503 153. Tsai MY, Yeh SD, Wang RS, Yeh S, Zhang C, Lin HY, Tzeng CR, Chang C 2006 Differential effects of spermatogenesis and fertility in mice lacking androgen receptor in individual testis cells. Proc Natl Acad Sci USA 103:1897518980 154. Zhang C, Yeh S, Chen YT, Wu CC, Chuang KH, Lin HY, Wang RS, Chang YJ, Mendis-Handagama C, Hu L, Lardy H, Chang C 2006 Oligozoospermia with normal fertility in male mice lacking the androgen receptor in testis peritubular myoid cells. Proc Natl Acad Sci USA 103:17718 17723 155. Chang C, Chen YT, Yeh SD, Xu Q, Wang RS, Guillou F, Lardy H, Yeh S 2004 Infertility with defective spermatogenesis and hypotestosteronemia in male mice lacking the androgen receptor in Sertoli cells. Proc Natl Acad Sci USA 101:6876 6881 156. De Gendt K, Swinnen JV, Saunders PT, Schoonjans L, Dewerchin M, Devos A, Tan K, Atanassova N, Claessens F, Lecureuil C, Heyns W, Carmeliet P, Guillou F, Sharpe RM, Verhoeven G 2004 A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc Natl Acad Sci USA 101:13271332 157. Holdcraft RW, Braun RE 2004 Androgen receptor function is required in Sertoli cells for the terminal differentiation of haploid spermatids. Development 131:459 467 158. Denolet E, De Gendt K, Allemeersch J, Engelen K, Marchal K, Van Hummelen P, Tan KA, Sharpe RM, Saunders PT, Swinnen JV, Verhoeven G 2006 The effect of a Sertoli cell-selective knockout of the androgen receptor on testicular gene expression in prepubertal mice. Mol Endocrinol 20:321334 159. Eacker SM, Shima JE, Connolly CM, Sharma M, Holdcraft RW, Griswold MD, Braun RE 2007 Transcriptional profiling of androgen receptor (AR) mutants suggests instructive and permissive roles of AR signaling in germ cell development. Mol Endocrinol 21:895907 160. Wang RS, Yeh S, Chen LM, Lin HY, Zhang C, Ni J, Wu CC, di SantAgnese PA, deMesy-Bentley KL, Tzeng CR, Chang C 2006 Androgen receptor in Sertoli cell is essential for germ cell nursery and junctional complex formation in mouse testes. Endocrinology 147:5624 5633 161. ODonnell L, Pratis K, Stanton PG, Robertson DM, McLachlan RI 1999 Testosterone-dependent restoration of spermatogenesis in adult rats is impaired by a 5-reductase inhibitor. J Androl 20: 109 117 162. Kousteni S, Bellido T, Plotkin LI, OBrien CA, Bodenner DL, Han L, Han K, DiGregorio GB, Katzenellenbogen JA, Katzenellenbogen BS, Roberson PK, Weinstein RS, Jilka RL, Manolagas SC 2001 Nongenotropic, sex-nonspecific signaling through the estrogen or androgen receptors: dissociation from transcriptional activity. Cell 104:719 730 163. Kousteni S, Chen JR, Bellido T, Han L, Ali AA, OBrien CA, Plotkin L, Fu Q, Mancino AT, Wen Y, Vertino AM, Powers CC, Stewart SA, Ebert R, Parfitt AM, Weinstein RS, Jilka RL, Manolagas SC 2002 Reversal of bone loss in mice by nongenotropic signaling of sex steroids. Science 298:843 846 164. Kousteni S, Han L, Chen JR, Almeida M, Plotkin LI, Bellido T, Manolagas SC 2003 Kinase-mediated regulation of common transcription factors accounts for the bone-protective effects of sex steroids. J Clin Invest 111:16511664 165. Moggs JG, Deavall DG, Orphanides G 2003 Sex steroids, ANGELS and osteoporosis. Bioessays 25:195199 166. Windahl SH, Galien R, Chiusaroli R, Clement-Lacroix P, Morvan F, Lepescheux L, Nique F, Horne WC, Resche-Rigon M, Baron R 2006 Bone protection by estrens occurs through non-tissue-selective activation of the androgen receptor. J Clin Invest 116:2500 2509 167. Choong CS, Wilson EM 1998 Trinucleotide repeats in the human

488

Endocrine Reviews, June 2008, 29(4):465 493 androgen receptor: a molecular basis for disease. J Mol Endocrinol 21:235257 Chamberlain NL, Driver ED, Miesfeld RL 1994 The length and location of CAG trinucleotide repeats in the androgen receptor N-terminal domain affect transactivation function. Nucleic Acids Res 22:31813186 Gerber HP, Seipel K, Georgiev O, Hofferer M, Hug M, Rusconi S, Schaffner W 1994 Transcriptional activation modulated by homopolymeric glutamine and proline stretches. Science 263:808 811 Dejager S, Bry-Gauillard H, Bruckert E, Eymard B, Salachas F, LeGuern E, Tardieu S, Chadarevian R, Giral P, Turpin G 2002 A comprehensive endocrine description of Kennedys disease revealing androgen insensitivity linked to CAG repeat length. J Clin Endocrinol Metab 87:38933901 Giovannucci E, Platz EA, Stampfer MJ, Chan A, Krithivas K, Kawachi I, Willett WC, Kantoff PW 1999 The CAG repeat within the androgen receptor gene and benign prostatic hyperplasia. Urology 53:121125 Zitzmann M, Depenbusch M, Gromoll J, Nieschlag E 2004 Xchromosome inactivation patterns and androgen receptor functionality influence phenotype and social characteristics as well as pharmacogenetics of testosterone therapy in Klinefelter patients. J Clin Endocrinol Metab 89:6208 6217 Dowsing AT, Yong EL, Clark M, McLachlan RI, de Kretser DM, Trounson AO 1999 Linkage between male infertility and trinucleotide repeat expansion in the androgen-receptor gene. Lancet 354: 640 643 Tut TG, Ghadessy FJ, Trifiro MA, Pinsky L, Yong EL 1997 Long polyglutamine tracts in the androgen receptor are associated with reduced trans-activation, impaired sperm production, and male infertility. J Clin Endocrinol Metab 82:37773782 Lund A, Tapanainen JS, Lahdetie J, Savontaus ML, Aittomaki K 2003 Long CAG repeats in the AR gene are not associated with infertility in Finnish males. Acta Obstet Gynecol Scand 82:162166 Van Golde R, Van Houwelingen K, Kiemeney L, Kremer J, Tuerlings J, Schalken J, Meuleman E 2002 Is increased CAG repeat length in the androgen receptor gene a risk factor for male subfertility? J Urol 167:621 623 Rajender S, Rajani V, Gupta NJ, Chakravarty B, Singh L, Thangaraj K 2006 No association of androgen receptor GGN repeat length polymorphism with infertility in Indian men. J Androl 27:785789 Ferlin A, Bartoloni L, Rizzo G, Roverato A, Garolla A, Foresta C 2004 Androgen receptor gene CAG and GGC repeat lengths in idiopathic male infertility. Mol Hum Reprod 10:417 421 Srinivas-Shankar U, Wu FC 2006 Drug insight: testosterone preparations. Nat Clin Pract Urol 3:653 665 Nieschlag E, Behre HM, Bouchard P, Corrales JJ, Jones TH, Stalla GK, Webb SM, Wu FC 2004 Testosterone replacement therapy: current trends and future directions. Hum Reprod Update 10:409 419 Qoubaitary A, Meriggiola C, Ng CM, Lumbreras L, Cerpolini S, Pelusi G, Christensen PD, Hull L, Swerdloff RS, Wang C 2006 Pharmacokinetics of testosterone undecanoate injected alone or in combination with norethisterone enanthate in healthy men. J Androl 27:853 867 Buchter D, von Eckardstein S, von Eckardstein A, Kamischke A, Simoni M, Behre HM, Nieschlag E 1999 Clinical trial of transdermal testosterone and oral levonorgestrel for male contraception. J Clin Endocrinol Metab 84:1244 1249 Gonzalo IT, Swerdloff RS, Nelson AL, Clevenger B, Garcia R, Berman N, Wang C 2002 Levonorgestrel implants (Norplant II) for male contraception clinical trials: combination with transdermal and injectable testosterone. J Clin Endocrinol Metab 87:35623572 Hair WM, Kitteridge K, OConnor DB, Wu FC 2001 A novel male contraceptive pill-patch combination: oral desogestrel and transdermal testosterone in the suppression of spermatogenesis in normal men. J Clin Endocrinol Metab 86:52015209 Page ST, Amory JK, Anawalt BD, Irwig MS, Brockenbrough AT, Matsumoto AM, Bremner WJ 2006 Testosterone gel combined with depomedroxyprogesterone acetate is an effective male hormonal contraceptive regimen and is not enhanced by the addition of a GnRH antagonist. J Clin Endocrinol Metab 91:4374 4380 Bhasin S, Woodhouse L, Casaburi R, Singh AB, Bhasin D, Ber-

Page et al. Advances in Male Contraception man N, Chen X, Yarasheski KE, Magliano L, Dzekov C, Dzekov J, Bross R, Phillips J, Sinha-Hikim I, Shen R, Storer TW 2001 Testosterone dose-response relationships in healthy young men. Am J Physiol Endocrinol Metab 281:E1172E1181 Meriggiola MC, Marcovina S, Paulsen CA, Bremner WJ 1995 Testosterone enanthate at a dose of 200 mg/week decreases HDLcholesterol levels in healthy men. Int J Androl 18:237242 Bhasin S, Storer TW, Berman N, Callegari C, Clevenger B, Phillips J, Bunnell TJ, Tricker R, Shirazi A, Casaburi R 1996 The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. N Engl J Med 335:17 Brinster RL 2007 Male germline stem cells: from mice to men. Science 316:404 405 Kubota H, Brinster RL 2006 Technology insight: in vitro culture of spermatogonial stem cells and their potential therapeutic uses. Nat Clin Pract Endocrinol Metab 2:99 108 de Kretser DM 2002 Male reproductive endocrinology. In: McLachlan R, ed. Endocrinology of the male reproductive system. South Dartmouth, MA: MDText.com, Inc. Bremner WJ, Matsumoto AM, Sussman AM, Paulsen CA 1981 Follicle-stimulating hormone and human spermatogenesis. J Clin Invest 68:1044 1052 Matsumoto AM, Bremner WJ 1985 Stimulation of sperm production by human chorionic gonadotropin after prolonged gonadotropin suppression in normal men. J Androl 6:137143 Matsumoto AM, Karpas AE, Bremner WJ 1986 Chronic human chorionic gonadotropin administration in normal men: evidence that follicle-stimulating hormone is necessary for the maintenance of quantitatively normal spermatogenesis in man. J Clin Endocrinol Metab 62:1184 1192 Matsumoto AM, Karpas AE, Paulsen CA, Bremner WJ 1983 Reinitiation of sperm production in gonadotropin-suppressed normal men by administration of follicle-stimulating hormone. J Clin Invest 72:10051015 Matthiesson KL, McLachlan RI, ODonnell L, Frydenberg M, Robertson DM, Stanton PG, Meachem SJ 2006 The relative roles of follicle-stimulating hormone and luteinizing hormone in maintaining spermatogonial maturation and spermiation in normal men. J Clin Endocrinol Metab 91:39623969 McLachlan RI, ODonnell L, Meachem SJ, Stanton PG, de Kretser DM, Pratis K, Robertson DM 2002 Identification of specific sites of hormonal regulation in spermatogenesis in rats, monkeys, and man. Recent Prog Horm Res 57:149 179 Meachem SJ, McLachlan RI, Stanton PG, Robertson DM, Wreford NG 1999 FSH immunoneutralization acutely impairs spermatogonial development in normal adult rats. J Androl 20:756 762; discussion 755 Moudgal NR, Sairam MR, Krishnamurthy HN, Sridhar S, Krishnamurthy H, Khan H 1997 Immunization of male bonnet monkeys (M. radiata) with a recombinant FSH receptor preparation affects testicular function and fertility. Endocrinology 138:30653068 Shetty J, Marathe GK, Dighe RR 1996 Specific immunoneutralization of FSH leads to apoptotic cell death of the pachytene spermatocytes and spermatogonial cells in the rat. Endocrinology 137: 2179 2182 Saito K, ODonnell L, McLachlan RI, Robertson DM 2000 Spermiation failure is a major contributor to early spermatogenic suppression caused by hormone withdrawal in adult rats. Endocrinology 141:2779 2785 Chapin RE, Wine RN, Harris MW, Borchers CH, Haseman JK 2001 Structure and control of a cell-cell adhesion complex associated with spermiation in rat seminiferous epithelium. J Androl 22:1030 1052 Yan HH, Mruk DD, Cheng CY 2007 Junction restructuring and spermatogenesis: the biology, regulation, and implication in male contraceptive development. Curr Top Dev Biol 80:5792 Cooper TG 2007 Sperm maturation in the epididymis: a new look at an old problem. Asian J Androl 9:533539 Chauvin TR, Griswold MD 2004 Androgen-regulated genes in the murine epididymis. Biol Reprod 71:560 569 Jelinsky SA, Turner TT, Bang HJ, Finger JN, Solarz MK, Wilson E, Brown EL, Kopf GS, Johnston DS 2007 The rat epididymal

168.

187. 188.

169. 170.

189. 190. 191. 192. 193. 194.

171.

172.

173.

174.

175. 176.

195.

196.

177. 178. 179. 180.

197.

198.

199.

181.

200.

182.

201.

183.

202.

184.

203. 204. 205. 206.

185.

186.

Page et al. Advances in Male Contraception transcriptome: comparison of segmental gene expression in the rat and mouse epididymides. Biol Reprod 76:561570 Johnston DS, Turner TT, Finger JN, Owtscharuk TL, Kopf GS, Jelinsky SA 2007 Identification of epididymis-specific transcripts in the mouse and rat by transcriptional profiling. Asian J Androl 9:522527 Yuan H, Liu A, Zhang L, Zhou H, Wang Y, Zhang H, Wang G, Zeng R, Zhang Y, Chen Z 2006 Proteomic profiling of regionalized proteins in rat epididymis indicates consistency between specialized distribution and protein functions. J Proteome Res 5:299 307 Suzuki K, Yu X, Chaurand P, Araki Y, Lareyre JJ, Caprioli RM, Matusik RJ, Orgebin-Crist MC 2006 Epididymis-specific promoter-driven gene targeting: a transcription factor which regulates epididymis-specific gene expression. Mol Cell Endocrinol 250:184 189 Yeung CH, Barfield JP, Anapolski M, Cooper TG 2004 Volume regulation of mature and immature spermatozoa in a primate model, and possible ion channels involved. Hum Reprod 19:2587 2593 Yeung CH, Barfield JP, Cooper TG 2006 Physiological volume regulation by spermatozoa. Mol Cell Endocrinol 250:98 105 Heckel M 1939 Production of oligospermia in a man by the use of testosterone propionate. Proc Soc Exp Biol Med 40:658 659 Handelsman DJ 2003 Editorial: hormonal male contraceptionlessons from the East when the Western market fails. J Clin Endocrinol Metab 88:559 561 Handelsman DJ, Farley TM, Peregoudov A, Waites GM 1995 Factors in nonuniform induction of azoospermia by testosterone enanthate in normal men. World Health Organization Task Force on Methods for the Regulation of Male Fertility. Fertil Steril 63: 125133 Walton MJ, Kumar N, Baird DT, Ludlow H, Anderson RA 2007 7-Methyl-19-nortestosterone (MENT) vs testosterone in combination with etonogestrel implants for spermatogenic suppression in healthy men. J Androl 28:679 688 Wang C, Wang XH, Nelson AL, Lee KK, Cui YG, Tong JS, Berman N, Lumbreras L, Leung A, Hull L, Desai S, Swerdloff RS 2006 Levonorgestrel implants enhanced the suppression of spermatogenesis by testosterone implants: comparison between Chinese and non-Chinese men. J Clin Endocrinol Metab 91:460 470 Gu YQ, Wang XH, Xu D, Peng L, Cheng LF, Huang MK, Huang ZJ, Zhang GY 2003 A multicenter contraceptive efficacy study of injectable testosterone undecanoate in healthy Chinese men. J Clin Endocrinol Metab 88:562568 Turner L, Conway AJ, Jimenez M, Liu PY, Forbes E, McLachlan RI, Handelsman DJ 2003 Contraceptive efficacy of a depot progestin and androgen combination in men. J Clin Endocrinol Metab 88:4659 4667 Aaltonen P, Amory JK, Anderson RA, Behre HM, Bialy G, Blithe D, Bone W, Bremner WJ, Colvard D, Cooper TG, Elliesen J, Gabelnick HL, Gu YQ, Handelsman DJ, Johansson EA, Kersemaekers W, Liu P, Mackay T, Matlin S, Mbizvo M, McLachlan RI, Meriggiola MC, Mletzko S, Mommers E, Muermans H, Nieschlag E, Odlind V, Page ST, Radlmaier A, Sitruk-Ware R, Swerdloff R, Wang C, Wu F, Zitzmann M 2007 10th Summit meeting consensus: recommendations for regulatory approval for hormonal male contraception. J Androl 28:362363 Bagatell CJ, Heiman JR, Matsumoto AM, Rivier JE, Bremner WJ 1994 Metabolic and behavioral effects of high-dose, exogenous testosterone in healthy men. J Clin Endocrinol Metab 79:561567 Zitzmann M, Junker R, Kamischke A, Nieschlag E 2002 Contraceptive steroids influence the hemostatic activation state in healthy men. J Androl 23:503511 Zitzmann M, Erren M, Kamischke A, Simoni M, Nieschlag E 2005 Endogenous progesterone and the exogenous progestin norethisterone enanthate are associated with a proinflammatory profile in healthy men. J Clin Endocrinol Metab 90:6603 6608 Sjogren B, Gottlieb C 2001 Testosterone for male contraception during one year: attitudes, well-being and quality of sex life. Contraception 64:59 65 Ringheim K 1995 Evidence for the acceptability of an injectable hormonal method for men. Fam Plann Perspect 27:123128 Nieschlag E, Mauss J, Coert A, Kicovic P 1975 Plasma androgen

Endocrine Reviews, June 2008, 29(4):465 493 489 levels in men after oral administration of testosterone or testosterone undecanoate. Acta Endocrinol (Copenh) 79:366 374 Zhang GY, Gu YQ, Wang XH, Cui YG, Bremner WJ 1998 A pharmacokinetic study of injectable testosterone undecanoate in hypogonadal men. J Androl 19:761768 Behre HM, Abshagen K, Oettel M, Hubler D, Nieschlag E 1999 Intramuscular injection of testosterone undecanoate for the treatment of male hypogonadism: phase I studies. Eur J Endocrinol 140:414 419 Meriggiola MC, Bremner WJ, Costantino A, Pavani A, Capelli M, Flamigni C 1997 An oral regimen of cyproterone acetate and testosterone undecanoate for spermatogenic suppression in men. Fertil Steril 68:844 850 Zhang GY, Gu YQ, Wang XH, Cui YG, Bremner WJ 1999 A clinical trial of injectable testosterone undecanoate as a potential male contraceptive in normal Chinese men. J Clin Endocrinol Metab 84:36423647 Kamischke A, Ploger D, Venherm S, von Eckardstein S, von Eckardstein A, Nieschlag E 2000 Intramuscular testosterone undecanoate with or without oral levonorgestrel: a randomized placebo-controlled feasibility study for male contraception. Clin Endocrinol (Oxf) 53:4352 Kamischke A, Venherm S, Ploger D, von Eckardstein S, Nieschlag E 2001 Intramuscular testosterone undecanoate and norethisterone enanthate in a clinical trial for male contraception. J Clin Endocrinol Metab 86:303309 Ahmed SR, Boucher AE, Manni A, Santen RJ, Bartholomew M, Demers LM 1988 Transdermal testosterone therapy in the treatment of male hypogonadism. J Clin Endocrinol Metab 66:546 551 Steidle C, Schwartz S, Jacoby K, Sebree T, Smith T, Bachand R 2003 AA2500 testosterone gel normalizes androgen levels in aging males with improvements in body composition and sexual function. J Clin Endocrinol Metab 88:26732681 Wang C, Berman N, Longstreth JA, Chuapoco B, Hull L, Steiner B, Faulkner S, Dudley RE, Swerdloff RS 2000 Pharmacokinetics of transdermal testosterone gel in hypogonadal men: application of gel at one site versus four sites: a General Clinical Research Center Study. J Clin Endocrinol Metab 85:964 969 Junaidi A, Luetjens CM, Wistuba J, Kamischke A, Yeung CH, Simoni M, Nieschlag E 2005 Norethisterone enanthate has neither a direct effect on the testis nor on the epididymis: a study in adult male cynomolgus monkeys (Macaca fascicularis). Eur J Endocrinol 152:655 661 Schearer SB 1978 Current efforts to develop male hormonal contraception. Stud Fam Plann 9:229 231 Barfield A, Melo J, Coutinho E, Alvarez-Sanchez F, Faundes A, Brache V, Leon P, Frick J, Bartsch G, Weiske WH, Brenner P, Mishell Jr D, Bernstein G, Ortiz A 1979 Pregnancies associated with sperm concentrations below 10 million/ml in clinical studies of a potential male contraceptive method, monthly depot medroxyprogesterone acetate and testosterone esters. Contraception 20:121 127 Bebb RA, Anawalt BD, Christensen RB, Paulsen CA, Bremner WJ, Matsumoto AM 1996 Combined administration of levonorgestrel and testosterone induces more rapid and effective suppression of spermatogenesis than testosterone alone: a promising male contraceptive approach. J Clin Endocrinol Metab 81:757762 Anawalt BD, Amory JK, Herbst KL, Coviello AD, Page ST, Bremner WJ, Matsumoto AM 2005 Intramuscular testosterone enanthate plus very low dosage oral levonorgestrel suppresses spermatogenesis without causing weight gain in normal young men: a randomized clinical trial. J Androl 26:405 413 Anawalt BD, Bebb RA, Bremner WJ, Matsumoto AM 1999 A lower dosage levonorgestrel and testosterone combination effectively suppresses spermatogenesis and circulating gonadotropin levels with fewer metabolic effects than higher dosage combinations. J Androl 20:407 414 Gui YL, He CH, Amory JK, Bremner WJ, Zheng EX, Yang J, Yang PJ, Gao ES 2004 Male hormonal contraception: suppression of spermatogenesis by injectable testosterone undecanoate alone or with levonorgestrel implants in Chinese men. J Androl 25:720 727 Anawalt BD, Herbst KL, Matsumoto AM, Mulders TM, Coelingh-Bennink HJ, Bremner WJ 2000 Desogestrel plus testosterone

207.

226. 227.

208.

228.

209.

229.

210.

230.

211. 212. 213. 214.

231.

232. 233.

215.

234.

216.

235.

217.

236. 237.

218.

219.

238.

220. 221. 222.

239.

240.

223. 224. 225.

241.

242.

490

Endocrine Reviews, June 2008, 29(4):465 493 effectively suppresses spermatogenesis but also causes modest weight gain and high-density lipoprotein suppression. Fertil Steril 74:707714 Wu FC, Balasubramanian R, Mulders TM, Coelingh-Bennink HJ 1999 Oral progestogen combined with testosterone as a potential male contraceptive: additive effects between desogestrel and testosterone enanthate in suppression of spermatogenesis, pituitarytesticular axis, and lipid metabolism. J Clin Endocrinol Metab 84: 112122 Anderson RA, Van Der Spuy ZM, Dada OA, Tregoning SK, Zinn PM, Adeniji OA, Fakoya TA, Smith KB, Baird DT 2002 Investigation of hormonal male contraception in African men: suppression of spermatogenesis by oral desogestrel with depot testosterone. Hum Reprod 17:2869 2877 Kinniburgh D, Zhu H, Cheng L, Kicman AT, Baird DT, Anderson RA 2002 Oral desogestrel with testosterone pellets induces consistent suppression of spermatogenesis to azoospermia in both Caucasian and Chinese men. Hum Reprod 17:1490 1501 Hay CJ, Brady BM, Zitzmann M, Osmanagaoglu K, Pollanen P, Apter D, Wu FC, Anderson RA, Nieschlag E, Devroey P, Huhtaniemi I, Kersemaekers WM 2005 A multicenter phase IIb study of a novel combination of intramuscular androgen (testosterone decanoate) and oral progestogen (etonogestrel) for male hormonal contraception. J Clin Endocrinol Metab 90:20422049 Anderson RA, Kinniburgh D, Baird DT 2002 Suppression of spermatogenesis by etonogestrel implants with depot testosterone: potential for long-acting male contraception. J Clin Endocrinol Metab 87:3640 3649 Brady BM, Amory JK, Perheentupa A, Zitzmann M, Hay CJ, Apter D, Anderson RA, Bremner WJ, Pollanen P, Nieschlag E, Wu FC, Kersemaekers WM 2006 A multicentre study investigating subcutaneous etonogestrel implants with injectable testosterone decanoate as a potential long-acting male contraceptive. Hum Reprod 21:285294 Neumann F, Topert M 1986 Pharmacology of antiandrogens. J Steroid Biochem 25:885 895 Meriggiola MC, Bremner WJ, Paulsen CA, Valdiserri A, Incorvaia L, Motta R, Pavani A, Capelli M, Flamigni C 1996 A combined regimen of cyproterone acetate and testosterone enanthate as a potentially highly effective male contraceptive. J Clin Endocrinol Metab 81:3018 3023 Meriggiola MC, Bremner WJ, Costantino A, Di Cintio G, Flamigni C 1998 Low dose of cyproterone acetate and testosterone enanthate for contraception in men. Hum Reprod 13:12251229 Meriggiola MC, Bremner WJ, Costantino A, Bertaccini A, Morselli-Labate AM, Huebler D, Kaufmann G, Oettel M, Flamigni C 2002 Twenty-one day administration of dienogest reversibly suppresses gonadotropins and testosterone in normal men. J Clin Endocrinol Metab 87:21072113 Kamischke A, Diebacker J, Nieschlag E 2000 Potential of norethisterone enanthate for male contraception: pharmacokinetics and suppression of pituitary and gonadal function. Clin Endocrinol (Oxf) 53:351358 Meriggiola MC, Costantino A, Saad F, DEmidio L, Morselli Labate AM, Bertaccini A, Bremner WJ, Rudolph I, Ernst M, Kirsch B, Martorana G, Pelusi G 2005 Norethisterone enanthate plus testosterone undecanoate for male contraception: effects of various injection intervals on spermatogenesis, reproductive hormones, testis, and prostate. J Clin Endocrinol Metab 90:20052014 Kamischke A, Heuermann T, Kruger K, von Eckardstein S, Schellschmidt I, Rubig A, Nieschlag E 2002 An effective hormonal male contraceptive using testosterone undecanoate with oral or injectable norethisterone preparations. J Clin Endocrinol Metab 87:530 539 Gu YQ, Tong JS, Ma DZ, Wang XH, Yuan D, Tang WH, Bremner WJ 2004 Male hormonal contraception: effects of injections of testosterone undecanoate and depot medroxyprogesterone acetate at eight-week intervals in Chinese men. J Clin Endocrinol Metab 89:2254 2262 Behre HM, Nashan D, Hubert W, Nieschlag E 1992 Depot gonadotropin-releasing hormone agonist blunts the androgen-induced suppression of spermatogenesis in a clinical trial of male contraception. J Clin Endocrinol Metab 74:84 90

Page et al. Advances in Male Contraception 258. Pavlou SN, Brewer K, Farley MG, Lindner J, Bastias MC, Rogers BJ, Swift LL, Rivier JE, Vale WW, Conn PM 1991 Combined administration of a gonadotropin-releasing hormone antagonist and testosterone in men induces reversible azoospermia without loss of libido. J Clin Endocrinol Metab 73:1360 1369 259. Tom L, Bhasin S, Salameh W, Steiner B, Peterson M, Sokol RZ, Rivier J, Vale W, Swerdloff RS 1992 Induction of azoospermia in normal men with combined Nal-Glu gonadotropin-releasing hormone antagonist and testosterone enanthate. J Clin Endocrinol Metab 75:476 483 260. Bagatell CJ, Matsumoto AM, Christensen RB, Rivier JE, Bremner WJ 1993 Comparison of a gonadotropin releasing-hormone antagonist plus testosterone (T) versus T alone as potential male contraceptive regimens. J Clin Endocrinol Metab 77:427 432 261. Swerdloff RS, Bagatell CJ, Wang C, Anawalt BD, Berman N, Steiner B, Bremner WJ 1998 Suppression of spermatogenesis in man induced by Nal-Glu gonadotropin releasing hormone antagonist and testosterone enanthate (TE) is maintained by TE alone. J Clin Endocrinol Metab 83:35273533 262. Behre HM, Kliesch S, Lemcke B, von Eckardstein S, Nieschlag E 2001 Suppression of spermatogenesis to azoospermia by combined administration of GnRH antagonist and 19-nortestosterone cannot be maintained by this non-aromatizable androgen alone. Hum Reprod 16:2570 2577 263. Tomera K, Gleason D, Gittelman M, Moseley W, Zinner N, Murdoch M, Menon M, Campion M, Garnick MB 2001 The gonadotropin-releasing hormone antagonist abarelix depot versus luteinizing hormone releasing hormone agonists leuprolide or goserelin: initial results of endocrinological and biochemical efficacies in patients with prostate cancer. J Urol 165:15851589 264. Herbst KL, Coviello AD, Page S, Amory JK, Anawalt BD, Bremner WJ 2004 A single dose of the potent gonadotropin-releasing hormone antagonist acyline suppresses gonadotropins and testosterone for 2 weeks in healthy young men. J Clin Endocrinol Metab 89:5959 5965 265. Lin DW, Coleman IM, Hawley S, Dumpit R, Gifford D, Kezele P, Hung H, Knudsen BS, Kristal AR, Nelson PS 2006 Influence of surgical manipulation on prostate gene expression: implications for molecular correlates of treatment effects and disease prognosis. J Clin Oncol 24:3719 37121 266. Struthers RS, Chen T, Campbell B, Jimenez R, Pan H, Yen SS, Bozigian HP 2006 Suppression of serum luteinizing hormone in postmenopausal women by an orally administered nonpeptide antagonist of the gonadotropin-releasing hormone receptor (NBI42902). J Clin Endocrinol Metab 91:39033907 267. Schlegel PN 2006 Efficacy and safety of histrelin subdermal implant in patients with advanced prostate cancer. J Urol 175:1353 1358 268. Westaby D, Ogle SJ, Paradinas FJ, Randell JB, Murray-Lyon IM 1977 Liver damage from long-term methyltestosterone. Lancet 2:262263 269. Bagchus WM, Hust R, Maris F, Schnabel PG, Houwing NS 2003 Important effect of food on the bioavailability of oral testosterone undecanoate. Pharmacotherapy 23:319 325 270. Daggett PR, Wheeler MJ, Nabarro JD 1978 Oral testosterone, a reappraisal. Horm Res 9:121129 271. Foss GL 1939 Clinical administration of androgens. Lancet 1:502 504 272. Johnsen SG, Bennett EP, Jensen VG 1974 Therapeutic effectiveness of oral testosterone. Lancet 2:14731475 273. Amory JK, Bremner WJ 2005 Oral testosterone in oil plus dutasteride in men: a pharmacokinetic study. J Clin Endocrinol Metab 90:2610 2617 274. Amory JK, Page ST, Bremner WJ 2006 Oral testosterone in oil: pharmacokinetic effects of 5 reduction by finasteride or dutasteride and food intake in men. J Androl 27:7278 275. Attardi BJ, Hild SA, Reel JR 2006 Dimethandrolone undecanoate: a new potent orally active androgen with progestational activity. Endocrinology 147:3016 3026 276. Gao W, Kim J, Dalton JT 2006 Pharmacokinetics and pharmacodynamics of nonsteroidal androgen receptor ligands. Pharm Res 23:16411658 277. Liu PY, Swerdloff RS, Christenson PD, Handelsman DJ, Wang C

243.

244.

245.

246.

247.

248.

249. 250.

251. 252.

253.

254.

255.

256.

257.

Page et al. Advances in Male Contraception 2006 Rate, extent, and modifiers of spermatogenic recovery after hormonal male contraception: an integrated analysis. Lancet 367: 14121420 World Health Organization 1999 WHO Laboratory manual for the examination of human semen and sperm-cervical mucus interaction. 4th ed. Cambridge, UK: Cambridge University Press Burkman R, Schlesselman JJ, Zieman M 2004 Safety concerns and health benefits associated with oral contraception. Am J Obstet Gynecol 190:S5S22 Bhasin S, Woodhouse L, Storer TW 2001 Proof of the effect of testosterone on skeletal muscle. J Endocrinol 170:2738 Behre HM, Kliesch S, Leifke E, Link TM, Nieschlag E 1997 Longterm effect of testosterone therapy on bone mineral density in hypogonadal men. J Clin Endocrinol Metab 82:2386 2390 Snyder PJ, Peachey H, Berlin JA, Hannoush P, Haddad G, Dlewati A, Santanna J, Loh L, Lenrow DA, Holmes JH, Kapoor SC, Atkinson LE, Strom BL 2000 Effects of testosterone replacement in hypogonadal men. J Clin Endocrinol Metab 85:2670 2677 Herbst KL, Anawalt BD, Amory JK, Matsumoto AM, Bremner WJ 2003 The male contraceptive regimen of testosterone and levonorgestrel significantly increases lean mass in healthy young men in 4 weeks, but attenuates a decrease in fat mass induced by testosterone alone. J Clin Endocrinol Metab 88:11671173 Campagnoli C, Clavel-Chapelon F, Kaaks R, Peris C, Berrino F 2005 Progestins and progesterone in hormone replacement therapy and the risk of breast cancer. J Steroid Biochem Mol Biol 96:95108 Brady BM, Amory JK, Perheentupa A, Zitzmann M, Hay CJ, Apter D, Anderson RA, Bremner WJ, Pollanen P, Nieschlag E, Wu FC, Kersemaekers WM 2006 A multicentre study investigating subcutaneous etonogestrel implants with injectable testosterone decanoate as a potential long-acting male contraceptive. Hum Reprod 21:285294 Parsons JK, Carter HB, Platz EA, Wright EJ, Landis P, Metter EJ 2005 Serum testosterone and the risk of prostate cancer: potential implications for testosterone therapy. Cancer Epidemiol Biomarkers Prev 14:22572260 Bhasin S, Singh AB, Mac RP, Carter B, Lee MI, Cunningham GR 2003 Managing the risks of prostate disease during testosterone replacement therapy in older men: recommendations for a standardized monitoring plan. J Androl 24:299 311 Morgentaler A, Bruning 3rd CO, DeWolf WC 1996 Occult prostate cancer in men with low serum testosterone levels. JAMA 276:1904 1906 Schatzl G, Madersbacher S, Thurridl T, Waldmuller J, Kramer G, Haitel A, Marberger M 2001 High-grade prostate cancer is associated with low serum testosterone levels. Prostate 47:5258 Cooper CS, MacIndoe JH, Perry PJ, Yates WR, Williams RD 1996 The effect of exogenous testosterone on total and free prostate specific antigen levels in healthy young men. J Urol 156:438 441; discussion 441 432 Amory JK, Watts NB, Easley KA, Sutton PR, Anawalt BD, Matsumoto AM, Bremner WJ, Tenover JL 2004 Exogenous testosterone or testosterone with finasteride increases bone mineral density in older men with low serum testosterone. J Clin Endocrinol Metab 89:503510 Page ST, Amory JK, Bowman FD, Anawalt BD, Matsumoto AM, Bremner WJ, Tenover JL 2005 Exogenous testosterone (T) alone or with finasteride increases physical performance, grip strength, and lean body mass in older men with low serum T. J Clin Endocrinol Metab 90:15021510 Thompson IM, Goodman PJ, Tangen CM, Lucia MS, Miller GJ, Ford LG, Lieber MM, Cespedes RD, Atkins JN, Lippman SM, Carlin SM, Ryan A, Szczepanek CM, Crowley JJ, Coltman Jr CA 2003 The influence of finasteride on the development of prostate cancer. N Engl J Med 349:215224 Sundaram K, Kumar N 2000 7-Methyl-19-nortestosterone (MENT): the optimal androgen for male contraception and replacement therapy. Int J Androl 23(Suppl 2):1315 Cummings DE, Kumar N, Bardin CW, Sundaram K, Bremner WJ 1998 Prostate-sparing effects in primates of the potent androgen 7-methyl-19-nortestosterone: a potential alternative to testosterone for androgen replacement and male contraception. J Clin Endocrinol Metab 83:4212 4219

Endocrine Reviews, June 2008, 29(4):465 493 491 296. Suvisaari J, Sundaram K, Noe G, Kumar N, Aguillaume C, Tsong YY, Lahteenmaki P, Bardin CW 1997 Pharmacokinetics and pharmacodynamics of 7-methyl-19-nortestosterone after intramuscular administration in healthy men. Hum Reprod 12:967973 297. Noe G, Suvisaari J, Martin C, Moo-Young AJ, Sundaram K, Saleh SI, Quintero E, Croxatto HB, Lahteenmaki P 1999 Gonadotrophin and testosterone suppression by 7-methyl-19-nortestosterone acetate administered by subdermal implant to healthy men. Hum Reprod 14:2200 2206 298. von Eckardstein S, Noe G, Brache V, Nieschlag E, Croxatto H, Alvarez F, Moo-Young A, Sivin I, Kumar N, Small M, Sundaram K 2003 A clinical trial of 7 -methyl-19-nortestosterone implants for possible use as a long-acting contraceptive for men. J Clin Endocrinol Metab 88:52325239 299. Walton MJ, Kumar N, Baird DT, Ludlow H, Anderson RA 2007 7-Methyl-19-nortestosterone (MENT) vs. testosterone in combination with etonogestrel implants for spermatogenic suppression in normal men. J Androl 28:679 688 300. Diel P, Friedel A, Geyer H, Kamber M, Laudenbach-Leschowsky U, Schanzer W, Thevis M, Vollmer G, Zierau O 2007 Characterisation of the pharmacological profile of desoxymethyltestosterone (Madol), a steroid misused for doping. Toxicol Lett 169:64 71 301. Labrie F, Luu-The V, Calvo E, Martel C, Cloutier J, Gauthier S, Belleau P, Morissette J, Levesque MH, Labrie C 2005 Tetrahydrogestrinone induces a genomic signature typical of a potent anabolic steroid. J Endocrinol 184:427 433 302. Death AK, McGrath KC, Kazlauskas R, Handelsman DJ 2004 Tetrahydrogestrinone is a potent androgen and progestin. J Clin Endocrinol Metab 89:2498 2500 303. Ostrowski J, Kuhns JE, Lupisella JA, Manfredi MC, Beehler BC, Krystek Jr SR, Bi Y, Sun C, Seethala R, Golla R, Sleph PG, Fura A, An Y, Kish KF, Sack JS, Mookhtiar KA, Grover GJ, Hamann LG 2007 Pharmacological and x-ray structural characterization of a novel selective androgen receptor modulator: potent hyperanabolic stimulation of skeletal muscle with hypostimulation of prostate in rats. Endocrinology 148:4 12 304. Miner JN, Chang W, Chapman MS, Finn PD, Hong MH, Lopez FJ, Marschke KB, Rosen J, Schrader W, Turner R, van Oeveren A, Viveros H, Zhi L, Negro-Vilar A 2007 An orally active selective androgen receptor modulator is efficacious on bone, muscle, and sex function with reduced impact on prostate. Endocrinology 148: 363373 305. Higuchi RI, Arienti KL, Lopez FJ, Mani NS, Mais DE, Caferro TR, Long YO, Jones TK, Edwards JP, Zhi L, Schrader WT, Negro-Vilar A, Marschke KB 2007 Novel series of potent, nonsteroidal, selective androgen receptor modulators based on 7H-[1,4]oxazino[3,2g]quinolin-7-ones. J Med Chem 50:2486 2496 306. Chen J, Hwang DJ, Bohl CE, Miller DD, Dalton JT 2005 A selective androgen receptor modulator for hormonal male contraception. J Pharmacol Exp Ther 312:546 553 307. Gao W, Reiser PJ, Coss CC, Phelps MA, Kearbey JD, Miller DD, Dalton JT 2005 Selective androgen receptor modulator treatment improves muscle strength and body composition and prevents bone loss in orchidectomized rats. Endocrinology 146:4887 4897 308. Amory JK, Anawalt BD, Bremner WJ, Matsumoto AM 2001 Daily testosterone and gonadotropin levels are similar in azoospermic and nonazoospermic normal men administered weekly testosterone: implications for male contraceptive development. J Androl 22:10531060 309. Wallace EM, Gow SM, Wu FC 1993 Comparison between testosterone enanthate-induced azoospermia and oligozoospermia in a male contraceptive study. I: Plasma luteinizing hormone, follicle stimulating hormone, testosterone, estradiol, and inhibin concentrations. J Clin Endocrinol Metab 77:290 293 310. Pakarainen T, Zhang FP, Makela S, Poutanen M, Huhtaniemi I 2005 Testosterone replacement therapy induces spermatogenesis and partially restores fertility in luteinizing hormone receptor knockout mice. Endocrinology 146:596 606 311. Foresta C, Bettella A, Vinanzi C, Dabrilli P, Meriggiola MC, Garolla A, Ferlin A 2004 A novel circulating hormone of testis origin in humans. J Clin Endocrinol Metab 89:59525958 312. Bay K, Matthiesson KL, McLachlan RI, Andersson AM 2006 The effects of gonadotropin suppression and selective replacement on

278. 279. 280. 281. 282.

283.

284. 285.

286.

287.

288. 289. 290.

291.

292.

293.

294. 295.

492

Endocrine Reviews, June 2008, 29(4):465 493 insulin-like factor 3 secretion in normal adult men. J Clin Endocrinol Metab 91:1108 1111 Kawamura K, Kumagai J, Sudo S, Chun SY, Pisarska M, Morita H, Toppari J, Fu P, Wade JD, Bathgate RA, Hsueh AJ 2004 Paracrine regulation of mammalian oocyte maturation and male germ cell survival. Proc Natl Acad Sci USA 101:73237328 Amory JK, Page ST, Anawalt BD, Coviello AD, Matsumoto AM, Bremner WJ 2007 Elevated end-of-treatment serum INSL3 is associated with failure to completely suppress spermatogenesis in men receiving male hormonal contraception. J Androl 28:548 554 Eckardstein SV, Schmidt A, Kamischke A, Simoni M, Gromoll J, Nieschlag E 2002 CAG repeat length in the androgen receptor gene and gonadotrophin suppression influence the effectiveness of hormonal male contraception. Clin Endocrinol (Oxf) 57:647 655 Yu B, Handelsman DJ 2001 Pharmacogenetic polymorphisms of the AR and metabolism and susceptibility to hormone-induced azoospermia. J Clin Endocrinol Metab 86:4406 4411 Liu GZ, Lyle KC 1987 Clinical trial of gossypol as a male contraceptive drug. Part II. Hypokalemia study. Fertil Steril 48:462 465 Liu GZ, Lyle KC, Cao J 1987 Clinical trial of gossypol as a male contraceptive drug. Part I. Efficacy study. Fertil Steril 48:459 461 Meng GD, Zhu JC, Chen ZW, Wong LT, Zhang GY, Hu YZ, Ding JH, Wang XH, Qian SZ, Wang C 1988 Recovery of sperm production following the cessation of gossypol treatment: a two-centre study in China. Int J Androl 11:111 Waites GM, Wang C, Griffin PD 1998 Gossypol: reasons for its failure to be accepted as a safe, reversible male antifertility drug. Int J Androl 21:8 12 Gu ZP, Mao BY, Wang YX, Zhang RA, Tan YZ, Chen ZX, Cao L, You GD, Segal SJ 2000 Low dose gossypol for male contraception. Asian J Androl 2:283287 Qian SZ 1987 Tripterygium wilfordii, a Chinese herb effective in male fertility regulation. Contraception 36:335345 Matlin SA, Belenguer A, Stacey VE, Qian SZ, Xu Y, Zhang JW, Sanders JK, Amor SR, Pearce CM 1993 Male antifertility compounds from Tripterygium wilfordii Hook f. Contraception 47:387 400 Huynh PN, Hikim AP, Wang C, Stefonovic K, Lue YH, Leung A, Atienza V, Baravarian S, Reutrakul V, Swerdloff RS 2000 Longterm effects of triptolide on spermatogenesis, epididymal sperm function, and fertility in male rats. J Androl 21:689 699 Lue Y, Sinha Hikim AP, Wang C, Leung A, Baravarian S, Reutrakul V, Sangsawan R, Chaichana S, Swerdloff RS 1998 Triptolide: a potential male contraceptive. J Androl 19:479 486 Lue Y, Wang C, Liu YX, Hikim AP, Zhang XS, Ng CM, Hu ZY, Li YC, Leung A, Swerdloff RS 2006 Transient testicular warming enhances the suppressive effect of testosterone on spermatogenesis in adult cynomolgus monkeys (Macaca fascicularis). J Clin Endocrinol Metab 91:539 545 Lue YH, Lasley BL, Laughlin LS, Swerdloff RS, Hikim AP, Leung A, Overstreet JW, Wang C 2002 Mild testicular hyperthermia induces profound transitional spermatogenic suppression through increased germ cell apoptosis in adult cynomolgus monkeys (Macaca fascicularis). J Androl 23:799 805 Kandeel FR, Swerdloff RS 1988 Role of temperature in regulation of spermatogenesis and the use of heating as a method for contraception. Fertil Steril 49:123 Lue YH, Hikim AP, Swerdloff RS, Im P, Taing KS, Bui T, Leung A, Wang C 1999 Single exposure to heat induces stage-specific germ cell apoptosis in rats: role of intratesticular testosterone on stage specificity. Endocrinology 140:1709 1717 Wang C, McDonald V, Leung A, Superlano L, Berman N, Hull L, Swerdloff RS 1997 Effect of increased scrotal temperature on sperm production in normal men. Fertil Steril 68:334 339 Wang C, Cui YG, Wang XH, Jia Y, Sinha Hikim A, Lue YH, Tong JS, Qian LX, Sha JH, Zhou ZM, Hull L, Leung A, Swerdloff RS 2007 Transient scrotal hyperthermia and levonorgestrel enhance testosterone induced spermatogenesis suppression in men through increased germ cell apoptosis. J Clin Endocrinol Metab 92:3292 3304 Hild SA, Marshall GR, Attardi BJ, Hess RA, Schlatt S, Simorangkir DR, Ramaswamy S, Koduri S, Reel JR, Plant TM 2007 Development of l-CDB-4022 as a nonsteroidal male oral contra-

Page et al. Advances in Male Contraception ceptive: induction and recovery from severe oligospermia in the adult male cynomolgus monkey (Macaca fascicularis). Endocrinology 148:1784 1796 Hild SA, Reel JR, Dykstra MJ, Mann PC, Marshall GR 2007 Acute adverse effects of the indenopyridine CDB-4022 on the ultrastructure of Sertoli cells, spermatocytes, and spermatids in rat testes: comparison to the known Sertoli cell toxicant di-n-pentylphthalate (DPP). J Androl 28:621 629 Hild SA, Reel JR, Larner JM, Blye RP 2001 Disruption of spermatogenesis and Sertoli cell structure and function by the indenopyridine CDB-4022 in rats. Biol Reprod 65:17711779 Hild SA, Attardi BJ, Reel JR 2004 The ability of a gonadotropinreleasing hormone antagonist, acyline, to prevent irreversible infertility induced by the indenopyridine, CDB-4022, in adult male rats: the role of testosterone. Biol Reprod 71:348 358 Grima J, Silvestrini B, Cheng CY 2001 Reversible inhibition of spermatogenesis in rats using a new male contraceptive, 1-(2,4dichlorobenzyl)-indazole-3-carbohydrazide. Biol Reprod 64:1500 1508 Lui WY, Lee WM, Cheng CY 2003 Sertoli-germ cell adherens junction dynamics in the testis are regulated by RhoB GTPase via the ROCK/LIMK signaling pathway. Biol Reprod 68:2189 2206 Siu MK, Mruk DD, Lee WM, Cheng CY 2003 Adhering junction dynamics in the testis are regulated by an interplay of 1-integrin and focal adhesion complex-associated proteins. Endocrinology 144:21412163 Mruk DD, Wong CH, Silvestrini B, Cheng CY 2006 A male contraceptive targeting germ cell adhesion. Nat Med 12:13231328 Davies B, Baumann C, Kirchhoff C, Ivell R, Nubbemeyer R, Habenicht UF, Theuring F, Gottwald U 2004 Targeted deletion of the epididymal receptor HE6 results in fluid dysregulation and male infertility. Mol Cell Biol 24:8642 8648 Gottwald U, Davies B, Fritsch M, Habenicht UF 2006 New approaches for male fertility control: HE6 as an example of a putative target. Mol Cell Endocrinol 250:49 57 Roberts KP, Ensrud KM, Wooters JL, Nolan MA, Johnston DS, Hamilton DW 2006 Epididymal secreted protein Crisp-1 and sperm function. Mol Cell Endocrinol 250:122127 Roberts KP, Johnston DS, Nolan MA, Wooters JL, Waxmonsky NC, Piehl LB, Ensrud-Bowlin KM, Hamilton DW 2007 Structure and function of epididymal protein cysteine-rich secretory protein-1. Asian J Androl 9:508 514 Turner RM 2006 Moving to the beat: a review of mammalian sperm motility regulation. Reprod Fertil Dev 18:2538 Fujimoto H, Tadano-Aritomi K, Tokumasu A, Ito K, Hikita T, Suzuki K, Ishizuka I 2000 Requirement of seminolipid in spermatogenesis revealed by UDP-galactose: ceramide galactosyltransferase-deficient mice. J Biol Chem 275:2262322626 Takamiya K, Yamamoto A, Furukawa K, Zhao J, Fukumoto S, Yamashiro S, Okada M, Haraguchi M, Shin M, Kishikawa M, Shiku H, Aizawa S, Furukawa K 1998 Complex gangliosides are essential in spermatogenesis of mice: possible roles in the transport of testosterone. Proc Natl Acad Sci USA 95:1214712152 Yildiz Y, Matern H, Thompson B, Allegood JC, Warren RL, Ramirez DM, Hammer RE, Hamra FK, Matern S, Russell DW 2006 Mutation of -glucosidase 2 causes glycolipid storage disease and impaired male fertility. J Clin Invest 116:29852994 Cox TM, Aerts JM, Andria G, Beck M, Belmatoug N, Bembi B, Chertkoff R, Vom Dahl S, Elstein D, Erikson A, Giralt M, Heitner R, Hollak C, Hrebicek M, Lewis S, Mehta A, Pastores GM, Rolfs A, Miranda MC, Zimran A 2003 The role of the iminosugar Nbutyldeoxynojirimycin (miglustat) in the management of type I (non-neuronopathic) Gaucher disease: a position statement. J Inherit Metab Dis 26:513526 Elstein D, Hollak C, Aerts JM, van Weely S, Maas M, Cox TM, Lachmann RH, Hrebicek M, Platt FM, Butters TD, Dwek RA, Zimran A 2004 Sustained therapeutic effects of oral miglustat (Zavesca, N-butyldeoxynojirimycin, OGT 918) in type I Gaucher disease. J Inherit Metab Dis 27:757766 Pastores GM, Barnett NL, Kolodny EH 2005 An open-label, noncomparative study of miglustat in type I Gaucher disease: efficacy and tolerability over 24 months of treatment. Clin Ther 27:1215 1227

313.

333.

314.

334. 335.

315.

316. 317. 318. 319.

336.

337. 338.

320. 321. 322. 323.

339. 340.

341. 342. 343.

324.

325. 326.

344. 345.

346.

327.

347.

328. 329.

348.

330. 331.

349.

350.

332.

Page et al. Advances in Male Contraception 351. van der Spoel AC, Jeyakumar M, Butters TD, Charlton HM, Moore HD, Dwek RA, Platt FM 2002 Reversible infertility in male mice after oral administration of alkylated imino sugars: a nonhormonal approach to male contraception. Proc Natl Acad Sci USA 99:1717317178 352. Amory JK, Muller CH, Page ST, Leifke E, Pagel ER, Bhandari A, Subramanyam B, Bone W, Radlmaier A, Bremner WJ 2007 Miglustat has no apparent effect on spermatogenesis in normal men. Hum Reprod 22:702707 353. Bone W, Walden CM, Fritsch M, Voigtmann U, Leifke E, Gottwald U, Boomkamp S, Platt FM, van der Spoel AC 2007 The sensitivity of murine spermiogenesis to miglustat is a quantitative trait: a pharmacogenetic study. Reprod Biol Endocrinol 5:1 354. Ren D, Navarro B, Perez G, Jackson AC, Hsu S, Shi Q, Tilly JL, Clapham DE 2001 A sperm ion channel required for sperm motility and male fertility. Nature 413:603 609 355. Liu J, Xia J, Cho KH, Clapham DE, Ren D 2007 CatSperbeta, a novel transmembrane protein in the CatSper channel complex. J Biol Chem 282:1894518952 356. Qi H, Moran MM, Navarro B, Chong JA, Krapivinsky G, Krapivinsky L, Kirichok Y, Ramsey IS, Quill TA, Clapham DE 2007 All four CatSper ion channel proteins are required for male fertility and sperm cell hyperactivated motility. Proc Natl Acad Sci USA 104: 1219 1223 357. Klein T, Cooper TG, Yeung CH 2006 The role of potassium chloride cotransporters in murine and human sperm volume regulation. Biol Reprod 75:853 858 358. Yeung CH, Cooper TG 2001 Effects of the ion-channel blocker quinine on human sperm volume, kinematics and mucus penetration, and the involvement of potassium channels. Mol Hum Reprod 7:819 828 359. Kim YH, Haidl G, Schaefer M, Egner U, Mandal A, Herr JC 2007 Compartmentalization of a unique ADP/ATP carrier protein SFEC (sperm flagellar energy carrier, AAC4) with glycolytic enzymes in the fibrous sheath of the human sperm flagellar principal piece. Dev Biol 302:463 476 360. Miki K, Qu W, Goulding EH, Willis WD, Bunch DO, Strader LF, Perreault SD, Eddy EM, OBrien DA 2004 Glyceraldehyde 3-phosphate dehydrogenase-S, a sperm-specific glycolytic enzyme, is required for sperm motility and male fertility. Proc Natl Acad Sci USA 101:1650116506 361. Welch JE, Brown PL, OBrien DA, Magyar PL, Bunch DO, Mori C, Eddy EM 2000 Human glyceraldehyde 3-phosphate dehydrogenase-2 gene is expressed specifically in spermatogenic cells. J Androl 21:328 338 362. Chen Y, Cann MJ, Litvin TN, Iourgenko V, Sinclair ML, Levin LR, Buck J 2000 Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor. Science 289:625 628 363. Esposito G, Jaiswal BS, Xie F, Krajnc-Franken MA, Robben TJ, Strik AM, Kuil C, Philipsen RL, van Duin M, Conti M, Gossen JA 2004 Mice deficient for soluble adenylyl cyclase are infertile because of a severe sperm-motility defect. Proc Natl Acad Sci USA 101:29932998 364. Xie F, Garcia MA, Carlson AE, Schuh SM, Babcock DF, Jaiswal BS, Gossen JA, Esposito G, van Duin M, Conti M 2006 Soluble adenylyl cyclase (sAC) is indispensable for sperm function and fertilization. Dev Biol 296:353362 365. Hess KC, Jones BH, Marquez B, Chen Y, Ord TS, Kamenetsky M, Miyamoto C, Zippin JH, Kopf GS, Suarez SS, Levin LR, Williams CJ, Buck J, Moss SB 2005 The soluble adenylyl cyclase in sperm mediates multiple signaling events required for fertilization. Dev Cell 9:249 259 366. Wang D, King SM, Quill TA, Doolittle LK, Garbers DL 2003 A new sperm-specific Na/H exchanger required for sperm motility and fertility. Nat Cell Biol 5:11171122

Endocrine Reviews, June 2008, 29(4):465 493 493 367. Wang D, Hu J, Bobulescu IA, Quill TA, McLeroy P, Moe OW, Garbers DL 2007 A sperm-specific Na/H exchanger (sNHE) is critical for expression and in vivo bicarbonate regulation of the soluble adenylyl cyclase (sAC). Proc Natl Acad Sci USA 104:9325 9330 368. Inoue N, Ikawa M, Isotani A, Okabe M 2005 The immunoglobulin superfamily protein Izumo is required for sperm to fuse with eggs. Nature 434:234 238 369. Guha SK, Singh G, Anand S, Ansari S, Kumar S, Koul V 1993 Phase I clinical trial of an injectable contraceptive for the male. Contraception 48:367375 370. Guha SK, Singh G, Ansari S, Kumar S, Srivastava A, Koul V, Das HC, Malhotra RL, Das SK 1997 Phase II clinical trial of a vas deferens injectable contraceptive for the male. Contraception 56: 245250 371. Chaudhury K, Bhattacharyya AK, Guha SK 2004 Studies on the membrane integrity of human sperm treated with a new injectable male contraceptive. Hum Reprod 19:1826 1830 372. Guha SK 2007 Biophysical mechanism-mediated time-dependent effect on sperm of human and monkey vas implanted polyelectrolyte contraceptive. Asian J Androl 9:221227 373. Kumar S, Chaudhury K, Sen P, Guha SK 2007 Quantitative analysis of surface micro-roughness alterations in human spermatozoa using atomic force microscopy. J Microsc 227:118 123 374. Naz RK 2006 Antisperm immunity for contraception. J Androl 27:153159 375. ORand MG, Widgren EE, Sivashanmugam P, Richardson RT, Hall SH, French FS, VandeVoort CA, Ramachandra SG, Ramesh V, Jagannadha Rao A 2004 Reversible immunocontraception in male monkeys immunized with eppin. Science 306:1189 1190 376. ORand MG, Widgren EE, Wang Z, Richardson RT 2006 Eppin: an effective target for male contraception. Mol Cell Endocrinol 250: 157162 377. Naz RK, Gupta SK, Gupta JC, Vyas HK, Talwar AG 2005 Recent advances in contraceptive vaccine development: a mini-review. Hum Reprod 20:32713283 378. Kumar S 1998 Research into anti-fertility vaccine continues despite protests. Lancet 352:1528 379. Moudgal NR, Jeyakumar M, Krishnamurthy HN, Sridhar S, Krishnamurthy H, Martin F 1997 Development of male contraceptive vaccinea perspective. Hum Reprod Update 3:335346 380. Khan MA, Ferro VA, Koyama S, Kinugasa Y, Song M, Ogita K, Tsutsui T, Murata Y, Kimura T 2007 Immunisation of male mice with a plasmid DNA vaccine encoding gonadotrophin releasing hormone (GnRH-I) and T-helper epitopes suppresses fertility in vivo. Vaccine 25:3544 3553 381. Guerin JF, Rollet J 1988 Inhibition of spermatogenesis in men using various combinations of oral progestagens and percutaneous or oral androgens. Int J Androl 11:187199 382. Seidman SN, Araujo AB, Roose SP, McKinlay JB 2001 Testosterone level, androgen receptor polymorphism, and depressive symptoms in middle-aged men. Biol Psychiatry 50:371376 383. Pollanen P, Nikkanen V, Huhtaniemi I 2001 Combination of subcutaneous levonorgestrel implants and transdermal dihydrotestosterone gel for male hormonal contraception. Int J Androl 24: 369 380. 384. Meriggiola MC, Costantino A, Cerpolini S, Bremner WJ, Huebler D, Morselli-Labate AM, Kirsch B, Bertaccini A, Pelusi C, Pelusi G 2003 Testosterone undecanoate maintains spermatogenic suppression induced by cyproterone acetate plus testosterone undecanoate in normal men. J Clin Endocrinol Metab 88:5818 5826 385. Brady BM, Walton M, Hollow N, Kicman AT, Baird DT, Anderson RA 2004 Depot testosterone with etonogestrel implants result in induction of azoospermia in all men for long-term contraception. Hum Reprod 19:2658 2667

Endocrine Reviews is published by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.

You might also like