You are on page 1of 23

nature reviews urology https://doi.org/10.

1038/s41585-023-00820-4

Expert recommendation Check for updates

Frequency, morbidity and


equity — the case for increased
research on male fertility
Sarah Kimmins1,2,3, Richard A. Anderson 4, Christopher L. R. Barratt5, Hermann M. Behre 6, Sarah R. Catford 7,8,
Christopher J. De Jonge9, Geraldine Delbes 10, Michael L. Eisenberg 11, Nicolas Garrido 12, Brendan J. Houston 13,
Niels Jørgensen 14, Csilla Krausz 15, Ariane Lismer1, Robert I. McLachlan16,17, Suks Minhas18, Tim Moss19, Allan Pacey 20
,
Lærke Priskorn 14, Stefan Schlatt 21, Jacquetta Trasler22, Leonardo Trasande23, Frank Tüttelmann 24,
Mónica Hebe Vazquez-Levin 25, Joris A. Veltman 26, Feng Zhang 27 & Moira K. O’Bryan 13
Abstract Sections

Currently, most men with infertility cannot be given an aetiology, which Introduction

reflects a lack of knowledge around gamete production and how it is Methods


affected by genetics and the environment. A failure to recognize the burden The most pressing questions
of male infertility and its potential as a biomarker for systemic illness facing andrology
exists. The absence of such knowledge results in patients generally being Recommendations
treated as a uniform group, for whom the strategy is to bypass the causality
Conclusions
using medically assisted reproduction (MAR) techniques. In doing so,
opportunities to prevent co-morbidity are missed and the burden of MAR
is shifted to the woman. To advance understanding of men’s reproductive
health, longitudinal and multi-national centres for data and sample
collection are essential. Such programmes must enable an integrated view
of the consequences of genetics, epigenetics and environmental factors
on fertility and offspring health. Definition and possible amelioration of
the consequences of MAR for conceived children are needed. Inherent in
this statement is the necessity to promote fertility restoration and/or use
the least invasive MAR strategy available. To achieve this aim, protocols
must be rigorously tested and the move towards personalized medicine
encouraged. Equally, education of the public, governments and clinicians
on the frequency and consequences of infertility is needed. Health options,
including male contraceptives, must be expanded, and the opportunities
encompassed in such investment understood. The pressing questions
related to male reproductive health, spanning the spectrum of andrology
are identified in the Expert Recommendation.

A full list of affiliations appears at the end of the paper. e-mail: moira.obryan@unimelb.edu.au

Nature Reviews Urology


Expert recommendation

Introduction can be a primary determinant of offspring health is not reflected in


Infertility affects at least one in seven heterosexual couples of repro- current health-care policy or social awareness. Concerted basic and
ductive age, globally1. Up to middle age (from 40 years to 65 years old), epidemiological research to test this possibility, then inform policy,
the incidence of infertility is almost equal in women and men, 35% and social programmes and environmental and lifestyle remediation
30%, respectively; in 20% of instances combined factors are involved strategies, are required.
and in 15% of instances no attribution can be made2. For most men The clinical integration of genetic analyses is already identifying
with infertility, aetiology remains undefined, in part owing to a lack of bona fide causes of male infertility8. However, such applications are
foundational knowledge around the processes of gamete production narrowly applied and lag considerably behind other areas of clinical
and quality and how they are affected by environmental and lifestyle medicine. When they are used, they generally only consider a portion
factors. This difficulty is compounded because of the failure of health of the genome and a tiny fraction of genes known to be expressed in
systems to recognize the burden of male infertility at a population spermatogenesis and the male reproductive tract. A shift in clinical
level and its potential as a biomarker of systemic illness. Equally, the approach and health funding to facilitate the adoption of genetic
opportunities to be reaped by improving men’s reproductive health technologies pioneered in other medical fields to the field of andrology
broadly, including improving medically assisted reproduction (MAR) will unequivocally improve the diagnostic rate. In turn, the provision of
technologies, are not currently recognized. The absence of a research molecular diagnoses will, in many circumstances, provide an evidence
focus on defining the precise molecular causes of male infertility base for patients and clinicians to choose the MAR strategy with the
results in men with infertility generally being treated as a uniform best risk:reward profile. Equally, it will enable realistic predictions of
group, for whom the strategy is to bypass a physiological deficit with- the consequences of genetic variation for the health of both the man
out treating the underlying causality. In doing so, opportunities to and the offspring, and ultimately a pathway for the development of
prevent co-morbidity are missed and the burden of MAR is shifted targeted treatment strategies.
to the woman3. Lifestyle and environmental factors are known to contribute to
Alarmingly, tests to provide a precise diagnosis of male infertility poor male reproductive health and infertility in a spectrum ranging
are rare and, consequently, few targeted treatment options exist. For from definitively causal to potential risk factors, such as the abuse
the majority of men, the designation of ‘infertility’ is assigned based of anabolic steroids compared with environmental exposures to
on family history, physical examination, semen analyses and hormone plasticizers or pharmaceuticals9. Emerging data suggest that factors
profiles (that is, surrogate markers) and without systematic rigor- such as sedentary lifestyle, poor nutrition, adiposity, exposure to
ous molecular analysis4. As a consequence, men presenting for MAR chemicals, alcohol, tobacco and cannabis increase the chances of
are grouped into broad categories based on semen phenotype, such male infertility; however, such correlations must be tested. Encourag-
as azoospermia, oligozoospermia, asthenozoospermia, teratozoo- ingly, many lifestyle factors are modifiable or correctable. However,
spermia (or a combination of these descriptors)5, each of which can for such changes to occur, men must first be made aware of any del-
have multiple causes. Importantly, the designation of many of these eterious inter-relationships and then be provided with the options to
terms is based on normative population data that are largely unlinked effect change. Accumulating evidence indicates that fertility can be
to fertility potential. As a result, the identification of who has infertility influenced throughout the lifespan9. Thus, men in late adolescence and
and is in need of medical intervention, even if treatments were to exist, early adulthood, for example, need to be armed with knowledge about
is surprisingly difficult. the risks to their reproductive and overall health if they participate in
For most cases of suspected male infertility, the default MAR unhealthy behaviours through educational and supportive social pro-
strategy requires the woman to carry a large burden3: a woman with grammes. Widespread implementation of such programmes requires
no identified reproductive disease or history becomes the patient to effective policies and the appropriation of funds by governments to
‘treat’ a presumed male fertility issue. The misnomer of ‘treatment’ for ensure awareness and access for boys and men of peri-reproductive
male infertility is emotionally taxing for the man and woman, involves and reproductive age (>13 years old). Furthermore, men must be
considerable time and financial commitment, is invasive, and includes a encouraged to increase self-awareness about their health and adopt
risk to the woman’s health. Although unintended and the consequence active health-care-seeking routines. In return, but within a long time
of a complex history, this occurrence is a stark example of gender frame, such behavioural changes should decrease infertility rates and
inequity in medical treatment, and one that extends in both directions. other diseases.
Treating one partner to circumvent a medical deficiency in the other is This recommendation document was requested by the Male
questionable. Equally, the failure to develop male-centred treatment Reproductive Health Initiative (MRHI), which is a working group of
options is a failure to accept male infertility as a substantial medical the European Society of Human Reproduction and Embryology, in
condition that affects a notable portion of the global population. an effort to provide clarity for the field, governments and the public
Increased emphasis on developing protocols that directly address the on the current status of andrology research and medicine and where
aetiologies of male infertility would address this inequity and probably future resources can be most gainfully deployed. Within this Expert
lead to improved clinical outcomes. Recommendation we have identified the most pressing questions
We argue that an urgent need to determine the underlying causes related to male reproductive health. The formulation of this guidance
of male infertility exists, including the interplay between genetics, document comes from experts from around the globe, spanning the
epigenetics, the environment and lifestyle. This imperative extends spectrum of andrology research, clinical practice and public policy.
well beyond the ability to conceive a child. An increasing and concern- These questions encompass and highlight many opportunities to influ-
ing body of evidence indicates that men with infertility have a higher ence the wellness of future generations. For each question, we briefly
disease burden and die younger than fertile men6, and that infertility summarize the current state of knowledge, and identify resource gaps
and ill-health can be transmitted to children conceived using MAR and opportunities, to deliver a roadmap to improve male reproductive
techniques7. The realization that a man’s health beyond his genetics health and treatment.

Nature Reviews Urology


Expert recommendation

Methods To date, most studies on the frequency of male infertility have


Following a period of consultation between MHRI members via online reported on high-income countries, and often those referred to MAR
meetings and e-mail, 13 questions were formulated. Lead authors for programmes. A need exists to greatly expand epidemiological studies
each question were appointed and asked to contact any additional outside the MAR field, which ought to feature a thorough and stand-
experts (or emerging leaders) within the field they felt would add to the ardized male evaluation, aetiological classification and to consider
quality of the section. Each section or topic was written by two or more environmental and geographical factors and ethnicity. This limita-
internationally recognized experts in the particular sub-area. Authors tion aside, we can definitively agree that male infertility is common,
were free to review all forms of literature from any source. Sections were accounts for a high percentage of MAR interventions and carries an
integrated by the first and last listed authors before several rounds of enormous personal and economic burden.
review by the entire author team. For each question, we briefly sum-
marize the current state of knowledge and identify resource gaps and The diagnosis of male infertility
opportunities, to deliver a roadmap to improve male reproductive The second question that needs addressing is how male infertility is
health and treatment. diagnosed (Box 1, question 2). A comprehensive infertility diagnostic
process including both partners is essential to provide informed predic-
The most pressing questions facing andrology tion, counselling and management. Despite widely available guidelines
In total, 13 questions were devised concerning pressing issues in male for male evaluation14–17, treating physicians in the MAR setting might
infertility (Box 1). To improve diagnosis, research and treatment, these not be male fertility specialists18. An inescapable need exists for a clini-
questions need to be answered. cal history focusing on the reproductive system, physical examination
(including of the testes), a semen analysis and guided laboratory testing
The proportion of men with infertility (endocrine profile, genetics, imaging and histology). Such an evalua-
The first question to answer is, what proportion of men are infertile tion could enable identification of reversible causes permitting natural
(Box 1, question 1)? Surprisingly, relatively little information exists conception, or serious comorbidities (Box 1, questions 3 and 8).
on the prevalence of male infertility at a global level (Fig. 1). In an epi- The diagnostic yield for a small number of male infertility sub-types
demiological study including 15,162 couples in the UK, 1 in 8 women is good (for example, chromosomal abnormalities and hypogonado-
and 1 in 10 men had experienced infertility10. However, these data are tropic hypogonadism), but for the majority of men, the aetiology
almost certainly underestimated. Determining an accurate percentage of the infertility remains unknown, that is, a physiological or cellu-
of men who have infertility is challenging for a number of reasons: first, lar deficit will be identified but the underlying causality is unknown.
the possibility of a female factor will confound a clinician’s ability to
attribute causality to either partner, that is, infertility is by definition
a two-person condition; second, there is a dearth of precise diagnostic
tests, so men are frequently not thoroughly examined; and third, any
Box 1
calculation or prediction of prevalence and incidence is dependent on
the population examined. For example, the incidence of male infertility The most pressing questions
in heterosexual couples presenting at clinics for MAR will differ from
those first attempting a pregnancy naturally. Whole-population-based facing andrology
assessments of male infertility are rare, with the vast majority of studies
on male infertility deriving data from infertility treatment centres or the 1. What proportion of men are infertile?
assessment of men in at-risk populations (such as those living in regions 2. How is male infertility diagnosed?
with high toxicant concentration). Addressing these challenges through 3. What treatments are available for male infertility and what
high-quality studies at global, regional and national levels is crucial, as treatments are needed?
these data might ultimately contribute to the establishment of holis- 4. What are the genetic causes of male infertility?
tic health-care systems with increased efficiency. Moreover. without 5. Environmental and lifestyle factors impact male fertility: what is
understanding the incidence of a disease, providing resources, estimat- needed to prevent and reduce their impacts?
ing effect, making effective health economic arguments, presenting 6. Is male fertility declining?
rational research questions and managing patients are difficult11. 7. What is the economic burden of male infertility?
The most commonly used data points to assess male fertility come 8. Do men with infertility have a higher disease burden than fertile
from a semen analysis (Fig. 1). Clinicians and patients should be aware men and do they die younger?
of its limited prognostic accuracy, and that natural conception might 9. Is it possible to develop robust gamete storage and restoration
or might not occur across a range of semen analysis values. Data from protocols for boys and men prior to other medical interventions?
before the widespread use of modern MAR (that is, before intracyto- 10. Do changes in epigenetic processes lead to male infertility or
plasmic sperm injection (ICSI)) showed that ~30% of couples for whom have inter-generational consequences?
the man had a sperm concentration of 1–5 million/ml and the woman 11. What are the long-term health outcomes for children born to
was healthy conceived naturally over a 2-year period, usually in the first men with compromised fertility and do these consequences
18 months12. This reality notwithstanding, in an unselected popula- vary with conception via natural or via medically assisted
tion, conception rates rise until a plateau at a sperm concentration of reproduction?
~40 million/ml (ref. 13). Thus, many men with what might be regarded 12. Can we develop additional male-based contraceptive methods?
as impaired spermatogenesis achieve fertility without medical inter- 13. How can andrology as a discipline communicate effectively with
ventions, or only come for medical attention when their partner also the public, health professionals and funding agencies?
has reduced fertility.

Nature Reviews Urology


Expert recommendation

Germany Denmark Finland Poland


16 23 15 15 7.3 9.1 13 8.9

Switzerland Czechia
17 21

USA Spain
15.8 23.1 15.2 16.5 12 17.5 Japan
9 8.8 6.7

4.65 19.7 5.1


China
Study year Number of participants 18.1

1980 100 4.8 6.2 8

500 10,000
1990

2000 1,000

30,000 New Zealand


2010 19.8 12.2
5,000 Tunisia Libya
39.8 14

Lower limit sperm counts


<20 million/ml <15 million/ml

Fig. 1 | Worldwide, limited data exist on sperm concentration in men. participants from which the data are derived (Supplementary Table 1), and a
A summary of sperm concentration from a selection of available studies (blue) solid versus dashed circle indicates that the average sperm concentration in that
for world regions spanning the years 1992 to 2019. Studies were selected to population in relation to reference limits at the time of the study <20 million/ml
exclude patients with infertility or men in regions associated with toxicant (lower limit 4th edition of WHO andrology laboratory manual, 1999 (ref. 383)) or
exposures. Comparative interpretation with regard to worldwide fertility trends <15 million/ml (lower limit, 5th edition of WHO andrology laboratory manual and
is difficult owing to a lack of available data (grey), variation in sample size and based on male partners who had a time to pregnancy of ≤12 months384). Data from
population diversity. The size of the circle represents the number of study refs. 168,170,171,385–406.

This limitation does not detract from the value of the examination as it to remain a key tool for assessing male infertility, its use in isolation is
will inform MAR strategy. In addition to probable physiological causes considerably limited.
of male infertility being identified through a comprehensive examina- Beyond a semen analysis, serum follicle-stimulating hormone
tion of the man, other conditions that are risk factors for male infertility (FSH) is the best, albeit imperfect, endocrine marker of spermatogenic
might be identified and addressed. For example, varicocele or high output: serum FSH levels rise as spermatogenic failure worsens owing
levels of white blood cells in semen are twice as prevalent in men with to falling serum inhibin B levels21. The upper limit of FSH in carefully
infertility as in those who are fertile, and obesity and chronic disease characterized, healthy men is 8.4 IU/l (ref. 22); the higher upper limits
are also more prevalent in patients with infertility19. (for example, 12 IU/l) suggested by some FSH assay manufacturers are
A fundamental consideration when analysing results from because of the inclusion of older men (>70 years) and of those with
a semen analysis is that the WHO set the lower 5th percentile of unrecognized reproductive defects, potentially leading to a distor-
whole-population levels as the level under which semen composition tion of the ‘normal’ range23–27. Serum luteinizing hormone (LH) and
is classified as abnormal20. Thus, by definition 5 in 100 men will be testosterone concentrations are an indication of testicular endocrine
classified as having an abnormal result for any particular parameter. function: around 1 in 8 men presenting for fertility care show evidence
As such, problems arise when studies simplistically use semen param- of biochemical (if not clinical) hypogonadism28. As evidence of the
eters as a standalone fertility end point. For example, an assumption value of a full reproductive work-up, low LH and testosterone levels
that a set percentage of men with one or more parameters below the often indicate secondary testicular failure and require investigations
WHO ‘reference ranges’20 will be infertile is inaccurate. However, and (such as iron studies and pituitary imaging) that demand specific care
providing that the analysis process was reproducible and accurate, for fertility and broader health14–16,29–31.
a semen analysis can provide an opportunity for comparisons between A holistic approach that goes beyond semen analysis is endorsed
populations. Importantly, an abnormal semen result is only a descrip- by many clinical practice guidelines (for example, American Urologi-
tive marker of an underlying disease. For example, oligoasthenozoo- cal Association–American Society for Reproductive Medicine guide-
spermia is not a diagnosis per se. Thus, although semen analysis is likely lines Part II and European Association of Urology guidelines on male

Nature Reviews Urology


Expert recommendation

infertility15,31). The range of potential underlying molecular aetiologies hormonal therapy in 15–20% of patients with congenital HH40. For these
is expansive, and has relevant implications for the health of the patient men, hypothetically, either the treatment was suboptimal or they have
and future offspring (Box 1, question 11). To improve the diagnosis of mutations in genes that are essential both for the central hormonal
male infertility, suggestions for clinical practice for data collection regulation of the testis function and for spermatogenesis, or that coex-
and research relevance will be of value for therapeutic innovation and isting but distinct genetic disorders are involved at the two sites. As HH
ultimately to improve long-term patient outcomes: the development is a rare disease accounting for 1–2% of men with infertility42, multicen-
of national MAR data collection that includes male infertility char- tre studies are required to unravel the reason for non-responsiveness.
acteristics following clinical practice guidelines as above — inherent If non-responsive genotypes do exist, genetic screening would help
in this goal is that MAR unit participation should be enforced, with to avoid prolonged (on average, 12 months) hormonal treatments in
auditing of a minimum data set and evaluation approaches for the these patients.
men (this possibility is feasible, as exemplified by the Australia and Other rational treatments include the surgical removal of obstruc-
New Zealand Assisted Reproduction Database 3.0 data collection)32; tions in some instances of obstructive azoospermia43–48, and the
the development of fertility tests with improved predictive value, treatment of varicocele in highly selected men16,49. However, these treat-
including tests that reduce subjectivity and/or make use of artificial ments are only applicable for a small fraction of men with infertility
intelligence technologies, which are already showing promise 33 and symptomatic management of male infertility is by far the most
(in order to develop improved fertility tests to the highest standards, common approach taken16. In most circumstances, MAR strategies
long-term clinical outcomes for men and children born as a result of are aimed at bypassing obstructions or physiological barriers to
using MAR techniques will be required)34; improved and increasingly fertility. For example, for men with obstructive azoospermia, a near
effective education in male health at all levels of society, analogous 100% sperm retrieval rate is obtained using surgery16,50. For men with
to the current state for women and children’s health (for example, non-obstructive azoospermia (NOA), surgical TESE is typically the
in medical schools, an andrology curriculum should be integrated only clinical option available and sperm are retrieved in approximately
with urology, internal medicine, endocrinology and gynaecology ~50% of procedures, which is impressive as they have no sperm in their
(fertility)); increased awareness of the frequency and consequences ejaculate51. However, the aetiologies of NOA are diverse and although
of male infertility by primary care physicians to accelerate referral to TESE success can be partially predicted based on genetic analyses
specialist clinics, avoiding delay in diagnosis and treatment (this latter (for example, virtually no success in men with 46,XX and those with
point is increasingly important owing to societal changes in advancing complete azoospermia factor a (AZFa) or AZFb deletions on the
parental age at conception)35,36; and an awareness, action plan, and data Y chromosome), in many men, the prediction is imperfect, with similar
collection protocol to detect the effects of environmental and lifestyle outcomes achieved from a range of aetiologies, including NOA caused
factors on the male gamete, fertility and offspring outcomes. From a by chemotherapy, Klinefelter syndrome and idiopathic instances51.
discovery perspective, the first and final points should be accompanied Notably, success only modestly correlates with testis size, serum FSH
by an expansive biospecimen collection that can be linked back to the or inhibin B14. Thus, the development of improved predictive tests to
clinical features and fertility outcome data37. indicate use of or avoidance of such highly invasive surgical procedures
are desperately needed14,51–53.
What treatments are available or needed? A plethora of empirical strategies for male infertility are avail-
The third question that needs to be considered is what treatments are able for which the rationale and evidence are limited or non-existent.
available for male infertility and what treatments are needed (Box 1, Administration of exogenous hormones and anti-oxidants are two
question 3)? Disappointingly and frustratingly, given the frequency of notable categories of empirical treatments because of their frequency
male infertility, evidence-based therapies to restore natural fertility are of use49. The clinical evidence for hormonal stimulation in men with HH
uncommon and many clinical interventions are based on low-level, or is established, but pharmacological application in men with idiopathic
no, evidence. Although often described as a treatment for male infer- oligoasthenoteratozoospermia is not. The rationale for this strategy
tility, most MAR strategies neither address the underlying pathology is presumably aimed at increasing baseline gonadotrophin levels to
nor provide a treatment for the man. Rather, they use the most mature promote sperm output, but the strategy has a low evidence base54–56.
germ cell type from anywhere in the reproductive tract in procedures Other examples, such as selective oestrogen receptor modulators
to achieve fertilization in vitro38. With notable exceptions, such as tes- (clomiphene, tamoxifen) and aromatase inhibitors57–60, are proposed
ticular sperm extraction (TESE), for the most part, such strategies place to enhance gonadotrophin stimulation and consequently spermato-
the majority of the burden on the woman. genesis, but also have a low evidence base. Similarly, hCG, human
Management strategies for male infertility can be divided into menopausal gonadotropin (hMG) or recombinant hFSH are used with
‘rational’ (based on a good understanding of the pathophysiology the same goal61. Even in men with NOA, presurgical hormones are given
and strong evidence of effectiveness), ‘symptomatic’ (bypassing the in the hope of inducing spermatogenesis or increasing the probability
pathology through the use of MAR) and ‘empirical’ (with potential merit of sperm extraction at the time of TESE62. FSH administration in idi-
but lacking evidence)39. opathic oligoasthenoteratozoospermia with FSH in the normal range
The best example of a rational medical therapy is the treatment is approved in a few countries by their national health-care system
of men with hypogonadotropic hypogonadism (HH) using a combina- (for example, Italy63), but selective oestrogen receptor modulators
tion of FSH and human chorionic gonadotropin (hCG, as a surrogate and aromatase inhibitors are considered off-label treatments for male
for LH) as a replacement for endogenous deficiencies. In many men infertility in most countries64. The FDA does not approve and practice
this treatment leads to fertility40. However, this approach is not a ‘cure guidelines do not recommend these treatments, pointing out the lim-
all’: approximately 50% of men with HH can be provided with a precise ited randomized, controlled trial data concerning their effectiveness
molecular diagnosis for their hormone deficiency41, but, for reasons and warning about a delayed transition to proven effective MAR15,60.
that are unclear, the germinal epithelium remains unresponsive to the Despite such warnings, 65.9% of urologists in the USA prescribed some

Nature Reviews Urology


Expert recommendation

form of empirical gonadotrophin therapy to treat idiopathic male unregulated antioxidant use. The same comments equally apply for any
infertility65,66. Clearly, such indiscriminately applied strategies are a form of unproved pharmacological intervention and herbal remedies.
burden on the health-care system and could actually harm fertility or In summary, evidence of efficacy exists for some treatment
delay using more effective interventions. Thus, an urgent need exists approaches to male infertility, but additional high-quality, compre-
within the discipline of andrology to train health providers on the risks hensive, randomized trials are required. The field urgently needs to
associated with these practices and to establish randomized clinical move towards the precise diagnosis of male infertility, the realization
trials to resolve any uncertainty. that many hundreds of aetiologies could exist and that different types
In some instances a rationale for the use of such strategies exists, of infertility will require different treatment strategies, that is, personal-
suggesting that when targeted correctly, the application of exoge- ized medicine. Equally, a great opportunity exists for pharmacogenom-
nous hormones or hormone regulators might enhance fertility, avoid- ics in defining subgroups of currently ‘idiopathic’ male infertility that
ing MAR or improving MAR success. The difficulty arises when the might be amenable to pharmacological treatment.
approach is applied too broadly and in the absence of data. For exam-
ple, with regard to FSH administration, the possibility of pre-selecting The genetic causes of male infertility
responders based on distinct genetic polymorphisms in FSHB and Fourth, what the genetic causes of male fertility are needs addressing
FSHR has been proposed as a potential pharmacogenetic approach67–69. (Box 1, question 4). Genetic factors have an important role in the aeti-
However, to date, results of this approach are contradictory and based ology of male infertility. Well-accepted genetic causes of human male
on relatively small study populations67–69. For this question, along with infertility include variant karyotypes (such as 47,XXY for Klinefelter
many others, patient outcomes could be improved by transnational syndrome) and submicroscopic genomic deletions on the Y chromo-
collaborative trials. some (Yq)87. As an indication of the incidence of these particular types
The other major agents used in the ‘treatment’ of male fac- of infertility, in a 2011 study of 1,583 consecutive men with azoospermia
tor infertility are antioxidants and nutritional supplements70. In a (comprising ~10% of the total patient population of a tertiary fertility
questionnaire-based study involving 1,327 reproductive-specialist centre), 21% had a molecular diagnosis, including 15% with Klinefelter
participants from six continents, antioxidants were prescribed by syndrome and 2% with Yq deletions88. A further 15% had malignancy,
85.6% of clinicians, with a marked variation of types, including zinc, 11% had obstruction and 7% endocrine or chronic disease. The remain-
vitamins E, C, B1, B9, B12 and D, l-arginine and co-enzyme Q, to name a ing 46% of patients did not receive an adequate causal diagnosis. The
few71. These agents are attractive for patients owing to their relatively latter was most likely substantially caused by the fact that sequencing
low costs, accessibility and favourable safety profile. The proposed of genes was not and is still not widely used for the diagnosis of male
beneficial effects of these agents are related to their potential positive infertility. Specifically, in many clinics the only sequencing undertaken
effects on spermatogenesis, sperm capacitation and fertilization via a is of CFTR when obstructive azoospermia owing to congenital absence
number of mechanisms of action, including their antioxidant effects72. of the vas deferens is found89. As evidence of the potential benefit of
However, caution must be exercised as overuse of supplements could moving to a comprehensive genome analysis, results showed that
actually harm spermatogenic output and sperm function73. For exam- applying an exome-based panel of only 21 genes strongly linked to
ple, over-supplementation with folic acid or zinc could harm male human male infertility provides a diagnostic yield of 8.5% in previously
fertility74. One particular area of interest is the use of antioxidants to unexplained idiopathic azoospermic men and that the results have
reduce sperm DNA fragmentation, secondary to the effects of oxidative predictive power for TESE90.
stress induced by reactive oxygen species75. Reactive oxygen species are With the advances of genomic technologies, mutations in
a normal part of many biological systems and are required for sperm hundreds of new candidate genes have been reported as potential
functions including capacitation76–78; however, in excess they outweigh monogenic causes of human male infertility8. Of these genes, 120 are
the minimal antioxidant defences of sperm and are damaging for sperm currently supported by compelling evidence, including having been
motility and DNA integrity79. Equally, endogenous antioxidant activity replicated in multiple cohorts8. This number is certainly a consider-
within the male reproductive tract can modulate oxidative germ cell able under-estimate of the reality. The molecular diagnosis of mul-
DNA damage and thereby reproductive outcomes80,81. The degree to tiple morphological abnormalities of the sperm flagella (a sub-type
which this activity operates within the seminiferous epithelium is of teratozoospermia) is a notable success story, in which the major-
unclear; however, after removal of the cytoplasm and most of its anti- ity of instances seem to be genetic in origin and can be diagnosed at
oxidant content during the process of spermiation, the epididymis and a molecular level if genome or exome sequencing is undertaken91.
the female reproductive tract are the mostly likely enactors of sperm Despite these advances, the vast majority (60–70%) of instances of male
DNA oxidative damage76,82,83. In this setting, the beneficial role of these infertility remain unexplained. Uncovering these missing aetiologies,
supplements (or dietary changes) to supplement endogenous antioxi- many of which are probably genetic in origin, requires researchers to
dant levels or in the treatment of male factor infertility remains to be resolve a number of substantial challenges, including extreme genetic
established. The utility of antioxidants, or not, is also confounded by heterogeneity, the existence of dominant causes of male infertility,
the fact that oxidative stress and sperm DNA fragmentation meas- non-coding variation and complex regions in our genome, and the
urements are not robust84. The lack of clear data in this area is evi- varying consequences of rare and common genomic variation and
denced by Cochrane analyses70,85, in which only low-quality evidence gene–environment interactions.
from a handful of highly heterogenous studies showed that antioxi-
dants improved pregnancy and live birth rates for couples experi- Extreme genetic heterogeneity. Individual genetic variants result-
encing subfertility. Thus, the use of antioxidants in the treatment of ing in human infertility are expected to be extremely rare in human
male infertility is controversial and an open question60,72,75,85. Large populations, owing to purifying selection. Thus, large cohorts and
and well-controlled trials are clearly needed74,86. Similarly, clinicians and multicentre collaborations across populations are essential to facilitate
patients need to be informed about the potential complications of the identification of novel infertility-related genes and variants that are

Nature Reviews Urology


Expert recommendation

shared between men with infertility. Similarly, the validation of causal- of MAR technology are generally healthy but have risk alleles. For
ity using animal models might be required. Although considerable example, for autosomal-dominant loci, ~50% of boys will have the
progress has been made in this area, including by the International Male paternal pathogenic mutation and will probably experience male
Infertility Genomics Consortium92,93, the Genetics of Male Infertility infertility. For Y-linked loci, almost 100% of boys born as a result of MAR
consortium94,95 and several individual laboratories96–99, many opportu- intervention will be affected by the paternal mutation112. How much of
nities remain. As other genetic causes of male infertility are discovered, an effect MAR practices are having on the propagation and expansion
early interventions might be realized, for example, the early collection of male infertility remains unknown. However, patients and clinicians
of sperm samples from men with damaging genetic variants in genes must consider this reality.
required for spermatogonial stem cell renewal or similar. In summary, severe forms of male infertility have a strong genetic
component that is currently understudied, limiting personalized medi-
The existence of dominant causes of male infertility. To date, most cine approaches in this field. To fully understand the role of genetics
attention regarding genetic causes of male infertility has been on in male infertility large-scale, genome-wide studies are required and
studying recessive and X-linked causes. The study of dominant causes genomics needs to become part of routine patient diagnostics work-up.
of male infertility, either maternally inherited or, more likely, occurring
de novo, requires patient–parent trio-based genetic studies100–103. This Negative environmental and lifestyle effects
area of research is crucial and will require a clinical shift from con- Fifth, environmental and lifestyle factors impact male fertility; thus,
sidering just the patient with infertility to also including his parents. what is needed to prevent and reduce their effects needs elucidating
Adoption of this strategy is essential if the field hopes to develop a com- (Box 1, question 5). Humans are exposed to thousands of factors that can
prehensive understanding of the genotype–phenotype relationship be damaging to reproductive health, beginning in utero and continuing
for human male infertility. throughout life. These factors include sub-optimal diet, pesticides, cos-
metics, plastics, cannabis, medicines, alcohol and cigarette smoking,
Non-coding variation and complex regions in our genome. Based among others113–117. Spermatogenesis is a complex process involving
on data from other disease conditions, the likelihood that some forms the coordinated action of thousands of genes and is sensitive to envi-
of male infertility will be caused by variations (for example, mutations) ronmental factors118. In utero and early life, exposures can affect the
in the non-coding regions of the genome is high, but this possibility establishment and programming of gonadal cells and the developing
is virtually unexplored. Improved genomic coverage using low-cost, reproductive tract, which can influence hormonal homeostasis, cell
whole-genome sequencing has strong potential to be used to iden- differentiation, gamete production and function119. This concept is
tify such pathogenic variants in non-coding regions, and a detailed extremely difficult to prove in humans, but evidence does exist.
analysis of the role of structural genomic variation in male infertility. Widespread use of and exposure to endocrine-disrupting chemi-
For example, the male-specific region on the human Y chromosome cals (EDCs, compounds that disrupt the body’s own hormones) is a
is full of long genomic repeats but multi-copy genes in this region particularly insidious risk with adverse effects on fertility120. Such
are difficult to sequence and haplotype104. The analysis of such com- exposures can be exogenous and/or anabolic steroids or EDCs found
plex regions requires the use of relatively new technologies such as in compounds used as plasticizers, pesticides for agriculture, industry
long-read sequencing for accurate analysis. or via environmental contamination121. Experimental evidence from
animal models combined with epidemiological studies involving men
Rare and common genomic variation. Interactions will occur between indicate that exposure to EDCs is associated with reproductive impair-
rare (mutations) and common (polymorphisms) variants. Currently, ment, including testicular cancer, disorders of sex development, crypto­
very few examples are known, but the widely divergent effects on rchidism, hypospadias and decreased sperm count115,121–123. Early life is a
infertility of the gr/gr Y chromosome microdeletion in European versus crucial environmentally sensitive period, but exposures throughout
Japanese men might serve as one. Specifically, gr/gr deletions appear to a man’s life might also have damaging and even irreversible conse-
cause infertility in European men but permit fertility in Japanese men105. quences for fertility. For example, exposures to the EDC insecticide
dichlorodiphenyltrichloroethane (DDT), which is used for malaria vec-
Gene–environment interactions. Environmental and lifestyle expo- tor control in South Africa, is associated with impaired semen quality124.
sures (Box 1, question 5) are linked with infertility in some populations Toxicology research in relation to male fertility remains difficult and
or genotypes but not others106. Equally, as is observed in several other controversial owing to the complexities of epidemiological studies
diseases, including kidney, neurological and urological diseases107–109, a that have a multitude of confounding factors, such as behavioural and
percentage of instances of male infertility is highly likely be the result of socioeconomic factors. These challenges are compounded by much
genetic variants in multiple genes interacting to cause infertility (poly- of what is known regarding repro-toxicant effects, most of which are
genic). This possibility is virtually unexplored in the field of andrology. based on animal models and single, rather than complex, exposures.
The corollary of these complex types of male infertility is that a predis- Specifically, regulation of environmental chemicals based on linear
position to male infertility might be inherited via the paternal germ line dose–response and assessing one compound at a time is not reflective
(in addition to the maternal germ line). Although poorly understood, of real-life exposure125,126. This limitation is illustrated by the fact that
evidence of this possibility already exists for Y chromosome variants European men have been found to have combined exposures to chemi-
in the AZFc region — most men with variants in this region are infertile, cals known to affect semen quality, namely bisphenol A, bisphenol F and
yet rare examples of fertility exist110. bisphenol S, polychlorinated dibenzodioxins and paracetamol, among
An additional consideration is that although damaging others. Of great concern is that tolerable exposures were exceeded for
genetic variants frequently block natural fertility, MAR might allow these chemicals127.
the transmission of such variants to the next generation 111. For In addition to environmental contaminants, which arguably men
autosomal-recessive loci of male infertility, children born as a result have little control over, recreational drug use, stress, diet and some

Nature Reviews Urology


Expert recommendation

types of physical activity are modifiable lifestyle factors that might alternatives. Such alternatives must undergo testing before introduc-
negatively effect male fertility, and potentially the health of their chil- tion to the market. Second, meaningful testing requirements for male
dren, which men have some control over15,128–132. For example, stress reproductive safety need defining, and assays capable of reflecting the
has a psychological and a physiological effect that includes alteration complexity of human behaviour and spermatogenesis need develop-
to the function of the hypothalamic–pituitary–gonadal axis and has ing. Ideally, toxicity testing would include specific assessment of germ
been implicated in male factor infertility in both humans and animal cell toxicity and end points including genotoxicity and epigenetic
models133. Major life events, such as job loss, war and bereavement, responses including intergenerational transmission126,147–149. Third,
correlate with reduced semen quality134,135. Chronic stress elevates glu- experimental models that better replicate the reality of multiple or
cocorticoids, reduces LH secretion and in turn can lower testosterone transgenerational exposures to repro-toxicants than current models
production by Leydig cells. Moreover, stress, obesity and metabolic need developing. Fourth, epidemiological studies should be inclusive
dysfunction activate inflammation and immunomodulatory responses, of women, men and their offspring to determine the consequences of
all of which disrupt the hypothalamic–pituitary–gonadal axis and exposures beginning in utero, throughout life and into future genera-
spermatogenesis136,137. Equally, emerging data suggest that such stress tions. Finally, knowledge translation should be improved for general
might have negative transgenerational consequences129,138,139. practitioners and the public on lifestyle and environmental factors that
As such exposures, physiological changes and lifestyle factors can influence fertility in men throughout their life (Box 2).
could negatively influence fertility, whether the effect is permanent
or reversible is a pertinent question. The use of intervention strategies Is male fertility declining?
such as weight-loss, diet, micronutrient supplementation, antioxidants Whether male fertility is declining is the sixth pressing question (Box 1,
or folic acid supplementation remain controversial140. Nonetheless, question 6). Worldwide, fertility rates defined as the number of chil-
positive effects of weight loss on sperm concentration and count in dren born per woman have been, or are, declining9,150. In Denmark, the
men with obesity demonstrate the potential of lifestyle intervention proportion of childless men has increased across birth cohorts151. How-
for improved fertility141. Similarly, data from animal models indicate ever, such rates are poor proxies of male fertility (the biological ability
that changes in diet can modify the composition of seminal plasma to conceive or the time frame within which conception is achieved)
and that such changes have functional consequences for immune and socioeconomic factors can largely explain these trends. Nonethe-
responses in the female tract and offspring health142. Similarly, inter- less, indications suggest that male fertility and reproductive health in
ventions to reduce stress in couples with infertility showed a positive general have declined, including a decrease in semen quality and an
association with pregnancy rates143. increase in testicular cancer and congenital urogenital malformations
However, frequently, human trials examining these factors are (cryptorchidism and hypospadias)150. However, any conclusions remain
poorly targeted and controlled, and use a wide range of end points, controversial152, and should be rigorously tested.
often not including key ones such as pregnancy and live birth rates A couple’s chances of conceiving are decreased when the man has
rather than sperm functional or integrity assessment144. However, if a sperm concentration below 40 million/ml (resulting in a prolonged
the trial quality can be improved, strong prospects exist to develop average time-to-pregnancy)13,153,154. Thus, semen quality is an indirect,
evidence-based pre-conception guidelines for men to improve albeit imperfect, measure of fertility. Of direct relevance to the poten-
reproductive health, fertility status and the health of children. tial of environmental factors that affect male fertility (Box 1, question 5)
Valid concerns exist that the use of cannabis products (another are data suggesting that sperm output in men has declined in past
example of a modifiable risk factor) could negatively affect fertility. Can- decades. A potential decline was suggested in the 1970s155,156. This
nabis is the third most widely used recreational drug worldwide, and is observation was strengthened in 1992 in an analysis of sperm count
predominantly used by men of reproductive age145. Detrimental effects data from studies published between 1938 and 1990 that suggested
of regular cannabis use (more than once per week) on fertility include that sperm counts had indeed declined157. In a meta-analysis pub-
reduced sperm motility, and concentration that is associated with lished in 2017 (ref. 158), including data from 42,935 men who provided
impaired functioning of the hypothalamic–pituitary–testicular axis146. semen samples between 1973 and 2011, a 1.4% per year decrease in
A particular worry is that with the growing legalization of cannabis, use sperm concentration and 1.6% per year decrease in total sperm count
could increase and whether these negative effects on fertility are per- was observed among men from the general Western populations.
manent or will cease with use is not known. At a minimum, appropriate The trends did not differ among studies from Europe, Australia and
health warnings should include potential for impaired fertility. America and continued up to the end of the study period, indicating
The focus of the field to date has largely been on EDCs, but the same that the decline is contemporary158. Other studies were subsequently
arguments apply to any environmental pollutant within the environ- published159–161, among them a meta-analysis of Chinese data show-
ment or food chain. An emphasis on answering these questions has ing that sperm concentration and total sperm count within an ejacu-
added benefits: the effects might be reversible, providing a genuine late from healthy Chinese men had decreased between 1981 and 2019
hope of improving human (and other animals) male reproductive (ref. 159). The decline was primarily observed in men from Northern
health and the possibility could exist to remove the contaminating China, suggesting regional variations within the same country. Data
factors. from multiple countries within Africa showed evidence that sperm
Considerable challenges exist for the field of andrology, govern- count decreased in the African population from 1965 to 2015 (ref. 162)
ment regulators and funding agencies that need to be addressed: first, and results of a study including fertile men in India showed a small
implement restricted use, replacement or bans on chemicals that downward trend in sperm concentration during a 37-year period
have been demonstrated to compromise male fertility. For example, between 1993 and 2005 (ref. 163). For the latter two studies, and several
policies need to be enacted that tightly regulate the use of EDCs, or others, the potential selection biases (such as including men with infer-
other repro-toxicants, in products and their contamination of the tility in trend analyses) might be pronounced. However, in a follow-up
environment, while investing in research to develop safe chemical analysis of the meta-analysis from 2017, a sufficient number of studies

Nature Reviews Urology


Expert recommendation

Box 2

Considerations for men’s health and fertility throughout life


1. The developing male (conception to birth) offered in school should include information on lifestyle factors
During embryo and fetal development several crucial periods of that can influence health and future fertility.
gonad differentiation occur that are susceptible to environmental 4. Young adults (~18 to ~24 years old)
exposures in utero and postnatally. Primordial germ cells, the Annual check-ups are often missed at this age, yet are key for
precursors of gonocytes, form during week 7 and persist during long-term health including fertility. General practitioners should
the fetal and neonatal stages. Male sexual differentiation is include discussion of factors that influence health and fertility,
dependent on androgen production by the fetal Leydig cells including obesity, cannabis use, smoking and alcohol.
and the spermatogenic support cells, the Sertoli cells. The fetal 5. Future fathers (~25 to ~40 years old)
period is marked by epigenome reprogramming of the germ Future fathers should consider lifestyle factors (such as diet,
cells and cellular differentiation. These cellular processes are prescription and recreational drug use, alcohol and hot tub use)
susceptible to environmental perturbation by factors such as that could influence fertility and time to conception. Men who
maternal diet or endocrine disruptor exposure. Such exposures in have or will engage in occupations associated with exposures
turn could affect lifetime spermatogenesis, susceptibility to testis that are hazardous to fertility (for example, firefighters, pilots,
cancer and fertility. Long-term follow-up studies are needed for armed forces, welders) should be informed of the potential
babies conceived by medically assisted reproduction. negative effects and be offered sperm analysis to establish
2. Childhood (birth to 12 years old) baseline fertility. Sperm freezing as a means of protecting future
Childhood is a period of testicular growth. Research on fertility should be discussed.
determinants of testicular growth and the prevention of 6. Middle age (~40 to ~50 years old)
spermatogenic failure is needed including the influence Men have a biological clock, with fertility declining past the age
of factors such as childhood obesity. Prepubertal boys treated of 40 years. Reduced sperm quality is associated with increased
for cancer are at increased risk of impaired fertility in adulthood. time to conception and rates of miscarriage. Reduced fertility
Chemotherapeutic agents can be gonadotoxic and can be can be associated with comorbidities, be an indicator of poor
causative of adult infertility or sterility. Methods are currently overall health and cardiovascular disease, and is associated with
being developed for fertility preservation in prepubertal and increased risks of prostate cancer.
peripubertal boys. 7. Older men (>50 years old)
3. Teenagers (13 to ~17 years old) Pregnancies conceived when the father is >50 years old are at a
Puberty marks the onset of spermatogenesis, which is fuelled high risk of miscarriage and obstetric complications. Children
by the differentiation of adult Leydig cells that are responsible conceived when their father is older are at an increased risk of
for androgen production throughout adult life. Health education autism and schizophrenia.

from Asian countries have now been published and a decline in sperm of MAR is reasonable166. Thus, as a field, establishing well-designed
count among unselected men from Asia was shown. Furthermore, studies that can be used to describe the current situation and longitu-
the data also showed that the decline in Western countries continued dinal future measures and investigating potential reasons for changes
and perhaps actually at an accelerated pace compared with what was in sperm numbers are essential. Equally, ‘surveillance populations’ in
previously suggested164. Given the rapidity of this change, the causality which semen samples are systematically analysed using reproducible
must be driven by environmental and lifestyle factors; if so, the hope methods from defined geographic regions need to be established
of them being reversed exists. across the globe. Recognizing that population profiles do not remain
Although not ideal with regard to study design, the existing analy- static over time152, assessing cross-sectional regional differences in
ses of historical data have been, up to this point, the best that could sperm output might further enable the indication of environmental
be done, and the results point to adverse temporal trends in sperm and lifestyle factors that compromise or improve reproductive health.
numbers and, by extension, a decline in male reproductive health, at Since the 1990s, cross-sectional, standardized and coordinated
least among Western and Asian men. However, this issue continues studies have been conducted to investigate the sperm counts of men
to be an area of debate and ongoing careful analysis is required165. from the general population (unselected regarding fertility status)
Furthermore, and as we have attempted to emphasize, such declines primarily in western Europe, the USA and Japan19,167–171, and minor geo-
cannot be strictly translated to the binary definition of fertile or infer- graphical differences were detected in medians of sperm concentration
tile but are rather an alarming suggestion that environmental factors and total sperm counts. When these studies are interpreted according
are adversely affecting the chances and/or frequency of infertility and to the association between sperm number and the likelihood of achiev-
other forms of reproductive tract disease (Box 1, question 5). Given the ing pregnancy13,153,154, they indicate that a substantial proportion of men
normal range of human sperm output even in healthy populations, from general populations now have sperm outputs that raise concerns
and the increased age at which men and women are now starting their for their fertility or infertility150,166,172,173. Given the association between
families, assuming that an increased number of men will be in need the number of sperm, their structure and function in an ejaculate, the

Nature Reviews Urology


Expert recommendation

disease burden, and early mortality174 (Box 1, question 8), these data applicable. Overall, determining the health economic impact of poor male
also raise alarm about the potential consequences for other areas of reproductive health is urgent.
the health system.
Answering the question of whether male fertility is declining, or Is disease burden and mortality comparable?
hopefully with intervention improving, will require the establishment Whether men with infertility have a higher disease burden than fertile
and analysis of longitudinal prospective studies to document the men and whether they die younger is the eighth question to address
current situation and assess causes of potential decline in male fertil- (Box 1, question 8). Growing evidence indicates that male infertility is an
ity. These studies need to include men from geographically diverse indicator of poor overall health and/or lifestyle and could be predictive
locations, and take into account migration patterns of the human of future disease. Study results have demonstrated higher incidences of
population. cancer, metabolic and cardiovascular disease in infertile men, who have
more comorbidity at the time of evaluation, than fertile men or men with
The economic burden of male infertility semen quality within normal ranges6,181–183.
Seventh, what the economic burden of male fertility is needs to be The mechanisms linking male fertility and health remain unre-
discerned (Box 1, question 7). The provision of a comprehensive solved, but study results have demonstrated that men with infertility
health-care system is expensive, and inevitably governments will be are at increased risk of certain conditions: investigations have linked
forced to make decisions as to which treatments and procedures should impaired semen quality or a diagnosis of male infertility with increased
be funded, and what research priorities should be supported based on risk of cancers including testis184–186, prostate186–188, melanoma and
economic imperatives. Thus, a fundamental challenge is to determine lymphoma189–195. Non-malignant diseases have also been examined.
the economic health effects of poor male reproductive health, which Increased incidence of diabetes, hypertension, and heart disease have
is a difficult challenge. been reported in the years following an infertility evaluation even
Limited economic assessments of fertility broadly (such as man- after accounting for any baseline comorbidities196–199. In addition, an
aging population declines using MAR techniques in Japan175) and increased risk of hospitalization has been reported among men with
estimates of the multibillion-dollar value of the MAR industry exist. reduced semen quality200. Moreover, the association between semen
However, for male infertility, only one economic impact assessment has quality and hospitalization was independent of, and stronger than,
been published and this study relates to the effect of EDCs176. This analy- the links between hospitalization and obesity, smoking and educa-
sis is important as it enabled identification of a 40–69% probability of tion, which are all established drivers of health201. Finally, results of
phthalates leading to 618,000 additional MAR procedures in Europe, several studies have demonstrated an association between male infer-
costing €4.71 billion annually. In the USA, 240,100 MAR procedures tility and early mortality202–206: a dose–response relationship exists,
annually were attributed to EDCs at a cost of US$2.5 billion177. These whereby the increasingly severe male infertility (reflected by poor
analyses are limited by the available human studies of individual EDCs sperm parameters) increases the risk of death.
and the uncertainty of the effects of EDCs in mixtures. However, col- Given these links, several important questions arise. Most impor-
lectively, the economic impact data suggest that reducing preventable tant is the question of the clinical implications of these observations.
EDC exposures will result in substantial economic benefit. As associations do not prove causation, understanding the aetiology
Little accurate information exists on the economic impact assess- of the association between a man’s reproductive and overall health is
ment of male infertility in all of its forms, whether it be for a given imperative before giving definitive guidance. Disentangling causality
country, region or globally. Important for addressing this void is assess- from reverse causality of the links will be crucial as we develop strate-
ment related to the Global Burden of Disease modelling178. The global gies to incorporate fertility testing, including genetic testing, into the
cost of male infertility, as measured by disability-adjusted life years broad context of health for men. For example, spermatogenesis could
(DALYs), currently ranks at 232 (321,829 DALYs) on a list of 272 condi- possibly provide an easily measured assay for genetic syndromes or
tions affecting men, between hookworm (336,015 DALYs) and breast sensitivity to environmental exposures for men, in which case, early
cancer (315,126). Among countries with high sociodemographic index, semen testing might be helpful.
male infertility ranks 204 of 266 in DALYs. However, the current DALY Several plausible cause–effect and effect–cause pathway hypoth-
assessment for male infertility is suboptimal as it only presents idi- eses have been proposed to explain how a man’s reproductive poten-
opathic instances of male infertility in individuals who were unable to tial could be linked to morbidity207. One hypothesis is based on the
conceive a child after 5 years of unprotected sexual intercourse — an idea of shared genetic links, including alterations in the expression of
estimate that is not an accurate representation of the disease. DALYs are disease genes connecting fertility and health. Of the ~20,000 protein-
used to assess the effect of a disease, and achieving a comprehensive coding genes in men, 84% are expressed in the testis208–210. Of these
analysis is important. An integral part of making a high-quality global genes, at least 2,274 (11.5%) are notably enriched within the testis208, rais-
burden of disease assessment must also include evaluation of the ing the possibility that genetic variants in these genes, which alter gene
mental health effects of infertility on men179. Information is increas- expression or function, will lead to male infertility. However, few of these
ing through the use of tools such as structured surveys, for example, genes are solely expressed within the testis, raising the possibility that
Fertility Quality of Life tools180, documenting the negative effect of spermatogenesis might be the first process to fail, but it will not be the
male infertility on the individual, couple, immediate family and society. only one to fail. We predict that the timing of somatic tissue failure will
Given the diversity in cultural, socioeconomic and political be modified based on compounding factors, including environmental
structures between regions of the world, considerable variations in exposures and/or the presence of other disease risk gene variants, but
the health economic impact of male reproductive health are likely. this hypothesis is poorly explored. This idea underpins the concept of
Moreover, this effect is likely to depend on context, such as low-income male infertility as an early indicator of long-term health at an individual
versus middle-income resource settings. Thus, assessment for one as well as a population level. Validated examples of this phenomenon
region is important and informative, but it might not be globally occurring include mutations in DNA mismatch repair genes identified

Nature Reviews Urology


Expert recommendation

in men with spermatogenic dysfunction and cancer, mutations asso- for fertility preservation in patients at risk of losing their fertility owing
ciated with infertility and cystic fibrosis, primary ciliary dyskinesia to gonadotoxic treatments232. Men with no active spermatogenesis,
(including Kartagener syndrome) associated with respiratory infec- specifically prepubertal boys, cannot have semen cryopreserved.
tions, and infertility owing to flagellar dysfunction211–214. Comparable These patients might benefit from bioptic collection of immature
links between these cellular pathways and reproductive health and tissue containing spermatogonial stem cells. Testis tissue fragments
long-term somatic health are also becoming evident in women215. can be cryobanked using dimethyl sulfoxide-based cryopreserva-
Conversely, perturbations in systemic health could influence tes- tion protocols233,234. Testicular biobanking was initiated in specialized
ticular function. Systemic inflammation is understood to have direct centres and is being further developed in research networks (such as
and indirect effects on spermatogenesis136,216. These effects are affected NordFertil and Androprotect235) storing hundreds of samples from new-
by, at least in part, the key roles that cytokines have in the regulation born to adolescent ages232. The nature of the testicular stem cells and
of spermatogenesis and testosterone production, including in the their somatic niches have been extensively explored and various experi-
maintenance of an immunologically privileged state217. mental strategies have been successfully applied in animal models.
Regardless of whether a man presents with primary or secondary Germ cell transplantation, testicular grafting and testicular organoids
infertility, additional studies are needed to provide improved predic- seem promising tools for providing future avenues for stem cell-based
tive power for men, including robust mechanistic data to improve fertility preservation233. Despite this progress, several challenges remain
characterization of their metabolic, immune and genetic state. As before spermatogonial stem cell transplantation becomes routine236.
precision medicine continues to provide novel insights into disease The risk of tumour cell contamination and the low efficiency of this
trajectory, early predictions into later health problems might enable strategy renders it necessary to improve the protocols before clinical
the formulation of clinical interventions to improve fertility and avoid applications237. Techniques are needed for highly accurate detection
extra-testicular morbidity. as well as a selection of tumour cell contaminants. However, testicular
Thus, men with infertility need to be assessed comprehensively grafting and germ cell transplantation have been successfully applied
and such information needs to be included in an accessible database. in primates and their successful preclinical application suggests that a
This information should include genomic data, information on environ- positive outcome is possible in the clinical setting238,239.
mental exposures, medical history and fertility characteristics. In addi- Alternative strategies using germline stem cells for fertility pres-
tion, information should be gathered from multiple locations around ervation have been developed. The target cells can be pluripotent pre-
the globe to enable detection of broad geographical and temporal cursors, spermatogonial stem cells or induced pluripotent stem cells.
trends in fertility and health. Protocols for in vitro culture and expansion of such cells are available
and strategies for their subsequent differentiation into germ cell-like
Gamete storage and restoration protocols cells have been established233,234. To generate gametes from germline
The ninth question is whether developing robust gamete storage and stem cells, it seems necessary to bring germline stem cells into contact
restoration protocols for boys and men before other medical interven- with gonadal cells233,234. Testicular organoids have been successfully
tions is possible (Box 1, question 9). Gamete or spermatogonial stem used to generate offspring from primary cells from rodents240–242. In
cell storage is broadly aimed at providing fertility preservation (that is, mouse models, protocols to achieve full spermatogenesis ex vivo have
insurance) for fertility restoration or creation protocols for men and been established243. The strategies for in vitro spermatogenesis offer
children to avoid the consequences of either predictable or unpredict- promise for clinical application, but also concern for its safe and legal
able losses of fertility. Foreseeable losses of fertility include patients application244.
receiving chemotherapy and/or radiotherapy218 or men undergoing Another aspect of gamete storage to consider is sperm freez-
orchidectomy during the final stages of gender reassignment219,220. ing, that is, gamete for fertility preservation unrelated to a medical
A considerable percentage of these men would become infertile or condition245. Probably the most frequent application of non-medical
sterile and might not be able to benefit from standardized procedures freezing is pre-vasectomy, for which about 20% of men might sub-
for fertility preservation221. From the preventive viewpoint, when indi- sequently use their frozen sperm246,247. In this case, men can avoid a
viduals can produce an ejaculate and spermatozoa are available, or if reversal operation or a TESE followed by an in vitro fertilization (IVF) or
sperm can be extracted from tissue obtained by testicular tissue biopsy, ICSI procedure224. Other potential users of non-medical sperm freezing
the option of sperm banking is desirable, well-established and provides a include men in positions for which a high risk of injury exists, such as
good cost–benefit equation222. Sperm quality declines with cryopreser- the armed forces, firefighters, and other professions with related toxic
vation and thawing, but almost all banked semen samples provide a good exposures248. In addition, compelling data indicate that pregnancy
chance of achieving fertilization and resulting in a live birth when ICSI outcomes and mutation load are increased in older men and their
is used223, and the same is true for spermatozoa obtained using TESE224. children249–253. These risks could be related to age-induced changes
The main limitation with sperm cryopreservation is misinfor- in the heritable sperm epigenome254–256. Conversely, any use of such
mation, despite evidence-based and standardized protocols for use technologies must be accompanied by a discussion about the possibil-
by oncologists having been put forward to cryopreserve human ity that gamete storage and MAR are also associated with conception,
spermatozoa225–227. Understanding of the potential value of such pro- pregnancy and health risks for future offspring257. Thus, the risks of
cedures is limited and treatment plans fail to include oncology and social sperm freezing must be carefully balanced.
fertility specialists specifically when offering and performing fertility Complex ethical and legal questions around gamete preservation
preservation procedures in young patients225,228. Continuous improve- remain in some parts of the world. For children, informed consent for
ments in these aspects of patient care are being implemented, but tissue retrieval surgery when the protocols to use that stored tissue
ongoing efforts are needed221,229–231. to produce sperm have not yet been developed is an issue232. Unan-
Concerning testicular stem cell-based strategies for male fertility swered ethical questions remain related to the length of cryostorage;
preservation and restoration, spermatogonial stem cells are target cells healthy live births have been attained from samples frozen beyond

Nature Reviews Urology


Expert recommendation

two decades258. Last, the experimental nature of fertility restoration was associated with differential methylation of regions of DNA that
or creation approaches in combination with the lack of consensus were in what is referred to as ‘the intermediate range of methylation’
as to what evidence should be available before granting clinical use (20–80%), and occurred at genes implicated in neurological devel-
remains a problem. opment and function285. Similarly, urine phthalate concentration
In summary, within the area of gamete preservation many ques- (a biomarker of phthalate exposure) has been connected with dif-
tions remain relating to protocol development, patient and clinician ferential methylation of sperm DNA at genes implicated in develop-
education, and the ethics and legal regulation of such procedures. This ment and cell function117. Moreover, a modest number of differentially
debate should be led by the field of andrology and preferably occur methylated regions are correlated with low blastocyst quality117. These
across borders and continents. studies in men, among others, offer proof-of-principle that the sperm
methylome is responsive to the environment.
The consequences of epigenetic change In humans, research regarding the intergenerational heritability
The tenth question is, do changes in epigenetic processes lead to male of complex diseases (such as diabetes, metabolic disorders and almost
infertility or have inter-generational consequences (Box 1, question 10)? certainly infertility) via the sperm epigenome in relation to pollutants,
The epigenome refers to the biochemical modifications linked to and diet and health is limited to epidemiological studies and is compli-
associated with DNA that affect chromatin (DNA) organization and gene cated by population variability286. However, considerable advances
expression259. The epigenome includes three known layers of biochemi- have been made using rodent models287–289. Conservation of the epi-
cal information: first, the modification of the nucleosome proteins genetic landscape in mice and men indicates that mice are a reliable
(the histones including the protamines) by the addition or removal of model for identifying epigenetic factors in sperm that are sensitive to
methylation, acetylation, and phosphorylation among others; second, environmental exposures and escape epigenetic reprogramming in
DNA methylation that occurs at the 5′-position of cytosine residues the embryo, for example, those likely to affect offspring health289–292.
within CpG dinucleotides; and third, small non-coding RNAs260–262. A range of environmental stressors, including poor diet, toxicants
DNA can be thought of as storing genetic information (analogous to and trauma, affect multiple facets of the sperm epigenome and have
computational hardware) and epigenetic processes provide the instruc- been associated with negative effects on offspring health286–289. As an
tions and architecture to read the genome (analogous to computational extension of the conservation of the mechanism between humans and
software) and are crucial for development, through the provision of rodents, studies conducted in mice have demonstrated that the sperm
cell-specific gene expression. Changes in epigenetics are associated epigenome is responsive to diet at functional loci that correlate with
with numerous human diseases, including cancer, diabetes and male offspring phenotypes. In male mice, diets with altered micronutrients,
infertility263,264. including either folate deficiency or folate supplementation293,294,
Studies in mouse models have clearly demonstrated that the com- affected gene-activating histone methylation in sperm (histone H3
plete disruption of pathways that affect histone modification, DNA lysine 4 trimethylation; H3K4me3) and DNA methylation in sperm.
methylation and each of the piwi-interacting RNA, microRNA and Moreover, these alterations to histone methylation have been shown to
small RNA pathways lead to sterility261,265–269. Genetic studies are rare be retained in the pre-implantation embryo and to be linked to aberrant
in humans and in need of replication, but available results suggest embryo transcriptomes and developmental abnormalities288. Similarly,
that similar losses of function in men can cause infertility270–274. Of a low-protein diet in male mice was associated with variation in sperm
great relevance, evidence indicates that environmental insults perturb non-coding RNA content295,296, gene-silencing histone methylation
the establishment of the sperm epigenome during spermatogenesis (histone H3 lysine 4 trimethylation (H3K9me3)) in sperm296 and DNA
and that such changes are associated with infertility and poor MAR methylation297. High-fat diets influence sperm H3K4me1 (ref. 298),
or natural conception outcomes, but this possibility is difficult to H3K4me3, non-coding RNAs288 and DNA methylation,299,300; further-
investigate in men275. Indeed, differences in sperm DNA methylation, more, offspring sired by obese males showed metabolic disturbances,
histone modifications and sperm RNA content have been linked to reduced lean muscle mass, increased fat deposition and in some studies
poor fertility276,277. For example, a subgroup of men presenting with these paternal effects were transmitted transgenerationally, showing
oligozoospermia and infertility produced sperm with altered DNA that paternal factors can affect child metabolism298–300.
methylation278–280. How these changes arise during spermatogenesis Investigations have begun to identify specific mechanisms that
or epididymal sperm maturation is unknown; however, evidence con- underpin the epigenetic transmission of environmental exposures
necting paternal health to an altered sperm epigenome and infertil- from the father to the embryo288,289,295,301. Importantly, these mecha-
ity is accumulating. In a study investigating the connection between nisms seem to be conserved between mice and men. For example,
obesity and the sperm epigenome, DNA methylation was assessed in retained histones are conserved across species and found at important
sperm before and after bariatric surgery. Remarkably, an altered sperm regulatory genes involved in housekeeping, spermatogenesis and
epigenome was detected after weight loss from bariatric surgery281,282. development and have been determined to be responsive to envi-
This study did not explore the connections of an obesity-altered ronmental signals and implicated in epigenetic inheritance289,290,292.
sperm methylome with fertility283, but an elevated BMI is known to be Likewise, DNA methylation has been shown to be responsive to envi-
associated with an increased risk of infertility284. Importantly, and ronmental exposures in sperm from rodents and men. For example,
given that production of sperm is continuous, these observations in a rodent model of DDT exposure DNA methylation and nucleosome
suggest that a potential opportunity exists to rapidly modify the sperm retention was altered in sperm from F1 males exposed in utero302.
epigenome and improve fertility through the development of lifestyle Similarly in two DDT-exposed populations of men, Greenlandic Inuit
interventions. and Vhavenda South Africans, common alterations were evident in
Additional evidence that DNA methylation of the sperm epig- the sperm epigenome at the level of DNA methylation and chromatin,
enome could be responsive to diet was demonstrated when men con- at genes implicated in fertility and embryo development. Moreover,
sumed supplemental folic acid285. High-dose folic acid supplementation epimutations occurred in regions that are predicted to persist in the

Nature Reviews Urology


Expert recommendation

early embryo and were associated with embryonic gene expression and studying long-term health outcomes and could ultimately encourage
population health (based on data discussed in one preprint article)303. the use of less invasive MAR strategies than ICSI. Some long-term health
The relevance of intergenerational and potentially transgenerational outcomes, such as neurodevelopment and growth, seem similar in
epigenetic inheritance for human health remains unresolved and children conceived using ICSI and in naturally conceived children330.
should be a priority to understand. First reports on the reproductive health of men conceived using ICSI
In summary, alterations to the sperm epigenome are associated suggested impaired spermatogenesis compared with those conceived
with human infertility and might have negative consequences for off- naturally331,332, whereas results of a larger subsequent study are reas-
spring conceived naturally or via MAR techniques. This area is crucial suring, showing similar sperm output and excluding major differences
to study as improved understanding of environmental triggers and in reproductive health333. However, all studies to date have contained
mechanisms will offer an opportunity to avoid, and potentially reverse, relatively small numbers of participants, meaning that concluding
infertility. Questions that are currently outstanding include whether anything about disease transmission from fathers to sons is difficult.
epigenome signatures are consistent among men with infertility and if In relation to female fertility, antral follicle count and reproductive
can they be used to diagnose or predict infertility; whether the changes hormone levels were similar in young adult women conceived using ICSI
to the sperm epigenome are a direct consequence of lifestyle and/or and those conceived naturally in one study334. Some studies including
environmental exposures and whether causality can be established; children conceived using IVF have suggested poorer cardiovascular and
what the effects are of an abnormal sperm epigenome on the embryo metabolic profiles than those conceived naturally257,335, but long-term
and long-term offspring health; how an abnormal sperm epigenome and data are sparse and few studies have examined the metabolic health of
the heritability of disease are connected; and whether an abnormal children conceived using ICSI336,337. The comparison groups for almost
sperm epigenome can be reversed to improve fertility. An extension all of these studies are children of fertile parents, making it difficult to
of this concept is whether changes (shifts towards the norm) could be differentiate between underlying genetic factors, exposures associated
used as a biomarker for improved gamete quality before MAR. with infertility and effects of the MAR procedure. Few studies have
explored the contribution of underlying infertility compared with the
Health outcomes for children MAR procedure338–341, and no studies have examined paternal fertility
What the long-term health outcomes for children born to men with status as a source of potential health differences.
compromised fertility are and whether these consequences vary with At present, whether observed differences in health outcomes
conception via natural or via MAR is the 11th question to answer (Box 1, between those conceived using ICSI and those conceived naturally
question 11). The long-term health consequences of MAR into adult- are caused by treatment factors, parental characteristics, or related to
hood remain unknown. Moreover, few studies have considered the adverse obstetric outcomes associated with MAR treatment is unclear.
relative contribution of parental infertility and the MAR procedure to Careful examination of the potential role that underlying paternal
offspring health outcomes or the effects of paternal infertility. These infertility and MAR procedures have on outcomes at both the molecular
questions are crucial, the answers to which might alter clinical practice. and the phenotypic level is required. Male infertility is a multifactorial
A large percentage of male infertility is likely to be genetic in origin heterogenous condition with potential genetic and environmental
and environmental exposures will be risk factors for infertility and/or aetiologies8. Thus, consequences for children’s health will depend on
alter sperm epigenetic content, each of which could affect offspring the underlying cause. Large prospective longitudinal studies with unbi-
health. Thus, concerns for children born as a result of MAR techniques ased control groups that stratify men by underlying molecular cause
include potential intergenerational inheritance of infertility beyond of infertility will be crucial in untangling this issue. Whole-genome and
the genetic examples7 (Box 1, questions 3 and 4) and the broad effects epigenome analyses of parents and children born as a result of using
of poor-quality spermatozoa on somatic aspects of offspring health, MAR techniques will almost certainly be needed. To distinguish the ICSI
including aspects beyond fertility. Complicating the picture are epige- technique from background paternal infertility effects, two naturally
netic changes arising from MAR procedures themselves304–307. What the conceived control groups, ideally from the same source population,
consequences of ICSI compared with IVF are for epigenetic composition are required: naturally conceived children from men with subfertility
in embryos and offspring is a notable question. men and those from age-matched fertile men. Detailed medical and
Changes in imprinted gene methylation and expression have been clinical histories for both parents are required, including evidence of
identified in cord blood, buccal swabs and placental tissue of children exposures to toxins and drugs, reproductive histories and individual
conceived using MAR techniques308–313, but few high-resolution molecu- MAR procedures used.
lar studies have been performed and their results are conflicting314–319; To improve understanding of the mechanistic underpinning of
furthermore, epigenetic variation at birth could resolve later in late-onset health outcomes (for example, metabolic disease and can-
life306,320. Epigenetic changes linked to MAR have been associated with cer), prospective studies should be accompanied by the collection
underlying parental infertility and the technique itself, but no definite of appropriate biological samples to enable high-resolution genetic
answers have been obtained307,321,322. and epigenetic studies to be done. Samples to be considered include
In terms of short-term health outcomes, children conceived using triad DNA (mother, father and baby), sperm, cord blood and placenta,
IVF or ICSI are at an increased risk of adverse perinatal outcomes, with some samples taken at multiple time-points (such as from the
congenital malformations and imprinting disorders compared with child throughout life). For epigenomic studies, the cell make-up of
naturally conceived children305,323–328. Assessments of short-term health samples must be considered: for instance, lack of contamination
outcomes associated with MAR and male factor infertility using large of sperm samples with somatic cells is crucial for the interpretation of
cohort studies have started. For example, evidence of increases in birth any comparison of DNA methylation profiles between fertile men and
defects associated with MAR has been reported, which was further those with infertility342,343. Effects of paternal age on the sperm genome
increased with the use of ICSI and again still with ICSI and male factor and epigenome344 suggest that having a baseline semen analysis (with
infertility329. These results strongly support the rationale and need for sperm cryopreserved) carried out routinely in early adulthood might

Nature Reviews Urology


Expert recommendation

help clinicians to distinguish between genetic, epigenetic, age and/or particularly in the mature human spermatozoon, need to be identified,
exposure-related effects when men return later in life with fertility which will greatly facilitate development of compounds manipulating
issues. Animal studies are necessary to help to extract combinatorial function355. For this possibility to be realized, considerable investment
effects (for example, individual techniques, interactions between in defining the processes that underpin normal male fertility is crucial.
parental infertility and techniques such as culture to blastocyst or Thus, availability of new male methods of contraception remains an
cryopreservation) and design preventive strategies (such as methyl ambition rather than an imminent reality. The hormonal approach has
donor supplementation of men with subfertility). a track record in clinical trials, but non-hormonal approaches might
be more successful if they have a rapid-onset, universally active but
Additional male contraceptives reversible approach. Global contraceptive development initiatives
Whether we can develop additional male contraceptive methods is the such as Family Planning 2030 can minimize or even fail to recognize
12th question to answer (Box 1, question 12). The development of steroid the involvement of men as vital participants in both existing and future
pharmacology since the 1950s has led to a wide and growing range of contraceptive availability and development, despite its clear alignment
highly effective hormonal methods of contraception for women, with with United Nations Sustainable Development Goals 3 and 5. As with
hormonal intrauterine systems and progestogen-only oral and implant female contraception, clearly, a huge unmet need and market exist,
preparations joining older methods in providing choices for women345. which requires the application of modern scientific approaches356.
This progress is in stark contrast to the absence of new contraceptive However, the outlook is optimistic, as, following substantial invest-
methods for men, despite the development of hormonal approaches ment by the Bill and Melinda Gates Foundation and the emergence of
to male contraception having a comparably long history346. The only organizations such as the Male Contraceptive Initiative, funding in this
commercially available reversible male method, condoms, remains area is improving, which will accelerate translation. What is required
an invaluable option both for contraception and for the prevention of now is a substantial research and development funding portfolio from
sexually transmitted infection, and in terms of ‘ever use’ can be con- a wider collection of organizations than currently support this field.
sidered the most widely used method of contraception that exists.
Surveys continue to demonstrate that a considerable population of men Effective communication
would welcome and use novel methods, and perhaps equally impor- Finally, the 13th question is how andrology as a discipline can com-
tantly, women would support and welcome their partner in using a municate effectively with the public, health professionals, and funding
male contraception method347. Undeniably, men are much involved agencies (Box 1, question 13). Men, unlike women, are often not encour-
in contraception, with the likelihood of increased engagement in new aged from a young age to be conscious of their future fertility and role as
methods that might additionally offer health benefits analogous to the parents, making approaches to fatherhood circumstantial357. Temporal
non-contraceptive benefits of many female methods347. isolation of men’s reproductive lives from their development of health
Whether new male-based contraceptive methods can be devel- literacy, health-care-seeking behaviours and lifestyle choices prob-
oped is a pertinent question. Trials of hormonal contraceptive ably contribute to behaviour that compromises fertility (for example,
approaches have been conducted for several decades, and the land- the perception of gains from androgen abuse at the potential cost to
mark WHO-supported studies of the early 1990s showed that a hormo- fertility358). However, behaviours that place future reproduction at risk
nal approach could provide effective and reversible contraception348. (such as smoking, sexual practices that increase the risk of contracting
Current approaches are based on progestogen and testosterone com- sexually transmitted infections and increasing age at parenthood) are
binations, with the progestogen providing most of the necessary gon- evident in both sexes251,359. An understanding of the motivations for
adotrophin suppression with ‘add-back’ physiological testosterone such behaviour and how to mitigate them is needed.
replacement to support androgen-dependent functions in men349. The Health services can improve provision of men’s needs in various
National Institute of Child Health and Human Development in the USA ways360, but men’s own behaviour needs to change if their fertility is
now leads the way in funding major trials of existing compounds and to be optimized. Men tend to monitor disease symptoms themselves
early studies of novel steroids350. The involvement of pharmaceutical for a period before seeking health care361, which delays presentation
companies in large trials in the early 2000s351 provided encourage- until late stages of disease. Ailments that are debilitating, or considered
ment that this approach was finally going to result in a product as well serious or amenable to effective treatment are most likely to lead to
as improving methodological rigour (double-blinded, randomized, help-seeking behaviour361. For example, Australian men consider infor-
controlled trials) and technical innovations (such as centralized mation about fertility unnecessary unless a problem arises362. Thus,
semen analysis and fluorescent detection of sperm), but unfortunately consideration of male infertility as an acute problem that is solvable
changing business priorities meant that this hope was short lived. using MAR is likely to appeal to men. This approach might achieve a
Given the obvious downside of interfering with the function of successful pregnancy, but it could be a missed opportunity to address
testosterone in male non-reproductive physiology, non-hormonal underlying behavioural issues that probably damage multiple aspects
approaches would probably have substantial advantages. of health in addition to fertility.
Such approaches could target spermatogenesis, sperm maturation Consideration of male infertility as a chronic, rather than acute,
in the epididymis, ejaculation, and/or sperm function352 in the female disease state might help to facilitate deliberate approaches to
reproductive tract (and so, include methods taken by the woman, or health-care provision that empower and motivate changes, such as
even by either men or women). Almost certainly because of a virtual the Adaptive Leadership Framework for Chronic Disease, to improve
absence of funding for this area, progress with non-hormonal contra- men’s overall health and well-being, and their roles as fathers363.
ceptive approaches has been slow and although several well-established Information about men’s attitudes to becoming parents, their
targets exist, such as retinoic acid receptor-α353, minimal progress fertility knowledge and responses to infertility is lacking364. Men and
has been made in developing compounds for testing in clinical trials. women desire parenthood equally but their knowledge about fertility
Moreover, high-confidence targets are sparse350,354 and more targets, in men, as a generalization, is deficient365–368. The limitations of men’s

Nature Reviews Urology


Expert recommendation

knowledge, and the misconceptions they hold, probably influence and quality resources they can recommend to their patients should,
delays to fatherhood367. therefore, be a priority381. The necessity for education of health pro-
Men’s fertility awareness (understanding of reproduction, risk fessionals is illustrated by evidence suggesting that the language and
factors for infertility and family planning) seems to be positively asso- approach used by them could confuse men rather than inform them382.
ciated with their education level369 and many men are interested in A reproductive life plan-based counselling approach (Box 2),
improving their knowledge of male fertility and reproductive health370. which centres on identifying reproductive intentions and strategies
Improving men’s knowledge about their fertility, and awareness of to achieve them, is effective for raising men’s awareness of reproduc-
the risks associated with delays in starting a family would be likely to tive health and fertility379, but is reliant on investment in education for
motivate behavioural changes to optimize fertility and family plan- health professionals’ development and rollout of patient education
ning. Notably, female fertility declines more rapidly than male fertility and resources.
with age371. Changes in health policy and research funding will be needed to
The effects of social expectations, career paths and individuals’ properly address male infertility, including the provision of additional
economic circumstances on reproductive behaviours must also be funding to define the fundamental mechanism underpinning male fer-
addressed365, but the best ways of achieving the changes required in tility. These require a comprehensive assessment of economic impact,
these areas are unknown. Fertility awareness might be improved by without which making structured arguments for investments in diagno-
the online delivery of fertility education372, which could cater to young sis, research, prevention and treatment will be difficult. A coordinated
men’s use of irony and humour as central learning mechanisms for effort from all interested parties is needed for such change11.
health-related social media373. Public information that causes alarm
about infertility is less effective at educating people than less sensa- Recommendations
tionalized messages, and can create feelings of susceptibility and worry To improve the diagnosis and treatment of male infertility and, ulti-
in individuals374. The suggestion by young Danish men that fertility mately, prevent infertility, we make ten recommendations for action. The
awareness should be a mandatory component of education in primary global implementation of such strategies promises to revolutionize
and secondary schools, with more personalized education provided the understanding and practice of andrology and improve male health
closer to when it is relevant, is sensible375. Unfortunately, education at a population level.
about fertility is not well covered in sex education programmes for 1. The formal acknowledgement by health-care systems, insur-
school children376. Certainly, the knowledge that individuals’ general ance companies and ultimately the public that male infertility
health and well-being influences fertility should be delivered before is a common and serious medical condition and that patients have
young people make decisions about behaviours that influence their a right to meaningful diagnoses and treatments. Inherent in this
chances of becoming parents later. statement is the acceptance that many hundreds of aetiologies
We need to understand what happens before men become patients could exist and that different types of infertility will require
with infertility. Part of this awareness relies on understanding basic different treatment strategies, that is, personalized medicine.
reproductive physiology, the origins of infertility and removing precon- 2. The establishment of a network of national registries and
ception risk. Awareness and effort in the provision of pre-conception biobanks containing standardized clinical and lifestyle informa-
care to men is increasing377,378, but it is still in its infancy. An important tion, and tissue from fertile men and those with infertility, their
feature of current recommendations is a reproductive life plan, which partners and children. Such registries should be linked to health
can improve men’s fertility awareness379 and provides an opportunity data within national health-care systems. They should be dis-
for assessment of risk factors for infertility, and timely intervention if tributed across the globe, co-ordinated to enable data sharing,
needed377. A shift to preconception care for men380 presents myriad and with time, expanded to include information and tissue from
research opportunities to improve understanding of male infertility. multiple generations. Such repositories should become epicen-
Such information will provide key indicators of male reproductive tres for studying the effects of epigenetic, genetic, environmen-
health, including opportunities to continually assess fertility pre- tal and lifestyle factors on men’s health, and for the monitoring
diction and examination of diagnostic markers in different regional of temporal changes in men’s health, including sperm output.
populations20. 3. The implementation of protocols and incentives by health-care
Effective ways of optimizing men’s fertility before conception are systems and insurance companies for clinics to adhere to clinical
largely unexplored, and the reproductive outcomes of any resulting guidelines and the centralization of de-identified information
interventions are unknown. Research funding agencies, health insti- to accelerate discovery.
tutions and governments must respond to the need for knowledge in 4. The expanded use of international collaborative research net-
this area to ensure that interventions are evidence based, acceptable, works to undertake epigenomic, genomic, lifestyle and environ-
effective and affordable. An example of the work required is provided mental exposure studies to define aetiologies of male infertility.
by the Australian Male Infertility Exposure Study, funded by the Austral- Such studies should include a diverse range of patients to provide
ian Medical Research Future Fund. The aim of the study is to understand improved interpretations of cause–effect relationships.
how health, lifestyle and environmental factors can influence male 5. The rapid integration of whole-exome and whole-genome sequ­
infertility and treatment outcomes for male infertility. enc­ing into the diagnosis of male infertility. The results of such
Young men and women consider that their general practitioners screening should be pooled globally to accelerate discovery and,
are an appropriate and desirable source of fertility information367; ultimately, the formulation of medical interventions.
thus, education and resources for primary care providers are essential. 6. The formulation of additional tests capable of enabling the dis-
Unfortunately, many general practitioners have limited knowledge cernment of the aetiology of male infertility and/or the success
about male fertility, which they appreciate is a barrier to discussing of highly invasive MAR procedures with increased accuracy and
fertility with their patients: education and information for clinicians, precision.

Nature Reviews Urology


Expert recommendation

7. The rigorous testing of compounds for negative effects on male other animals, is affected by genetics and environmental exposures,
fertility and the sharing of results globally. The most urgent need often in combination, is a necessity; also, depending on the timing of
is in the area of EDCs, including the need to test complex cocktails such exposures (acute and chronic), fertility could be compromised
of compounds. In parallel, regulatory policies that tightly regu- to differing extents.
late the use of EDCs and other repro-toxicants in products, the The size of the tasks is considerable, but we are not starting from
workplace and the environment must be enacted and adhered zero. Many laboratories have made impressive advances in defining
to, while investing in research to develop safe chemical alter- how sperm are produced, including the development of in vitro systems
natives. Such alternatives must undergo rigorous testing using that recapitulate many of the in vivo pathways, facilitating the rapid
model systems of relevance to human reproduction before testing of compounds on germ cell health. The adoption of exome
introduction on the market. and genome sequencing technologies is revealing a rapidly expanding
8. The rigorous testing of MAR treatment strategies via high-quality number of genetic causes of human infertility and the use of animal
clinical trials before their integration into standard clinical prac- models is shifting discoveries from association to proof. If we are able
tice. Such testing should span the gamut of pharmaceutical to merge these data with those of environmental, lifestyle-induced
products, dietary supplements and MAR ‘add-on’ protocols. epigenetic changes, the chance of developing interventions to avoid
9. The development of education and public health campaigns to or reverse infertility is good, or at a minimum, realistic predictions of
normalize the discussion of male fertility and infertility across the consequences of such changes on the health of children conceived
diverse population demographics. Such campaigns should span using MAR and the long-term health of the men with infertility will be
the life course of men, be easily understandable and accessible possible. As exemplified by genetic studies, many of the necessary
and linked into health-care systems to encourage earlier and technologies already exist.
more frequent engagement of men in health seeking than is Arguably the two biggest challenges to progress are the necessity to
currently the case. generate large tissue biobanks and datasets across the globe — certainly
10. Improved and continuous education of the health-care work- beyond those that currently exist, which are skewed towards men of
force in the broad area of male reproductive health. This endeav- European descent — and for the ‘re-education’ of the medical profes-
our includes an emphasis on training during medical school, the sion and the public to consider male reproductive health as a staple
provision of on-going training and resources to primary care of health care, rather than something to be considered only when
physicians and nurses and the integration of services across infertility is present. Overcoming these challenges can be achieved
urology, internal medicine, endocrinology, gynaecology and through coordinated action by professional societies, engagement with
paediatrics. education at all ages and effective communication with governments
and the public.
Conclusions Men’s health, from fetal development to old age, is affected by
In summary, in this Expert Recommendation, considerable gaps in what they eat, drink, breathe and touch (Box 2). Some of these inter-
scientific and medical databases, socio-economic impact data and actions are beyond an individual’s control (for example, exposure to
social awareness and education regarding male reproductive health environmental oestrogens), whereas other exposures can be controlled
and infertility are identified; we believe that if investment is made in (such as avoidance of heavy drinking and recreational drug use). Thus,
these factors, several aspects of society will improve. Scoping of the improving education around reproductive health will be a considerable
literature by acknowledged experts in the field for each question cul- tool for teenagers and young adults beginning sexual relations and
minated in the overarching conclusion that an urgent need exists for broad social interactions, for young couples considering conception
an increased focus on male fertility and infertility at the discovery level, (natural or assisted) and for adults who have delayed starting a family.
diagnostics, the development of targeted treatments and education. Educational content should be tailored to demographic and socio­
To achieve each of these formidable goals in an optimal manner, economic groups and presented in an understandable and practical
male infertility must be recognized as a common, serious and expen- way, with an emphasis on self-control, self-care and self-seeking for
sive disease, made up of many sub-types. The global andrology disci- health care. For men, these positive behaviour practices have histori-
pline, in its totality, must also work together to undertake definitive, cally been particularly challenging, so determining what men need in
well-controlled experiments to generate robust outcomes reflective order to ‘buy-in’ as active participants is important.
of diverse geographic and socio-economic populations. Doing so will With success in research leading to improved diagnostics and
have many benefits, including an ability to directly address the root treatment of male reproductive disorders, implementation into stand-
causes of infertility and potentially preventing somatic disease later ardized global health-care practices, men, their partners and future
in life. Doing so should also improve the long-term health outcomes of generations will live long and healthy lives.
children conceived naturally or using MAR techniques, and of women
for whom invasive medical interventions will be avoided. Benefits to Published online: xx xx xxxx
the health-care system, with time, will involve a decreased frequency
References
of interventions to achieve fertility (MAR). Equally, changes should 1. WHO. Infecundity, infertility, and childlessness in Developing Countries - DHS
lead to decreased somatic disease, or, at a minimum, the detection Comparative reports no. 9. WHO https://www.who.int/publications/m/item/
infecundity-infertility-and-childlessness-in-developing-countries---dhs-
of infertility-associated somatic disease at an earlier time point in
comparative-reports-no.-9 (2004).
progression than currently. Shifting from a descriptive (phenotypic) 2. Forti, G. & Krausz, C. Clinical review 100: evaluation and treatment of the infertile couple.
understanding of male infertility to a molecular diagnosis is essen- J. Clin. Endocrinol. Metab. 83, 4177–4188 (1998).
3. De Jonge, C. & Barratt, C. L. R. The present crisis in male reproductive health: an urgent
tial, and ultimately, the development of targeted therapies that will
need for a political, social, and research roadmap. Andrology 7, 762–768 (2019).
address the root cause of infertility rather than its symptoms needs 4. Esteves, S. C. Evolution of the World Health Organization semen analysis manual: where
to be accelerated. Accepting that human male fertility, like that in are we? Nat. Rev. Urol. 19, 439–446 (2022).

Nature Reviews Urology


Expert recommendation

5. Boitrelle, F. et al. The sixth edition of the WHO manual for human semen analysis: 39. Tournaye, H., Krausz, C. & Oates, R. D. Concepts in diagnosis and therapy for male
a critical review and SWOT analysis. Life 11, 1368 (2021). reproductive impairment. Lancet Diabetes Endocrinol. 5, 554–564 (2017).
6. Eisenberg, M. L., Li, S., Behr, B., Pera, R. R. & Cullen, M. R. Relationship between semen 40. Young, J. et al. Clinical management of congenital hypogonadotropic hypogonadism.
production and medical comorbidity. Fertil. Steril. 103, 66–71 (2015). Endocr. Rev. 40, 669–710 (2019).
7. Lismer, A. & Kimmins, S. Emerging evidence that the mammalian sperm epigenome 41. Butz, H., Nyiro, G., Kurucz, P. A., Liko, I. & Patocs, A. Molecular genetic diagnostics of
serves as a template for embryo development. Nat. Commun. 14, 2142 (2023). hypogonadotropic hypogonadism: from panel design towards result interpretation in
8. Houston, B. J. et al. A systematic review of the validated monogenic causes of human clinical practice. Hum. Genet. 140, 113–134 (2021).
male infertility: 2020 update and a discussion of emerging gene-disease relationships. 42. Chudnovsky, A. & Niederberger, C. S. Gonadotropin therapy for infertile men with
Hum. Reprod. Update 28, 15–29 (2021). hypogonadotropic hypogonadism. J. Androl. 28, 644–646 (2007).
9. Skakkebaek, N. E. et al. Environmental factors in declining human fertility. Nat. Rev. 43. Avellino, G. J., Lipshultz, L. I., Sigman, M. & Hwang, K. Transurethral resection of the
Endocrinol. 18, 139–157 (2022). ejaculatory ducts: etiology of obstruction and surgical treatment options. Fertil. Steril.
10. Datta, J. et al. Prevalence of infertility and help seeking among 15 000 women and men. 111, 427–443 (2019).
Hum. Reprod. 31, 2108–2118 (2016). 44. Heidenreich, A., Altmann, P. & Engelmann, U. H. Microsurgical vasovasostomy versus
11. Barratt, C. L. R. et al. The diagnosis of male infertility: an analysis of the evidence to microsurgical epididymal sperm aspiration/testicular extraction of sperm combined with
support the development of global WHO guidance-challenges and future research intracytoplasmic sperm injection. A cost-benefit analysis. Eur. Urol. 37, 609–614 (2000).
opportunities. Hum. Reprod. Update 23, 660–680 (2017). 45. Aggour, A., Mostafa, H. & Maged, W. Endoscopic management of ejaculatory duct
12. Baker, H. W. G. in Endocrinology (eds Jameson J. L. & DeGroot, L. J.) Ch. 141, 2556–2579 obstruction. Int. Urol. Nephrol. 30, 481–485 (1998).
(Saunders Elsevier, 2010). 46. Modgil, V., Rai, S., Ralph, D. J. & Muneer, A. An update on the diagnosis and management
13. Bonde, J. P. et al. Relation between semen quality and fertility: a population-based study of ejaculatory duct obstruction. Nat. Rev. Urol. 13, 13–20 (2016).
of 430 first-pregnancy planners. Lancet 352, 1172–1177 (1998). 47. Wosnitzer, M., Goldstein, M. & Hardy, M. P. Review of azoospermia. Spermatogenesis 4,
14. Schlegel, P. N. et al. Diagnosis and treatment of infertility in Men: AUA/ASRM guideline e28218 (2014).
part I. J. Urol. 205, 36–43 (2021). 48. Practice Committee of the American Society for Reproductive Medicine in collaboration
15. Schlegel, P. N. et al. Diagnosis and treatment of infertility in Men: AUA/ASRM guideline with the Society for Male Reproduction and Urology. The management of obstructive
part II. J. Urol. 205, 44–51 (2021). azoospermia: a committee opinion. Fertil. Steril. 111, 873–880 (2019).
16. Minhas, S. et al. European Association of Urology guidelines on male sexual and 49. Colpi, G. M. et al. European Academy of Andrology guideline management of
reproductive health: 2021 update on male infertility. Eur. Urol. 80, 603–620 (2021). oligo-astheno-teratozoospermia. Andrology 6, 513–524 (2018).
17. Practice Committee of the American Society for Reproductive Medicine. Diagnostic 50. Nicopoullos, J. D. et al. Use of surgical sperm retrieval in azoospermic men:
evaluation of the infertile male: a committee opinion. Fertil. Steril. 103, e18–e25 (2015). a meta-analysis. Fertil. Steril. 82, 691–701 (2004).
18. Samplaski, M. K. et al. Reproductive endocrinologists are the gatekeepers for male 51. Punjani, N., Kang, C. & Schlegel, P. N. Two decades from the introduction of
infertility care in North America: results of a North American survey on the referral microdissection testicular sperm extraction: how this surgical technique has improved
patterns and characteristics of men presenting to male infertility specialists for infertility the management of NOA. J. Clin. Med. 10, 1374 (2021).
investigations. Fertil. Steril. 112, 657–662 (2019). 52. Krausz, C. et al. Genetic dissection of spermatogenic arrest through exome analysis:
19. Punab, M. et al. Causes of male infertility: a 9-year prospective monocentre study clinical implications for the management of azoospermic men. Genet. Med. 22,
on 1737 patients with reduced total sperm counts. Hum. Reprod. 32, 18–31 (2017). 1956–1966 (2020).
20. Campbell, M. J. et al. Distribution of semen examination results 2020 — A follow up of 53. Ji, C. et al. Potential of testis-derived circular RNAs in seminal plasma to predict the
data collated for the WHO semen analysis manual 2010. Andrology 9, 817–822 (2021). outcome of microdissection testicular sperm extraction in patients with idiopathic
21. Bohring, C. & Krause, W. Serum levels of inhibin B in men with different causes of non-obstructive azoospermia. Hum. Reprod. 36, 2649–2660 (2021).
spermatogenic failure. Andrologia 31, 137–141 (1999). 54. Rastrelli, G., Corona, G., Mannucci, E. & Maggi, M. Factors affecting spermatogenesis
22. Sikaris, K. et al. Reproductive hormone reference intervals for healthy fertile young men: upon gonadotropin-replacement therapy: a meta-analytic study. Andrology 2, 794–808
evaluation of automated platform assays. J. Clin. Endocrinol. Metab. 90, 5928–5936 (2005). (2014).
23. Waller, E.-J., Conceicao, J., Matson, P. & Yovich, J. Proposed age-stratified reference 55. Boeri, L., Capogrosso, P. & Salonia, A. Gonadotropin treatment for the male
intervals of FSH derived from normozoospermic men. Asian Pac. J. Reprod. 10, 162–167 hypogonadotropic hypogonadism. Curr. Pharm. Des. 27, 2775–2783 (2020).
(2021). 56. Ferlin, A. et al. Management of male factor infertility: position statement from the
24. Nahoul, K. & Roger, M. Age-related decline of plasma bioavailable testosterone in adult Italian Society of Andrology and Sexual Medicine (SIAMS): endorsing organization: Italian
men. J. Steroid Biochem. 35, 293–299 (1990). Society of Embryology, Reproduction, and Research (SIERR). J. Endocrinol. Invest. 45,
25. Neaves, W. B., Johnson, L., Porter, J. C., Parker, C. R. Jr. & Petty, C. S. Leydig cell numbers, 1085–1113 (2022).
daily sperm production, and serum gonadotropin levels in aging men. J. Clin. Endocrinol. 57. Lee, J. A. & Ramasamy, R. Indications for the use of human chorionic gonadotropic
Metab. 59, 756–763 (1984). hormone for the management of infertility in hypogonadal men. Transl. Androl. Urol. 7,
26. Bjørnerem, A. et al. Endogenous sex hormones in relation to age, sex, lifestyle factors, S348–S352 (2018).
and chronic diseases in a general population: the Tromsø Study. J. Clin. Endocrinol. 58. Prior, M., Stewart, J., McEleny, K., Dwyer, A. A. & Quinton, R. Fertility induction in
Metab. 89, 6039–6047 (2004). hypogonadotropic hypogonadal men. Clin. Endocrinol. 89, 712–718 (2018).
27. Gray, A., Feldman, H. A., McKinlay, J. B. & Longcope, C. Age, disease, and changing sex 59. Ramasamy, R., Stahl, P. J. & Schlegel, P. N. Medical therapy for spermatogenic failure.
hormone levels in middle-aged men: results of the Massachusetts Male Aging Study. Asian J. Androl. 14, 57–60 (2012).
J. Clin. Endocrinol. Metab. 73, 1016–1025 (1991). 60. Omar, M. I. et al. Benefits of empiric nutritional and medical therapy for semen
28. Andersson, A. M., Jorgensen, N., Frydelund-Larsen, L., Rajpert-De Meyts, E. & parameters and pregnancy and live birth rates in couples with idiopathic infertility:
Skakkebaek, N. E. Impaired Leydig cell function in infertile men: a study of 357 idiopathic a systematic review and meta-analysis. Eur. Urol. 75, 615–625 (2019).
infertile men and 318 proven fertile controls. J. Clin. Endocrinol. Metab. 89, 3161–3167 61. Behre, H. M. Clinical use of FSH in male infertility. Front. Endocrinol. 10, 322 (2019).
(2004). 62. Caroppo, E. & Colpi, G. M. Hormonal treatment of men with nonobstructive azoospermia:
29. Schlegel, P. N. et al. Diagnosis and treatment of infertility in men: AUA/ASRM guideline what does the evidence suggest? J. Clin. Med. 10, 387 (2021).
part II. Fertil. Steril. 115, 62–69 (2021). 63. Barbonetti, A. et al. The use of follicle stimulating hormone (FSH) for the treatment of
30. Schlegel, P. N. et al. Diagnosis and treatment of infertility in men: AUA/ASRM guideline the infertile man: position statement from the Italian Society of Andrology and Sexual
part I. Fertil. Steril. 115, 54–61 (2021). Medicine (SIAMS). J. Endocrinol. Invest. 41, 1107–1122 (2018).
31. Jungwirth, A et al. EAU Guidelines on Male Infertility https://d56bochluxqnz.cloudfront. 64. Del Giudice, F. et al. A systematic review and meta-analysis of clinical trials implementing
net/media/EAU-Guidelines-on-Male-Infertility-2019.pdf (EAU, 2019). aromatase inhibitors to treat male infertility. Asian J. Androl. 22, 360–367 (2020).
32. Australia and New Zealand Assisted Reproduction Database ANZARD https://www. 65. Ko, E. Y., Siddiqi, K., Brannigan, R. E. & Sabanegh, E. S. Jr. Empirical medical therapy for
fertilitysociety.com.au/anzard/#anzard (2019). idiopathic male infertility: a survey of the American Urological Association. J. Urol. 187,
33. Nixon, B. et al. New horizons in human sperm selection for assisted reproduction. 973–978 (2012).
Front. Endocrinol. 14, 1145533 (2023). 66. Thaker, H. et al. Empirical medical therapy for idiopathic male infertility. F. S Rep. 1, 15–20
34. You, J. B. et al. Machine learning for sperm selection. Nat. Rev. Urol. 18, 387–403 (2021). (2020).
35. Khandwala, Y. S., Zhang, C. A., Lu, Y. & Eisenberg, M. L. The age of fathers in the USA is 67. Ferlin, A. et al. Toward a pharmacogenetic approach to male infertility: polymorphism
rising: an analysis of 168 867 480 births from 1972 to 2015. Hum. Reprod. 32, 2110–2116 of follicle-stimulating hormone beta-subunit promoter. Fertil. Steril. 96, 1344–1349.e2
(2017). (2011).
36. Hviid Malling, G. M. et al. ‘Doing it in the right order’: childless men’s intentions regarding 68. Simoni, M. et al. Treatment with human, recombinant FSH improves sperm DNA
family formation. Hum. Fertil. 25, 188–196 (2020). fragmentation in idiopathic infertile men depending on the FSH receptor polymorphism
37. Priskorn, L. et al. RUBIC (ReproUnion Biobank and Infertility Cohort): a binational p.N680S: a pharmacogenetic study. Hum. Reprod. 31, 1960–1969 (2016).
clinical foundation to study risk factors, life course, and treatment of infertility and 69. Casamonti, E. et al. Short-term FSH treatment and sperm maturation: a prospective
infertility-related morbidity. Andrology 9, 1828–1842 (2021). study in idiopathic infertile men. Andrology 5, 414–422 (2017).
38. Mazzilli, R. et al. Severe male factor in in vitro fertilization: definition, prevalence, and 70. de Ligny, W. et al. Antioxidants for male subfertility. Cochrane Database Syst. Rev. 5,
treatment. An update. Asian J. Androl. 24, 125–134 (2022). CD007411 (2022).

Nature Reviews Urology


Expert recommendation

71. Agarwal, A. et al. A global survey of reproductive specialists to determine the clinical 106. Romerius, P. et al. Estrogen receptor alpha single nucleotide polymorphism modifies
utility of oxidative stress testing and antioxidant use in male infertility. World J. Mens. the risk of azoospermia in childhood cancer survivors. Pharmacogenet Genomics 21,
Health 39, 470–488 (2021). 263–269 (2011).
72. Barati, E., Nikzad, H. & Karimian, M. Oxidative stress and male infertility: current 107. Visscher, P. M., Yengo, L., Cox, N. J. & Wray, N. R. Discovery and implications
knowledge of pathophysiology and role of antioxidant therapy in disease management. of polygenicity of common diseases. Science 373, 1468–1473 (2021).
Cell Mol. Life Sci. 77, 93–113 (2020). 108. Townsley, K. G., Brennand, K. J. & Huckins, L. M. Massively parallel techniques for
73. Aitken, R. J. Antioxidant trials — the need to test for stress. Hum. Reprod. Open. 2021, cataloguing the regulome of the human brain. Nat. Neurosci. 23, 1509–1521 (2020).
hoab007 (2021). 109. Groopman, E. E., Povysil, G., Goldstein, D. B. & Gharavi, A. G. Rare genetic causes of
74. Schisterman, E. F. et al. Effect of folic acid and zinc supplementation in men on semen complex kidney and urological diseases. Nat. Rev. Nephrol. 16, 641–656 (2020).
quality and live birth among couples undergoing infertility treatment: a randomized 110. Zhu, X. B. et al. Vertical transmission of the Yq AZFc microdeletion from father to son over
clinical trial. JAMA 323, 35–48 (2020). two or three generations in infertile Han Chinese families. Asian J. Androl. 12, 240–246
75. Zini, A., San Gabriel, M. & Baazeem, A. Antioxidants and sperm DNA damage: a clinical (2010).
perspective. J. Assist. Reprod. Genet. 26, 427–432 (2009). 111. Smits, R. M. et al. De novo mutations in children born after medical assisted
76. Aitken, R. J. Reactive oxygen species as mediators of sperm capacitation and reproduction. Hum. Reprod. 37, 1360–1369 (2022).
pathological damage. Mol. Reprod. Dev. 84, 1039–1052 (2017). 112. Yu, X. W., Wei, Z. T., Jiang, Y. T. & Zhang, S. L. Y chromosome azoospermia factor
77. Houston, B., Curry, B. & Aitken, R. J. Human spermatozoa possess an IL4I1 l-amino acid region microdeletions and transmission characteristics in azoospermic and severe
oxidase with a potential role in sperm function. Reproduction 149, 587–596 (2015). oligozoospermic patients. Int. J. Clin. Exp. Med. 8, 14634–14646 (2015).
78. Lee, D., Moawad, A. R., Morielli, T., Fernandez, M. C. & O’Flaherty, C. Peroxiredoxins 113. Beaud, H., Tremblay, A. R., Chan, P. T. K. & Delbes, G. Sperm DNA damage in cancer
prevent oxidative stress during human sperm capacitation. Mol. Hum. Reprod. 23, patients. Adv. Exp. Med. Biol. 1166, 189–203 (2019).
106–115 (2017). 114. Payne, K. S., Mazur, D. J., Hotaling, J. M. & Pastuszak, A. W. Cannabis and male fertility:
79. Aitken, R. J. & Baker, M. A. Oxidative stress, sperm survival and fertility control. a systematic review. J. Urol. 202, 674–681 (2019).
Mol. Cell Endocrinol. 250, 66–69 (2006). 115. Diamanti-Kandarakis, E. et al. Endocrine-disrupting chemicals: an Endocrine Society
80. Aitken, R. J. & Roman, S. D. Antioxidant systems and oxidative stress in the testes. scientific statement. Endocr. Rev. 30, 293–342 (2009).
Oxid. Med. Cell Longev. 1, 15–24 (2008). 116. Salas-Huetos, A., James, E. R., Aston, K. I., Jenkins, T. G. & Carrell, D. T. Diet and sperm
81. Gharagozloo, P. & Aitken, R. J. The role of sperm oxidative stress in male infertility and the quality: nutrients, foods and dietary patterns. Reprod. Biol. 19, 219–224 (2019).
significance of oral antioxidant therapy. Hum. Reprod. 26, 1628–1640 (2011). 117. Wu, H. et al. Preconception urinary phthalate concentrations and sperm DNA
82. Kowalczyk, A. The role of the natural antioxidant mechanism in sperm cells. Reprod. Sci. methylation profiles among men undergoing IVF treatment: a cross-sectional study.
29, 1387–1394 (2022). Hum. Reprod. 32, 2159–2169 (2017).
83. O’Flaherty, C. Orchestrating the antioxidant defenses in the epididymis. Andrology 7, 118. Skakkebæk, N. E. et al. Environmental factors in declining human fertility. Nat. Rev.
662–668 (2019). Endocrinol. 18, 139–157 (2021).
84. Aitken, R. J. & Bakos, H. W. Should we be measuring DNA damage in human 119. Sharpe, R. M. Environmental/lifestyle effects on spermatogenesis. Philos. Trans. R. Soc.
spermatozoa? New light on an old question. Hum. Reprod. 36, 1175–1185 (2021). Lond. B Biol. Sci. 365, 1697–1712 (2010).
85. Smits, R. M. et al. Antioxidants for male subfertility. Cochrane Database Syst. Rev. 3, 120. Bergman, A. et al. State of the science of endocrine disrupting chemicals 2012
CD007411 (2019). (eds Bergman, Å et al.) (World Health Organization, 2012).
86. Steiner, A. Z. et al. The effect of antioxidants on male factor infertility: the Males, 121. Gore, A. C. et al. EDC-2: the Endocrine Society’s second scientific statement on
Antioxidants, and Infertility (MOXI) randomized clinical trial. Fertil. Steril. 113, 552–560.e3 endocrine-disrupting chemicals. Endocr. Rev. 36, E1–E150 (2015).
(2020). 122. Istvan, M. et al. Maternal occupational exposure to endocrine-disrupting chemicals
87. Tuttelmann, F., Ruckert, C. & Ropke, A. Disorders of spermatogenesis: perspectives for during pregnancy and semen parameters in adulthood: results of a nationwide
novel genetic diagnostics after 20 years of unchanged routine. Med. Genet. 30, 12–20 cross-sectional study among Swiss conscripts. Hum. Reprod. 36, 1948–1958 (2021).
(2018). 123. Bonde, J. P. et al. The epidemiologic evidence linking prenatal and postnatal exposure
88. Tuttelmann, F. et al. Clinical experience with azoospermia: aetiology and chances for to endocrine disrupting chemicals with male reproductive disorders: a systematic review
spermatozoa detection upon biopsy. Int. J. Androl. 34, 291–298 (2011). and meta-analysis. Hum. Reprod. Update 23, 104–125 (2016).
89. Krausz, C. & Riera-Escamilla, A. Genetics of male infertility. Nat. Rev. Urol. 15, 369–384 124. Aneck-Hahn, N. H., Schulenburg, G. W., Bornman, M. S., Farias, P. & de Jager, C. Impaired
(2018). semen quality associated with environmental DDT exposure in young men living in a
90. Wyrwoll, M. J. et al. Genetic architecture of azoospermia-time to advance the standard malaria area in the Limpopo Province, South Africa. J. Androl. 28, 423–434 (2007).
of care. Eur. Urol. 83, 452–462 (2022). 125. Krewski, D. et al. Toxicity testing in the 21st century: progress in the past decade and
91. Toure, A. et al. The genetic architecture of morphological abnormalities of the sperm tail. future perspectives. Arch. Toxicol. 94, 1–58 (2020).
Hum. Genet. 140, 21–42 (2021). 126. Barrow, P. & Schmitt, G. Regulatory approaches to nonclinical reproductive toxicity
92. Salas-Huetos, A. et al. Disruption of human meiotic telomere complex genes TERB1, TERB2 testing of anti-cancer drugs. Anticancer. Agents Med. Chem. 17, 1171–1183 (2017).
and MAJIN in men with non-obstructive azoospermia. Hum. Genet. 140, 217–227 (2021). 127. Kortenkamp, A. et al. Combined exposures to bisphenols, polychlorinated dioxins,
93. Wyrwoll, M. J. et al. Bi-allelic mutations in M1AP are a frequent cause of meiotic arrest and paracetamol, and phthalates as drivers of deteriorating semen quality. Env. Int. 165,
severely impaired spermatogenesis leading to male infertility. Am. J. Hum. Genet. 107, 107322 (2022).
342–351 (2020). 128. Marcho, C., Oluwayiose, O. A. & Pilsner, J. R. The preconception environment and sperm
94. Kasak, L. et al. Bi-allelic recessive loss-of-function variants in FANCM cause epigenetics. Andrology 8, 924–942 (2020).
non-obstructive azoospermia. Am. J. Hum. Genet. 103, 200–212 (2018). 129. Short, A. K. et al. Elevated paternal glucocorticoid exposure alters the small noncoding
95. Hardy, J. J. et al. Variants in GCNA, X-linked germ-cell genome integrity gene, identified RNA profile in sperm and modifies anxiety and depressive phenotypes in the offspring.
in men with primary spermatogenic failure. Hum. Genet. 140, 1169–1182 (2021). Transl. Psychiatry 6, e837 (2016).
96. Liu, C. et al. Deleterious variants in X-linked CFAP47 induce asthenoteratozoospermia 130. Komninos, D. et al. High fat diet-induced obesity prolongs critical stages of the
and primary male infertility. Am. J. Hum. Genet. 108, 309–323 (2021). spermatogenic cycle in a Ldlr−/−.Leiden mouse model. Sci. Rep. 12, 430 (2022).
97. Zhu, F. et al. Biallelic SUN5 mutations cause autosomal-recessive acephalic spermatozoa 131. Nassan, F. L. et al. Association between intake of soft drinks and testicular function in
syndrome. Am. J. Hum. Genet. 99, 942–949 (2016). young men. Hum. Reprod. 36, 3036–3048 (2021).
98. Coutton, C. et al. Bi-allelic mutations in ARMC2 lead to severe astheno-teratozoospermia 132. Samplaski, M. K. & Nangia, A. K. Adverse effects of common medications on male fertility.
due to sperm flagellum malformations in humans and mice. Am. J. Hum. Genet. 104, Nat. Rev. Urol. 12, 401–413 (2015).
331–340 (2019). 133. Sominsky, L. et al. Linking stress and infertility: a novel role for ghrelin. Endocr. Rev. 38,
99. Tang, S. et al. Biallelic mutations in CFAP43 and CFAP44 cause male infertility with 432–467 (2017).
multiple morphological abnormalities of the sperm flagella. Am. J. Hum. Genet. 100, 134. Abu-Musa, A. A., Nassar, A. H., Hannoun, A. B. & Usta, I. M. Effect of the Lebanese civil war
854–864 (2017). on sperm parameters. Fertil. Steril. 88, 1579–1582 (2007).
100. Veltman, J. A. & Brunner, H. G. De novo mutations in human genetic disease. Nat. Rev. 135. Fenster, L. et al. Effects of psychological stress on human semen quality. J. Androl. 18,
Genet. 13, 565–575 (2012). 194–202 (1997).
101. Hodzic, A. et al. De novo mutations in idiopathic male infertility — a pilot study. 136. Leisegang, K., Henkel, R. & Agarwal, A. Obesity and metabolic syndrome associated with
Andrology 9, 212–220 (2021). systemic inflammation and the impact on the male reproductive system. Am. J. Reprod.
102. Oud, M. S. et al. Exome sequencing reveals variants in known and novel candidate genes Immunol. 82, e13178 (2019).
for severe sperm motility disorders. Hum. Reprod. 36, 2597–2611 (2021). 137. Leisegang, K., Sengupta, P., Agarwal, A. & Henkel, R. Obesity and male infertility:
103. Oud, M. S. et al. A de novo paradigm for male infertility. Nat. Commun. 13, 154 (2022). mechanisms and management. Andrologia 53, e13617 (2021).
104. Xavier, M. J., Salas-Huetos, A., Oud, M. S., Aston, K. I. & Veltman, J. A. Disease gene 138. Kleeman, E. A., Gubert, C. & Hannan, A. J. Transgenerational epigenetic impacts of
discovery in male infertility: past, present and future. Hum. Genet. 140, 7–19 (2021). parental infection on offspring health and disease susceptibility. Trends Genet. 38,
105. Krausz, C. et al. Phenotypic variation within European carriers of the Y-chromosomal 662–675 (2022).
gr/gr deletion is independent of Y-chromosomal background. J. Med. Genet. 46, 21–31 139. Gapp, K. et al. Alterations in sperm long RNA contribute to the epigenetic inheritance
(2009). of the effects of postnatal trauma. Mol. Psychiatry 25, 2162–2174 (2020).

Nature Reviews Urology


Expert recommendation

140. Maciejewski, R., Radzikowska-Büchner, E., Flieger, W., Kulczycka, K., Baj, J., Forma, A. & 172. Jorgensen, N., Asklund, C., Carlsen, E. & Skakkebaek, N. E. Coordinated European
Flieger, J. An overview of essential microelements and common metallic nanoparticles investigations of semen quality: results from studies of Scandinavian young men
and their effects on male fertility. Int. J. Env. Res. Public. Health 19, 11066 (2022). Sep 4. is a matter of concern. Int. J. Androl. 29, 54–61 (2006). discussion 105–108.
141. Andersen, E. et al. Sperm count is increased by diet-induced weight loss and maintained 173. Smarr, M. M. et al. Is human fecundity changing? A discussion of research and data gaps
by exercise or GLP-1 analogue treatment: a randomized controlled trial. Hum. Reprod. 37, precluding us from having an answer. Hum. Reprod. 32, 499–504 (2017).
1414–1422 (2022). 174. Belladelli, F., Muncey, W. & Eisenberg, M. L. Reproduction as a window for health in men.
142. Schjenken, J. E. et al. High fat diet alters male seminal plasma composition to impair Fertil. Steril. 120, 429–437 (2023).
female immune adaptation for pregnancy in mice. Endocrinology 162, bqab123 (2021). 175. Economist Intelligence Unit. Fertile ground. How can Japan raise its fertility rate?
143. Maleki-Saghooni, N., Amirian, M., Sadeghi, R. & Latifnejad Roudsari, R. Effectiveness Economist Intelligence Unit https://www.eiu.com/graphics/marketing/pdf/
of infertility counseling on pregnancy rate in infertile patients undergoing assisted Fertility-in-Japan-EIU.pdf (2018).
reproductive technologies: a systematic review and meta-analysis. Int. J. Reprod. 176. Hauser, R. et al. Male reproductive disorders, diseases, and costs of exposure to
Biomed. 15, 391–402 (2017). endocrine-disrupting chemicals in the European Union. J. Clin. Endocrinol. Metab. 100,
144. Rimmer, M. P. et al. Outcome reporting across randomized controlled trials evaluating 1267–1277 (2015).
potential treatments for male infertility: a systematic review. Hum. Reprod. Open. 2022, 177. Attina, T. M. et al. Exposure to endocrine-disrupting chemicals in the USA: a
hoac010 (2022). population-based disease burden and cost analysis. Lancet Diabetes Endocrinol. 4,
145. World Drug Report 2021. United Nations Office on Drugs and Crime https://www.unodc.org/ 996–1003 (2016).
unodc/en/data-and-analysis/wdr2021.html (2021). 178. Institute for Health Metrics and Evaluation. Global Burden of Disease. IHME https://www.
146. Fronczak, C. M., Kim, E. D. & Barqawi, A. B. The insults of illicit drug use on male fertility. healthdata.org/research-analysis/gbd (2023).
J. Androl. 33, 515–528 (2012). 179. Arya, S. T. & Dibb, B. The experience of infertility treatment: the male perspective.
147. Sakib, S., Voigt, A., Goldsmith, T. & Dobrinski, I. Three-dimensional testicular organoids Hum. Fertil. 19, 242–248 (2016).
as novel in vitro models of testicular biology and toxicology. Env. Epigenet 5, dvz011 180. Koert, E., Takefman, J. & Boivin, J. Fertility quality of life tool: update on research and
(2019). practice considerations. Hum. Fertil. 24, 236–248 (2019).
148. Martus, H. J. et al. Summary of major conclusions from the 7th International Workshop 181. Salonia, A. et al. Are infertile men less healthy than fertile men? Results of a prospective
on Genotoxicity Testing (IWGT), Tokyo, Japan. Mutat. Res. 852, 503134 (2020). case-control survey. Eur. Urol. 56, 1025–1031 (2009).
149. Greally, J. M. Endocrine disruptors and the epigenome (OECD review) (Organisation for 182. Ventimiglia, E. et al. Infertility as a proxy of general male health: results of a
Economic Co-operation and Development, 2011). cross-sectional survey. Fertil. Steril. 104, 48–55 (2015).
150. Skakkebaek, N. E. et al. Male reproductive disorders and fertility trends: influences of 183. Eisenberg, M. L. et al. The relationship between male BMI and waist circumference
environment and genetic susceptibility. Physiol. Rev. 96, 55–97 (2016). on semen quality: data from the LIFE study. Hum. Reprod. 29, 193–200 (2014).
151. Priskorn, L. et al. Increasing trends in childlessness in recent birth cohorts — a 184. Jacobsen, R. et al. Risk of testicular cancer in men with abnormal semen characteristics:
registry-based study of the total Danish male population born from 1945 to 1980. Int. J. cohort study. BMJ 321, 789–792 (2000).
Androl. 35, 449–455 (2012). 185. Walsh, T. J., Croughan, M. S., Schembri, M., Chan, J. M. & Turek, P. J. Increased risk
152. Boulicault, M. et al. The future of sperm: a biovariability framework for understanding of testicular germ cell cancer among infertile men. Arch. Intern. Med. 169, 351–356
global sperm count trends. Hum. Fertil. 25, 888–902 (2021). 1-15. (2009).
153. Guzick, D. S. et al. Sperm morphology, motility, and concentration in fertile and infertile 186. Del Giudice, F. et al. Association between male infertility and male-specific malignancies:
men. N. Engl. J. Med. 345, 1388–1393 (2001). systematic review and meta-analysis of population-based retrospective cohort studies.
154. Slama, R. et al. Time to pregnancy and semen parameters: a cross-sectional study Fertil. Steril. 114, 984–996 (2020).
among fertile couples from four European cities. Hum. Reprod. 17, 503–515 (2002). 187. Al-Jebari, Y. et al. Risk of prostate cancer for men fathering through assisted
155. Nelson, C. M. & Bunge, R. G. Semen analysis: evidence for changing parameters of male reproduction: nationwide population based register study. BMJ 366, l5214 (2019).
fertility potential. Fertil. Steril. 25, 503–507 (1974). 188. Walsh, T. J. et al. Increased risk of high-grade prostate cancer among infertile men.
156. MacLeod, J. & Wang, Y. Male fertility potential in terms of semen quality: a review of the Cancer 116, 2140–2147 (2010).
past, a study of the present. Fertil. Steril. 31, 103–116 (1979). 189. Hanson, H. A. et al. Subfertility increases risk of testicular cancer: evidence from
157. Carlsen, E., Giwercman, A., Keiding, N. & Skakkebaek, N. E. Evidence for decreasing population-based semen samples. Fertil. Steril. 105, 322–328.e1 (2016).
quality of semen during past 50 years. BMJ 305, 609–613 (1992). 190. Eisenberg, M. L., Li, S., Brooks, J. D., Cullen, M. R. & Baker, L. C. Increased risk of cancer
158. Levine, H. et al. Temporal trends in sperm count: a systematic review and in infertile men: analysis of U.S. claims data. J. Urol. 193, 1596–1601 (2015).
meta-regression analysis. Hum. Reprod. Update 23, 646–659 (2017). 191. Eisenberg, M. L., Betts, P., Herder, D., Lamb, D. J. & Lipshultz, L. I. Increased cancer risk
159. Lv, M. Q. et al. Temporal trends in semen concentration and count among 327 373 and azoospermia. Fertil. Steril. 100, 681–685 (2013).
Chinese healthy men from 1981 to 2019: a systematic review. Hum. Reprod. 36, 1751–1775 192. Swerdlow, A. J., Bruce, C., Cooke, R., Coulson, P. & Jones, M. E. Infertility and risk of breast
(2021). cancer in men: a national case-control study in England and Wales. Breast Cancer Res.
160. Vahidi, S., Moein, M. R., Yazdinejad, F., Ghasemi-Esmailabad, S. & Narimani, N. Iranian 24, 29 (2022).
temporal changes in semen quality during the past 22 years: a report from an infertility 193. Man, Y. et al. Whole-exome sequencing identifies the VHL mutation (c.262T > C,
center. Int. J. Reprod. Biomed. 18, 1059–1064 (2020). p.Try88Arg) in non-obstructive azoospermia-associated cystic renal cell carcinoma.
161. Rosa-Villagrán, L., Barrera, N., Montes, J., Riso, C. & Sapiro, R. Decline of semen quality Curr. Oncol. 29, 2376–2384 (2022).
over the last 30 years in Uruguay. Basic. Clin. Androl. 31, 8 (2021). 194. Chen, P. C. et al. Male infertility increases the risk of cardiovascular diseases: a
162. Sengupta, P., Nwagha, U., Dutta, S., Krajewska-Kulak, E. & Izuka, E. Evidence for nationwide population-based cohort study in Taiwan. World J. Mens. Health 40, 490–500
decreasing sperm count in African population from 1965 to 2015. Afr. Health Sci. 17, (2022).
418–427 (2017). 195. Elenkov, A., Melander, O., Nilsson, P. M., Zhang, H. & Giwercman, A. Impact of genetic risk
163. Mishra, P., Negi, M. P. S., Srivastava, M., Singh, K. & Rajender, S. Decline in seminal quality score on the association between male childlessness and cardiovascular disease and
in Indian men over the last 37 years. Reprod. Biol. Endocrinol. 16, 103 (2018). mortality. Sci. Rep. 11, 18526 (2021).
164. Levine, H. et al. Temporal trends in sperm count: a systematic review and 196. Brubaker, W. D., Li, S., Baker, L. C. & Eisenberg, M. L. Increased risk of autoimmune
meta-regression analysis of samples collected globally in the 20th and 21st centuries. disorders in infertile men: analysis of US claims data. Andrology 6, 94–98 (2018).
Hum. Reprod. Update 29, 157–176 (2023). 197. Eisenberg, M. L., Li, S., Cullen, M. R. & Baker, L. C. Increased risk of incident chronic
165. Jorgensen, N. et al. Are worldwide sperm counts declining? Fertil. Steril. 116, 1457–1463 medical conditions in infertile men: analysis of United States claims data. Fertil. Steril.
(2021). 105, 629–636 (2015).
166. Tiegs, A. W., Landis, J., Garrido, N., Scott, R. T. Jr. & Hotaling, J. M. Total motile sperm 198. Glazer, C. H. et al. Male factor infertility and risk of multiple sclerosis: a register-based
count trend over time: evaluation of semen analyses from 119,972 men from subfertile cohort study. Mult. Scler. 24, 1835–1842 (2017).
couples. Urology 132, 109–116 (2019). 199. Glazer, C. H. et al. Risk of diabetes according to male factor infertility: a register-based
167. Jorgensen, N. et al. East-West gradient in semen quality in the Nordic-Baltic area: a study cohort study. Hum. Reprod. 32, 1474–1481 (2017).
of men from the general population in Denmark, Norway, Estonia and Finland. Hum. Reprod. 200. Latif, T. et al. Semen quality as a predictor of subsequent morbidity: a Danish cohort
17, 2199–2208 (2002). study of 4,712 men with long-term follow-up. Am. J. Epidemiol. 186, 910–917 (2017).
168. Paasch, U. et al. Semen quality in sub-fertile range for a significant proportion of young 201. Latif, T. et al. Semen quality associated with subsequent hospitalizations — can the effect
men from the general German population: a co-ordinated, controlled study of 791 men be explained by socio-economic status and lifestyle factors? Andrology 6, 428–435
from Hamburg and Leipzig. Int. J. Androl. 31, 93–102 (2008). (2018).
169. Mendiola, J., Stahlhut, R. W., Jorgensen, N., Liu, F. & Swan, S. H. Shorter anogenital 202. Jensen, T. K., Jacobsen, R., Christensen, K., Nielsen, N. C. & Bostofte, E. Good semen
distance predicts poorer semen quality in young men in Rochester, New York. Env. Health quality and life expectancy: a cohort study of 43,277 men. Am. J. Epidemiol. 170,
Perspect. 119, 958–963 (2011). 559–565 (2009).
170. Fernandez, M. F. et al. Semen quality and reproductive hormone levels in men from 203. Glazer, C. H. et al. Male factor infertility and risk of death: a nationwide record-linkage
Southern Spain. Int. J. Androl. 35, 1–10 (2012). study. Hum. Reprod. 34, 2266–2273 (2019).
171. Iwamoto, T. et al. Semen quality of 1559 young men from four cities in Japan: 204. Eisenberg, M. L. et al. Semen quality, infertility and mortality in the USA. Hum. Reprod.
a cross-sectional population-based study. BMJ Open 3, e002222 (2013). 29, 1567–1574 (2014).

Nature Reviews Urology


Expert recommendation

205. Del Giudice, F. et al. The association between mortality and male infertility: systematic 237. Abdelaal, O., Barber, H., Atala, A. & Sadri-Ardekani, H. Purging of malignant cell
review and meta-analysis. Urology 154, 148–157 (2021). contamination prior to spermatogonia stem cell autotransplantation to preserve
206. Del Giudice, F. et al. Increased mortality among men diagnosed with impaired fertility: fertility: progress & prospects. Curr. Opin. Endocrinol. Diabetes Obes. 26, 166–174
analysis of US claims data. Urology 147, 143–149 (2021). (2019).
207. Choy, J. T. & Eisenberg, M. L. Male infertility as a window to health. Fertil. Steril. 110, 238. Shetty, G. et al. Postpubertal spermatogonial stem cell transplantation restores
810–814 (2018). functional sperm production in rhesus monkeys irradiated before and after puberty.
208. Uhlen, M. et al. Proteomics. Tissue-based map of the human proteome. Science 347, Andrology 9, 1603–1616 (2021).
1260419 (2015). 239. Fayomi, A. P. et al. Autologous grafting of cryopreserved prepubertal rhesus testis
209. Fagerberg, L. et al. Analysis of the human tissue-specific expression by genome-wide produces sperm and offspring. Science 363, 1314–1319 (2019).
integration of transcriptomics and antibody-based proteomics. Mol. Cell Proteom. 13, 240. Alves-Lopes, J. P., Soder, O. & Stukenborg, J. B. Use of a three-layer gradient system
397–406 (2014). of cells for rat testicular organoid generation. Nat. Protoc. 13, 248–259 (2018).
210. Djureinovic, D. et al. The human testis-specific proteome defined by transcriptomics and 241. Mincheva, M. et al. Reassembly of adult human testicular cells: can testis cord-like
antibody-based profiling. Mol. Hum. Reprod. 20, 476–488 (2014). structures be created in vitro. Mol. Hum. Reprod. 24, 55–63 (2018).
211. Bonadona, V. et al. Cancer risks associated with germline mutations in MLH1, MSH2, and 242. Oliver, E. et al. Self-organising human gonads generated by a Matrigel-based gradient
MSH6 genes in Lynch syndrome. JAMA 305, 2304–2310 (2011). system. BMC Biol. 19, 212 (2021).
212. Ji, G. et al. Common variants in mismatch repair genes associated with increased risk of 243. Ishikura, Y. et al. In vitro reconstitution of the whole male germ-cell development from
sperm DNA damage and male infertility. BMC Med. 10, 49 (2012). mouse pluripotent stem cells. Cell Stem Cell 28, 2167–2179.e9 (2021).
213. Mak, V. et al. Proportion of cystic fibrosis gene mutations not detected by routine testing 244. Saitou, M. & Hayashi, K. Mammalian in vitro gametogenesis. Science 374, eaaz6830
in men with obstructive azoospermia. JAMA 281, 2217–2224 (1999). (2021).
214. Gunes, S. et al. Microtubular dysfunction and male infertility. World J. Mens. Health 38, 245. Pennings, G., Couture, V. & Ombelet, W. Social sperm freezing. Hum. Reprod. 36,
9–23 (2020). 833–839 (2021).
215. Day, F. R. et al. Large-scale genomic analyses link reproductive aging to hypothalamic 246. Degraeve, A. et al. European countries have different rates of sperm cryopreservation
signaling, breast cancer susceptibility and BRCA1-mediated DNA repair. Nat. Genet. 47, before vasectomy and at the time of reversal. Andrology 10, 1286–1291 (2022).
1294–1303 (2015). 247. Marinaro, J., Hayden, R. P., Shin, P. & Tanrikut, C. The utility of sperm cryopreservation at
216. O’Bryan, M. K. & Hedger, M. P. Inflammatory networks in the control of spermatogenesis : the time of vasectomy reversal. J. Urol. 205, 236–240 (2021).
chronic inflammation in an immunologically privileged tissue? Adv. Exp. Med. Biol. 636, 248. Ravitsky, V. & Kimmins, S. The forgotten men: rising rates of male infertility urgently
92–114 (2008). require new approaches for its prevention, diagnosis and treatment. Biol. Reprod. 101,
217. Meinhardt, A. & Hedger, M. P. Immunological, paracrine and endocrine aspects of 872–874 (2019).
testicular immune privilege. Mol. Cell Endocrinol. 335, 60–68 (2011). 249. Kong, A. et al. Rate of de novo mutations and the importance of father’s age to disease
218. Meseguer, M. et al. Sperm cryopreservation in oncological patients: a 14-year follow-up risk. Nature 488, 471–475 (2012).
study. Fertil. Steril. 85, 640–645 (2006). 250. Taylor, J. L. et al. Paternal-age-related de novo mutations and risk for five disorders.
219. Liu, W., Schulster, M. L., Alukal, J. P. & Najari, B. B. Fertility preservation in male to female Nat. Commun. 10, 3043 (2019).
transgender patients. Urol. Clin. North. Am. 46, 487–493 (2019). 251. Sartorius, G. A. & Nieschlag, E. Paternal age and reproduction. Hum. Reprod. Update 16,
220. Cooper, H. C., Long, J. & Aye, T. Fertility preservation in transgender and non-binary 65–79 (2010).
adolescents and young adults. PLoS ONE 17, e0265043 (2022). 252. Couture, V., Delisle, S., Mercier, A. & Pennings, G. The other face of advanced paternal
221. Lambertini, M. et al. Fertility preservation and post-treatment pregnancies in age: a scoping review of its terminological, social, public health, psychological, ethical
post-pubertal cancer patients: ESMO clinical practice guidelines(dagger). Ann. Oncol. and regulatory aspects. Hum. Reprod. Update 27, 305–323 (2021).
31, 1664–1678 (2020). 253. Urhoj, S. K. et al. Advanced paternal age and childhood cancer in offspring: a nationwide
222. Lee, S. J. et al. American Society of Clinical Oncology recommendations on fertility register-based cohort study. Int. J. Cancer 140, 2461–2472 (2017).
preservation in cancer patients. J. Clin. Oncol. 24, 2917–2931 (2006). 254. Cao, M. et al. High-resolution analyses of human sperm dynamic methylome reveal
223. Kuczynski, W. et al. The outcome of intracytoplasmic injection of fresh and thousands of novel age-related epigenetic alterations. Clin. Epigenetics 12, 192 (2020).
cryopreserved ejaculated spermatozoa-a prospective randomized study. Hum. Reprod. 255. Salas-Huetos, A. et al. The combined effect of obesity and aging on human sperm DNA
16, 2109–2113 (2001). methylation signatures: inclusion of BMI in the paternal germ line age prediction model.
224. Hervas, I. et al. TESE-ICSI outcomes per couple in vasectomized males are negatively Sci. Rep. 10, 15409 (2020).
affected by time since the intervention, but not other comorbidities. Reprod. Biomed. 256. Laurentino, S. et al. A germ cell-specific ageing pattern in otherwise healthy men.
Online 43, 708–717 (2021). Aging Cell 19, e13242 (2020).
225. Newton, H. L. et al. Inconsistencies in fertility preservation for young people with cancer 257. Berntsen, S. et al. The health of children conceived by ART: ‘the chicken or the egg?’.
in the UK. Arch. Dis. Child. 107, 265–270 (2022). Hum. Reprod. Update 25, 137–158 (2019).
226. Anazodo, A. et al. The development of an international oncofertility competency 258. Horne, G. et al. Live birth with sperm cryopreserved for 21 years prior to cancer
framework: a model to increase oncofertility implementation. Oncologist 24, treatment: case report. Hum. Reprod. 19, 1448–1449 (2004).
e1450–e1459 (2019). 259. Rotondo, J. C., Lanzillotti, C., Mazziotta, C., Tognon, M. & Martini, F. Epigenetics of male
227. Anazodo, A. et al. How can we improve oncofertility care for patients? A systematic infertility: the role of DNA methylation. Front. Cell Dev. Biol. 9, 689624 (2021).
scoping review of current international practice and models of care. Hum. Reprod. 260. Erkek, S. et al. Molecular determinants of nucleosome retention at CpG-rich sequences
Update 25, 159–179 (2019). in mouse spermatozoa. Nat. Struct. Mol. Biol. 20, 868–875 (2013).
228. Delgouffe, E., Braye, A. & Goossens, E. Testicular tissue banking for fertility 261. Siklenka, K. et al. Disruption of histone methylation in developing sperm impairs
preservation in young boys: which patients should be included? Front. Endocrinol. 13, offspring health transgenerationally. Science 350, aab2006 (2015).
854186 (2022). 262. Sendler, E. et al. Stability, delivery and functions of human sperm RNAs at fertilization.
229. Martinez, F. & International Society for Fertility Preservation. Update on fertility Nucleic Acids Res. 41, 4104–4117 (2013).
preservation from the Barcelona International Society for Fertility Preservation-ESHRE- 263. Cavalli, G. & Heard, E. Advances in epigenetics link genetics to the environment and
ASRM 2015 expert meeting: indications, results and future perspectives. Fertil. Steril. 108, disease. Nature 571, 489–499 (2019).
407–415.e11 (2017). 264. Janssen, S. M. & Lorincz, M. C. Interplay between chromatin marks in development and
230. Mulder, R. L. et al. Fertility preservation for male patients with childhood, adolescent, disease. Nat. Rev. Genet. 23, 137–153 (2021).
and young adult cancer: recommendations from the PanCareLIFE Consortium and the 265. Lin, H. piRNAs in the germ line. Science 316, 397 (2007).
International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet 266. Trasler, J. M. Epigenetics in spermatogenesis. Mol. Cell Endocrinol. 306, 33–36 (2009).
Oncol. 22, e57–e67 (2021). 267. Glaser, S. et al. The histone 3 lysine 4 methyltransferase, Mll2, is only required briefly in
231. Ghidei, L. et al. Current gaps in fertility preservation for men: how can we do better? development and spermatogenesis. Epigenetics Chromatin 2, 5 (2009).
J. Clin. Oncol. 40, 2524–2529 (2022). 268. Kimmins, S. et al. Differential functions of the Aurora-B and Aurora-C kinases in
232. Goossens, E. et al. Fertility preservation in boys: recent developments and new insights. mammalian spermatogenesis. Mol. Endocrinol. 21, 726–739 (2007).
Hum. Reprod. Open. 2020, hoaa016 (2020). 269. Peters, A. H. et al. Loss of the Suv39h histone methyltransferases impairs mammalian
233. Sharma, S., Wistuba, J., Pock, T., Schlatt, S. & Neuhaus, N. Spermatogonial stem cells: heterochromatin and genome stability. Cell 107, 323–337 (2001).
updates from specification to clinical relevance. Hum. Reprod. Update 25, 275–297 270. Nagirnaja, L. et al. Variant PNLDC1, defective piRNA processing, and azoospermia.
(2019). N. Engl. J. Med. 385, 707–719 (2021).
234. Wyns, C., Curaba, M., Vanabelle, B., Van Langendonckt, A. & Donnez, J. Options for 271. Fellmeth, J. E., Ghanaim, E. M. & Schindler, K. Characterization of
fertility preservation in prepubertal boys. Hum. Reprod. Update 16, 312–328 (2010). macrozoospermia-associated AURKC mutations in a mammalian meiotic system.
235. Kliesch, S. [Androprotect and prospects for fertility treatment]. Urol. A 55, 898–903 Hum. Mol. Genet. 25, 2698–2711 (2016).
(2016). 272. Dieterich, K. et al. Homozygous mutation of AURKC yields large-headed polyploid
236. Kanbar, M., Delwiche, G. & Wyns, C. Fertility preservation for prepubertal boys: are we spermatozoa and causes male infertility. Nat. Genet. 39, 661–665 (2007).
ready for autologous grafting of cryopreserved immature testicular tissue? 273. Eloualid, A. et al. Prevalence of the Aurora kinase C c.144delC mutation in infertile
Ann. Endocrinol. 83, 210–217 (2022). Moroccan men. Fertil. Steril. 101, 1086–1090 (2014).

Nature Reviews Urology


Expert recommendation

274. Ben Khelifa, M. et al. Identification of a new recurrent aurora kinase C mutation in both 306. Novakovic, B. et al. Assisted reproductive technologies are associated with limited
European and African men with macrozoospermia. Hum. Reprod. 27, 3337–3346 (2012). epigenetic variation at birth that largely resolves by adulthood. Nat. Commun. 10, 3922
275. Tremblay, A., Beaud, H. & Delbes, G. Transgenerational impact of chemotherapy: would (2019).
the father exposure impact the health of future progeny [French]? Gynecol. Obstet. 307. Chen, W. et al. Integrated multi-omics reveal epigenomic disturbance of assisted
Fertil. Senol. 45, 609–618 (2017). reproductive technologies in human offspring. EBioMedicine 61, 103076 (2020).
276. Carrell, D. T. Epigenetics of the male gamete. Fertil. Steril. 97, 267–274 (2012). 308. Katari, S. et al. DNA methylation and gene expression differences in children conceived
277. Santiago, J., Silva, J. V., Howl, J., Santos, M. A. S. & Fardilha, M. All you need to know in vitro or in vivo. Hum. Mol. Genet. 18, 3769–3778 (2009).
about sperm RNAs. Hum. Reprod. Update 28, 67–91 (2021). 309. Katagiri, Y. et al. Effects of assisted reproduction technology on placental imprinted gene
278. Kobayashi, H. et al. Aberrant DNA methylation of imprinted loci in sperm from expression. Obstet Gynecol. Int. 2010, 437525, (2010).
oligospermic patients. Hum. Mol. Genet. 16, 2542–2551 (2007). 310. Lou, H. et al. Assisted reproductive technologies impair the expression and methylation
279. Marques, C. J. et al. Abnormal methylation of imprinted genes in human sperm is of insulin-induced gene 1 and sterol regulatory element-binding factor 1 in the fetus and
associated with oligozoospermia. Mol. Hum. Reprod. 14, 67–74 (2008). placenta. Fertil. Steril. 101, 974–980.e972 (2014).
280. Minor, A., Chow, V. & Ma, S. Aberrant DNA methylation at imprinted genes in testicular 311. Loke, Y. J., Galati, J. C., Saffery, R. & Craig, J. M. Association of in vitro fertilization with
sperm retrieved from men with obstructive azoospermia and undergoing vasectomy global and IGF2/H19 methylation variation in newborn twins. J. Dev. Orig. Health Dis. 6,
reversal. Reproduction 141, 749–757 (2011). 115–124 (2015).
281. Donkin, I. et al. Obesity and bariatric surgery drive epigenetic variation of spermatozoa in 312. Sakian, S. et al. Altered gene expression of H19 and IGF2 in placentas from ART
humans. Cell Metab. 23, 369–378 (2016). pregnancies. Placenta 36, 1100–1105 (2015).
282. Velotti, N. et al. Effect of bariatric surgery on in vitro fertilization in infertile men with 313. Choux, C. et al. The epigenetic control of transposable elements and imprinted genes in
obesity. Surg. Obes. Relat. Dis. 17, 1752–1759 (2021). newborns is affected by the mode of conception: ART versus spontaneous conception
283. Keyhan, S. et al. Male obesity impacts DNA methylation reprogramming in sperm. Clin. without underlying infertility. Hum. Reprod. 33, 331–340 (2018).
Epigenetics 13, 17 (2021). 314. Tierling, S. et al. Assisted reproductive technologies do not enhance the variability of
284. Stokes, V. J., Anderson, R. A. & George, J. T. How does obesity affect fertility in men – and DNA methylation imprints in human. J. Med. Genet. 47, 371–376 (2010).
what are the treatment options? Clin. Endocrinol. 82, 633–638 (2015). 315. Zechner, U. et al. Quantitative methylation analysis of developmentally important genes
285. Chan, D. et al. Customized methylC-capture sequencing to evaluate variation in the in human pregnancy losses after ART and spontaneous conception. Mol. Hum. Reprod.
human sperm DNA methylome representative of altered folate metabolism. Env. Health 16, 704–713 (2010).
Perspect. 127, 87002 (2019). 316. Li, L. et al. Evaluation of DNA methylation status at differentially methylated regions in
286. Pembrey, M., Saffery, R. & Bygren, L. O. Human transgenerational responses to early-life IVF-conceived newborn twins. Fertil. Steril. 95, 1975–1979 (2011).
experience: potential impact on development, health and biomedical research. J. Med. 317. Feng, C. et al. General imprinting status is stable in assisted reproduction-conceived
Genet. 51, 563–572 (2014). offspring. Fertil. Steril. 96, 1417–1423.e1419 (2011).
287. Radford, E. J. et al. In utero effects. In utero undernourishment perturbs the adult sperm 318. Oliver, V. F. et al. Defects in imprinting and genome-wide DNA methylation are not
methylome and intergenerational metabolism. Science 345, 1255903 (2014). common in the in vitro fertilization population. Fertil. Steril. 97, 147–153.e147 (2012).
288. Chen, Q. et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired 319. Rancourt, R. C., Harris, H. R. & Michels, K. B. Methylation levels at imprinting control
metabolic disorder. Science 351, 397–400 (2016). regions are not altered with ovulation induction or in vitro fertilization in a birth cohort.
289. Lismer, A. et al. Histone H3 lysine 4 trimethylation in sperm is transmitted to the embryo Hum. Reprod. 27, 2208–2216 (2012).
and associated with diet-induced phenotypes in the offspring. Dev. Cell 56, 1–16 (2021). 320. Penova-Veselinovic, B. et al. DNA methylation patterns within whole blood of
290. Hammoud, S. S. et al. Distinctive chromatin in human sperm packages genes for embryo adolescents born from assisted reproductive technology are not different from
development. Nature 460, 473–478 (2009). adolescents born from natural conception. Hum. Reprod. 36, 2035–2049 (2021).
291. Brykczynska, U. et al. Repressive and active histone methylation mark distinct promoters 321. Litzky, J. F. et al. Placental imprinting variation associated with assisted reproductive
in human and mouse spermatozoa. Nat. Struct. Mol. Biol. 17, 679–687 (2010). technologies and subfertility. Epigenetics 12, 653–661 (2017).
292. Lambrot, R. et al. Whole-genome sequencing of H3K4me3 and DNA methylation in 322. Song, S. et al. DNA methylation differences between in vitro- and in vivo-conceived
human sperm reveals regions of overlap linked to fertility and development. Cell Rep. children are associated with ART procedures rather than infertility. Clin. Epigenetics 7, 41
36, 109418 (2021). (2015).
293. Ly, L. et al. Intergenerational impact of paternal lifetime exposures to both folic acid 323. Hansen, M., Bower, C., Milne, E., de Klerk, N. & Kurinczuk, J. J. Assisted reproductive
deficiency and supplementation on reproductive outcomes and imprinted gene technologies and the risk of birth defects — a systematic review. Hum. Reprod. 20,
methylation. Mol. Hum. Reprod. 23, 461–477 (2017). 328–338 (2005).
294. Lambrot, R. et al. Low paternal dietary folate alters the mouse sperm epigenome and 324. Albertini, D. F. et al. Birth defects and congenital health risks in children conceived
is associated with negative pregnancy outcomes. Nat. Commun. 4, 2889 (2013). through assisted reproduction technology (ART): a meeting report. J. Assist. Reprod.
295. Sharma, U. et al. Biogenesis and function of tRNA fragments during sperm maturation Genet. 31, 947–958 (2014).
and fertilization in mammals. Science 351, 391–396 (2016). 325. Hansen, M. & Bower, C. The impact of assisted reproductive technologies on intra-uterine
296. Yoshida, K. et al. ATF7-dependent epigenetic changes are required for the growth and birth defects in singletons. Semin. Fetal Neonatal Med. 19, 228–233 (2014).
intergenerational effect of a paternal low-protein diet. Mol. Cell 78, 445–458 e446 326. Qin, J. et al. Assisted reproductive technology and risk of congenital malformations:
(2020). a meta-analysis based on cohort studies. Arch. Gynecol. Obstet. 292, 777–798 (2015).
297. Watkins, A. J. et al. Paternal diet programs offspring health through sperm- and seminal 327. Boulet, S. L. et al. Assisted reproductive technology and birth defects among liveborn
plasma-specific pathways in mice. Proc. Natl Acad. Sci. USA 115, 10064–10069 (2018). infants in Florida, Massachusetts, and Michigan, 2000–2010. JAMA Pediatr. 170, e154934
298. Terashima, M. et al. Effect of high fat diet on paternal sperm histone distribution and male (2016).
offspring liver gene expression. Epigenetics 10, 861–871 (2015). 328. Palomba, S., Homburg, R., Santagni, S., La Sala, G. B. & Orvieto, R. Risk of adverse
299. de Castro Barbosa, T. et al. High-fat diet reprograms the epigenome of rat spermatozoa pregnancy and perinatal outcomes after high technology infertility treatment:
and transgenerationally affects metabolism of the offspring. Mol. Metab. 5, 184–197 a comprehensive systematic review. Reprod. Biol. Endocrinol. 14, 76 (2016).
(2016). 329. Luke, B. et al. The risk of birth defects with conception by ART. Hum. Reprod. 36, 116–129
300. Pepin, A. S., Lafleur, C., Lambrot, R., Dumeaux, V. & Kimmins, S. Sperm histone H3 lysine 4 (2021).
tri-methylation serves as a metabolic sensor of paternal obesity and is associated with 330. Catford, S. R., McLachlan, R. I., O’Bryan, M. K. & Halliday, J. L. Long-term follow-up
the inheritance of metabolic dysfunction. Mol. Metab. 59, 101463 (2022). of ICSI-conceived offspring compared with spontaneously conceived offspring:
301. Rodgers, A. B., Morgan, C. P., Leu, N. A. & Bale, T. L. Transgenerational epigenetic a systematic review of health outcomes beyond the neonatal period. Andrology 6,
programming via sperm microRNA recapitulates effects of paternal stress. Proc. Natl 635–653 (2018).
Acad. Sci. USA 112, 13699 (2015). 331. Belva, F. et al. Semen quality of young adult ICSI offspring: the first results. Hum. Reprod.
302. Beck, D., Ben Maamar, M. & Skinner, M. K. Integration of sperm ncRNA-directed 31, 2811–2820 (2016).
DNA methylation and DNA methylation-directed histone retention in epigenetic 332. Belva, F. et al. Reproductive hormones of ICSI-conceived young adult men: the first
transgenerational inheritance. Epigenetics Chromatin 14, 6 (2021). results. Hum. Reprod. 32, 439–446 (2017).
303. Lismer, A., et al. Exposure of Greenlandic Inuit and South African VhaVenda men to 333. Catford, S. R. et al. Reproductive function in men conceived with in vitro fertilization
the persistent DDT metabolite is associated with an altered sperm epigenome at and intracytoplasmic sperm injection. Fertil. Steril. 117, 727–737 (2022).
regions implicated in paternal epigenetic transmission and developmental disease — 334. Belva, F. et al. Serum reproductive hormone levels and ultrasound findings in female
a cross-sectional study. Preprint at bioRxiv https://doi.org/10.1101/2022.08.15.504029 offspring after intracytoplasmic sperm injection: first results. Fertil. Steril. 107, 934–939
(2022). (2017).
304. Amor, D. J. & Halliday, J. A review of known imprinting syndromes and their association 335. Guo, X. Y. et al. Cardiovascular and metabolic profiles of offspring conceived by assisted
with assisted reproduction technologies. Hum. Reprod. 23, 2826–2834 (2008). reproductive technologies: a systematic review and meta-analysis. Fertil. Steril. 107,
305. Lazaraviciute, G., Kauser, M., Bhattacharya, S., Haggarty, P. & Bhattacharya, S. A 622–631.e5 (2017).
systematic review and meta-analysis of DNA methylation levels and imprinting disorders 336. Belva, F. et al. Body fat content, fat distribution and adipocytokine production and
in children conceived by IVF/ICSI compared with children conceived spontaneously. their correlation with fertility markers in young adult men and women conceived by
Hum. Reprod. Update 20, 840–852 (2014). intracytoplasmic sperm injection (ICSI). Clin. Endocrinol. 88, 985–992 (2018)

Nature Reviews Urology


Expert recommendation

337. Belva, F. et al. Metabolic syndrome and its components in young adults conceived by 370. Daumler, D., Chan, P., Lo, K. C., Takefman, J. & Zelkowitz, P. Men’s knowledge of their
ICSI. Int. J. Endocrinol. 2018, 8170518 (2018). own fertility: a population-based survey examining the awareness of factors that are
338. Zhu, J. L., Basso, O., Obel, C., Hvidtjorn, D. & Olsen, J. Infertility, infertility treatment associated with male infertility. Hum. Reprod. 31, 2781–2790 (2016).
and psychomotor development: the Danish National Birth Cohort. Paediatr. Perinat. 371. Victoria State Government Department of Health. Infertility in women. Victoria State
Epidemiol. 23, 98–106 (2009). Government https://www.betterhealth.vic.gov.au/health/conditionsandtreatments/
339. Diop, H. et al. Early autism spectrum disorders in children born to fertile, subfertile, infertility-in-women (2021).
and ART-treated women. Matern. Child. Health J. 23, 1489–1499 (2019). 372. Daniluk, J. C. & Koert, E. Fertility awareness online: the efficacy of a fertility education
340. Hargreave, M. et al. Association between fertility treatment and cancer risk in children. website in increasing knowledge and changing fertility beliefs. Hum. Reprod. 30,
JAMA 322, 2203–2210 (2019). 353–363 (2015).
341. Schendelaar, P. et al. Increased time to pregnancy is associated with less optimal 373. Goodyear, V. & Quennerstedt, M. #Gymlad — young boys learning processes and
neurological condition in 4-year-old singletons, in vitro fertilization itself is not. health-related social media. Qual. Res. Sport. Exerc. Health 12, 18–33 (2020).
Hum. Reprod. 29, 2773–2786 (2014). 374. Sangster, S. L. & Lawson, K. L. Is any press good press? the impact of media portrayals
342. Jenkins, T. G., Liu, L., Aston, K. I. & Carrell, D. T. Pre-screening method for somatic cell of infertility on young adults’ perceptions of infertility. J. Obstet. Gynaecol. Can. 37,
contamination in human sperm epigenetic studies. Syst. Biol. Reprod. Med. 64, 146–155 1072–1078 (2015).
(2018). 375. Berthelsen, A. S. N., Gamby, A. L. N., Christensen, U., Schmidt, L. & Koert, E. How do
343. Leitao, E. et al. The sperm epigenome does not display recurrent epimutations in patients young men want to receive information about fertility? Young men’s attitudes towards
with severely impaired spermatogenesis. Clin. Epigenetics 12, 61 (2020). a fertility campaign targeting men in Copenhagen, Denmark. Hum. Reprod. Open. 2021,
344. Asenius, F., Danson, A. F. & Marzi, S. J. DNA methylation in human sperm: a systematic hoab027 (2021).
review. Hum. Reprod. Update 26, 841–873 (2020). 376. Goldfarb, E. S. & Lieberman, L. D. Three decades of research: the case for comprehensive
345. Economidis, M. A. & Mishell, D. R. Jr. Pharmacological female contraception: an overview sex education. J. Adolesc. Health 68, 13–27 (2021).
of past and future use. Expert. Opin. Investig. Drugs 14, 449–456 (2005). 377. Warner, J. N. & Frey, K. A. The well-man visit: addressing a man’s health to optimize
346. Thirumalai, A. & Amory, J. K. Emerging approaches to male contraception. Fertil. Steril. pregnancy outcomes. J. Am. Board. Fam. Med. 26, 196–202 (2013).
115, 1369–1376 (2021). 378. Choiriyyah, I. et al. Men aged 15–44 in need of preconception care. Matern. Child. Health
347. Reynolds-Wright, J. J., Cameron, N. J. & Anderson, R. A. Will men use novel male J. 19, 2358–2365 (2015).
contraceptive methods and will women trust them? A systematic review. Ann. Rev. Sex. 379. Bodin, M., Tydén, T., Käll, L. & Larsson, M. Can reproductive life plan-based counselling
Res. 58, 838–849 (2021). increase men’s fertility awareness? Ups. J. Med. Sci. 123, 255–263 (2018).
348. World Health Organisation Task Force on Methods for the Regulation of Male Fertility. 380. Grau Grau, M. & Riley Bowles, H. in Engaged Fatherhood for Men, Families and Gender
Contraceptive efficacy of testosterone-induced azoospermia and oligozoospermia in Equality: Healthcare, Social Policy, and Work Perspectives (eds Grau Grau, M, las Heras
normal men. Fertil. Steril. 65, 821–829 (1996). Maestro, M. & Bowles, H. R.) 1–12 (Springer International Publishing, 2022).
349. Behre, H. M. et al. Efficacy and safety of an injectable combination hormonal 381. Hogg, K., Rizio, T., Manocha, R., McLachlan, R. I. & Hammarberg, K. Men’s preconception
contraceptive for men. J. Clin. Endocrinol. Metab. 101, 4779–4788 (2016). health care in Australian general practice: GPs’ knowledge, attitudes and behaviours.
350. Long, J. E., Lee, M. S. & Blithe, D. L. Update on novel hormonal and nonhormonal Aust. J. Prim. Health 25, 353–358 (2019).
male contraceptive development. J. Clin. Endocrinol. Metab. 106, e2381–e2392 382. Barnes, L. W. Conceiving Masculinity. Male Infertility, Medicine, and Identity
(2021). (Temple University Press, 2014).
351. Mommers, E. et al. Male hormonal contraception: a double-blind, placebo-controlled 383. WHO. Laboratory Manual for the examination of human semen and sperm-cervical mucus
study. J. Clin. Endocrinol. Metab. 93, 2572–2580 (2008). interaction (Cambridge University Press, 1999).
352. North, B. B. et al. Evaluation of the novel vaginal contraceptive agent PPCM in preclinical 384. WHO. WHO laboratory manual for the examination and processing of human semen
studies using sperm hyaluronan binding and acrosome status assays. Andrology 10, 5th edn (World Health Organization, 2010).
367–376 (2021). 385. Rodprasert, W. et al. An update on semen quality among young Finnish men and
353. Noman, M. A. A., Kyzer, J. L., Chung, S. S. W., Wolgemuth, D. J. & Georg, G. I. Retinoic acid comparison with Danish data. Andrology 7, 15–23 (2019).
receptor antagonists for male contraception: current status. Biol. Reprod. 103, 390–399 386. Jorgensen, N. et al. Recent adverse trends in semen quality and testis cancer incidence
(2020). among Finnish men. Int. J. Androl. 34, e37–e48 (2011).
354. Male Contraceptive Initiative. The Drug Development Pipeline. MCI https://www. 387. Jorgensen, N. et al. Human semen quality in the new millennium: a prospective
malecontraceptive.org/the-drug-development-pipeline.html (2023). cross-sectional population-based study of 4867 men. BMJ Open 2, e000990
355. Gruber, F. S., Johnston, Z. C., Barratt, C. L. & Andrews, P. D. A phenotypic screening (2012).
platform utilising human spermatozoa identifies compounds with contraceptive activity. 388. Gyllenborg, J. et al. Secular and seasonal changes in semen quality among young
Elife 9, e51739 (2020). Danish men: a statistical analysis of semen samples from 1927 donor candidates during
356. Chamberlain, S. G. et al. Reboot contraceptives research — it has been stuck for 1977-1995. Int. J. Androl. 22, 28–36 (1999).
decades. Nature 587, 543–545 (2020). 389. Huang, C. et al. Decline in semen quality among 30,636 young Chinese men from 2001
357. Hinton, L. & Miller, T. Mapping men’s anticipations and experiences in the reproductive to 2015. Fertil. Steril. 107, 83–88.e2 (2017).
realm: (in)fertility journeys. Reprod. Biomed. Online 27, 244–252 (2013). 390. Rao, M. et al. Evaluation of semen quality in 1808 university students, from Wuhan,
358. Mossman, J. A. & Pacey, A. A. The fertility fitness paradox of anabolic-androgenic steroid Central China. Asian J. Androl. 17, 111–116 (2015).
abuse in men. J. Intern. Med. 286, 231–232 (2019). 391. Zou, Z. et al. Semen quality analysis of military personnel from six geographical areas
359. Practice Committee of the American Society for Reproductive Medicine. Smoking and of the People’s Republic of China. Fertil. Steril. 95, 2018–2023.e3 (2011).
infertility: a committee opinion. Fertil. Steril. 110, 611–618 (2018). 392. Li, Y. et al. Semen quality of 1346 healthy men, results from the Chongqing area of
360. Healthy Male. Engaging Men in Primary Health Care. Healthy Male https://www. southwest China. Hum. Reprod. 24, 459–69 (2009).
healthymale.org.au/health-professionals/engaging-men-primary-health-care (2023). 393. Junqing, W. et al. Reference value of semen quality in Chinese young men. Contraception
361. Smith, J. A., Braunack-Mayer, A., Wittert, G. & Warin, M. “It’s sort of like being a detective”: 65, 365–368 (2002).
understanding how Australian men self-monitor their health prior to seeking help. 394. Gao, J. et al. Semen quality in a residential, geographic and age representative sample
BMC Health Serv. Res. 8, 56 (2008). of healthy Chinese men. Hum. Reprod. 22, 477–484 (2007).
362. Pearson, L., Holton, S., McLachlan, R. & Hammarberg, K. Australian men’s fertility 395. Wang, C. et al. Cross-sectional study of semen parameters in a large group of normal
information seeking attitudes and behaviour: a qualitative investigation. Sex. Reprod. Chinese men. Int. J. Androl. 8, 257–274 (1985).
Healthc. 29, 100621 (2021). 396. Itoh, N. et al. Have sperm counts deteriorated over the past 20 years in healthy, young
363. Stevenson, E. L. & McEleny, K. R. Male subfertility as a chronic illness: the role of adaptive Japanese men? Results from the Sapporo area. J. Androl. 22, 40–44 (2001).
challenges. Hum. Fertil. 20, 148–154 (2017). 397. Medras, M. et al. The quality of semen among a sample of young, healthy men from
364. Hadley, R. & Hanley, T. Involuntarily childless men and the desire for fatherhood. Lower Silesia (AndroLS). Endokrynol. Pol. 68, 668–675 (2017).
J. Reprod. Infant. Psychol. 29, 56–68 (2011). 398. Rahban, R. et al. Semen quality of young men in Switzerland: a nationwide
365. Hammarberg, K., Collins, V., Holden, C., Young, K. & McLachlan, R. Men’s knowledge, cross-sectional population-based study. Andrology 7, 818–826 (2019).
attitudes and behaviours relating to fertility. Hum. Reprod. Update 23, 458–480 (2017). 399. Lopez-Teijon, M., Elbaile, M. & Alvarez, J. G. Geographical differences in semen quality in
366. Sørensen, N. O. et al. Fertility awareness and attitudes towards parenthood among a population of young healthy volunteers from the different regions of Spain. Andrologia
Danish university college students. Reprod. Health 13, 146 (2016). 40, 318–328 (2008).
367. Prior, E., Lew, R., Hammarberg, K. & Johnson, L. Fertility facts, figures and future plans: 400. Mendiola, J. et al. Reproductive parameters in young men living in Rochester, New York.
an online survey of university students. Hum. Fertil. 22, 283–290 (2019). Fertil. Steril. 101, 1064–1071 (2014).
368. De Jonge, C. J., Gellatly, S. A., Vazquez-Levin, M. H., Barratt, C. L. R. & 401. Levine, R. J. et al. Air-conditioned environments do not prevent deterioration of human
Rautakallio-Hokkanen, S. Male attitudes towards infertility: results from a global semen quality during the summer. Fertil. Steril. 57, 1075–1083 (1992).
questionnaire. World J. Mens. Health 41, 204–214 (2022). 402. Uhler, M. L. et al. Relationship between sperm characteristics and hormonal parameters
369. Pedro, J., Brandão, T., Schmidt, L., Costa, M. E. & Martins, M. V. What do people know in normal couples. Fertil. Steril. 79 (Suppl. 3), 1535–1542 (2003).
about fertility? A systematic review on fertility awareness and its associated factors. 403. Selevan, S. G. et al. Semen quality and reproductive health of young Czech men exposed
Ups. J. Med. Sci. 123, 71–81 (2018). to seasonal air pollution. Environ. Health Perspect. 108, 887–894 (2000).

Nature Reviews Urology


Expert recommendation

404. Sheriff, D. S. & Legnain, M. Evaluation of semen quality in a local Libyan population. infertility, and identifying treatment strategies. A.P. has undertaken paid consultancy for
Indian J. Physiol. Pharmacol. 36, 83–87 (1992). Cryos International, Cytoswim Ltd, Exceed Health, Meals Group, and Merck Serono in the
405. Shine, R., Peek, J. & Birdsall, M. Declining sperm quality in New Zealand over 20 years. last 2 years, but all monies have been paid to the University of Sheffield (former employer).
N. Z. Med. J. 121, 50–56 (2008). M.K.O. is supported in part by the Bill and Melinda Gates Foundation in the area of
406. Hadjkacem Loukil, L. et al. Relation between male obesity and male infertility in a contraceptive development. All other authors declare no competing interests.
Tunisian population. Andrologia 47, 282–285 (2015).
Additional information
Acknowledgements Supplementary information The online version contains supplementary material available at
Thanks goes to R. Agnew, E. Heighton and A. Mergo from The University of Dundee for https://doi.org/10.1038/s41585-023-00820-4.
their assistance with data collection for Fig. 1. Thanks also goes to A. O’Connor from the
University of Melbourne for assistance with referencing. This research was supported in part Peer review information Nature Reviews Urology thanks Marco Alves and Gerhard Haidl for
by funding by the National Health and Medical Research Council of Australia to M.K.O., R.I.M. their contribution to the peer review of this work.
and J.A.V. (APP1120356); S.K. is a Canada Research Chair in Epigenomics, Reproduction and
Development and funding for this study is provided by the Canada Research Chairs program, Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
and the Canadian Institutes of Health Research grants (DOHaD Team grant 358654 and published maps and institutional affiliations.
Operating grant 350129). The authors thank ESHRE for financial and logistical support of
the MRHI. Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to
this article under a publishing agreement with the author(s) or other rightsholder(s); author
Author contributions self-archiving of the accepted manuscript version of this article is solely governed by the
All authors researched data for the article. S.K., R.A.A., C.L.R.B., C.J.D.J. and M.K.O. contributed terms of such publishing agreement and applicable law.
substantially to discussion of the content. All authors wrote the article. S.K., R.A.A., C.L.R.B.,
H.M.B., S.R.C., C.J.D.J., G.D., M.L.E., N.G., N.J., C.K., A.L., R.I.M., S.M., T.M., A.P., L.P., S.S., J.T., L.T.,
F.T., M.H.V.-L., J.A.V., F.Z. and M.K.O. reviewed and/or edited the manuscript before submission. Related links
Australian Male Infertility Exposure Study: http://www.mcri.edu.au/research/projects/
Competing interests australian-male-infertility-exposure-amie-study
C.L.R.B. is supported in part by the Bill and Melinda Gates Foundation in the area of European Society of Human Reproduction and Embryology: http://www.eshre.eu/
contraceptive development, Chief Scientist Office (Scotland), NHS and Genus Healthcare. Specialty-groups/Special-Interest-Groups/Andrology/MRHI
C.L.R.B. has received fees from Cooper Surgical for lectures on scientific research methods Family Planning 2030: https://fp2030.org/
outside the submitted work and Ferring for a lecture on the future of male reproductive health. Male Contraceptive Initiative: http://www.malecontraceptive.org
M.L.E. is an adviser to Ro, Doveras, VSeat, and Next. R.A.A. reports grants and personal fees NordFertil: https://nordfertil.org
from Roche Diagnostics, personal fees from Ferring Pharmaceuticals, IBSA, Merck Serono, United Nations Sustainable Development Goals 3 and 5: https://sdgs.un.org/goals
outside the submitted work. S.K. is an inventor on a Provisional patent 14647-P68337CA00
10 June 2022: Methods for identifying epigenetic modifications in sperm, diagnosing © Springer Nature Limited 2023

1
Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada. 2The Centre de Recherche du Centre
Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada. 3The Département de Pathologie et Biologie Cellulaire, Université de Montréal,
Montreal, Quebec, Canada. 4MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK. 5Division of Systems Medicine, School of
Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK. 6Center for Reproductive Medicine and Andrology, University
Hospital, Martin Luther University Halle-Wittenberg, Halle, Germany. 7Hudson Institute of Medical Research, Melbourne, Victoria, Australia. 8Department
of Obstetrics and Gynaecology, The Royal Women’s Hospital, Melbourne, Victoria, Australia. 9Department of Urology, University of Minnesota,
Minneapolis, MN, USA. 10Institut National de la Recherche Scientifique, Centre Armand-Frappier Sante Biotechnologie, Laval, Quebec, Canada.
11
Department of Urology and Obstetrics and Gynecology, Stanford University, Stanford, CA, USA. 12IVI Foundation, Instituto de Investigación Sanitaria La
Fe, Valencia, Spain. 13School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia. 14Department of Growth and
Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen
University Hospital, Rigshospitalet, Copenhagen, Denmark. 15Department of Experimental and Clinical Biomedical Sciences, ‘Mario Serio’, University
of Florence, University Hospital of Careggi Florence, Florence, Italy. 16Hudson Institute of Medical Research and the Department of Obstetrics and
Gynaecology, Monash University, Melbourne, Australia. 17Monash IVF Group, Richmond, Victoria, Australia. 18Department of Surgery and Cancer Imperial,
London, UK. 19Healthy Male and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia. 20Faculty of Biology,
Medicine and Health, University of Manchester, Manchester, UK. 21Centre for Reproductive Medicine and Andrology, University of Münster, Münster,
Germany. 22Departments of Paediatrics, Human Genetics and Pharmacology & Therapeutics, McGill University and Research Institute of the McGill
University Health Centre, Montreal, Quebec, Canada. 23Center for the Investigation of Environmental Hazards, Department of Paediatrics, NYU Grossman
School of Medicine, New York, NY, USA. 24Institute of Reproductive Genetics, University of Münster, Münster, Germany. 25Instituto de Biología y Medicina
Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Fundación IBYME, Buenos Aires, Argentina. 26Biosciences
Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK. 27Obstetrics and Gynecology Hospital, Institute of Reproduction
and Development, Fudan University, Shanghai, China.

Nature Reviews Urology

You might also like