Download as pdf or txt
Download as pdf or txt
You are on page 1of 67

REVIEW

Aging and the Male Reproductive System


Jean-Marc Kaufman,1 Bruno Lapauw,1 Ahmed Mahmoud,1 Guy T’Sjoen,1
and Ilpo Tapani Huhtaniemi2,3

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


1
Department of Endocrinology, Ghent University Hospital, 9000 Ghent, Belgium; 2Department of Surgery and
Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, London W12 0NN,
United Kingdom; and 3Department of Physiology, Institute of Biomedicine, University of Turku, 20520 Turku,
Finland
ORCiD numbers: 0000-0002-1400-6812 (J.-M. Kaufman); 0000-0002-1584-4965 (B. Lapauw);
0000-0001-6200-5817 (A. Mahmoud); 0000-0003-0457-9673 (G. T’Sjoen);
0000-0001-9092-7886 (I. T. Huhtaniemi).

ABSTRACT This narrative review presents an overview of current knowledge on fertility and reproductive hormone changes in aging
men, the factors driving and modulating these changes, their clinical consequences, and the benefits and risks of testosterone (T)
therapy. Aging is accompanied by moderate decline of gamete quality and fertility. Population mean levels show a mild total T
decline, an SHBG increase, a steeper free T decline, and a moderate LH increase with important contribution of comorbidities (e.g.,
obesity) to these changes. Sexual symptoms and lower hematocrit are associated with low T and are partly responsive to T therapy.
The relationship of serum T with body composition and metabolic health is bidirectional; limited beneficial effects of T therapy on
body composition have only marginal effects on metabolic health and physical function. Skeletal changes are associated primarily
with estradiol and SHBG. Cognitive decline is not consistently linked to low T and is not improved by T therapy. Although limited
evidence links moderate androgen decline with depressive symptoms, T therapy has small beneficial effects on mood, depressive
symptoms, and vitality in elderly patients with low T. Suboptimal T (and/or DHT) has been associated with increased risk of stroke,
but not of ischemic heart disease, whereas an association with mortality probably reflects that low T is a marker of poor health.
Globally, neither severity of clinical consequences attributable to low T nor the nature and magnitude of beneficial treatment effects
justify the concept of some broadly applied “T replacement therapy” in older men with low T. Moreover, long-term safety of T
therapy is not established. (Endocrine Reviews 40: 906 – 972, 2019)

A part from the physiological cessation of ovarian


function at menopause, the clinically most
important and best characterized aging-related changes
Testosterone (T) is the major anabolic hormone in
men as well as a key player in maintaining normal
reproductive and sexual function. The population mean
in the endocrine system concern pancreatic and thyroid of serum T levels declines with aging as a consequence
function. Other endocrine systems, for example, adrenal of the aging process per se and/or lifestyle changes and
function and GH/IGF- secretion, also undergo changes comorbidities, although the changes in serum T vary
upon aging, caused either directly by chronological aging greatly among individuals. Clinical changes with aging,
ISSN Print: 0163-769X
ISSN Online: 1945-7189
or indirectly by aging-associated changes in general which also occur at a variable pace among individual
Printed: in USA health, body composition, physical activity, and life- men, are in several aspects reminiscent of signs and
Copyright © 2019 style. Function of the hypothalamic–pituitary–testicular symptoms of hypogonadism in younger men (). It is
Endocrine Society (HPT) axis is not an exception, and the extent, clinical therefore not surprising that possible links between
Received: 16 July 2018
Accepted: 27 December 2018
significance, and treatment options of its age-related these hormonal and clinical changes have been the
First Published Online: deterioration are currently a topical and controversial subject of investigation and speculation. Indeed, a causal
19 March 2019 issue. relationship would in turn hold the appealing promise

906 https://academic.oup.com/edrv doi: 10.1210/er.2018-00178


REVIEW

ESSENTIAL POINTS
· In aging men there is a mild to moderate decline of gamete quality and fertility, which is accompanied by a relative
increase of the risk of adverse offspring outcome, but absolute risk remains low
· Hormonal changes in aging men are individually variable, with mean levels in the population showing a mild
decline of total testosterone (T), an increase of SHBG, a more pronounced decline of free T, and a moderate
increase of LH
· Comorbidities and, in particular, the increased prevalence of obesity play an important contributory role in the hormonal
changes in aging men: hypogonadism related to comorbidities tends to be of the secondary type, whereas changes related
to aging appear to be primarily testicular
· Among the most consistent associations with low T in elderly men are the occurrence of sexual symptoms and lower
hematocrit, and both seem at least partly responsive to T therapy
· The association of low T with mortality most likely reflects reverse causality, with low T a marker of poorer health

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


status
· Overall, neither the severity of clinical adverse effects attributable to low T, nor the nature and magnitude of beneficial
treatment effects justify the concept of a broadly applied T replacement therapy in older men with low T
· Long-term (cardiovascular and prostate) safety of T therapy has not been established

of possible beneficial antiaging effects of T “re- has become available. Whereas observational data
placement” therapy. In fact, the appeal of such ten- available for the  review () were mostly from
tative benefits of T therapy has been such that it has retrospective and cross-sectional studies, since then
translated during the last two decades into an prospectively obtained data are now available from
exploding increase of T use by middle-aged and elderly several large cohorts. Also, significantly contributing
men. This trend, which is particularly alarming in the new data from RCTs have become available. Second,
United States (, ), has apparently not been dis- changes in gonadal function in aging men and their
couraged by major knowledge gaps as to the benefit/ potential clinical consequences are of increasing clinical
risk profile of T therapy in these men (), and it is not relevancy, not only because of demographics with an
in line with the much more cautious and restrictive increasing number and proportion of elderly men in
recommendations by scientific societies (, ) and the the population, but even more so because the assertive
US Food and Drug Administration (FDA) () on generation of aging baby boomers has high expecta-
indications for T prescriptions. An apparent decline in tions as to quality of life and functional fitness in all
T prescriptions seen since  (, ) may be linked to domains, encompassing cognitive, physical, and sexual
the warning by the FDA and the voluntary cessation function. Moreover, there is the reality of the wide-
by drug companies of direct-to-consumer marketing spread misuse of, and publicity for, T therapy in the
of T products. management of physical performance. Finally, reports
The decline of androgen levels in elderly men and suggesting increased cardiovascular (CV) risk associ-
its clinical and therapeutic implications were ex- ated with T therapy (, ) and the FDA March 
tensively reviewed in this journal in  (). As to safety announcement () have generated discussion
clinical implications, the conclusion was then nu- and controversy. The FDA announced that manu-
anced, acknowledging that the hormonal changes facturers are required to change labeling of T products
might play a contributory role in part of the clinical to add information on possible increased risk of
alterations that accompany aging in some men, but heart attack and stroke in patients taking T and to
concluding that for many clinical signs and symp- clarify that prescription of T products is approved only
toms reminiscent of the clinical picture in young for therapy in men with low T due to certain condi-
hypogonadal men, the data remained inconclusive as tions, that is, due to disorders of the testicles, pituitary
to the causal role of age-related decline of androgen gland, or brain that cause hypogonadism. Moreover,
levels. The conclusion was frankly negative as to the the FDA states explicitly that, “The benefit and safety of
place of T therapy outside established pronounced these medications have not been established for the
symptomatic hypogonadism, because of lack of data treatment of low testosterone levels due to aging, even
from randomized controlled trials (RCTs) on hard if a man’s symptoms seem related to low testosterone”.
clinical endpoints and because the scale of performed This narrative review presents an updated overview
RCTs would clearly not allow for establishing clinical of the state of knowledge on changes in testicular
benefit and, even less so, long-term safety. function in aging men, the clinical consequences, and
An updated review of the evidence seems timely for the benefits and risks of T therapy. We focus primarily
several reasons. First, a substantial amount of new data on human data and largely on studies published since

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 907


REVIEW

the  review. For clinical data, we gave as much retained only RCTs. Thus, albeit comprehensive, this
priority as possible to studies with stronger design; for review does not intend to exhaustively cover all aspects
example, for evaluation of efficacy of T therapy we of the topic.

Spermatogenesis and Reproductive Capacity in an average maximum of . cm  between  and


Aging Men  years of age to  cm  between  and  years
of age (), again in line with the relatively well-
Parenthood is increasingly postponed, especially in preserved spermatogenic function of the aged
developed countries. Advanced age of childbearing testis. Histomorphometric studies on testes of
has been related to sociodemographic factors (). men between young adulthood and old age de-
Data from the United States indicated that mean tected clear age-related changes, albeit with large
paternal age has increased on average by . years individual variation (, ). The most common
during the past  years ( to ) from . to

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


feature of an aging testis is a mosaic of seminif-
. years of age (). Advanced paternal age is most erous tubular lesions ranging from complete
commonly (but not universally) defined as . years spermatogenesis to complete sclerosis of the
of age (). seminiferous epithelium. The changes include
narrowing of tubular diameter, thickening of basal
membrane with arrested spermatogenesis and fi-
Age-related changes in spermatogenesis brosis, and reduction of number of Sertoli and
A recent systematic review and meta-analysis evalu- spermatogenic cells, with vacuolization of the
ated semen data from almost , men extracted former and multinucleation of the latter. How-
from  studies (). The results indicated a consistent ever, spermatozoa are observed even in testes of
age-dependent decline in semen quality. Age was the oldest men. Thickening and herniation of the
associated with small to moderate declines in semen basal membrane with dilatation of seminiferous
volume, total sperm count per ejaculate, percentage tubules can also be observed (–), as well as
motility, progressive motility, and normal morphol- increased fibrosis and weight of tunica albuginea
ogy. Sperm concentration did not decline with ().
increasing age. A significant increase in sperm DNA
fragmentation has also been observed. Overall,
declined progressive motility and increased DNA Age-related changes in fertilizing capacity
fragmentation were the most conspicuous age- Several studies indicate that increasing male age is
related changes. Some studies have reported age significantly associated with rising time to spontane-
thresholds for the onset of decline in sperm quality, ous pregnancy and declining fertility rate [reviewed in
with the earliest decline starting from the age of  ()]. For example, a study of  planned preg-
to  years (, ). nancies indicated that, compared with men , years
The observed small decline in total sperm count of age, the ORs, adjusted for maternal age, for con-
with age in the meta-analysis discussed above () is ception in # months were ., ., and . in
corroborated by the results of a cross-sectional study men aged  to ,  to , and $ years, re-
of testicular volume (mostly representing sper- spectively (). The oldest reported age at biologically
matogenic tissue) and inhibin B (a marker of Sertoli confirmed parenthood is . years (). There have
cell function and spermatogenesis) in elderly been reports of fatherhood at an older age, but these
community-dwelling men  to  years of age (). were not confirmed with the now available tools to
The study indicated that the mean testicular volume biologically establish paternity.
in men . years of age is % smaller than in - Data from a prospective study of almost ,
to -year-old men. This difference in testicular pregnant women () indicated that pregnancies fa-
volume is accompanied by an almost twofold thered by a man aged $ years had almost twice the
elevation of serum FSH levels. Serum inhibin B risk of ending in a fetal loss compared with preg-
was slightly but significantly lower in the elderly nancies with younger fathers [hazard ratio (HR),
men by %, resulting in a marked decrease in the .]. The paternal age effect tended to be stronger for
inhibin B/FSH ratio. The latter findings together late fetal death. There are also a few studies suggesting
with the reduced testicular volume are consistent that male age may play a negative role in the success of
with the concept that in elderly men Sertoli cell intrauterine insemination cycles [reviewed in ()].
mass is reduced (), with a compensatory in- Conflicting results are reported on the effect of pa-
crease of FSH stimulation allowing for global ternal age on the outcome of in vitro fertilization
Sertoli cell function (and spermatogenesis) to (IVF) with or without intracytoplasmic sperm in-
remain relatively stable. Another study also jection (ICSI) [reviewed in (, , )]. A recent
reported a modest decrease of testicular size from study by Wu et al. () revealed that there were no

908 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

differences in the implantation and pregnancy rates in and . years of age sired offspring with lesser mean
conventional IVF cycles between the different pa- intelligence scores (). Relative risks (RRs) are high
ternal age groups when maternal age was , and  for an adverse health outcome in the offspring (RR,
to  years. However, implantation and pregnancy . to .), but absolute risks remain small. For
rates decreased with paternal age in the - to -year- example, for a father . years of age, the RR for
old maternal age group. Ma et al. () recently achondroplasia in the offspring is . with an ab-
reported a lower OR of live birth and a higher risk of solute risk of  out of  [for details, see Ram-
abortion in the paternal age group of $ years in asamy et al. ()]. Counseling regarding the risks of
IVF/ICSI cycles. Wu et al. () found that increased advanced paternal age should still be conducted with
paternal age negatively influences the number of high- all couples. Sperm cryopreservation is a possible
quality embryos, but it has no effect on pregnancy option. No current guidelines recommend the
outcomes in couples undergoing IVF/ICSI cycles. routine use of preimplantation genetic diagnosis/
Paternal age did not affect ongoing pregnancy rates in screening based on advanced paternal age alone ().
the first IVF/ICSI cycles as reported by Meijerink et al.

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


(), but live births were not taken into account. Few Conclusion
data are available on the outcome of testicular sperm Unlike the situation in the female, the decline in
extraction in advanced paternal age. One study gamete quality and fertility in the male is only mild
found that paternal age as an independent factor did to moderate and gradual, possibly starting at the age
not affect embryo quality and pregnancy outcomes of  to  years. This decline is accompanied by an
of IVF/ICSI using testicular spermatozoa (). In increase in the RRs of adverse outcomes affecting
summary, the bulk of evidence supports, although physical and mental health of the offspring. The
not unequivocally, a decline in male fertility with absolute risks, however, remain small, precluding a
increasing age in both unassisted and assisted screening strategy or dissuading fatherhood at a
reproduction. particular age (). Counseling concerning these effects
is, however, recommended (, , ). Semen cryopres-
ervation at a young age is a possible option, but there are
Pathophysiologic mechanisms underlying changes
currently no studies reporting on the outcome. The effect
in reproductive capacity
of treatments aiming at improving male fertility, for “Unlike for total serum T, the
Several local urogenital and systemic changes oc-
example, varicocele correction () and therapy with findings on free T are quite
curring with advancing age [see Table  (, , ,
antioxidants (), are largely unexplored in men . uniform, with all reporting an
, –)] can contribute to decreased coital age-related decline….”
years of age.
frequency, decreased semen quality and impaired
sperm genetic integrity and reproductive capacity,
increased fetal loss, and adverse health outcomes in
Sex Steroids in Aging
the offspring.
Influence of aging on blood concentrations
Offspring health
Although older paternal age does not appear to pose Testosterone
an independent risk of adverse perinatal outcomes, in Most studies report a slow aging-related decline in
pregnancies achieved either with or without assisted the circulating concentration of T, starting during
reproductive technology (), negative effects on the third to fourth decade of life and continuing
physical and mental health in later life have been gradually until extreme longevity (). The decline is
reported as summarized below [Table  (, , –)]. not only due to chronological aging, but it is also
The age-related (epi)genetic and genomic changes that strongly influenced by the concomitant weight gain
occur in the male germ line have been recently and other common aging-associated comorbidities
reviewed (). The authors documented an increase in (Fig. ). Longitudinal studies report annualized
congenital birth defects and neurologic disorders and a decreases of T with a range of .% to .% (–).
statistically significant increase in -year offspring Cross-sectional studies showed somewhat smaller
mortality. changes (, ). Cohort and secular effects, labo-
Reviews of studies on mental health indicate a ratory drift, and stability of samples upon storage
tripling of the risk of schizophrenia and autistic may explain the difference. One clear confounder is
spectrum disorders in children of fathers . years the expanding epidemic of obesity, which is bound
of age compared with fathers aged  to  years to differently affect T levels at the two ends of an
(), a % increased risk of bipolar disease for observation period, with effects of weight gain
children whose fathers were . years of age at their augmenting the apparent age effect. The European
birth compared with children of younger fathers Male Aging Study (EMAS) found the cross-sectional
(), as well as lower intelligence and learning decline of T in men between  and  years of age
abilities with older paternal age. Fathers , of age to be .% per year (), and the longitudinally

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 909


REVIEW

including ethnicity, cohort selection, and analytical


Table 1. Factors That May Contribute to Decreased factors. Importantly, however, T concentration in
Reproductive Capacity in Advanced Paternal Age
most aging men remains within the range of young
Hormonal and metabolic changes, for example, decreased T men, and the levels infrequently drop to the clearly
(see text in this review) hypogonadal range.
Testicular changes, decreased testicular volume Unlike for total serum T, the findings on free T
are quite uniform, with all reporting an age-related
Body weight increase decline, usually of the order of .% to .% per
Decreased physical activity and lean muscle mass annum [e.g., see ()]. This is explained by the
constant finding of an aging-related increase of
Decreased bulk and strength of pelvic floor muscles SHBG by ~% per year, which results in a decline of
Systemic disease (e.g., CVD, hypertension, malignancies) free T. However, in real life, the increase of SHBG
can be counterbalanced by an opposite trend caused
Urogenital disease (including infections, prostatic disease,
by obesity and other health problems (see below).

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


varicocele)
Therefore, the real impact of T changes has to be
Therapy (for prostatic disease, hypertension, hypogonadism, assessed on the basis of increased SHBG due to aging
malignancy) and the concomitant decrease caused by comor-
Erectile and ejaculatory dysfunction, decreased libido, and bidities. As a representative example of longitudinal
coital frequency changes, in the EMAS the annual longitudinal de-
crease of total T was .%, the increase of SHBG
Cumulative effects of lifelong exposures to toxins and was .%, and calculated free T decreased by .%
pollutants
per year (). The levels of bioavailable, that is,
Excessive generation of reactive oxygen species (DNA non–SHBG-bound T (the sum of free T and
damage) albumin-bound T), decline in parallel with free T
Genetic changes (e.g., mutations, aneuploidy)
upon aging (, ). The mechanisms of aging- and
obesity-dependent changes in SHBG are discussed
Epigenetic changes (DNA methylation, histone modifications, below.
and miRNA expression)
Active T metabolites
Besides T, aging-related changes also occur in
concentrations of its two active metabolites, that is,
assessed decline during a .-year follow-up was DHT and estradiol (E). Although well-validated
.% per year (). The annualized decline of immunoassay methods are able to reliably monitor
testicular steroidogenesis appears to accelerate with male serum T levels (see below), only the results of
advancing age, as an annualized % decline of T and newer mass spectrometry (MS)–based measure-
.% of DHT was recently reported in men between ments of DHT and E can be considered reliable (,
 and  years of age (). Another study in men  ). DHT levels are ~- to -fold lower than those
to  years of age (mean,  years of age) at baseline of T, and there is high correlation (r 5 .) between
reported an annualized .% T decline during  concentrations of the two androgens (). One study
years (). has established the reference range (mean 6  SD)
The decreasing trend of T can be found in of DHT for healthy men  to  years of age as .
random community-dwelling populations, but when to . nmol/L (). Handelsman et al. () ob-
men in exceptionally good health are studied, some served in a large population-based sample (n .
(, ), but not all (), cross-sectional studies have ,) that DHT levels change minimally before
found no significant age-related decline of T. Also, the age of  years but decrease gradually thereafter
several older studies were unable to document the from a mean of . to . nmol/L in men . years
predominant finding of aging-related decline in of age (i.e., by ~.% per year). However, it is ap-
serum total T (). In the absence of convincing parent that circulating concentrations of DHT are a
longitudinal data, the effect of aging alone on T poor indicator of action of this hormone, whose
remains open. Interestingly, recent cross-sectional action in target tissues (e.g., prostate) is largely
and longitudinal studies from Japan, Korea, and independent of serum levels but is determined by
China (–) reported either no aging-related in situ conversion of T to DHT (). Also, the
change or increases in T, which persisted after ad- concentration of a weak androgen and substrate
justment for usual confounders, thus suggesting of T production, androstenedione, decreases with
ethnic differences in this phenomenon. In conclu- aging ().
sion, the majority of studies show a slight gradual Data on aging-related changes in E are in-
decline in total T levels in men upon aging. The consistent [reviewed in ()] due at least in part to
disparate findings may have multiple explanations, poor accuracy of the immunoassays used in older

910 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

Table 2. Adverse Health Outcomes in the Offspring in Advanced Paternal Age


Disorder RR (or Other) Reference
Reviewed in (9, 11, 39–43) .
Craniosynostotic diseases Abbreviation: NA, not available.

Apert syndrome 9.5 (11)

Crouzon syndrome 8 (11)

Pfeiffer syndrome 6 (11)

Affected growth/connective tissues

Achondroplasia 7.8 (11)

Thanatophoric dysplasia NA (strong evidence) (41)

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Osteogenesis imperfecta NA (strong evidence) (41)

Syndactyly NA (strong evidence) (41)

Cleft palate NA (medium evidence) (41)

Multiple endocrine neoplasia syndromes (MEN2A and MEN2B) NA (42)

Neurofibromatosis I 3.7 (11)

Marfan syndrome NA (strong evidence) (41)

Down syndrome 1.37 (11)

Klinefelter syndrome 1.6 (11)

Retinoblastoma 3 (11)

Brain tumors 1.69 (11)

Childhood leukemia 1.14 (11)

Non-Hodgkin lymphoma 1.51 (40)

Breast cancer 1.6 (11)

Increased 5-y mortality 1.65 (HR) (43)

Epilepsy 1.3 (11)

Bipolar disease 1.37 (40)

Schizophrenia 2.9 (39)

Autistic spectrum disorders 3.45 (HR) (39)

Lower intelligence NA (40)

Lower learning abilities 1.70 (OR) (40)

studies. By MS, the concentrations of E are, year) and estrone (.% per year), and that the
according to Handelsman et al. (), relatively stable, changes were only partly explained by body mass
showing a minor .% annual decrease. Hsu et al. index (BMI), diabetes status, and comorbidities.
() found in a -year follow-up study in a group of Thus, several studies have found E levels to increase
apparently healthy men, from the baseline age of  during aging, unlike the age-related decreasing
years a % annual increase of E and a % decrease trends of T and DHT. A higher conversion rate of T
in estrone. In a very recent .-year longitudinal to E (in adipose tissue) and increased SHBG levels
study on -year-old men E levels remained stable in aged men may explain the E findings. However,
(). Jasuja et al. () reported age-related increases calculated free E and non–SHBG-bound E still
between  to  and 1 years in both E (.% per show an age-dependent decrease (, ).

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 911


REVIEW

Adrenal androgens immunoassays documented longitudinal annual


The adrenal gland also produces androgens and decreasing trends of .% for DHEA, .% to .% for
their precursor steroids, most notably dehydro- DHEAS, and .% for androstanediol glucuronide
epiandrosterone (DHEA) and its sulfate conjugate (, , ). Some discrepant findings, demon-
(DHEAS). Their production and secretion clearly strating no age-related changes in adrenal andro-
decrease with aging in both sexes. This decrease is gens, are most likely due to regression to the mean
functionally significant in women, but because of [reviewed in ()]. The concentration of circulating
their weak androgenic activity, their overall contri- T in castrated men is ~% of that in intact men,
bution to male androgens is of lesser importance. which apparently represents the contribution of
Several earlier studies have monitored the aging- adrenal androgens to serum T ().
related changes in adrenal androgen production in A comprehensive study of Labrie et al. ()
men [e.g., see (, , )]. Earlier studies using showed a dramatic decline (% to %) in the

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022

Figure 1. Relationship between age, BMI, total and free T, LH, and SHBG of 3220 men aged 40 to 79 y, from the EMAS. The cohort was
stratified according to BMI into men who are nonobese (BMI ,25 kg/m2), overweight (BMI of 25 to 30 kg/m2), and obese (BMI .30 kg/
m2). The graphs present mean (with 95% CI in shaded area and vertical lines) levels of total and free T, LH, and SHBG. (a) Total T
concentration decreases in men who are overweight/obese compared with nonobese men at all ages. (b) Free T, similar to total T, is
reduced in men who are overweight/obese compared with nonobese men at all ages. (c) LH increases with age, but is not associated
with BMI, and it was not significantly different among the three BMI groups at the median age of 60 y. LH was higher in nonobese
patients aged .70 y compared with the overweight and obese groups, owing to a negative age interaction. (d) SHBG increases with age.
For total T and SHBG, the age trends in the three BMI categories were similar, indicating lack of interaction between BMI and age. The
free T vs age trend in the group with obesity was less steep than in the other two groups, indicating an interaction between BMI and age.
Male reference ranges are as follows: T, 288 to 1009 ng/dL (10 to 35 nmol/L); free T, 7.2 to 20.2 ng/dL (250 to 700 pmol/L); LH, 1.5 to 8.0
IU/L; SHBG, 13 to 62 nmol/L. [Reproduced with permission from Wu FC, Tajar A, Pye SR, et al. Hypothalamic-pituitary-testicular axis
disruptions in older men are differentially linked to age and modifiable risk factors: the European Male Aging Study. J Clin Endocrinol
Metab 2008;93(7):2737–2745.]

912 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

circulating levels of several adrenal androgens [DHEA, Afro-Caribbean than European men, but the dif-
DHEAS, -androstene-b,b-diol (-diol), -diol- ference disappeared in - to -year-old men,
sulfate, -diol-fatty acid esters, and androstenedione] and a reverse difference prevailed in DHEA and
in a cross-sectional study between the ages of  to  DHEAS (). A meta-analysis demonstrated that, after
years. Most of the decline took place before the age of adjustment for age, black men have a modestly but
 years. Solvent extraction and chromatography were significantly .% to .% higher level of free T, but not
used to improve specificity of the immunoassay of total T, E, or SHBG, compared with white men
measurements. It was calculated that the adrenals (). In contrast, a large study reported higher E, but
contribute to % to % of the molar content of not T, levels in black men than white men (). A large
androgens in the circulation of a middle-aged man. international study reported differences between older
However, the adrenal contribution to androgen bio- men of different ethnic origins. The most conspicuous
activity is much less, because most adrenal androgens finding was the significantly higher serum T con-
have low or marginal bioactivity. centrations, compared with European and African
A recent study () measured by liquid American men, of Asian men living in Hong Kong and

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


chromatography–tandem MS (LC-MS/MS) the ref- Japan, but not of Asian men living in the United States.
erence intervals for  steroids (pregnenolone, pro- Furthermore, the differences were much narrower
gesterone, -deoxycorticosterone, corticosterone, after adjustment for BMI (). Hence, besides genuine
aldosterone, -oxycortisol, -hydroxycortisol, ethnic differences, lifestyle factors may be a major
-hydroxyprogesterone, -deoxycortisol, -deoxy- contributing factor to the apparent ethnic differences
cortisol, cortisol, cortisone, DHEA, DHEAS, andros- in hormone levels as well as to the large variation of the
tenedione, and T) in men and women between  findings.
and  years of age. Strong negative correlation with A recent study () found that South Asian men
age was observed with all steroids, both of testicu- have lower T levels (and higher LH levels) than do
lar and adrenal origin (with the exception of white European and Afro-Caribbean men, but there
-deoxycortisol). Of note was that the age-related were no differences in free T levels after controlling
decrease of T lost significance in this study after ad- for covariables (i.e., body fat). Total and free T levels
justment for BMI. were not strongly related to adiposity in South Asian
men in contrast to the other two ethnic groups. These
Factors affecting blood levels of T in the elderly findings may be important for the assessment of
Many of the factors discussed below are not directly suspected hypogonadism and for understanding its
related to aging, but rather they contribute to the large mechanisms in different ethnic groups. They may
individual variability ( to  nmol/L) prevailing in also explain why the aging-related decline of T has
circulating total T levels. However, their importance not been found in some Asian populations (see
may vary during a man’s lifespan and become more above).
important in old age when the body’s compensatory
capacity is approaching its limits. Genetic differences
A number of candidate genes regulating androgen
Ethnic differences synthesis, kinetics in circulation, metabolism, and
Limited and variable data exist on the effects of action in target cells have been found to contribute to
ethnicity on T levels. Besides genetics, regional and the actual circulating T concentration. Studies in twins
ethnic differences in lifestyle factors may confound and siblings have shown a strong .% heritability in
these findings. For example, Asian men have pro- serum T concentrations (–). Some genetic vari-
portionally more visceral fat compared with Euro- ants (polymorphisms) contributing to serum T levels
pean men (), which may impinge differently on have already been detected, but most genetic factors
their HPT function. Total T and calculated free T contributing to the wide variation of physiological
concentrations have been reported to be lower in T concentrations at all ages still remain unknown.
Pakistani compared with white European and Af- Earlier studies used candidate gene approaches, but
rican Caribbean men, all living in the United also data from genome-wide association studies, are
Kingdom (). A similar study compared serum T now available.
and several end manifestations of androgen action With respect to individual genes, those most ex-
[prostate-specific antigen (PSA), lean body mass, tensively studied have been androgen receptor (AR)
bone density] in groups of Indian and European men and SHBG. The polymorphic AR gene exon  CAG
living in the United States, and it found total and free repeat variant, whose length correlates inversely with
T and phenotypic androgen manifestations to be receptor activity, has minor effects on circulating T
lower in Indian men (). A larger United States– levels, but they reach statistical significance with large
based study could not find differences in T between sample sizes (, ). A report from the EMAS cohort
white, black, and Hispanic men (). Another study confirmed direct associations of this repeat poly-
found higher T and DHT levels in - to -year-old morphism with circulating levels of total and free T

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 913


REVIEW

and E (). Also, another AR gene repeat poly- magnitude of T changes caused by changes in weight. In
morphism (GGN) has a small effect on circulating T the EMAS (Fig. ), in middle-aged and older men, total
levels (). Several studies have reported ethnic dif- and free T in those with BMI $ kg/m were ~ nmol/L
ferences in the length of the CAG repeat with an and  pmol/L lower, respectively, in comparison with
average of  in whites and  in blacks (). Another nonobese men. The former difference is more than that
study found the following ethnic differences of the caused by aging from  to  years, and the latter equals
CAG repeat length: Afro-Caribbean (mean repeat ~ years of aging. No concomitant increase in LH levels
length, .) ,white (.) ,Hispanic (.) ,Thai occurs in men with obesity, indicating that their hypo-
(.) (). The CAG repeat length of Indian men has gonadism is of the secondary hypothalamic–pituitary
been reported to be similar to European men (). type.
Two studies have shown that the AR gene CAG repeat A reciprocal interaction prevails between T and
affects serum T levels only in older men (, ), but obesity. On the one hand, T therapy reduces the fat
this finding could not be repeated in other studies (, mass (, ), and androgen deprivation therapy
). (ADT) results in weight gain and increased adiposity

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Several polymorphisms of SHBG have been found (). In turn, changes of body weight affect T levels,
to associate with circulating androgen and/or es- which increase upon weight loss, either through diet
trogen levels in men (–). A genome-wide as- or bariatric surgery (, ), and decrease upon
sociation study identified two single-nucleotide weight gain, which is the most important single
polymorphisms (SNPs) at the SHBG locus and one reason for aging-related decrease of serum T. The
near FAMB on the X chromosome that were as- effects of obesity on T appear more marked than vice
sociated with low T concentration (). The SNP in versa as illustrated by limited effects of severe T
exon  affected the affinity of SHBG for binding T lowering on BMI during  months in a controlled
and the measured free T fraction. The polymorphism study () (see also “Adipose tissue” under “Adipose
is rare, but it could in principle influence calculations tissue and metabolic health” below). The fact that T
of free T based on the molar concentrations of T and and LH rise after weight loss supports the causative
SHBG. The individual effects of the three SNPs on role of obesity in T suppression rather than the
serum T concentration were .%, .%, and .%. opposite (, ). The weight loss–related improve-
Another SHBG polymorphism has been found to ment of HPT function is more effective in younger
associate with increased T (.%) and SHBG (.%) men, highlighting the multiplicity of mechanisms (in-
levels (). cluding deteriorating health) influencing the aging HPT
A number of polymorphisms have been reported axis ().
to affect circulating T levels in following genes: CYP What exactly causes the secondary hypogonadism
(), NCOA (), CYPA (), BHSD (), with gonadotropin suppression in men with obesity is
UGTB (), FSHB (), JMJDC (), ESR and still incompletely understood. It is apparent that the
ESR (), MMP and CD (), GSTM (), expanding adipose tissue and the multiple signaling
FSHR (, ), ACTN (), VDR (), CLOCK molecules produced by fat cells play a role. Leptin is
(), GPCRA (), MAMLD () SRDA (), one candidate peptide produced by fat cells (). A
and GnRH (). Many potentially important T–leptin link is suggested by the finding that T
polymorphic influences have apparently not yet been treatment of men decrease their leptin levels ().
identified, and only a small fraction of the large Most studies, however, demonstrate direct correlation
between-subject variation of serum T can so far be between leptin levels and gonadotropins, and leptin
explained by known polymorphisms. Whether ge- resistance in obesity has been offered as a mechanism
netic factors affect the aging-related decline of T has for the tenuous involvement of this hormone in the
only been studied to a limited extent. Hence, pathogenesis of obesity-associated secondary hypo-
whether there is a genetic component affecting the gonadism (). Other candidates/mechanisms are the
rate and depth of T changes upon aging remains proinflammatory cytokines produced by fat tissue (e.g.,
unclear. TNFa, IL-, and IL-), whose increased levels in
obesity might suppress gonadotropins (, ), as
Lifestyle-related factors well as central nervous system endocannabinoids ()
Overweight and obesity. Overweight and and central insulin resistance (). Furthermore,
obesity are the most obvious and most common adiponectin can modulate GnRH and gonadotropin
lifestyle factors that have important influence on cir- secretion ().
culating T levels (, ). In men who are overweight/ The long-held theory of increased adipose tissue
obese, total T decreases concomitantly with decreased estrogen production and its increased feedback in-
SHBG, whereas free T decreases less or remains initially hibition of GnRH/gonadotropin secretion may not
unaltered and becomes clearly suppressed only in hold true in light of recent findings. Estrogen levels in
massive obesity (, ). Data from the EMAS (, ) men with obesity are in fact low, rather than high,
and Massachusetts Male Aging Study () describe the (–) and increase only in men with extreme

914 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

obesity (, ). A recent study demonstrated that production is found in animal experiments during
hyperinsulinemia combined with elevated lipids chronic stress (). Deslypere and Vermeulen ()
suppresses gonadotropin secretion through direct found that younger men respond to stress such as
pituitary action, which provides another candidate hypoglycemia or cardiac infarction with suppression of
mechanism for the obesity-associated hypogonado- T, whereas such a response was not found in older
tropic hypogonadism (). Finally, the obesity- men. Low free T has also been associated with de-
associated decrease in SHBG production (see pression (see below).
below) might play a role by lowering the set point of Physical stress and exercise. Short intense
the hypothalamic–pituitary feedback inhibition in exercise increases serum T levels independent of go-
relationship to circulating total T. Because in men nadotropin stimulation (). In contrast, T is sup-
with obesity mainly total T, and to a lesser extent free pressed during more prolonged exercise stress,
T, is suppressed, negative feedback inhibition of probably through the combined effects of suppressed
gonadotropins can be achieved at a lower concen- gonadotropins and increased cortisol and cathecol-
tration of total T. In conclusion, apparently multiple amine levels. Endurance training in men induces

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


simultaneous mechanisms in obesity participate in subclinical hypothalamic–pituitary inhibition of the
the suppression of gonadotropins. HPT axis (, ).
Diet. A relatively minor influence of dietary Numerous studies have examined the effects of
factors has been reported on serum T levels (). aging on the response of the HPT axis and T pro-
Macronutrient distribution in the diet has been shown duction to exercise. The increase of T following acute
to affect hormone levels. A low-fat diet (#%), in- physical stress has been found suppressed in -year-
dependent of weight reduction, is associated with old men in comparison with younger men (, ).
lower serum T concentrations when compared with Studies on whether lifelong exercise training increases
higher fat diet in men (–), and dietary intake of circulating levels of T have yielded conflicting results
energy in the form of fat and protein increases T levels (–). Some studies indicate that lifelong physical
(). Weight loss in men who are overweight/obese activity attenuates the aging-related decline of T (,
with high-protein or high-carbohydrate diets resulted ), but there are also negative findings (, ).
in an equal increase in free and total T, as well as Although lifelong exercise may not affect T level, when
SHBG (). The reason for lower T production high-intensity training is initiated in sedentary men, “…healthy lifestyle, including
upon a low-fat diet is not known, but fiber may reduce their total T, free T, and SHBG show significant in- good physical condition, may
the reabsorption of steroids excreted through the creases (, , ). Taken together, these findings reduce the aging-associated
biliary tract, or lower cholesterol may provide less indicate that healthy lifestyle, including good physical decline of HPT function.”
substrate for steroidogenesis. When the same weight condition, may reduce the aging-associated decline of
loss was achieved by diet alone and diet combined with HPT function. In support of this conclusion, Yeap
excercise, no difference in T elevation was observed et al. () reported that engagement of men .
(). years of age in healthy behaviors, with one of these
The short intermittent fasting of Ramadan sig- being physical exercise, predicts higher total T and
nificantly reduces body weight and sexual desire and SHBG, but not free T. When a man engages in ex-
activity without affecting serum T levels (). Upon ercise, he most likely has also other health behaviors,
controlled nutritional intervention, argan and olive and the role of exercise in isolation is difficult to single
oil consumption was reported to increase serum T out.
and LH concentrations in conditions with unaltered Smoking. A recent meta-analysis summarized
body weight (). Some (), but not all (), information on the effect of smoking on T levels ().
studies have shown that soy supplements in the diet It was found that in  studies on , men, with a
reduce serum T levels. Collectively, it is apparent mean age of  to  years, smokers had on average
that the quality of diet, which may change during a . nmo/L higher total T than did nonsmokers. The
man’s lifespan, does not have major effects on HPT increase has been found to parallel with increased
function. SHBG levels and with no significant change in free T (,
Psychological stress. Numerous studies have , ). The elevation of T is expected because nicotine
reported a negative effect of psychological stress on and/or its metabolites share the same disposal pathway
semen quality (, ), but data on the HPT axis are with androgens (). The SHBG elevation occurs
variable. A recent study reported that in men with possibly because of the toxic effects of tobacco smoke
elevated cortisol and norepinephrine levels, but not components on liver function. No studies exist on
with low levels, life events were associated with in- possible age-related differences in the effects of smoking.
creased serum LH and T (). Besides the Alcohol. Acute alcohol intoxication suppresses
hypothalamic–pituitary–adrenal axis, acute psycho- serum T levels, probably through direct testicular
logical stress has been shown to activate the HPT axis, effects, but concomitantly increased adrenal secretion
leading to increased T levels (). An opposite of cortisol may contribute to the effect (). Most
negative effect of adrenocortical activation on T studies do not find an effect of moderate alcohol

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 915


REVIEW

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022

916 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

Figure 2. (a) Relationships between age, weight changes, and hormone changes in the EMAS during an average 4.3 y follow-up
period. Values are the unadjusted mean changes of 10-y age bands with 95% confidence interval. The cohort was stratified according
to weight change into 3 groups: $10% loss (closed square), stable (within 10% of baseline, closed circle) and $10% gain (closed
triangle). Weight changes appeared to influence hormone levels similarly at all ages. [Reproduced with permission from Camacho
EM, Huhtaniemi IT, O’Neill TW, et al. Age-associated changes in hypothalamic-pituitary-testicular function in middle-aged and older
men are modified by weight change and lifestyle factors: longitudinal results from the European Male Ageing Study. Eur J Endocrinol
2013;168(3):445–455.] (b) Mean hormone changes by percentage weight change in the same EMAS population as in (a). The cohort
was stratified (x-axis) according to weight gain or loss into seven separate groups from .15% loss to .15% gain. The number 0 on
the x-axis represents the “within 5%” group. Error bars represent 61 SEM. Data are adjusted for baseline age and center, changes in
smoking status, alcohol consumption, comorbidities, and physical activity. Total and free T showed a cubic relationship, SHBG a
linear relationship, and LH a cubic relationship with weight change. Free T changed only with the highest level of weight change
(15%), which is a reflection of its derivation from T and SHBG. The linear change of SHBG buffers the changes of free T, which only
responds to extreme changes in weight. Hypothalamic–pituitary inhibition, reflected by LH levels, is lifted only following major
weight loss. Significantly different from (referent): *P , 0.05; **P , 0.01. [Reproduced with permission from Camacho EM,
Huhtaniemi IT, O’Neill TW, et al. Age-associated changes in hypothalamic-pituitary-testicular function in middle-aged and older

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


men are modified by weight change and lifestyle factors: longitudinal results from the European Male Ageing Study. Eur J Endocrinol
2013;168(3):445–455.]

consumption on serum T levels (, ). One study action, bring about substantial suppression of circu-
reporting on drinkers found increased total and free T lating T concentrations. Spironolactone, an inhibitor
levels, as well as decreased SHBG (), but another of several steroidogenic enzymes, also suppresses T
study found lower T levels in drinkers (). No production, besides having an AR antagonistic action.
correlation of alcohol consumption with serum T was In contrast to findings in experimental animals,
found in the EMAS (). A surprising finding was cimetidine, a weakly antiandrogenic H antagonist,
made in a recent study on . men from Europe has not been found to suppress T levels in men treated
and the United States with a linear association between for peptic ulcer.
the amount of alcohol consumption and serum total Opioids cause secondary hypogonadism by inhib-
and free T, in the face of unaltered SHBG (). This iting GnRH secretion. Opioid users have a higher
effect was explained by alcohol detoxification leading likelihood of having low T (OR, .), and after con-
to changed steroid metabolism in the liver. Alcohol trolling for opioid use, the men . years of age have
oxidation increases the NADH/NAD1 ratio and an OR of . for low T compared to the men  to 
changes the redox state in favor of androstenedione years of age, and the men with more than two
conversion to T (). comorbidities have an OR of . compared to the men
Serum T levels are suppressed, and those of go- with no comorbidities (). Dopamine agonists such
nadotropins are elevated, in chronic alcoholics but as bromocriptine increase, and antagonists such as
both normalize during abstinence, indicating that metoclopramide decrease, serum T concentrations
ethanol-induced hypogonadism is primary in origin, (). It is well documented that chronic glucocorti-
functional in nature, and transient in its evolution coid treatment suppresses T production by combined
(). Whether the HPT axis responds differently to inhibiton of gonadotropin secretion and testicular
alcohol in older than younger men has not been steroidogenesis (). A recent study demonstrated
studied. that ibuprofen reduces the Leydig cell sensitivity to LH
stimulation, inducing a condition reminiscent of
Medications compensated hypogonadism with normal T and an
Several drugs interfere with T production, metabolism, increased LH/T ratio (). Hypothetically, the use of
or actions. Although little is known about the possible COX inhibitor–type pain killers may therefore amplify
effects of aging on these actions, they play an in- the aging-related deterioration of Leydig cell function
creasingly important role in T action in the elderly, and accelerate the transit from compensated to real
because most of them are used in the treatment of hypogonadism.
comorbidities occurring in old age. Statins and
-hydroxy--methylglutaryl coenzyme A reductase Disease-related factors
inhibitors decrease cholesterol production, and be- A major question with respect to functional hypo-
cause it is the substrate of T production, there is some gonadism is the direction of causality between low T
concern on T suppression in statin users. There is and comorbidities. The question is important because
some evidence that this is the case, but the clinical of its major impact on treatment strategies. In the
significance of the small suppression (average of ~.%, relationship with some symptoms or diseases, for
i.e., 2. nmol/L; % CI, 2. to 2. nmol/L; in a example, impaired sexual function, osteoporosis, and
meta-analysis) is not clear (). Steroid sythesis anemia, low T seems obviously the causal factor (see
inhibitors (aminoglutethimide, ketoconazole) and below). In those with other diseases (e.g., critical
GnRH analogs, by virtue of their mechanisms of illness), low T is most likely the response to

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 917


REVIEW

pathophysiological stress caused by disease. Often, but not in ankylosing spondylitis or lupus eryth-
however, the causality can work similar to a vicious ematosus disseminatus, serum total T and free T
cycle in both directions. It is also uncertain whether concentrations are consistently low, but both elevated
the comorbidity-associated suppression of HPT ac- () and normal/suppressed () gonadotropin
tivity is a pathological, neutral, or beneficial homeo- levels have been reported. Hemochromatosis is a rare
static adaptation response to disease. cause of male hypogonadism where both T and go-
The physiological aging process is overlapped nadotropins are suppressed due to combined hypo-
by pathological mechanisms leading to aging-related thalamic and pituitary malfunction (). Men with
comorbidities, and many of them affect the HPT HIV have reduced serum total T concentrations, no
function more dramatically than does chronological change or increase in SHBG, and decreased free
aging (). Low serum T in acute and chronic disease T levels (, ). Findings on gonadotropins are
is fairly common and has a multifactorial cause, not uniform among studies, and apparently both
including a combination of acute weight loss, stress, hypogonadotropic and hypergonadotropic forms of
specific medications, and infections. The most com- hypogonadism occur in patients with HIV (,

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


mon and most important cause of aging-related –). Of note, in a long-term longitudinal study,
hypogonadism is overweight/obesity (), which although free T levels were lower, the rate of age-
predisposes to CV diseases (CVDs), diabetes, mus- related decline of free T was similar in men infected
culoskeletal disorders, and to some cancers. The re- with HIV compared with uninfected men ().
lationship between T levels and CVD is discussed in
“The CV system” below. Unlike the primary testicu- Mechanisms underlying age-associated changes
lar failure due to chronological aging, the obesity-
associated aging is often associated with secondary Leydig and Sertoli cell function
hypothalamic–pituitary failure with low T and sup- Although the mechanisms of the aging-related T
pressed or inappropriately normal gonadotropin levels decline are difficult to address in men, one animal
(see above). The metabolic syndrome (MetS), type  model in particular, the brown Norway rat, appears to
diabetes mellitus (TDM), and ischemic heart diseases display similar age-related primary decline of testicular
(IHDs) can be considered a continuum of the same function as in men (, ). In aged brown Norway
pathogenic mechanisms initiated by obesity-associated rats, Leydig cells produce reduced levels of T in re-
insulin resistance, dyslipidemias, and chronic in- sponse to maximal LH stimulation both in vitro and in
flammation of fat tissue, all connected to secondary vivo (, ), which appears to be due to reduced
hypogonadism. A similar decrease in T is found in steroidogenic capacity per Leydig cell rather than to
hepatic steatosis, which can be considered a hepatic reduced cell number (). T production decreases in
manifestation of the MetS (). In men with the more the face of increasing gonadotropin levels, indicating
advanced form of hepatic cirrhosis, androgen (T and primary testicular failure. In these animals, the
DHT) levels are decreased and estrogen levels are mechanisms of reduced steroidogenesis include a
increased (, ). The T levels keep falling as the decreased number of functional plasma membrane LH
disease advances (), and low T levels are also in- receptors and a decreased cAMP response to LH
dependently associated with nonalcoholic fatty liver stimulation, probably due to impaired LH receptor/G
disease (). The low T in liver cirrhosis can be caused protein coupling (). Another in vitro study on
by direct testicular injury evoked by alcohol, but more senescent Wistar rat Leydig cells confirmed these
frequently it is associated with central hypogonadism findings and, additionally, demonstrated an aging-
probably caused by inflammatory cytokines, with associated decrease in MAPK activation and expres-
subsequent secondary testicular failure (). The sion levels of several steroidogenic enzymes ().
hypogonadism is amplified by increased hepatic SHBG Reduced cholesterol import, synthesis, and transport
production and consequently lower free T. to mitochondria may ultimately be the main mech-
Gonadal dysfunction is common in men with anism, including reduced TSPO, StAR, and --
chronic kidney disease and with end-stage renal protein activities, behind the aging-related decline of
disease. T deficiency is usually of the primary type, Leydig cell steroidogenesis (, ).
accompanied by elevated serum gonadotropin con- Experimental studies, especially on the brown
centrations (). Low T levels in patients on he- Norway rat, have also elucidated the extrinsic factors
modialysis may predict increased mortality risk (, that may participate in the aging-related decline of
). Serum T concentrations are low in chronic Leydig cell function. They include hydrogen perox-
obstructive pulmonary disease, and the situation is ide produced by testicular macrophages () and
aggravated by glucocorticoid treatment (). Also, endocrine-disrupting chemicals, many of which affect
this situation seems to be of the primary type, because Leydig cell function in a similar fashion with aging
the patients have elevated gonadotropin levels (). (). In particular, increased oxidative stress is shared
The mechanism may be direct inhibitory effect of by both: an imbalance between pro-oxidants and
hypoxia on testicular function. In rheumatoid arthritis, antioxidants is typical of aging cells (). Aged Leydig

918 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

cells produce reactive oxygen species from several very similar responses were observed in a more
sources, including the mitochondrial electron trans- physiological setting where, after GnRH antagonist-
port chain and mitochondrial and microsomal cyto- suppressed gonadotropin secretion, elderly men re-
chrome P enzyme reactions (). T decreases with leased ~% less T than did young men in response to
aging, along with reduced Leydig cell antioxidant the same pulsatile LH clamp (). In response to
capacity (), which may be responsible for the recombinant LH challenge and sampling from the
decreased steroidogenesis. Experimental manipula- spermatic vein, the same authors observed a reduced T
tions support the cause-and-effects involvement of this response with aging and in obesity, as well as aug-
mechanism beyond correlations (). The specific mented Leydig cell downregulation (), with both
targets of an altered redox environment, as well as the supporting the decreased Leydig cell capacity to
mechanisms of how the pro-oxidant environment produce T. Young and elderly men exhibit similar LH
affects Leydig cell steroidogenesis, remain unclear. responses to GnRH stimulation (, ), and al-
The total number of Leydig cells in men between though age is associated with increased LH pulse
 to  and  to  years of age was found to be frequency and reduced pulse size (, ), relatively

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


reduced by % (), which explains the primary small changes occur in the T feedback inhibition of LH
hypogonadism associated with aging. Ultrastructur- secretion (). Hence, aging per se has only minor
ally, many Leydig cells of the aging testis preserve their effects on functions at the central level of the HPT axis.
normal appearance, whereas others acquire cytoplas- The situation is different with obesity (see below). The
mic or intranuclear Reinke crystals or paracrystal- altered LH secretory pattern was interpreted to in-
line inclusions, multiple vacuoles, lipofuscin granules, dicate reduced hypothalamic GnRH secretion, possi-
and lipid-laden cytoplasm, showing signs of de- bly due to the milieu of lowered sex steroids, and more
differentiation and involution (, ). There is an generally to aging-associated failure of the integrative
apparent correlation between the amount of deformed neurohormone outflow.
Leydig cells and the decrease of T production. The The sequence of events upon male aging is that the
decreased testicular perfusion is caused by athero- gradual loss of testicular capacity to produce T is
sclerotic alterations in testicular arterioles (). These initially compensated for by increased gonadotropin
changes may be the primary cause for the degenerating secretion, which is able to maintain normal T levels. A
“T decreases with aging along
changes in Leydig cells and their reduced steroidogenic situation termed “compensated hypogonadism” first
with reduced Leydig cell
capacity. arises, which is hallmarked by elevated gonadotropin antioxidant capacity, which
The number of Sertoli cells per testis decreases secretion in the face of unaltered T levels (). Men may be responsible for the
between  to  and  to  years of age by nearly with this condition are initially asymptomatic (), decreased steroidogenesis.”
% (), but the germ cell/Sertoli cell ratio did not but as testicular capacity to maintain normal T se-
change with age in this study. Ultrastructurally, aging cretion in response to elevated LH is gradually lost, the
Sertoli cells contain abundant lipid droplets and cy- men develop the typically age-related primary hypo-
toplasmic vacuoles with amorphous material, multiple gonadism (). With a prevalence of .% of men in
nuclei, mitochondria with tubular cristae, immature the EMAS population, compensated hypogonadism
nuclei, and sparse development of cytoplasmic or- appears to be a relatively common, but in many cases
ganelles (). Many of the signs are indicative of reversible, condition.
dedifferentiation. Another component in HPT suppression often
coincides with, but is independent of, chronologi-
Gonadotropin secretion and action cal aging, that is, secondary hypothalamic–pituitary
Gonadotropin secretion changes as the male ages. Part hypogonadism, characterized by low T and low or
of it is due to chronological aging, and partly it is due inappropriately normal gonadotropin levels. It is more
to aging-associated comorbidities, of which weight common, encompassing about % of the hypo-
gain is most important. gonadism of aging men in the EMAS, and it is as-
According to the EMAS, primary gonadal failure sociated with overweight and obesity, but not with
represents the pure form of aging-related hypo- chronological age (, ). As described above, other
gonadism (, ). A failure of testicular T pro- common causes for HPT suppression are aging-
duction ensues, partly due to an aging-associated associated chronic illnesses, for example, diabetes
decrease of Leydig cell number and the capacity to mellitus, the MetS, and IHD, where both the central
respond to LH and to produce steroids (see above). and testicular functions are variably affected. In many
Older studies showed decreased testicular T responses cases, both the central and testicular impairment
to human chorionic gonadotropin (hCG) stimulation overlap.
upon aging (, ), although they may not mimic What still has not been thoroughly addressed is
the physiological situation because of the long half-life how the function of kisspeptin in the HPT regulation
of hCG compared with LH, as well as the conse- responds to aging and associated comorbidities. An
quential desensitization of Leydig cell steroidogenesis immunohistochemical study on human hypothalamus
in response to supraphysiological stimulation. However, detected an aging-related robust enhancement in

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 919


REVIEW

central kisspeptin signaling and a moderate en- elucidate further kisspeptin function in aging and
hancement in central neurokinin B signaling (). obesity in aging men.
Such changes are compatible with the known reduced In conclusion, it seems that the primary hypo-
T negative feedback to kisspeptin neurons and may gonadism specifically associated with aging is caused
participate in the elevated LH levels in the primary by impaired Leydig cell function, with reduced gonadal
hypogonadism of aging men. Similar studies on feedback and consequent increase in LH secretion,
obesity do not exist in humans, but rodent studies have whereas the hypothalamic–pituitary function remains
demonstrated an expected obesity-associated sup- largely unaffected. It leads initially to a state of
pression of Kiss expression in the hypothalamus and compensated hypogonadism (), where T remains
impaired LH response to kisspeptin (). Recent normal but LH is inappropriately elevated. When the
studies in rats (), but not in monkeys (), support reserve stimulated testicular capacity is exhausted, the
that kisspeptin neuronal activity is reduced in aging outcome is primary hypogonadism (low T, high LH).
males. It is likely that in the near future research will Obesity and several other comorbidities cause pre-
dominantly secondary hypogonadism where multi-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


ple mechanisms impair the hypothalamic–pituitary
function.

SHBG levels
SHBG levels increase upon aging, but this is coun-
teracted by the concomitant decrease evoked by
obesity. Because SHBG is produced by the liver, it is
not inconsequential that the two opposite responses
seen upon aging and weight gain somehow reflect
changes in the metabolic state of this organ.
The reasons why SHBG levels increase in men
upon aging are not clearly understood. One reason
could be the aging-associated increase in levels of the
anti-inflammatory cytokine adiponectin (, ),
which has been shown to stimulate SHBG production
in cultured human liver cells by activating AMP-
activated protein kinase, reducing lipid content, and
activating hepatocyte nuclear factor (HNF)-a ()
(Fig. ). Lewerin et al. () observed that the aging-
related increase in adiponectin levels was inversely
associated with Hb. We could therefore speculate that
as adiponectin stimulates SHBG levels it indirectly
suppresses free T, which could then be responsible for
the suppressed Hb, a sign of hypogonadism. There
could even exist an SHBG–adiponectin–T regulatory
link, because men with hypogonadism have a high
adiponectin level that is suppressed by T treat-
ment (). Also, a decreased inhibitory effect of the
aging-related decrease of IGF- action as the cause
of increased SHBG with aging has been proposed
(–), and there are also in vitro data on cultured
liver cells indicating that IGF- can be a direct negative
regulator of SHBG synthesis ().
Recent research has emphasized that the obesity-
associated SHBG decrease is related to monosaccha-
ride (glucose and fructose)-induced liver lipogenesis
and fat content rather than to the body’s total and
Figure 3. Molecular mechanisms involved in obesity-associated decrease of hepatic SHBG visceral fat (, ) (Fig. ). SHBG may even
synthesis. Cytokines produced by fat tissue act as negative (TNFa, IL-1b) or positive (adiponectin) be a primary determinant of the MetS and TDM,
regulators of SHBG expression by interacting with various signal cascades resulting in alterations of
because a Mendelian randomization analysis has
HNF-4a levels that stimulate SHBG expression. Dietary factors such as carbohydrates or olive oil are
also able to regulate SHBG expression by affecting HNF-4a or PPARg levels, respectively. Thyroid
shown that a genetic variant of SHBG that affects the
hormone levels can also regulate SHBG production by affecting HNF-4a expression. [Reproduced levels of this protein reciprocally influences the risk of
with permission from Simó R, Saez-Lopez C, Barbosa-Desongles A, et al. Novel insights in SHBG these diseases (, ). Obesity is associated with
regulation and clinical implications. Trends Endocrinol Metab 2015;26(7):376–383.] increased production of proinflammatory cytokines by

920 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

fat tissue and with low SHBG (). In vivo and in vitro in particular fructose, inhibit SHBG synthesis in
studies have demonstrated that TNFa reduces SHBG cultured hepatic cells by increasing de novo lipogenesis
production of HepG cells indirectly through nuclear (). An opposite effect was found with the olive oil
factor kB–mediated downregulation of HNF-a component oleate, which increases SHBG levels by
(). A similar effect has been demonstrated for IL- downregulating PPARg ().
b (). An opposite relationship with obesity is
found with the anti-inflammatory cytokine adipo- Testosterone metabolism
nectin (, ), which stimulates SHBG production Some older studies have compared endogenous ste-
in HepG cells (). The long-held concept that roid profiles in testis tissue of young and elderly men as
insulin negatively and directly regulates hepatic SHBG potential signs of aging-related alterations in androgen
production through an unknown mechanism does not metabolism. In one study, the concentrations of nine
hold true in light of more recent research [reviewed in endogenous steroids were measured in the testis tissue
(, )]. A recent study showed that low SHBG is of men aged  to  years (oligozoospermia and
correlated with high leptin and low adiponectin levels, varicocele) and  to  years (prostatic carcinoma)

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


irrespective of T levels (). (Table ) (). The sum concentration of the steroids
The twofold higher level of SHBG in women than was significantly higher (.%) in the young men
men () occurs apparently because of estrogen- than in the elderly men, whose elevated LH levels
induced downregulation of hepatic peroxisome revealed a primary testicular defect. Because the
proliferator-activated receptor g (PPARg) expression conversion of [H]pregnenolone to downstream
(). Whether there is an additional sex-specific metabolites was similar in both age groups, it was
reason for the lower SHBG levels in men remains concluded that aging-related impairment in the mi-
largely unknown. The human SHBG promoter does tochondrial pregnenolone production, rather than in
not have androgen response elements. One explana- the subsequent microsomal steps of steroid meta-
tion could be the lower adiponectin level in male bolism, was the cause of reduced steroidogenesis.
circulation (). However, there are also data showing that the activity
Several components in the diet appear to have direct of microsomal steroidogenic enzymes is decreased in
regulatory effects on SHBG synthesis. Monosaccharides, aging testes (, ). Furthermore, decreased levels

Table 3. Concentrations of Endogenous Steroids Detected in the Testis Tissue (in nmol/L) of Young and Old Men in Two Studies
Study Takahashi et al. Vermeulen and Deslypere

Age groups, y 25–35 61–85 ,50 .65

Steroids

Pregnenolone 590 6 64 230 6 29a 793 6 93 532 6 70b

17-Hydroxypregnenolone 110 6 12 49 6 5.4a 41.6 6 5.4 23.2 6 3.5b

Progesterone 81 6 6.7 66 6 7.4 30.5 6 29 40.7 6 5.1b

17-Hydroxyprogesterone 54 6 5.3 30 6 4.7b 122 6 18 122 6 1.6

Dehydroepiandrosterone 290 6 23 220 6 31 10.8 6 0.9 8.5 6 1.0

5-Androstene-3b,17b-diol 270 6 30 140 6 20 c


59.3 6 7.0 37.6 6 5.8b

Androstenedione 99 6 14 72 6 12 16.5 6 2.0 17.0 6 4.1

Testosterone 1700 6 194 860 6 119 c


451 6 57 447 6 63

5a-Dihydrotestosterone 20.4 6 5.1 16.9 6 1.6

5a-Androstane-3a,17b-diol 20.0 6 4.2 18.5 6 2.2

Estradiol 19 6 1.9 9 6 1.6a 5.1 6 0.62 4.91 6 0.58

Total 3680 6 340 1940 6 210 c


1596 6 134 1267 6 128b

[Data from Takahashi J, Higashi Y, LaNasa JA, et al. Studies of the human testis. XVIII. Simultaneous measurement of nine intratesticular steroids: evidence for reduced mitochondrial
function in testis of elderly men. J Clin Endocrinol Metab 1983;56(6):1178–1187; and from Vermeulen A, Deslypere JP. Intratesticular unconjugated steroids in elderly men. J Steroid
Biochem 1986;24(5):1079–1083.]
aP , 0.001, vs young men.
bP , 0.05, vs young men.
cP , 0.01, vs young men.

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 921


REVIEW

of D, but not D, steroids have been found, including effects of T deficiency can be fully prevented by
increased levels of progesterone and -hydrox- treatment with an estrogenic compound acting on
yprogesterone, in the aging testis (, ) (Table ). ERa (, ). A predominant role of estrogens
Bélanger et al. () measured circulating con- rather than androgens in the maintenance of skeletal
centrations of a number of adrenal (see above) and integrity in aging men is further indicated by the lack
testicular steroids and their conjugates in men in the of an effect on bone metabolism of antiandrogen
age range of  to  years. Apart from the clear monotherapy in patients with prostate cancer (,
decline of adrenal steroids and their metabolites, ). Treatment of aging men with a a-reductase
several age-related changes of testicular steroids were inhibitor, which inhibits the conversion of T to the
observed. The decline of T (.% per year) was par- more potent androgen DHT, appears also not to affect
alleled by a similar decrease (.% per year) of another bone metabolism (–).
testis-derived steroid, -hydroxyprogesterone. Unlike Findings from observational studies also concur
the marked decline in the serum concentrations of in indicating that E is important for maintaining
adrenal or testicular C steroids DHEA (.% per skeletal integrity in aging men. Cross-sectional

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


year), -diol (.% per year), DHEAS (.% per year), studies have consistently shown a direct associa-
-diol-sulfate (.% per year), T (.% per year), and tion of serum E and in particular its non–SHBG-
androstenedione (.% per year), serum DHT con- bound fractions with the areal BMD (aBMD) at
centration did not change, which could mean that different skeletal sites and, less extensively docu-
extratesticular a-reductase activity increases with age. mented, an inverse association with biochemical
The fragmentary information available, often from markers of bone resorption [() and reviewed in
small studies with heterogeneous samples, suggests (, )]. Lower (non–SHBG-bound) E levels have
that aging reduces the activity of all steroidogenic also been associated with less favorable volumetric
enzymes with no specific aging-sensitive steps. Animal BMD (vBMD), bone geometric variables, trabecular
experiments have demonstrated impaired LH receptor structure, and cortical porosity (–). Longi-
function, possibly due to the accumulating load of free tudinal studies have shown inverse association of
oxygen radicals with age, as an important reason for (non–SHBG-bound) E with prospectively assessed
the impairment of Leydig cell steroidogenesis (). bone loss (–). Differences in prospectively
There is in vivo evidence in humans that impaired assessed BMD changes in older men have also been
responsiveness of aged Leydig cells to LH stimulation associated with a polymorphism of the CYP gene
could be the primary cause for their weakened ste- encoding the aromatase enzyme (, ). Although
roidogenesis (, ). The ultimate reason for the not seen in all studies, there are indications for a
aging-related primary impairment of testicular func- threshold for serum (non–SHBG-bound) E above
tion could be decreased perfusion and oxygen supply. which bone mass in aging men is better preserved
(). This threshold may lie at ~ to  pg/mL ( to
 pmol/L) bioavailable E or ~ to  pg/mL ( to
Clinical Correlates of Sex Steroid Levels in  pmol/L) total E (, ), which is higher than
Elderly Men the  pg/mL ( pmol/L) that may be sufficient to
maintain bone mass in younger men as derived from
Bone metabolism and skeletal integrity the results of an experimental intervention study
Sex steroids are major players in the regulation of bone ().
homeostasis and the preservation of skeletal integrity As to the relationship of serum T and its non–
in adult men. Their levels were reported to be asso- SHBG-bound fractions with bone metabolism,
ciated with several aspects of bone health in aging men, findings from observational studies in community-
including bone mineral density (BMD), bone loss, and dwelling aging men have been inconsistent and fail to
fracture risk. Direct sex steroid effects on bone appear convincingly document an association independent
to be exerted predominantly by E, the major aro- of E with either prevalent BMD, bone turnover
matization product of T, via estrogen receptor (ER)a. markers, or BMD changes [() and reviewed in
An androgenic action of T and its a-reduced me- (, )]. Interestingly, a number of observational
tabolite DHT in the adult male skeleton via the AR is studies in aging men have shown association of se-
exerted both directly on bone tissue and mainly in- rum SHBG levels, independent of serum T and E,
directly through effects on extraskeletal tissues, in with prevalent BMD, levels of bone turnover markers,
particular muscle [reviewed in ()]. and BMD changes (, –). In the MrOS US
study, the highest rate of bone loss was observed in the
Quantitative and structural skeletal changes men presenting the combined findings of the lowest
Acquired profound hypogonadism in aging men is serum bioavailable T, lowest bioavailable E, and
associated with high bone turnover bone loss as is highest levels of SHBG (). In this study there was a
illustrated by the effects of ADT in patients with threshold effect for serum SHBG at ~ nmol/L,
prostate cancer (, ). These adverse skeletal above which faster bone loss was observed.

922 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

Fracture risk BMD changes or incident fractures did provide evi-


In men with ADT for prostate cancer, a model for dence for interaction between T and E with an ad-
acquired profound hypogonadism, the high bone ditive effect on fracture risk of low T in the presence of
turnover bone loss translates into increased fracture low E (, ) or for interaction between low
risk (). In community-dwelling aging men, in line bioavailable T, low bioavailable E, and high SHBG
with the well-documented role of E in restraining (). Serum T might influence fracture risk through
bone turnover and preserving BMD, and notwith- extraskeletal androgenic actions such as on muscle
standing some discordant findings (–), there is mass and function. In the MrOS US and Sweden
substantial evidence linking low serum (non–SHBG- cohorts, low T and bioavailable T were indeed asso-
bound) E with increased fracture risk as reviewed ciated with an increased risk of falls (, ).
by Vanderschueren et al. (). Low serum E has In several cohort studies (, , , ), but
been associated with increased hip fracture risk in not in all (, ), high serum SHBG was associated
aging men from the Framingham Study (), and independently of T and E levels with increased risk of
both low E and calculated free E were associated incident fractures and in particular also risk of in-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


with increased risk of clinical fractures in men from cidental vertebral fractures (, ). In the MrOS US
the EPIC Study (). In the MrOS Sweden cohort, cohort an interaction was observed between SHBG
calculated low free E was independently associated levels and bioavailable T, with substantial fracture risk
with clinical vertebral, nonvertebral, and hip fracture in men combining low bioavailable T with high SHBG,
risk (). In the MrOS US cohort, the lowest quartile whereas the highest fracture risk was in men with high
of calculated bioavailable E (,. pg/mL or  SHBG, low bioavailable T, and low bioavailable E
pmol/L) was associated with a higher risk of non- (). The pathophysiological basis for the association
vertebral fractures (). Also, in the MrOS Hong of serum SHBG with both maintenance of bone mass
Kong cohort () low E and calculated bioavailable and fracture risk remains to be established but may
E were associated with increased risk of all fractures, involve (differential) modulation of T, DHT and E
with the serum levels in the lowest quartiles being action. The association may also result from residual
associated with the highest risk. In these studies confounding, for example, as a reflection of low ac-
the associations between (non–SHBG-bound) E tivity of the somatotropic axis. Finally, the possibility
“Studies assessing…incident
and fracture risk were attenuated when adjusted for of some yet-to-be-uncovered physiologically relevant
IHD or myocardial infarction
BMD, but they remained mostly significant. This intrinsic action of SHBG or a SHBG–sex steroid failed overall to show an
suggests that skeletal factors not captured by aBMD complex cannot be excluded. Also note that in- independent association with
assessed by dual-energy X-ray absorptiometry (DXA), dependent associations between serum SHBG and baseline (free) T.”
such as bone geometry or cortical porosity (), or, bone phenotypes have also been observed in young
hypothetically, some extraskeletal factors contribute to men ().
the inverse association of serum E with fracture risk. In summary, E is the main sex steroid needed for
In most studies the association between E levels preservation of skeletal integrity in aging men. Low E
and fracture risk was nonlinear, with a suggestion of is associated with lower BMD, higher bone turnover,
markedly increased fracture risk when serum levels greater bone loss, qualitative bone changes, and in-
were below a threshold situated around  pg/mL ( creased risk of mainly nonvertebral fractures. Also,
pmol/L) for total E. This threshold is somewhat lower high SHBG, independently of sex steroid levels, is
than that reported as critical for accelerated bone loss associated with greater bone loss and increased frac-
(). Finally, note that the evidence for association of ture risk of both nonvertebral and vertebral fractures.
low E with fracture risk pertains almost exclusively to The androgenic actions of T appear to play a more
clinical and nonvertebral fractures. Two recent studies marginal role in skeletal health and fracture risk, and
assessed vertebral fractures systematically by serial this most likely mainly through indirect extraskeletal
radiographs in the MrOS US cohort () and in a effects. The associations of bone health variables with
combined analysis of men from the MrOS Sweden and both E and SHBG are nonlinear, with a suggestion
MrOS Hong Kong cohorts (), respectively. In of a low and high serum level threshold, respectively.
neither of these two studies was the risk of radio- Note, however, that many of the reported associations
graphically assessed vertebral fractures related to se- with E and SHBG in community-dwelling men ex-
rum E levels. plain only a limited fraction of the variability in BMD
In most studies no independent association was changes and fracture risk, as these men in general are
found between (non–SHBG-bound) T and risk of not markedly hypogonadal. Recently, in an analysis of
incident fractures (, –). In contrast to these the MrOS cohorts, Orwoll et al. () found that in
consistently negative findings, independent association elderly men (aged  to  years at baseline; around
of low serum T was reported for the Dubbo cohort, -year follow-up) measurement of E, SHBG, and T
even though BMD was related to serum E and not to did not contribute to prediction of BMD changes or
T (). Interestingly, findings of several studies not meaningfully to fracture risk assessment in a model
showing independent associations of T with either including clinical risk factors with BMD (FRAX ®

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 923


REVIEW

algorithm). From this analysis, the authors concluded ). Conversely, weight loss in men with obesity is
that there is limited clinical utility of serum E, T, and associated with rising total T, SHBG, and, if the weight
SHBG measurements for the evaluation of osteopo- loss is extensive, free T levels (). This reciprocity
rosis risk in elderly men. was addressed in a pooled cohort of studies [MrOS
Sweden, GOOD, Study of Health in Pomerania
Body composition, metabolic health, and (SHIP), SHIP Trend, and Inter] using a bidirectional
physical performance Mendelian randomization approach, which is sup-
Aging in men is associated with profound changes in posed to be free from residual confounding. Herein,
body composition. Starting from mid-adult age, the genetic risk score on BMI was inversely associated
healthy men on average gain fat and lose lean mass with serum total T levels (independent of the asso-
(–), which in part leads to concomitant declines ciations with SHBG levels), whereas no association was
in metabolic health and physical performance (, found between the genetic risk score on total T levels
, ). This is of importance, as resulting clinical and BMI (). In addition to androgen exposure,
entities such as the MetS and TDM, but also sar- there is some evidence suggesting a contribution of

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


copenia and frailty, are associated with quality of life, estrogen exposure to the regulation of fat mass and
morbidity, and mortality (–). As discussed metabolic health. For example, in men who are aro-
previously (), part of these changes in body com- matase deficient, the metabolic profile improves
position in aging men appear mediated by the decline during estrogen treatment (, ), and to prevent
in androgen exposure. However, these relationships fat mass gain in men with hypogonadism by T
are confounded by concurrent changes in lifestyle, treatment, minimal E exposure by aromatization of T
illness, and other hormonal axes. Moreover, especially seems required (). However, reports on the re-
for adiposity, this relationship is bidirectional in nature lationship between E levels and adiposity in aging
and modulated by SHBG. In the following sections, we men are equivocal. Although some cross-sectional
discuss recent publications addressing these questions studies reported positive associations between (free)
in population-based cohorts of aging men. E levels and indices of adiposity (, –, ),
this was not confirmed in other studies (, , ,
Adipose tissue and metabolic health , ), and longitudinal analyses from MrOS and
Adipose tissue. Men with profound hypo- EMAS did not find associations between E levels and
gonadism gain fat mass and are prone to develop changes in fat mass (, ).
related metabolic consequences such as the MetS or Metabolic health. The relationship between
TDM (). Correspondingly, most cross-sectional serum sex steroid levels and the MetS, insulin re-
studies in elderly men found inverse associations sistance, or TDM was addressed in many cross-
between T levels and indices of adiposity (, , , sectional studies in middle-aged and older men
, , –). However, many of these findings (, , , , , –). Given the in-
are probably affected by residual confounding or terrelationships between sex steroids, body composi-
obscured by the effects of adiposity to decrease SHBG tion, and metabolic health we discuss herein only
levels. For instance, in the EMAS, adiposity indices prospective studies. In the Rancho-Bernardo study,
were higher in all men with low total T (whether free T men with lower baseline total T levels were at higher
was low or normal) but not in men with low free T and risk to develop insulin resistance or TDM during
normal total T levels (). As total T levels are largely follow-up, whereas this was unrelated to bioavailable
determined by the SHBG concentration, solely relying T, total E, or bioavailable E levels (). Similar
on total T to define hypogonadism will thus lead to an findings were reported from a Finnish study (), the
overestimation of hypogonadism prevalence in sub- Massachusetts Male Aging Study (MMAS) (, ),
jects with obesity (, ). Longitudinal studies are the Baltimore Longitudinal Study of Aging (), and
also less convincing; for example, in the MrOS US the Danish Inter study (), as well as from an
cohort, neither total T, bioavailable T, nor SHBG levels individual participant data meta-analysis of  ob-
were associated with changes in fat mass during . servational studies () wherein men with low total T
years of follow-up (). Similarly, after adjusting for or SHBG levels at baseline had a higher incidence of
confounders, neither baseline total T, free T, nor the MetS or TDM, without finding associations with
SHBG levels were associated with changes in waist estimates of non–SHBG-bound T. In the Concord
circumference in the EMAS cohort (). Moreover, a Health and Ageing in Men Project (CHAMP) study,
possible relationship is most probably reciprocal, as moreover, only low baseline SHBG was associated
longitudinal studies found that declines in total T and with incident MetS after multivariate adjustment
free T are most pronounced in men remaining obese (). In the EMAS, men with lower total T, free T, or
or gaining weight with aging (in part mediated SHBG levels or a higher E/T ratio at baseline had an
through changes in SHBG levels) (, , , , increased risk of developing the MetS after . years
) and that larger declines in total T and free T levels (). However, in a secondary analysis categorizing
were associated with higher fat mass at follow-up (, participants according to total T and free T status, men

924 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

with low free T but normal total T levels had no might seem surprising, but they are corroborated by
increased risk as compared with men with normal total findings from the MrOS Hong Kong (), MrOS
T and free T levels. Moreover, even the higher in- Sweden (), and BACH/Bone () studies. In
cidence of the MetS in men with total T , ng/dL longitudinal analyses from the MrOS US study, men
(,. nmol/L) (whether free T was low or normal) having higher baseline total T or bioavailable T (but
was no longer significant after adjusting for baseline also SHBG) levels presented with less decline in ap-
confounders (). In several other studies in elderly pendicular lean mass. This finding, however, only
men, despite cross-sectional associations between sex applied to men who lost weight during follow-up, and
steroid levels and prevalent MetS and TDM, only low no associations between E levels and changes in lean
SHBG was predictive of incident MetS (, , ) mass were found (). In the MINOS study, no
and only total T in the SHIP () and estrone in the relationship between baseline total T or free T levels
Framingham Heart Study () was predictive of and loss of lean mass was found, although a more
incident TDM. Furthermore, in a Norwegian study rapid loss of lean mass was observed in men with total
(), associations between baseline total T and SHBG T or free T levels below the normal range for young

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


levels and incident TDM were no longer significant men (), whereas in the CHAMP study, -year
after adjusting for waist circumference, but they did longitudinal changes in total T, free T, DHT, SHBG,
find an independent association between total E and estrone (but not E) were all associated with
levels and incident TDM. Similar findings were re- contemporary changes in appendicular lean mass,
ported in the high-risk population from the Diabetes even after multivariate adjustment (). Finally,
Prevention Program (). However, neither of these changes in total T or free T levels were not associated
two studies corrected for baseline SHBG levels, and no with lean mass at the second study visit in a British
estimates of non–SHBG-bound E were evaluated. study ().
Only in a smaller Finnish cohort (using immunoas- Again, despite the established anabolic effects of
says) was incident TDM associated with low baseline androgens in hypogonadal men (, ), the con-
total T and free T but not SHBG levels (). tribution of androgen exposure to changes in lean
Taken together, despite strong and consistent in- mass in population-based cohorts of aging men seems
verse associations between T and SHBG levels and rather limited, and it cannot be excluded that lower T “…the role for moderate
indices of adiposity and metabolic health in cross- levels are a marker of mutual confounding factors. decline in androgen exposure
sectional studies, evidence from longitudinal studies is Additionally, and perhaps somewhat unexpectedly, in the increase of adiposity and
less convincing. If any, the role for moderate decline in most studies did not show stronger associations be- worsening of metabolic health
androgen exposure in the increase of adiposity and tween lean mass and estimates of non–SHBG-bound during normal aging is
probably limited.”
worsening of metabolic health during normal aging is T than with total T levels. Furthermore, although there
probably limited. Moreover, it cannot be excluded were limited reports on relationships with E and
that these results still are affected by residual con- DHT levels, and myocytes are known to express ERa,
founding and that low T and SHBG levels are more a ERb, and a-reductase, these findings are rather
risk marker rather than a true risk factor for these unexpected and do not concur with results from
changes. experimental studies (, ).
Physical performance. In addition to regula-
Lean mass and physical performance tion of lean mass, androgen exposure has been linked
Lean mass. From observations in hypogonadal to different measures of physical performance. For
men () and users of anabolic androgens (), a instance, several studies found total T, bioavailable T,
relationship between the aging-related declines in or free T levels to be positively associated with muscle
androgen exposure and lean mass could be expected. strength (, , ), and in MrOS (United States
Although positive associations between (appendicular) and composite cohorts), men with higher bioavailable
lean mass and T levels were reported in the MINOS T levels had slightly better muscle strength and
(), the BACH/Bone (), and the MrOS Hong physical performance (, , ). Similarly, in the
Kong cohorts (), other cross-sectional studies did Framingham Offspring Study cohort, free T (but not
not confirm this (, , , ). For instance, in total T or SHBG) levels were positively associated with
the MrOS US and Sweden cohorts, lean mass index physical performance battery score, walking speed, and
and appendicular lean mass did not differ according to grip strength (). In contrast, in the MMAS (),
bioavailable T or free T levels (, , ), whereas despite associations between total T, bioavailable T,
in another cohort of community-dwelling elderly men, and DHEA/DHEAS levels and the physical perfor-
the association between total T or free T and lean mass mance score, neither of these androgens was associated
indices was only revealed after adjusting for SHBG with grip strength and only DHEA levels below a
and/or total body weight (). In the CHAMP study, certain threshold were associated with chair stand
however, total T, free T, total E, and estrone levels score, whereas in the BACH/Bone study, associations
were all positively associated with appendicular lean of total T levels with physical performance were only
mass (). These associations with estrogen levels weak or nonsignificant (). In longitudinal analyses

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 925


REVIEW

from the MrOS US study, higher total T and bio- performance such as sarcopenia or frailty than to
available T (but not E or SHBG) levels were asso- direct muscle strength measurements. So, despite the
ciated with a lesser decrease in one measure of physical anabolic effects of T on muscle mass in men with
performance (timed chair stands) only in participants hypogonadism, the decline in androgen exposure in
who lost weight during follow-up, a finding that was aging men does not seem to fully account for their
partly explained by changes in lean mass (). In decreases in muscle strength and physical perfor-
contrast, in the Longitudinal Aging Study Amsterdam mance, and both processes are more likely to be related
and Health ABC cohorts, neither total T nor free T to the underlying physiologic processes of aging per se.
levels were associated with declines in physical per- Additionally, although most studies adjusted for
formance or muscle strength (), and although possible baseline confounders such as age, BMI, and
baseline free T levels in the Framingham Offspring smoking and comorbidities such as CV or renal
Study cohort were associated with lower risk of de- disease, most statistical models only explain part of the
veloping or worsening of mobility limitation, they observed variance and probably suffer from residual
were not associated with changes in grip strength or confounding.

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


walking speed, nor were there any relationships be- To summarize, changes in body composition in
tween E levels and changes in physical performance aging men are associated with important clinical en-
(). In the CHAMP study, self-reported functional tities and are thus of importance. Although a re-
decline was no longer associated with total T or free T lationship between androgen exposure and changes in
after adjusting for baseline muscle mass or strength body composition is suggested from many population-
(). This same study, however, recently reported based studies in elderly men, the absolute direct
associations between changes in total T, free T, and contribution in men during normal aging seems rather
DHT with changes in muscle strength during a -year limited, and many questions are still to be resolved.
period, independent from possible confounders, al- Issues to be addressed are, for instance, the modulating
though the authors did not adjust for baseline sex role of SHBG, as several studies reported associations
steroid levels or baseline muscle strength (). between SHBG levels and (changes in) indices of
With respect to clinical entities such as sarcopenia adiposity (, , , , ), muscle mass (,
and frailty, the available literature is difficult to ) and strength (), and physical performance
compare owing to the use of different operating (), but also the role of intratissular metabolization
definitions. In cross-sectional analyses, men from the toward DHT, E, and other metabolites merits fur-
MINOS study with sarcopenia had lower total T and ther research. Finally, as mentioned earlier, the above-
free T levels (), whereas in Japanese men only free mentioned relationships are also modulated by
T was associated with prevalent sarcopenia (). In changes in behavior, general health, and other hor-
the MMAS (), only SHBG (and not total T or free monal axes (, , , , –). Although
T) levels remained associated with prevalent frailty most of the cited studies tried to account for this by
after adjustment for possible confounders, whereas in adjusting for comorbidities and/or medication use,
the MrOS US cohort, this was only so for bioavailable the possibility of residual confounding cannot be
T (and not total T, E, or SHBG) levels (). Lon- excluded.
gitudinal analyses from the MrOS US study found that
men with low baseline bioavailable T levels had higher Erythropoiesis
odds to have a more advanced frailty status at -year Anemia is common in frank hypogonadism (), and
follow-up (). Furthermore, although both total T exogenous T administration is known for its stimu-
and free T levels were related to frailty status in the latory effects on erythropoiesis [reviewed in ()].
Health in Men study, only baseline free T levels were Although a considerable proportion of the elderly
predictive of frailty status after  to  years of follow-up present with unexplained anemia (–), it is
(). In the CHAMP study, despite associations unclear whether and to what extent the aging-related
between baseline total T, free T, DHT, total E, and decline in androgen exposure in men contributes to
estron levels and the number of frailty components, this phenomenon. In two specific studies in men with
-year changes in total T and free T, but not baseline TDM, anemia was more prevalent in those men with
levels, were associated with a higher risk to become low, as compared with those with normal, total or free
prefrail or frail according to one (Cardiovascular T levels (, ). Population-based data on this
Health Study) but not another (Study of Osteoporotic association are still rather scarce, although recently
Fractures) frailty index (). Finally, a recent analysis some evidence emerged supporting a relationship
from EMAS follow-up data showed that after ad- between low T exposure and a decline in erythro-
justment for BMI, only free T (and not total T or poiesis in aging men. In the InCHIANTI study, a
DHT) was related to a lesser worsening in the frailty positive association between total T, bioavailable T,
index (). and Hb levels was found, and men in the lowest
From these studies, androgen levels seem more quartile of bioavailable T levels were at higher odds to
related to (changes in) composite entities of physical be anemic. Moreover, among nonanemic participants,

926 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

those with both low total or bioavailable T levels at on erythroid progenitor cells (, ), or suggested
baseline were at higher risk to be anemic at the -year effects on iron bioavailability (, , ). However,
follow-up visit, independently from some common negative findings on the mechanisms underlying T
baseline confounders (). Similarly, a cross-sectional effects on erythropoiesis have been published as well
analysis of National Health and Nutrition Examina- (, ). Furthermore, as suggested in some studies
tion Survey data on adult men $ years of age but not in others, and only supported by circumstantial
showed lower hematocrit (Hct) in the tertile of men evidence, part of the effects of T on erythropoiesis might
with the lowest free T levels (). Although this be mediated through conversion into E (, , ,
finding persisted when limiting the study sample to ) or DHT (, ). Finally, contributing or
men $ years of age, there was insufficient power to modulating effects of other hormones, lifestyle, con-
account for residual confounding from concomitant comitant illnesses, and treatments might play a role
illnesses or treatments. This was recently more ac- both in aging-associated as well as hypogonadism-
curately addressed in the EMAS cohort, wherein it was associated anemia. For instance, a study in men un-
found that the proportion of elderly men diagnosed dergoing ADT for localized prostate cancer suggested

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


with moderate or severe late onset hypogonadism a contribution of impaired IGF- signaling in the ob-
(LOH; with low total T, low free T, and the presence of served decline in red blood cells (), and in the MrOS
three sexual symptoms) present with a . to  g/L Sweden study, smoking was associated with Hb
lower Hb level as compared with eugonadal men, level ().
differences that persisted after correcting for general
health status and comorbidities (). Also in the The CV system
EMAS, although unadjusted analyses suggested that Low T or diagnosis of LOH have been linked to a
Hb levels were lower only in those men with low free variety of risk factors for atherosclerosis and CVD,
T, multivariate correcting for age, BMI, and prevalent such as obesity, the MetS, TDM, increased blood
comorbidities also revealed lower Hb in men with low pressure, dyslipidemia, or endothelial dysfunction (,
total T but normal free T levels (). Furthermore, in , ). However, observed associations between
the Busselton Health Survey, total T, free T, DHT, and low serum T and a less favorable CV risk profile might
total E levels were all positively correlated with Hb in many instances reflect either reverse causality, with
“Overall, …data suggest that
level (), whereas in the MrOS Sweden cohort, total low T being the consequence rather than the cause of
hypogonadism in elderly men
E but not total T levels were independently associated the unfavorable clinical profile, or a bidirectional in- is an independent risk factor
with Hb levels, and total E levels were lower in men terrelationship between low T and CV risk factors. A for development of a lower Hb
with anemia (). However, in the latter study no number of cross-sectional studies looked at associa- level and anemia.”
estimates of the non–SHBG-bound sex steroid frac- tions between prevalent CVD () or preclinical
tions were evaluated, and the analyses were not ad- markers of atherosclerosis such as carotid artery
justed for SHBG levels. Prospective data from the intima–media thickness (IMT) (–), coronary
EMAS revealed that both men with persisting or in- artery and aortic calcifications (, ) or abdominal
cident primary hypogonadism presented with lower aorta aneurysm (), and endogenous sex steroid
Hb levels at .-year follow-up as compared with men levels. They reported mixed and overall inconclusive
who remained eugonadal (). Again, this finding findings of either an inverse relationship with serum
persisted after adjusting for age and chronic illnesses (free) T or no association independent of known CV
although the statistical power of these models was low risk factors. Moreover, positive reports carry a high
given the overall low incidence of primary hypo- risk of residual confounding, whereas some of the
gonadism in this cohort. Finally, longitudinal analyses negative studies suffer from low power.
from the CHAMP study showed that changes in Older observational studies that evaluated the
total T, free T, DHT, SHBG, E, and estron levels relationship between endogenous T levels and pro-
were all related to contemporary changes in Hb level, gression of atherosclerosis as assessed by a variety of
independent of baseline age, BMI, smoking, and parameters such as progression of carotid artery IMT,
comorbidities (). Overall, these data suggest that peripheral vascular disease, arterial stiffness, or pro-
hypogonadism in elderly men is an independent risk gression of abdominal aorta or carotid artery ath-
factor for development of a lower Hb level and anemia. erosclerosis showed either no association or only a
However, the origin of anemia in the elderly is mul- weak association of low T with progression of ath-
tifactorial (), and the decrease in androgen exposure erosclerosis, and they did not reveal a significant trend
probably only accounts for a minor part of the overall when meta-analyzed [reviewed in ()]. A more
decline in erythropoiesis in aging men. recent study from the German Study of Health in
Biomolecular mechanisms by which T stimulates Pomarania also reported negative findings for asso-
erythropoiesis remain incompletely resolved. Stimu- ciation of serum T with progression of parameters of
lation of erythropoietin production and/or sensitivity subclinical atherosclerosis ().
has been suggested by some researchers (–), As to prospective observational studies assessing
whereas others demonstrated direct effects of androgens CV events, those that focused specifically on incident

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 927


REVIEW

IHD or myocardial infarction (MI) failed overall to reporting associations of incident events with total
show an independent association with baseline (free) and/or free or bioavailable T, some reporting no as-
T. Indeed, mostly negative findings from earlier studies sociation with (free/bioavailable) T, and some reporting
[reviewed in (, )] are being confirmed by more associations with other sex steroids. In the MrOS
recent studies based on substantial numbers of ob- Sweden study, high T predicted a reduced risk of CV
served events and MS-based hormone assay tech- events (). In a case-cohort design analysis from the
nology. In the Western Australian Health in Men French Three-City prospective cohort study, there
Study (HIMS),  MIs occurred during a mean of . was a J-shaped relationship of serum total and bio-
years in  community-dwelling men aged  to  available T with incident ischemic arterial disease
years at baseline (). In analyses adjusting for age, (ischemic stroke, fatal and nonfatal CHD), with both
other risk factors, and comorbidities, the incidence of low and high T levels associated with increased CVD
MIs was not significantly related to either total T, risk in elderly men (). In the Cardiovascular Health
calculated free T, DHT, or E. In the United Study, total and free T were not associated with in-
States–based Atherosclerosis Risk in Communities cident CVD in analyses adjusting for CV risk factors,

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


study,  coronary heart disease (CHD) events (i.e., although there was a curvilinear association of DHT
definite or probable MI, definite coronary death, or with incident CVD (). In an analysis of the Fra-
coronary revascularization procedure) occurred in mingham Heart Study, serum T was not significantly
 men with a mean baseline age of . years associated with incident CVD, but men with higher
during a median follow-up of . years. Following E levels were at lower risk of CVD (). In a later
multivariable adjustment there was no association of analysis from the Framingham Heart Study there was
quartiles of T with incident CHD; no other T fractions no association of baseline sex steroids and gonado-
or sex steroids were tested in this study (). tropin levels or their trajectories with incident CVD
In contrast to the negative findings for association during  and  years, but the numbers of men and
with CHD, there is limited evidence indicating a re- events in this analysis were rather limited (). A
lationship between serum sex steroid levels and the systematic review with meta-analysis of  studies did
risk for stroke in aging men. It had previously been not provide evidence for a pronounced harmful or
reported from the Honolulu–Asia Aging Study that beneficial effect of E on incident CVD ().
men in the highest quintile of serum E had a twofold Finally, a number of studies addressed the re-
increased risk of stroke, with no association of stroke lationship between sex steroid levels and CVD mor-
risk with serum T (). However, in the HIMS, serum tality. A systematic review and meta-analysis of studies
total and free T in the lowest quartile [, ng/dL up to  looking at incident CVD mortality in-
(,. nmol/L) and ,. ng/dL (, pmol/L), cluding , men from seven studies followed for a
respectively] were found to predict a higher risk of mean of . years is suggestive of an association of
incident stroke or transient ischemic attack (). lower serum total T with increased CV mortality [RR,
Association of stroke risk with serum androgen levels .; % CI, . to .; for the lowest vs highest T
was subsequently confirmed in an updated analysis of tertile], but heterogeneity among studies limited the
the HIMS based on  events of stroke observed validity of the summary estimate (). Overall, the
during a mean .-year follow-up of  men (). findings from more recent studies, although rather
Men with higher total T, free T or DHT had a lower disparate, tend to confirm the existence of a modest
incidence of stroke in fully adjusted analyses: the HR inverse association of serum androgen levels with
for the highest vs the lowest quartile of serum level CVD mortality. In an analysis of the Third National
was ., ., and . for total T, free T, and DHT, Health and Nutrition Examination Survey (), 
respectively. In this study there was no association deaths from CVD occurred in  men during a
between stroke incidence and serum E levels. In the -year follow-up. For decreases equal to the differences
Cardiovascular Health Study () among  men between the th and th percentile of the sex
(mean age,  years),  had an incident ischemic hormone distribution, lower free T, total E, and free
stroke. In analyses adjusting for stroke risk factors, E, but not total T, were associated with increased risk
which had a somewhat limited statistical power, for CVD mortality. However, the young mean age of
total and free T were not associated with stroke risk.  years at baseline limits the relevancy of this study in
However, a curvilinear relationship between serum the context of aging. In  men from the German
DHT and stroke incidence was observed with the prospective Study of Health in Pomerania,  deaths
lowest risk of stroke at a serum DHT of  to  ng/ occurred during an average follow-up of . years, of
dL and a greater risk for levels . and , ng/dL. which  were attributed to CVD (). A total T
The findings from studies looking at more het- of , ng/dL (. nmol/L), but not serum total T as a
erogeneous composite endpoints of CVD, including continuous variable, was associated with increased risk
events of CHD, stroke, and CVD death, and in of all-cause mortality after multivariable adjustments
some studies also peripheral arterial disease and heart (HR, .; % CI, . to .) and was also predictive
failure, reported mixed findings with some studies of cause-specific death from CVD (HR, .; % CI,

928 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

. to .); the effect of low T on mortality was the basis of the combination of a low total T and free T
strongest in men $ years of age. The authors later with the presence of three sexual symptoms had a
reported that they could not establish a causal re- higher risk of all-cause and CVD mortality: the HR for
lationship between serum T and mortality when CVD mortality was . (% CI, . to .) for  men
performing a Mendalian randomization analysis (). categorized as moderate LOH [i.e., total T , ng/dL
In  men of the HIMS with a mean age of  years ( nmol/L) and $ ng/dL ( nmol/L), free
there were  deaths during a mean follow-up period T ,. ng/dL ( pmol/L), and three sexual
of . years, of which  were due to CVD (). symptoms] and was . (% CI, . to .) for 
Lower free T [. vs . ng/dL ( vs  pmol/L)] men with severe LOH [i.e., total T , ng/dL
(HR, .; % CI, . to .) and higher LH and ( nmol/L), free T ,. ng/dL ( pmol/L), and
SHBG were associated with all-cause mortality, three sexual symptoms]; although the low T con-
whereas lower free T (HR, .; % CI, . to .) tributed to the increased risk, the presence of the three
and higher LH also predicted cause-specific mortality sexual symptoms was associated with mortality irre-
from CVD. In a subsequent report of an updated spective of serum T when compared with asymp-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


analysis of the HIMS encompassing  deaths ( tomatic men.
from IHD) in  men  to  years of age during a In summary, although suggesting the existence
mean .-year follow-up (), the authors reported of a modest relationship between CVD and (non–
that after allowance for other risk factors, midrange SHBG-bound) T, the overall picture on the asso-
serum total T, free T, and DHT were associated with ciation between endogenous sex steroid levels and
the lowest all-cause mortality (lowest mortality for CVD is rather disparate. The variability of findings
values corresponding to the third quartile); similar and the many inconsistencies might indicate re-
nonsignificant trends were observed for the association sidual confounding and, in particular, the possibility
of total and free T with IHD mortality, whereas higher of reverse causality related to general health status.
DHT was significantly associated with lower IHD Yet, some facts appear to emerge. First, the data
mortality (third quartile/first quartile, adjusted HR, clearly do not support the existence of an association
.; fourth quartile/first quartile, HR, .). In the between endogenous sex steroid levels and incident
Cardiovascular Health Study,  deaths were re- IHD and MI in aging community-dwelling men.
“There is limited evidence
ported in  men during a median -year follow-up Second, the data suggest an association of endog-
indicating a relationship
(): the authors reported a curvilinear association of enous androgen levels with incident stroke and with between serum sex steroid
serum DHT with all-cause mortality, similar to the CVD-specific as well as all-cause mortality. Whether levels and the risk for stroke in
association of DHT with incident CVD, and no as- the nature of the relationship is either a threshold aging men.”
sociation of mortality with total or free T; an asso- effect, a linear or a curvilinear association needs
ciation with cause-specific CVD mortality was not further clarification. In view of the unclear physi-
reported. In an analysis of mortality in the Athero- ological role in men of circulating DHT, which
sclerosis Risk in Communities study ( deaths in originates predominantly from hepatic T meta-
 men during a median of . years), quartiles of bolism, there is a need to learn more on the sig-
total T were not associated with total mortality and not nificance of observed associations of stroke and
with cardiac-associated mortality, of which there were, mortality with DHT, in particular where this oc-
however, only  cases (). In an analysis of the curred in the absence of a parallel similar association
Danish Monitoring Trends and Determinants of with T. Do these findings indicate real differential
Cardiovascular Disease (MONICA) study, the authors pathophysiological relationships for circulating T
assessed the association of intraindividual hormone and DHT or do they rather result from methodo-
changes with mortality (). From  men ( to logical and statistical coincidences? Also needing
 years of age) in MONICA  who had a second further unraveling is the nature and underlying
examination  years later (MONICA ),  died mechanisms of reported curvilinear relationships
during a subsequent mean follow-up of . years. between serum androgens and both stroke and
Independently of baseline serum T, men with the most mortality. Finally, for none of the reported associ-
pronounced total T decline (T ,th percentile) had a ations can a causal relationship be considered as
higher all-cause mortality risk compared with men established. In this context it is interesting that
within the th to th percentile (HR, .; % CI, despite known metabolically undesirable effects of
. to .); there were no consistent associations for ADT for prostate cancer such as weight gain and
changes in free T, SHBG, or LH and not for changes in increased insulin resistance (), in a meta-analysis
total T and CVD-specific mortality. In an analysis of of randomized trials CVD-associated mortality was
the EMAS cohort involving  men aged  to  not different and overall mortality was lower in
years,  deaths were registered during a median nonmetastatic patients with prostate cancer treated
follow-up of . years, of which  were from CVD with ADT compared with controls in either studies
(). Although total and free T were not associated of at least  years’ duration or short-term studies of
with mortality,  men categorized as having LOH on  months or less ().

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 929


REVIEW

Sexual function Associations with sex steroids


Sexual problems are rarely discussed with doctors, The importance of T in the maintenance of normal
but they are common in older men and may have sexual function is well established in young eugonadal
physical and/or psychological causes (). For men and in men with hypogonadism (), but its role
physicians and researchers, it is important to make a in aging men’s sexual function is still controversial. In
distinction in the evaluation of impaired so-called cross-sectional studies, men with lower T tend to have
“overall sexual function” from lack of sexual desire, less sexual activity and desire, and more ED. It has
infrequent spontaneous erections or morning erec- been suggested from these studies that sexual symp-
tions, premature ejaculation, erectile dysfunction toms may be the most helpful in identifying men likely
(ED) during masturbation or intercourse, limited to have adult-onset hypogonadism. Decreased spon-
penile flow measured by Doppler, inability to reach taneous erections and low sexual desire were the most
climax, sexual function distress, or sexual pain. Re- prevalent clinical symptoms in both younger and older
search often ignores that some men will engage in men in the validation of the New England Research
sexual activity that does not involve penetration and Institute hypogonadism screener. In addition to these

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


are still sexually active. Also, the role of a (sexually two symptoms, difficulty achieving or maintaining an
attractive) partner willing or able to have sexual erection was a symptom that significantly discrimi-
intercourse cannot be underestimated, and there nated the low T group from the control group in both
is a lack of data on the sexual function of non- age categories (). In the EMAS, with  men aged
heterosexual older men.  to  years, low levels of total and free T were
Most available studies in aging men focus on modestly associated with less frequent sexual thoughts,
sexual desire and ED. In most research, an association ED, and poor morning erections. The authors pro-
of low serum T with low sexual desire is described. posed criteria for LOH based on the presence of these
Low sexual desire can also result from medical and three sexual symptoms, in addition to a total T
mental health conditions (). ED is similarly more level , ng/dL (, nmol/L) and a free T level ,
likely to be the result of atherosclerosis, neurologic pg/mL (, pmol/L) (). When adjusted for age,
impairment, medication use, or psychogenic causes. BMI, and medical comorbidity, the associations be-
The evidence that T treatment is beneficial for im- tween T levels and sexual symptoms did not persist
proving decreased sexual desire is not consistent but (). Moreover, there was ~% background preva-
is overall supportive, and for ED T supplementation lence in sexual symptoms irrespective of total T
is considered a second-in-line pharmacotherapy after concentration, suggesting other contributory factors
phosphodiesterase  (PDE) inhibitors (see “Treat- than only low T. In a consecutive series of  pa-
ment of Hypogonadism in Elderly Men” below). tients with sexual dysfunction (mean age, . years),
an inverse relationship between T levels and the
Prevalence of sexual problems in older men prevalence of severe ED and a positive relationship
In the HIMS, % of  men  to  years of age with intercourse frequency was reported for the older
provided data on sexual activity. Increasing age, quartile ( to  years of age). For this older group,
partner’s lack of interest, partner’s physical limitations, there was an association between higher T levels and
osteoporosis, prostate cancer, diabetes, antidepressant better penile flow at penile color Doppler ultrasound
use, and beta-blocker use were independently asso- (). In the MMAS cohort, at the population level,
ciated with reduced odds of sexual activity (). In sexual desire and serum T concentrations were
this study population, sexual problems were highly strongly related. However, a simple individual report
prevalent: overall, % reported at least one problem, of “low libido” had a low predictive value in indicating
.% reported ED, .% lacked interest in sexual low T levels below commonly applied total T
activity, .% of participants were unable to reach thresholds, with a positive predictive value for total T
climax, and .% were anxious about their ability to values , and  ng/dL (. and . nmol/L) of
perform sexually. Unpleasurable or painful sex was less % and %, respectively (). A recent systematic
frequently reported. Low T levels were associated with review on  ptients evaluated for ED and 
lack of interest, but not with any of the other com- patients evaluated for decrease in sexual desire showed
plaints. Medical comorbidity, rather than T, seemed to that the presence of these findings were associated with
be the main predictor of sexual problems in older men positive likelihood ratios of . (% CI, . to .) and
(). In the -year longitudinal MMAS cohort with . (% CI, . to .), respectively, for finding T levels
 men aged  to  years at baseline, significant below threshold T ().
negative changes were shown in all domains of sexual Some evidence suggests that metabolites of T, such
function (sexual intercourse, erection frequency, sex- as E and DHT, may play a role in (the age-related
ual desire, satisfaction with sex, and difficulty with decline of) sexual function (, ), but in the EMAS
orgasm), except for frequency of ejaculation with cohort it was only total and free T (, ), and not
masturbation. Adjusted for baseline sexual function, E or DHT, that were associated with overall sexual
all outcomes were strongly related to age (). function. In fully adjusted models, higher free T was

930 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

specifically associated with greater masturbation fre- not sensitive to detect subtle changes in relatively
quency and less ED. Higher serum E was associated healthy community-dwelling aging men, whereas
with greater sexual function–related distress. The role other tests have been used to assess different specific
of DHT in these relationships has been inadequately cognition domains such as verbal and visual mem-
explored. However, in an experimental setting in ory, concentration, verbal fluency, and visuospatial
healthy younger men with suppressed endogenous T and visuoperceptual function. Still other studies have
with a GnRH agonist and administration of graded focused on the association of sex steroids with
doses of exogenous T, inhibition of a-reduction of T Alzheimer’s dementia (AD). This makes it arduous to
to DHT did not modify T effects on sexual function reliably discern overall trends. Nevertheless, al-
(). In the  participants in the US–based Tes- though some studies reported negative or even op-
tosterone Trials (TTrials) aged $ years with evi- posite findings, several earlier cross-sectional studies
dence of sexual dysfunction, diminished vitality, and/ concurred, reporting an association of either higher
or mobility disability and with an average of two total or optimal levels of T and, in particular, levels of
T , ng/dL (,. nmol/L), and in particular in the non–SHBG-bound (free or bioavailable) T with

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


subgroup of  men participating in the Sexual better performance in MMSE or tests addressing
Function Trial [reduced libido; score # on the specific aspects of cognition. More limited evidence
Sexual Desire Domain of the Derogatis Interview for suggested that E levels might be inversely associ-
Sexual Function (DISF-SDD); partner willing to have ated with cognitive performance [see , ) for
sexual intercourse at least twice a month], baseline reviews].
total and free T, but not E or SHBG, had small As to longitudinal observational studies, in an
significant cross-sectional associations with measures analysis of the Baltimore Longitudinal Study of Aging,
of sexual desire, erectile function, and sexual activity  men aged  to  years at baseline where followed
(). for an average of  years (). A higher ratio of T/
As to longitudinal data in the HIMS for  men SHBG was associated with better scores on visual and
with sex hormone data, higher free and total T at verbal memory, visuospatial functioning, and visuo-
baseline ( to ) predicted higher sexual activity motor scanning and with a reduced rate of longitu-
in the  months preceding the follow-up examination dinal decline of visual memory, whereas there were no
“…the evidence supports the
( to ): in a multivariable-adjusted analysis, for associations of T or the ratio of T/SHBG with mea-
existence of an association of
each  SD increase of baseline free T the odds of sures of verbal knowledge. In a subsequent analysis lower total and free T levels
having sex in the  months preceding the follow-up from the Baltimore Longitudinal Study of Aging, with lower sexual desire and
visit increased by % (). In a longitudinal study among  community-dwelling men  to  years of function in aging men.”
from the Concord Health and Ageing in Men Project age followed for a mean of . years,  developed
study, with  men at baseline and  at -year AD (). In multivariable-adjusted analyses, a di-
follow-up, whereas baseline levels of total and calcu- agnosis of AD was inversely associated with the ratio of
lated free T, DHT, E, and estrone did not predict a T/SHBG at both baseline and last measurement; each
decline in sexual function, the decline of total and -unit (nmol/nmol) increase of the ratio of T/SHBG
calculated free T (but not of DHT, E, or estrone) was associated with an ~% lower risk for AD. When
during follow-up was associated with a decline in considered separately, T and SHBG did not predict
sexual activity and desire, but not in erectile function. risk of AD. In the Honolulu–Asia Aging Study, de-
However, this decrease in serum T was quantitatively velopment of dementia and decline of cognitive
minor (,%), being too small to fully account for the function according to the Cognitive Abilities Screening
decrease in sexual desire or activity (). Instrument was assessed during an average of  years in
In conclusion, overall, the evidence supports the  men  to  years of age without dementia ().
existence of an association of lower total and free T A total of  men developed AD, including  cases
levels with lower sexual desire and function in aging with contributing cerebrovascular disease. After ad-
men. To what extent the reported associations are justment for cofounders, bioavailable T was not asso-
explained by residual confounding rather than a causal ciated with incident dementia or cognitive decline.
relationship remains uncertain. Alternatively, higher bioavailable E was associated with
greater cognitive decline and with risk of AD (HR per
Mental health SD increase, .; % CI, . to .) and of AD with
cerebrovascular disease (HR, .; % CI, . to .).
Cognition In  men with mean age of  years participating in
The methodology of studies assessing relationships the Health ABC study, free T was not associated with
between sex steroid levels and cognition in aging men cognitive decline (). In  men with a mean age of
is heterogeneous with, in particular, a variety of dif- . years from the Medical Research Council Cognitive
ferent tests to evaluate cognition. Several studies Function and Ageing Study, the men with cognitive
evaluated global cognition with the Mini-Mental State decline after  years according to the MMSE had lower
Examination (MMSE), which is well established but total T and higher SHBG serum levels; there were no

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 931


REVIEW

significant associations with verbal memory (story re- to predict cognitive decline or incident AD. Overall,
call) or the modified MMSE (MS) scores (). In the the evidence from observational studies does not
MrOS US study cohort,  men $ years of age support an important causative role of altered sex
with sex steroid measurements by immunoassay, of steroid levels in the decline of cognitive function in
whom  randomly selected men also had sex older men.
steroid measurements by MS at baseline, were sub-
jected at baseline and . years later to two tests Depression
of cognitive function, that is, Trails B (executive The lower prevalence of depression in men compared
function and motor speed) and MS (global cognitive with women and findings of lower T levels in men
function) (). In analyses adjusting for con- with depression compared with controls in some
founders, baseline free T and free E as well as total T earlier reports have been suggested to indicate a
and E were not associated with cognitive perfor- protective role of T against depression. Compatible
mance or change in cognition. However, higher with such a protective role of T are findings of ob-
SHBG was associated with worse cognitive perfor- servational cross-sectional studies in aging men of an

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


mance at follow-up testing (Trails B and MS) and inverse association of (free or bioavailable) T with
with greater decline on MS performance in mul- depressive symptoms or prevalence of depression
tivariate adjusted models also including free T and and more in particular association of clearly low T
free E. Men $ years of age participating in the levels (e.g., lowest quartile or quintile) with de-
Australian CHAMP study were assessed at baseline pression. However, these studies cannot establish a
(n 5 ), -year follow-up (n 5 ), and -year causal relationship, and other studies failed to reveal
follow-up (n 5 ) with measurements by MS of sex such associations (see (, ) for reviews). In his-
steroids (T, DHT, E, and estrone) and cognitive torical longitudinal cohort studies in a health care
testing with the MMSE (). None of the baseline setting for US veterans, a low T level in the hypo-
hormone levels was predictive of cognitive decline gonadal range [total T # ng/dL (#. nmol/L) or
during  years. In multivariable-adjusted models free T #. ng/dL (# pmol/L)] in men $ years of
accounting for potential confounders, men with age () and a low total T # ng/dL (#. nmol/L)
declining levels of total T, calculated free T, DHT, or in men $ years of age (), respectively, were
estrone during the follow-up period were more associated with a higher -year incidence of depressive
likely to have cognitive decline according to the illness.
MMSE. The absence of association of baseline sex Prospective longitudinal studies have produced
steroid levels with cognitive decline might suggest mixed results. In volunteering men ( to  years of
the existence of common factors underlying the age) participating in the Baltimore Longitudinal Study
parallel temporal evolutions of hormone levels and of Aging and followed for an average of  years,
cognitive function rather than a direct causal re- baseline and mean values for total T and the ratio of T/
lationship between them. In a recent report from the SHBG were not associated with depressive symptoms
Study of Health in Pomerania (), baseline T and assessed with the Center for Epidemiologic Studies
androstenedione by MS as well as calculated free T Depression Scale (CES-D) either in cross-sectional
in  men (mean age, . years) were not as- (n 5  men) or longitudinal (n 5  men) an-
sociated with - and -year changes in MMSE alyses (). In community-dwelling elderly men
score. (mean age, . years) followed for  years (), total
Finally, although it has been suggested that ADT and free T were not associated with the score on the
for prostate cancer may be associated with adverse -item Geriatric Depression Scale (GDS) in cross-
effects on cognition (), a recent large population- sectional analyses either at baseline (n 5 ) or at
based British study did not reveal an increased risk of endpoint (n 5  men); presentation with a GDS
dementia (). Using the United Kingdom’s score . indicating possible depression at the end of
Clinical Practice Research Datalink, the authors follow-up was not related to baseline (free) T, and
assembled a cohort of , men newly diagnosed changes in (free) T levels during follow-up were not
with nonmetastatic prostate cancer. During a mean associated with GDS score. In ~ men with hor-
follow-up of . years,  patients were newly di- mone values participating in the Study of Health in
agnosed with dementia. Compared with nonuse, Pomerania (), total and free T and androstenedione
ADT use was not associated with an increased risk of levels were not associated with depressive symptoms
dementia (incidence, adjusted HR, .; % CI, . defined as two self-reported questionnaire items of the
to.). Composite International Diagnostic–Screener either
In conclusion, several studies do suggest that low or in a cross-sectional analysis or during - and -year
suboptimal (free) T may be associated with cognitive follow-up in a longitudinal multivariable adjusted
decline. However, in most prospective studies, in- analysis.
cluding more recent large cohort studies with MS- At variance with these negative findings, other
based hormone assays, baseline sex steroid levels failed studies have reported that low T is predictive of

932 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

incident depressive symptoms. In multivariable ad- or congenital pathology. It is irreversible but treatable
justed analyses in  men $ years of age followed by gonadotropin or androgen replacement. The other
in the Longitudinal Aging Study Amsterdam, baseline type is functional hypogonadism with no organic
free T below a threshold of . ng/dL ( pmol/L) was cause, and it is therefore potentially reversible when
associated with baseline depressive symptoms assessed the underlying etiology is treated. Hypogonadism of
with the Center for Epidemiologic Studies Depression the aging male is usually primary when due to
Scale, and baseline free T in the lowest quintile chronological aging alone, and secondary when as-
[i.e., ,. ng/dL ( pmol/L)] predicted the onset of sociated with obesity or other comorbidities common
depressive symptoms in a longitudinal analysis; total T in old age. The frequently used term “late-onset
was not associated with depressive symptoms (). In hypogonadism” is in fact a misnomer, because in
the HIMS,  men developed depression among  % of cases it is caused by obesity or other comor-
older men ( to  years of age) during a .-year bidities with no association with chronological age
median follow-up (). Men with depression were ().
older and more likely to have CVD. Low serum total T

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


[, ng/dL (. nmol/L)], as measured by LC-MS/ Clinical evaluation
MS, was associated with incident depression after Algorithms have been developed for the clinical
adjustment for potential confounders (HR, .; % workup of an older male patient with suspected
CI, . to .). Low levels of calculated free T, DHT, hypogonadism [see, e.g., Fig.  ()]. The first task
and E were not associated with risk of depression in the clinical evaluation is to define whether the
(). patient is genuinely hypogonadal, because many of the
In conclusion, the mixed findings leave open the symptoms are nonspecific and not necessarily caused
possibility that markedly low T levels in older men by androgen deficiency. Hypogonadal symptoms, of
are associated with increased prevalence and in- which the sexual ones are most specific (), com-
cidence of depressive symptoms. However, in view bined with low serum T are the starting point of
of a substantial body of negative results, the whole of making the diagnosis. If the man is androgen deficient
the findings from observational studies are in- (repeatedly low serum T), the next step is to dis-
conclusive. In particular, there is only limited evi- criminate whether he has organic or functional “…there is only limited
dence that moderate declines of androgen levels in hypogonadism. It is not unusual that a Klinefelter evidence that moderate
aging men play a role in the occurrence of depressive syndrome patient remains undiagnosed in young declines of androgen levels in
symptoms. age. We concentrate here only on hypogonadism aging men play a role in the
diagnosis of aging men, that is, the functional occurrence of depressive
symptoms.”
hypogonadism.
Diagnosis of Hypogonadism in Aging Men Diagnosis of functional hypogonadism should
only be made in men with unequivocally low T (see
The classical definition of male hypogonadism entails below) and specific clinical symptoms. The latter can
a clinical syndrome where the testes fail to produce be subdivided into physical, psychological, and sexual
physiological concentrations of T and/or a normal domains (Table ). Most typically they include re-
number of spermatozoa due to organic pathology of duced sexual function, loss of muscle mass and
one or more levels of the HPT axis (). Reduced T strength, declined physical activity and performance,
production almost always leads to reduced sper- and a decrease of some cognitive abilities and de-
matogenesis (with Pasqualini syndrome, i.e., fertile pressive symptoms (). From these, sexual symp-
eunuch, as an exception), but spermatogenic failure toms are more specific whereas specificity of
can occur in men with normal T production (e.g., decreased cognition and depressive symptoms is
obstructive and idiopathic azoospermia). Failure at questionable (see “Clinical Correlates of Sex Steroid
the testicular level (low T, high gonadotropins) Levels in Elderly Men” above for a more extensive
is termed primary hypogonadism, and failure at review of the clinical correlates of low T). In surveys
the hypothalamic–pituitary level (low T, low or in- of community-dwelling men, decreased sexual desire,
appropriately normal gonadotropins) is termed sec- and ED, as well as less specific symptoms such as
ondary hypogonadism. Hypothalamic–pituitary and reduced physical performance, insomnia, fatigue,
testicular impairment can also occur in the same indi- irritability, and low mood, have been associated with
vidual, typically in the hypogonadism of older men who low T (, ). In the EMAS, only three sexual
are obese. symptoms (poor morning erections, ED, and reduced
The causes of primary and secondary hypo- sexual thoughts) showed clear syndromic association
gonadism are listed in Table . They can be divided with low T (). Many of the symptoms are non-
into two etiologically distinct types. One is the specific and overlap with those of chronic diseases
“classical” organic hypogonadism caused by perma- common in old age (the MetS, TDM, and CV, renal,
nent dysfunction at the hypothalamic, pituitary, or and pulmonary diseases). Important metabolic pa-
testicular level due to intrinsic structural, destructive, rameters include BMI and waist circumference, as

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 933


REVIEW

Table 4. Causes of Organic and Functional Primary and Secondary Male Hypogonadism
[Adapted with permission from
Grossmann M, Matsumoto Organic
AM. A perspective on middle-
aged and older men with Primary Secondary
functional hypogonadism:
Klinefelter syndrome Hyperprolactinemia
focus on holistic management. J
Clin Endocrinol Metab 2017; Cryptorchidism Stalk section or disease
102(3):1067–1075.]
a
Combined primary and Cancer chemotherapy Hemochromatosis
secondary.
Irradiation to the testes Idiopathic hypogonadotropic hypogonadism

Testicular trauma Kallmann syndrome

Testicular torsion Hypothalamic or pituitary tumors or infiltrative diseases

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Infectious orchitis Pituitary surgery or radiation

HIV infectiona

Anorchia, castration

Myotonic dystrophy

Old age (no comorbidities)a

Functional

Primary Secondary

Medications (inhibitors of androgen synthesis) Hyperprolactinemia

Renal disease Use of opioids and anabolic steroids

Alcohol use Use of glucocorticoidsa

Old age (with comorbidities)a

Chronic illnessa

Obesity, type 2 diabetes mellitus

Excessive exercise, malnutrition, wastinga

Anorexia/excessive exercise

Androgen deprivation therapy with GnRH analogs

Anabolic-androgenic steroid withdrawal syndrome

many older men with documented hypogonadism structured interview (Androtest) concentrating on
are obese. It is also important to assess the man’s sexual symptoms may have higher specificity ().
general health with respect to systemic illness, eating These case-finding tools are not recommended for
disorders, exercise habits, and use of prescription and detecting T deficiency in men receiving health care for
recreational drugs that may affect T synthesis or unrelated reasons.
metabolism. More specific organic findings sug-
gesting androgen deficiency may include osteopo- Hormonal evaluation
rosis and anemia. The critical hormonal parameters in the diagnosis of
There are several self-reported questionnaires on male hypogonadism are total T, free T, SHBG, and
symptoms of hypogonadism, such as the Aging Males’ gonadotropins. Although the currently available im-
Symptoms scale (), the Androgen Deficiency in the munoassays for SHBG and gonadotropins appear
Aging Male scale (), the Massachusetts Male Aging adequate, a fair amount of uncertainty and contro-
Study scale (MMAS) (), and the New England versy still surrounds the approaches on how to
Research Insitute hypogonadism screener (), but monitor circulating T levels, both total and free.
they all are hallmarked by high sensitivity com- For the diagnosis of hypogonadism, serum T
bined with poor specificity (), thus potentially should be consistently and unequivocally low as
leading to overdiagnosis of hypogonadism. A measured on two separate occasions in blood

934 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

Figure 4. Algorithm for the diagnosis and treatment of LOH [Huhtaniemi and Forti (461)]. If the clinician suspects LOH, the diagnosis
and choice of treatment can be obtained by following the flowchart from top to bottom, and choosing the next alternative on the basis of
the findings. Patients who present with symptoms of LOH should have a morning total T measurement. The results of this test, and a
subsequent free T determination (measured or calculated), will determine whether the patient should be assessed for his suitability for T
replacement. The hormone levels will vary with local protocols, so those presented are merely illustrative, and the clinician should use
local reference ranges for the assays in making their diagnosis. DRE, digital rectal examination; PRL, prolactin. [Reproduced with
permission from Huhtaniemi I, Forti G. Male late-onset hypogonadism: pathogenesis, diagnosis and treatment. Nat Rev Urol 2011;8(6):
335–344.]

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022

samples obtained mandatorily in the morning and hypogonadism (), the only lower total T limit put
preferably in a fasting condition. For positive di- forward is a total T of  ng/dL (. nmol/L) that
agnosis, both values have to be subnormal. There is can be used for assays that are calibrated against the
no consensus among the guidelines on what is Centers for Disease Control and Prevention stan-
considered subnormal, with the cutoff values for dard (). This is based on a project supported by
total T varying from  to  ng/dL ( to the American Endocrine Society and the Partnership
. nmol/L) (, ). Moreover, reference ranges for the Accurate Testing of Hormones in which
vary between laboratories due to methodological harmonized T reference ranges were generated
issues related to both T assay standardization and from data from US- and Europe-based cohorts of
generation of reference ranges. Therefore, in a recent community-dwelling men by cross-calibrating the
update of the American Endocrine Society clinical LC-MS/MS T assays used in each cohort against a
practice guideline on T therapy in men with method and calibrator developed by the Centers for

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 935


REVIEW

Disease Control and Prevention: the harmonized decreased morning erections, , ng/dL (. nmol/
reference range (.th to .th percentile) for L) for ED, and , ng/dL (. nmol/L) for low sexual
nonobese men  to  years of age was  to thoughts. The free T inflection point for decreased
 ng/dL (. to . nmol/L) (). Furthermore, it sexual thoughts, fatigue and sadness was found
is considered that a patient with T , ng/dL at ,. ng/dL ( pmol/L) and for ED and decreased
(, nmol/L) is most likely hypogonadal and may morning erections at , ng/dL ( pmol/L). The lack
benefit from T replacement therapy, and those with of specificity of even the most specific symptoms was
T . ng/dL (.. nmol/L) do not (). In highlighted by their high background prevalence of
general, the lower limit of T of young men can be % to % irrespective of the T concentration
used as the limit, but access to a reliable assay with a measured. Multiple correspondence analysis revealed,
validated reference range is important. If T is re- most conspicuously, that the three sexual symptoms
peatedly and unequivocally low, gonadotropin were clustered with a low total T level , ng/dL
measurements should be conducted to discriminate ( nmol/L) and , ng/dL ( nmol/L), and a free T
between primary and secondary hypogonadism. level ,. ng/dL ( pmol/L) in syndromic associ-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Inflection points were identified in the EMAS for ations. The symptoms that were significantly associ-
the different symptoms to be significantly associated ated with a low T level included the aforementioned
with T (): , ng/dL ( nmol/L) for decreased three sexual symptoms and a total T , nmol/L (OR,
vigorous activity, , ng/dL ( nmol/L) for .; % CI, . to .). The addition of the
threshold of free T ,. ng/dL ( pmol/L) increased
the OR to . (% CI, . to .). These limits, that
is, three sexual symptoms, total T , nmol/L, and
free T , pmol/L, can be regarded as evidence-
Table 5. Signs and Symptoms Associated With T
Deficiency Syndrome based criteria for the LOH diagnosis. They are,
however, not sufficient without the ancillary general
Sexual health information. The association was attenuated by
Decreased libido adjustment for age, BMI, and the number of coexisting
illnesses. The practicability of applying these rather
ED complex criteria in daily clinical practice may be an
Decreased frequency of morning erections issue.
Decreased performance
T assay: by immunoassay or MS?
Somatic The recent onslaught of MS for steroid hormone
measurements, including unconditional demands for
Increased visceral body fat/obesity
them to be the only acceptable method (), has
Decreased lean muscle mass confused clinicians and scientists who have relied for
nearly  years on various immunoassay methods. The
Decreased strength
superior specificity of MS methods will undoubtedly
Fatigue/loss of energy make them the future mainstay in steroid hormone
measurement. However, for purely practical reasons,
Decreased physical activity/vitality
many clinical steroid measurements will continue to
Low BMD be carried out using immunoassays because of their
high sensitivity, technical ease, output speed, and
Anemia
capacity, as well as cost and availability of commercial
Hot flushes reagents. It is unrealistic and unnecessary to replace
Loss of facial, axillary, and pubic hair/slow beard growth
them immediately with MS-based assays, because their
availability is still limited by cost, need of expensive
Decline in general feeling of well-being equipment, technical demands, and limited com-
Frailty mercial applications.
The problems with immunoassays, including their
Psychological cross-reactivity with similar analytes and standardi-
Depression/depressed mood zation and sensitivity issues, are well recognized ().
The Endocrine Society Council established a “Sex
Mood changes Steroid Assays Reporting Task Force” (), which
Irritability defined the necessary performance criteria for
methods to be used in clinical and nonclinical studies
Lack of concentration and recognized the need of high-quality, well-validated
Insomnia/sleep disturbances assays, irrespective of the technique (MS or im-
munoassay), to improve the quality of work, including

936 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

the assay traceability to a certified standard (). The are fraught with some potential problems, including
pros and cons of MS and immunoassay in steroid poor precision, inaccuracy, and low specificity. Un-
analysis have been recently discussed (), but the certainties on the dynamics of T binding to albumin,
debate continues (). The poor quality of T assays orosomucoid, and CBG represent another caveat.
has been a long-time target of complaints (, ). Nevertheless, there are clinically relevant differences in
Important reasons for this are the various shortcuts their performance with, in particular, the algorithm
taken when the methodology was introduced to based on the allosteric T binding model (), pro-
routine clinical laboratories, exhibiting reduced sen- ducing results far off target compared with equilibrium
sitivity, hard-to-control matrix effects, interference dialysis ().
with plasma steroid binding proteins, and nonspecific For measurement of free T, the equilibrium dialysis
competition with the assay antibodies. method is considered the reference method. It is
Several recent studies on large sample numbers technically demanding, sensitive to errors, and seldom
have demonstrated that the new platform T immu- available for clinical diagnostics. Direct analog im-
noassays are fully adequate for samples from men at all munoassays are not recommended for free T mea-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


concentrations [see, e.g., (–)]. One of them surement because of their poor accuracy (). Salivary T
concerned a large comparative study on T measure- shows good correlation with serum free T when re-
ments by a well-standardized immunoassay and a liably measured, preferably by MS (), but pre-
comparator LC-MS/MS method in serum samples analytical issues, for example, blood contamination
of . men (). T measurements by immuno- and metabolism of T in salivary gland (), may
assay offered good accuracy at all concentrations reduce its accuracy to monitor serum free T. Mea-
found in eugonadal men and in men with hypo- surement of bioavailable T is based on precipitation of
gonadism. Importantly, the platform immunoassay for SHBG-bound T and assay of remaining T in the
T had sensitivity and specificity also in the hypo- supernatant. However, because of the high levels of
gonadal male range, although with somewhat lower, imprecision and inaccuracy of this method it is not
but still acceptable, accuracy than in the eugonadal recommended for clinical practice (); estimates of
range. Not all immunoassays used in clinical T bioavailable T can also be calculated with the algo-
measurements are likely to have the same high quality. rithms based on the law of mass action similarly as for
If MS assays are not available, a good practical ap- free T.
proach is to test the immunoassay’s accuracy by For the above reasons, computational algorithms
comparing T concentrations measured by immuno- for estimation of free and bioavailable T remain the
assay and MS from a representative subset of samples most practical alternatives for clinical diagnosis.
(e.g., n 5 ). Therefore, calculated free T using high-quality T and
SHBG assays has been considered the most useful
Free T measurement clinical marker (). The calculated free T algorithms
According to the free hormone hypothesis, still suffer from multiple inaccuracies, including ignoring
lacking robust experimental proof, only the un- the nonlinear nature of T–SHBG binding, using dif-
bound (free) hormone is representative of the bi- ferent association constants for SBHG and albumin
ological activity in the target tissues (, ). binding, and variable association with equilibrium
Consequently, it is important to be able to measure dialysis data (). However, much of the critique of
accurately the partitioning of T in plasma between calculated free T is not in keeping with the experi-
protein-bound and free fractions to assess its real ence from clinical practice, where the linear model
biological activity. T is bound partly (~%) with algorithms (in particular the most commonly used
high affinity to SHBG and with low affinity (~%) to Vermeulen equation) appear to give, despite their
albumin, to a lesser extent to cortisol-binding shortcomings, acceptable data for clinical praxis and
globulin and orosomucoid, and the remaining research (, ).
~.% is referred to as free T. The sum of free T and Proof for the usefulness of free T assesment in the
loosely albumin-bound T, which may dissociate at diagnosis of hypogonadism of elderly men was pro-
least in part in the microcirculation, is often referred vided by the EMAS data (, ) and some other
to as bioavailable T. studies (). They revealed in a cross-sectional
The free T fraction can be measured following analysis that men with normal total T but low free
equilibrium dialysis or ultrafiltration of serum (either T concentrations had higher LH levels, reported more
indirectly with use of a radioactive T tracer or by direct hypogonadism-associated sexual and physical symp-
MS-based T measurement), or estimated by calcula- toms, and had lower Hb values and BMD than did
tion from total T, SHBG, and albumin concentrations men with normal total and free T, and, conspicuously,
using published algorithms either based on the law of than did men with low total T but normal free T. Use
mass action (–), empirically derived (), or of total T alone is likely to result in overdiagnosis of
based on a recently proposed alternative dynamic hypogonadism, especially in men who are obese and
allosteric model of T binding (), All algorithms when the total T levels are borderline decreased. The

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 937


REVIEW

usefulness of free T measurements in fine-tuning the and men with obesity were compared, although the
diagnosis was apparent in the borderline range of  former had higher average levels of T. Hence, although
to  ng/dL ( to  nmol/L), but not below or above. there is minimal variation in T levels between Eu-
In a recent longitudinal analysis of the EMAS data ropean and American cohorts of men, more variability
(), the same finding was made in men developing has been found between and within multiethnic,
secondary hypogonadism during a .-year follow-up multiracial, and geographically diverse populations in
period: the agreement with hypogonadal symptoms some (, ), but not all (), studies. Whether the
was only found in men with low total and free T, but reference ranges should be established with men from
not in those with low total and normal free T. the general population or men in perfect health
All current guidelines (, , , , ) rec- remains a topic of discussion. Another issue is how to
ommend free T assessment only as a complementary take into account the known decreasing effect of
tool in symptomatic men whose total T is near the obesity on free and total T, SHBG, and gonadotropins
lower limit of the normal range and in conditions that in otherwise healthy men ().
affect SHBG concentrations and render total T a less

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


reliable index of gonadal function. The only exceptions
are a recommendation from Japan for preferential use Treatment of Hypogonadism in Elderly Men
of free T (), and from Australia against the use of
free T in clinical decision-making (). In younger men with low T and an established di-
agnosis of hypogonadism, owing to a defined con-
Reference range of T for aging men genital or acquired irreversible cause, T replacement
One of the problems in the diagnosis of hypogonadism therapy is a common clinical practice. The aim of such
of older men has been the lack of age-dependent replacement therapy is to reestablish “physiological”
reference ranges, and the diagnosis is usually made serum T levels within the reference range for young
on the basis of data from healthy young men. Rig- adult men. This practice is generally not challenged,
orously established age-dependent reference ranges for even though for historical and ethical reasons it is
T have been missing so far. It has also remained mostly based on a rational concept and clinical ex-
unclear whether the reference ranges differ between perience rather than controlled data. Any treatment
populations, and to what extent their variability is due involving T administration is a pharmacological
to analytical variation and to biological variation. treatment, and although probably acceptable in this
To improve the situation, Travison et al. () context in young men, the term “replacement therapy”
recently established harmonized reference ranges for should be avoided in the context of newly initiated T
total T, applicable across laboratories by cross- therapy in older men. Indeed, it is a misnomer because
calibration of assays to a reference method and stan- we do not really know what the physiological, desired,
dard. The data are based on serum samples from . or ideal serum T levels are in older men. Also, whereas
community-dwelling men from four cohort studies there appears to be a dose ceiling effect for some T
from Europe and the United States. The influence of actions with no additional effect for T levels above the
systematic methodological differences was minimized normal range, this is obviously not the case for some
by harmonizing all measurements to a higher order important actions of T. Indeed, for T effects such as on
standard prior to the estimation of reference ranges. muscle and erythropoiesis, there is a progressive re-
Preanalytical differences were minimized by using sponse from infraphysiological, over physiological, up
samples collected in a similar fashion (fasting morning to frankly supraphysiological dosages without obvious
samples, storage at 2°C). T concentrations were threshold or ceiling effects around the serum T ref-
measured by the reference LC-MS/MS method from a erence range. Moreover, a critical appraisal indicates
subset of samples from each cohort. Normalizing that in many studies on T therapy in older men, mean
equations were used to generate harmonized values, T exposure during T treatment tended to be supra-
which were used to derive standardized, age-specific physiological. This is certainly so relative to age-
reference ranges of the whole sample cohort. specific mean T levels, but also relative to serum T
The harmonization process reduced intercohort levels in young men when taking into account factors
variation, and the reference range calculated from such as the marked physiological circadian variation of
all serum samples in - to -year-old nonobese serum T in young men, preparation-specific T
(BMI , kg/m) European and American men was pharmacokinetics, and the slower T clearance in older
found to be  to  ng/mL (. to . nmol/L). men (). Finally, the term “replacement therapy”
Whereas the .th percentile remained stable upon tends to have a potentially misleading connotation of
aging, the median (th percentile) and .th per- something physiological and thus innocuous, which
centile decreased in older age groups (Table ). After may not need to be supported by the same level of
methodological harmonization, the differences be- evidence as other pharmacologic interventions. This
tween the different cohorts were minimal. The same long-prevailing misconception is likely to be one of
trends were observed when the data on nonobese men several explanations for the rather limited and mostly

938 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

Table 6. Model-Based Estimates of Population Centiles for Total T Concentrations (ng/dL) Based on Data From Nonobese
Men (N 5 6933) and All Men (N 5 9054) in Four Harmonized Cohorts
Age (y)
Percentile 19–39 40–49 50–59 60–69 70–79 80–99
To convert from ng/dL to
All nonobese men nmol/L, multiply by 0.0347.
[Reproduced with permission
2.5 267 235 219 218 218 157 from Travison TG, Vesper HW,
Orwoll E, et al. Harmonized
5.0 304 273 256 254 252 218 reference ranges for circulating
testosterone levels in men of
10.0 344 310 297 296 292 278 four cohort studies in the
United States and Europe. J Clin
25.0 424 386 374 374 372 362 Endocrinol Metab 2017;102(4):
1161–1173.]

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


50.0 531 481 477 477 477 476

75.0 643 608 605 604 604 604

90.0 774 749 749 749 749 749

95.0 850 839 839 839 839 839

97.5 929 929 929 929 926 913

All men

2.5 229 208 192 190 190 119

5.0 273 243 222 221 220 203

10.0 318 283 262 260 259 256

25.0 396 358 341 340 340 338

50.0 507 461 446 446 446 446

75.0 626 588 573 572 572 572

90.0 755 729 720 720 720 720

95.0 834 813 812 812 812 812

97.5 902 902 902 902 902 902

only recent evidence from controlled trials on T may suffer from publication bias. Overall, they nev-
therapy in older men. As for all pharmacological in- ertheless suggest a beneficial effect of T administration
terventions, clinical practice should preferably be on erectile function and sexual desire in those men
supported by sufficient information on safety and with initially low serum T, independently of age (,
efficacy of T therapy for defined clinical outcomes and ).
for treatment durations relevant for the intended Recently, Corona et al. () performed an updated
clinical use. To date, the evidence base from controlled meta-analysis of results of T therapy on sexual function
trials, besides being limited, is characterized by great based on International Index of Erectile Function (IIEF)
heterogeneity that pertains to inclusion criteria (age, scores, which included  studies enrolling  par-
serum T levels, presence of hypogonadism symptoms, ticipants with a mean follow-up of . weeks and mean
and/or comorbidities), evaluation criteria and tools, as age of . 6 . years. They found that T therapy
well as modality, dosage, and duration of T therapy. significantly (P , .) improved erectile function
Older studies mostly included only small numbers of compared with placebo according to the IIEF Erectile
subjects, and RCTs of .-year duration are rare. Function Domain, although the effect size was modest
[. (. to .) and . (. to .) using a fixed
Efficacy of T therapy and random model, respectively], and the absolute
changes in IIEF Erectile Function Domain score may
Sexual function not be clinically significant in many subjects, in par-
The findings from older studies on the effects of T ticular in those with more severe ED (). The
therapy on sexual function have been inconsistent and treatment effect was greater in studies including men

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 939


REVIEW

with baseline T , ng/dL (, nmol/L) compared aiming at T levels in the upper normal range for young
with those including men with T , ng/dL men ( to  ng/dL), failed to significantly improve
(, nmol /L). They found that T therapy also im- sexual function (sexual desire, erectile function, overall
proved scores for the other IIEF domains, including sexual function scores, partner intimacy) compared
sexual desire domain, intercourse satisfaction domain, with placebo. In this trial improvement of sexual
orgasmic domain, overall sexual satisfaction, and IIEF- function was a secondary outcome, and presence of
 total score (). sexual symptoms was not an inclusion criterion.
This meta-analysis included the results of the re- In the Sexual Function Trial () none of the
cent Sexual Function Trial, which is part of the co-  considered baseline patient characteristics pre-
ordinated set of T trials constituting the TTrials. This dicted responsiveness to T therapy. In particular, a
randomized placebo-controlled trial () is highly BMI . kg/m and/or diabetes, which were present
relevant in the context of this review, as it included in as many as % and % of the study population,
older men ($ years of age; mean age,  years) with respectively, were not predictive of outcome. This is at
low sexual desire (score , on the DISF-SDD, with a variance with findings of meta-analyses suggesting that

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


score range from  to ) and unequivocally low serum the effects of T therapy on sexual function may be
T [average of two early morning T , ng/dL somewhat less in men with obesity and diabetes (,
(,. nmol/L)] treated  year with placebo or T gel ).
(initial dosage of  mg daily by % gel), aiming at Studies addressing potential additive effects of T
serum T levels in the midrange for young men. The therapy to improve erectile function in men with ED
men treated with T (n 5 ) reported a significantly treated with a PDE inhibitor are heterogeneous in key
greater (P , .) increase of sexual activity (Psy- variables such as the gonadal status at inclusion.
chosexual Daily Questionnaire question ), increase of Findings are not univocal (, ). Overall, available
sexual desire (DISF-SDD), and improvement of evidence is inconclusive and does not support the use
erectile function (IIEF score) than did the men treated of T treatment to potentiate therapeutic effects of
with placebo gel (n 5 ). The men treated with T PDE inhibitors. In particular, meta-analysis has
were more likely than those treated with placebo to shown positive effects in uncontrolled studies but not
report improved sexual desire since the beginning of in controlled trials (). In a controlled trial in men
the trial (P , .). Additional analyses () in- with low serum T and ED who were first optimized
dicated that compared with placebo, T therapy in- with sildenafil, T therapy had no added beneficial effect
creased  out of  Psychosexual Daily Questionnaire on mood and general well-being compared with
question  measures of sexual activity, including sexual placebo ().
daydreams, anticipation of sex, sexual interactions In conclusion, T therapy can improve various
with partner, “flirting by you,” orgasm, ejaculation, aspects of sexual function, including sexual desire,
intercourse, masturbation, night spontaneous erec- erectile function, and overall sexual activity. These
tions, and erection in response to sexual activity; there beneficial effects are largely limited to men with un-
was a nonsignificant treatment effect for the items equivocally low serum T and prevalent symptoms of
“flirting with you by others” and “day spontaneous sexual dysfunction. The effect size is mostly only
erections.” Interestingly, in T-treated subjects, in- moderate, with effects on ED lesser than those re-
cremental increases in total T, free T, and E were ported for PDE inhibitors, and there is presently no
associated with improvements in sexual activity and convincing demonstration that cotreatment with T
desire, but not in erectile function. No threshold T potentiates the effect of PDE inhibitors on erectile
level was observed for any outcome. In general function.
treatment effect size was low to moderate, with the
effect size on erectile function less than what has been Body composition, metabolic profile, and
reported for treatment with PDE inhibitors (). physical performance
The efficacy results from this study are consistent with Body composition, muscle mass and
the findings of another recent trial involving  men strength, and physical performance. Healthy
with low sexual desire or low energy and low serum T older men ( to  years of age) with suppressed
(, ng/dL or . nmol/L), but of younger mean endogenous T production by administration of a long-
age of . years (). The men randomized to  mg acting GnRH agonist and treated with graded doses of
of topical % T solution daily for  months reported a T ( to  mg T enanthate weekly for  weeks)
greater increase of sexual drive, sexual activity, and respond with a dose- and blood T concentration–
erectile function compared with placebo-treated men. related increase of fat free mass, skeletal muscle mass,
In the Testosterone’s Effects on Atherosclerosis pro- and muscle strength and with a decrease of fat mass
gression in Aging Men (TEAAM) trial, involving  (). This important experiment, besides showing that
men $ years of age with low-normal serum T [total the anabolic response to T in older men is proportional
T,  to  ng/dL (. to  nmol/L); free T, , pg/ to the T dose up to frankly supraphysiological T levels,
mL (, pmol/L)], treatment with . g of % T gel, has also shown that the anabolic response to T in older

940 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

men is similar to that in younger men ( to  years of test, and the Tinetti gait and balance test, or for self-
age). In recent studies with a similar general design in reported physical activity (Physical Activity Scale of the
younger men, it was further shown that a-reduction of Elderly). In post hoc analyses, there was a significantly
T to DHT is not essential for mediating these anabolic greater improvement in the Aggregate Locomotor
effects of T () and that aromatization of T to E, Function test and PPT in the men with at least two
which does not intervene in the anabolic action of T on frailty criteria and of the PPT in men at least  years
skeletal muscle, appears to be required for full ex- of age. The authors subsequently reported that, not
pression of T action to reduce fat mass (). surprisingly, the treatment effects on muscle strength
In a meta-analysis of  earlier RCTs, including a and lean mass after the -month T treatment were not
total of  men [weighted mean age, . years; maintained  months after treatment ().
mean T,  ng/dL (. nmol/L); range,  to In older men on optimal medical therapy for stable
 ng/dL (. to  nmol/L)], of whom  were chronic heart failure [New York Heart Association
randomized to T treatment (), the authors found functional class II/III; median age,  years; % with
that T treatment decreased total body fat mass by a T , ng/dL (,. nmol/L)], parenteral T ther-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


mean of . kg (.% of initial body fat) and increased apy for  months with long-acting T undecanoate
fat-free mass by a mean of . kg (.%), whereas the ( mg at , , and  weeks) (n 5 ) compared
effects on muscle strength were heterogeneous, with a with placebo (n 5 ) increased quadriceps isometric
tendency toward improvement only for leg/knee ex- and isokinetic strength, peak oxygen consumption,
tension and grip strength in the dominant arm (pooled and peak work load during a cardiopulmonary ex-
effect size, .; % CI, . to .). Another meta- ercise test, as well as the distance walked in the
analysis of earlier trials ( trials;  men; mean age, -minute walk test. Baseline T was directly related to
. years) also concluded that treatment with T (or baseline isometric strength, peak oxygen consumption,
DHT) produced a small to moderate increase of and -minute walk test distance, and changes in serum
muscle strength (). T in T-treated subjects correlated with changes in peak
Several relevant randomized, controlled trials have oxygen consumption and isometric strength ().
more recently been reported. In older men (mean age, This finding of improved functional capacity in
 years) with at least one criterion of frailty, a low to T-treated men with heart failure was confirmed in a “The findings on the impact of
low-normal serum T and a hip BMD T-score # 2 or meta-analysis of this and three other trials (). T treatment on physical
less or a history of nontraumatic fracture, randomized In a subgroup analysis of the TOM trial, treat- function have…been rather
to treatment daily with  mg of transdermal T (% ment of mobility-limited men $ years of age with disappointing…with mostly
gel) (n 5 ) or placebo (n 5 ), T therapy for low T [total T,  to  ng/dL (. to . nmol/L) small effect size and of
uncertain clinical significance.”
 months resulted in a significant difference above or free T , ng/dL ( pmol/L)] with  mg of T
placebo of .% lean mass and .% appendicular daily by gel during  months (n 5 ) attenuated the
skeletal muscle mass, and a trend toward decreased age-related fall in aerobic function compared with
body fat (). There was no treatment effect for a placebo-treated subjects (n 5 ) (). In the
battery of tests of muscle strength and physical per- TEAAM study in community-dwelling men with low
formance. However, in this study baseline T levels to low-normal serum T [ to  ng/dL (. to
were rather high, and the achieved T increase during . nmol/L) or free T , ng/dL (, pmol/L)],
treatment appeared small. treatment with  mg of T (% gel) daily for  years
Srinivas-Shankar et al. () randomized (n 5 ; mean age, . years; mean BMI, . kg/
community-dwelling elderly intermediate-frail and m) compared with placebo treatment (n 5 ;
frail men [mean age, . years; T # ng/dL mean age, . years; mean BMI, . kg/m) was
( nmol/L) or free T #. ng/dL ( pmol/L); associated with a small increase in lean mass of . kg
% with only one or two out of five frailty criteria] (. to . kg; P , .), which in the T-treated men
to  months of transdermal treatment with  mg of T was related to the changes in total and free T (). T
daily (% gel) (n 5 ) or to placebo treatment (n 5 treatment compared with placebo was associated
). Treatment resulted in a mean increase of lean with significantly greater increases in chest-press
body mass by . kg and decrease of fat mass by strength and in chest-press and leg-press power,
. kg. Isometric knee extension peak torque (primary which were related to changes in T levels. T sup-
outcome) improved in the T-treated men and declined plementation was also associated with greater improve-
in the placebo group, resulting in a significant treat- ment in both unloaded and loaded stair-climbing
ment effect. The treatment effect was not significant power, which was associated with the improvement in
for either isokinetic knee extension peak torque, leg-press power. Although the visit-by-treatment in-
isometric and isokinetic knee flexion peak torque, or teraction was not statistically significant, numerically
handgrip strength. Moreover, there was no treatment the treatment effects were maximal at the -month
effect for any physical function test, including the visit and tended to wane with time. In general,
Aggregate Locomotor Function test (ALF), the treatment effect size was small and of uncertain clinical
Physical Performance Test (PPT), the -minute walk significance.

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 941


REVIEW

The Physical Function Trial, part of the TTrials, The small, but consistent improvements in lean mass
included men $ years of age with T , ng/dL are associated with more variable effects on muscle
(,. nmol/L), with self-reported difficulty walking or strength and power, with limited improvements or no
climbing stairs and with a gait speed of ,. m/s on effect depending on the considered study or within
the -minute walk test. Between men randomized to single studies depending on the considered muscle
 months of treatment with T gel aimed at achieving group and test. The findings on the impact of T
levels in the midrange for young men (n 5 ) treatment on physical function have in turn been
compared with men randomized to placebo treatment rather disappointing, with negative findings or positive
(n 5 ), there were no significant differences for findings with mostly small effect size and of uncertain
either the percentage of men whose -minute walk clinical significance. Although not formally estab-
distance increased by at least  m (primary outcome), lished, it appears from the whole of available data that
the changes from baseline of the -minute walk dis- higher T doses, longer treatment duration, lower
tance, or the percentage of men whose score on the baseline serum T, and the presence of baseline
-item Physical Functioning Scale (score range,  to functional capacity limitations are factors that may be

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


) of the SF- survey increased by at least  points. associated with greater treatment responses.
There was a difference between groups for increase of Body composition and metabolic profile.
the -item Physical Functioning Scale score from Corona et al. () performed a meta-analysis of 
baseline (. points; P 5 .). For these four efficacy RCTs ( placebo-controlled), with a mean trial du-
outcomes, there was a significant difference in favor of ration of . months and that included a total of 
T treatment when statistical power was increased by subjects in the T therapy group and  in the control
considering the larger combined group of participants group, respectively. Mean age, BMI, and baseline T
to the different TTrials (P 5 . to .) (). levels of enrolled men were . years, . kg/m, and
Hildreth et al. () randomized community-  ng/dL (. nmol/L), respectively. Studies differed
dwelling healthy and highly functioning men with a in administered T doses, T formulations, and cohort
mean age of  years and low-normal serum T ( to characteristics. The authors found T therapy was not
 ng/dL or  to . nmol/L) to treatment with associated with a decrease in weight ( trials), waist
transdermal T % gel [ doses aiming at  to  ng/ circumference, or BMI, but they confirmed a signifi-
dL (. to  nmo/L) and  to  ng/dL (. to cant decrease in fat mass ( studies) and increase in
. nmol/L) levels, respectively] or placebo, and to lean mass ( studies). T therapy was associated with a
either progressive resistance training (PRT) or no reduction in fasting glycemia and insulin resistance
exercise for  months. In the PRT group (n 5 ), according to the homeostatic model assessment
functional performance (a battery of tests including (HOMA) of insulin resistance (HOMA-IR): consid-
the -minute walk test) and upper and lower body ering only placebo-controlled RCTs, standardized
strength were not different between T- treated (n 5 means were 2. (2. to 2.) and 2.
) and placebo-treated (n 5 ) men, despite a (2. to 2.) for glycemia and the HOMA index,
greater reduction of fat mass and increase of fat free respectively (both P . .); the between-group
mass in the T-treated subjects. In non-PRT men (n 5 differences for glycemia and HOMA-IR were found
) T treatment (n 5 ) compared with placebo (n 5 to be related to modifications in lean mass but not in
) had no effect on functional performance, but it fat mass. The effect on fasting glycemia was greater in
reduced fat mass, increased fat-free mass, and im- RCTs enrolling subjects with metabolic disease (n 5
proved upper body strength but not lower body ) compared with those evaluating elderly subjects
strength. The findings in this study, although in- (n 5 ). When considering only placebo-controlled
dicating additive effects of T and exercise on changes RCTs, T therapy was associated with a decrease of total
in body composition, did not offer support to the cholesterol [2. (2. to 2.)] and triglycerides
hypothesis that T treatment might enhance the effects [2. (2. to 2.)]; there was no significant
of exercise on muscle strength or functional perfor- effect on high-density lipoprotein (HDL) cholesterol
mance in older men. More studies are needed to levels, systolic blood pressure, and diastolic blood
answer the question whether this may have resulted pressure ().
from suboptimal exercise intensity or required In a study in older men (median,  years of age)
achievement of higher, supraphysiological T levels with low-normal baseline T, treated with transdermal
(). T ( mg/d T patch) [n 5 ; baseline median T,
In summary, T therapy in older men with low to  ng/dL (. nmol/L)] or placebo patch [n 5 ;
low-normal serum T, whether they are generally baseline median T,  ng/dL (. nmol/L)] the
healthy or have physical function impairments, is subjects ingested a labeled mixed meal and under-
consistently associated with improvement of body went a frequently sampled labeled IV glucose tolerance
composition with a quantitatively modest decrease of test before and after a -year treatment. Compared
total body fat mass, increase in fat-free mass, and with placebo treatment, T did not improve carbo-
increase or prevented decrease of skeletal muscle mass. hydrate tolerance or alter insulin secretion, insulin

942 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

action, glucose effectiveness, hepatic insulin clearance,  ng/dL (. nmol/L)] to  weeks treatment
or the pattern of postprandial glucose metabolism with T undecanoate (-mg injections at , , and
(). In the previously discussed study showing that T  weeks) (n 5 ) or placebo injections (n 5 ). At
therapy improved muscle strength and functional the  weeks study endpoint there was a marginal
capacity in elderly men with heart failure, treatment decrease of waist circumference [estimated mean, 2.
also improved insulin sensitivity as assessed by (2. to 2.) cm] and total cholesterol [2.
HOMA-IR (). (2. to .) mmol/L], but there was no treatment
As to relevant recent trials, in the older men with effect on HbAc, HOMA-IR, fasting glucose, HDL
low serum T participating in the TTrials (% with cholesterol, low-density lipoprotein cholesterol, tri-
BMI . kg/m; % with diabetes) -month T glycerides, body weight, or BMI. Jones et al. ()
therapy (n 5 ) compared with placebo treatment randomized men (mean age, . years) with TDM
(n 5 ) resulted in a significant decrease of total and/or the MetS and baseline T # ng/dL (nmol/
cholesterol (adjusted mean difference, 2. mg/dL; L) or free T #. ng/dL ( pmol/L) to daily
P , .), HDL cholesterol (2. mg/dL; P , .), treatment with  mg of transdermal T (% gel)

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


and low-density lipoprotein cholesterol (2. mg/dL; (n 5 ) or placebo gel (n 5 ) for  monts (the
P 5 .), but not of triglycerides. There was a small TIMES study). There was a % reduction in
significant decrease in fasting insulin (2. mIU/mL; HOMA-IR in T-treated men compared with the
P 5 .) and HOMA-IR (2.; P 5 .), but not of placebo group and a reduction in lipoprotein a and
fasting glucose or HbAc levels. There was also no HDL cholesterol, but there was no treatment effect on
treatment effect on weight, BMI, or waist/hip ratio HbAc, fasting insulin and glucose levels, HOMA-B,
(). In the TEAAM trial a subset of  nondiabetic triglycerides, abdominal obesity, percentage body fat,
men (mean age, . years) underwent an octreotide waist circumference, or BMI. Gianatti et al. ()
insulin suppression test at baseline and at  and randomly assigned men (median age,  years) with
 months after randomization to T therapy ( mg TDM and serum T # ng/dL ( nmol/L) to
daily by % gel) or placebo. Insulin sensitivity, esti-  weeks of intramuscular T undecanoate injections
mated as the steady-state glucose concentration at (n 5 ; baseline median HbAc,  mmol/mol or
equilibrium during octreotide and insulin infusion, .%) or matching placebo (n 5 ; baseline HbAc, “…although T therapy in aging
was not significantly different between T-treated (n 5  mmol/mol or .%). Treatment reduced fat mass men with low to low-normal T
) and placebo-treated (n 5 ) men across the [mean adjusted difference, 2. kg (2. to 2.)], can modestly improve insulin
 months of treatment (P 5 .); stratification with a decrease of subcutaneous but not visceral ab- sensitivity, overall the data on
according to baseline T # ng/dL (. nmol/L) dominal fat, and increased lean mass [. kg (. to metabolic effects…are
inconsistent.”
and . ng/dL, or according to age # years or . .)], but T therapy failed to improve HOMA-IR
 years, did not show significant treatment effects for [2. (2. to .)] or glycemic control accord-
any subgroup (). ing to HbAc [.% (. to .)]. Grossmann et al.
Studies focusing more specifically on T effects in () performed a meta-analysis of seven RCTs (the
men with obesity, the MetS, or TDM have reported RCTs discussed above included) on the effects of T
inconsistent findings. In an RCT, middle-aged (me- therapy in men with TDM and the MetS, enrolling a
dian,  years of age) men with obesity (BMI $ kg/ total of  men. Although there was heterogeneity
m) on a hypocaloric diet were randomized to among trials depending on the method of calculation of
 weeks of -weekly intramuscular T undecanoate HOMA-IR, the meta-analysis suggests that T therapy
(n 5 ) or placebo (n 5 ). Although there was no modestly improves insulin resistance [standardized
difference in weight loss between groups, men re- mean difference, 2. (-. to 2.); P , .].
ceiving T showed a greater reduction in body fat mass There is no effect of treatment on glycemic control as
and visceral fat and less loss of lean mass. These assessed by HbAc [standardized mean difference,
relative differences in body composition changes did 2. (2. to .); P 5 .].
not translate in an effect of therapy on metabolic In conclusion, although T therapy in aging men
parameters, with no difference between groups for with low to low-normal T can modestly improve
fasting glucose, HOMA-IR, HbAc, triglycerides, or insulin sensitivity, overall the data on the metabolic
cholesterol fractions (). Aversa et al. () reported effects of T therapy are inconsistent. It does not appear
that treatment of middle-aged men (mean age,  that the evidence available supports the use of T
years) with the MetS and T # ng/dL ( nmol/L) therapy with the aim of improving metabolic status
for  months with T undecanoate injections ( mg either in older men with low to low-normal T in
every  weeks) (n 5 ) improved HOMA-IR general or more in particular in older men with TDM
compared with treatment with placebo gel (n 5 or the MetS.
), which resulted mainly from an increase in the
placebo group and only a marginal decrease in the T Bone health
group. Hackett et al. () randomized men with There are few controlled studies on the effects of T
TDM [mean age, . years; mean baseline T, therapy on the bone health of (older) men. Their focus

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 943


REVIEW

is on the effects on BMD as a surrogate marker of bone studies. An effect of T therapy to decrease the levels of
strength and they are mostly of only short duration. A bone resorption markers was confirmed in a meta-
meta-analysis by Tracz et al. () included  men analysis ().
from eight placebo-controlled trials (). Six of these As to more recent studies, Kenny et al. ()
trials included men with a mean age of  years or randomized  men (mean age,  years) to daily
older. However, in seven out of eight trials mean treatment with  mg of transdermal T (% gel) or
serum total T at baseline was within the normal range, placebo. Patients were recruited on the basis of
that is,  to  ng/dL (. to . nmol/l). Only age $ years, at least one criterion of frailty, a low to
one study included men with prevalent fractures, low-normal serum T, and a hip aBMD T-score of 2
whereas half of the studies included men at risk for or less or a history of nontraumatic fracture in the past
secondary osteoporosis (glucocorticoid use, rheuma-  years. They reported a small positive treatment effect
toid arthritis, AIDS wasting); two studies included at  months on aBMD at the lumbar spine, no effect
observations during  months, the others were at the hip, and a negative treatment effect at the
of #-year duration. Meta-analysis showed a small forearm. Interpretation of this study is, however,

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


significant effect of T treatment on lumbar spine hampered by a large patient dropout, mean baseline T
aBMD assessed by DXA (effect size, .; % CI, . level well within the normal range, and only limited
to .) corresponding to a % (.% to %) increase. serum T increases achieved during treatment. The
There was no significant treatment effect at the Bone Trial, part of the TTrials (), included men on
femoral neck [effect size, . (2. to .)]. The the basis of age $ years and low serum T , ng/
effect of T on spine aBMD was significant for five trials dL (. nmol/L), but not of low BMD or fracture
with intramuscular injection of T esters [effect size, history. A total of  men with a mean age of .
. (. to .)] corresponding to an % (% to years, mean BMI of . kg/m, and normal aBMD
%) aBMD increase, but it was not significant for mean T-scores (. at the lumbar spine, . at the total
three trials using transdermal treatment with patches hip, 2. at the femoral neck) were randomized to
[effect size, 2. (2. to .) ().  months of transdermal T treatment with adjusted
Three studies in this meta-analysis included older doses of T gel aiming at serum T in the normal range
men without secondary causes of osteoporosis, who for young men or with placebo. Compared with
are more representative of community-dwelling aging placebo, T treatment resulted in an increase of spine
men. Amory et al. () reported that T therapy in- trabecular bone vBMD by quantitative CT (QCT;
creased aBMD at the spine and total hip, but not at the treatment effect, .% (.% to .%); P , .] and
femoral neck, in men (mean age,  years) treated estimated strength by finite element analysis of QCT
 months with intramuscular injections of  mg of data [treatment effect, .% (.% to .%); P ,
T enanthate or placebo every  weeks. They also .]; increases of vBMD were significantly associated
showed that the T treatment effect was not altered by with the increases in serum T and E during treatment.
cotreatment with the a-reductase inhibitor finas- Compared with the effects on spine trabecular bone,
teride; the treatment effect was independent of increases of vBMD were slightly smaller and increases
baseline hormone levels but positively related to the of estimated strength were similar for spine peripheral
magnitude of the increase of both T and E serum bone and whole bone. Highly significant positive
levels during treatment. Kenny et al. () reported for treatment effects are also seen on vBMD and estimated
men (mean age,  years) treated for  year daily with strength at the hip trabecular bone, peripheral bone,
 mg of transdermal T or placebo a marginal beneficial and whole bone, although of clearly smaller amplitude
effect on femoral neck aBMD (.% increase from than at the spine. It is well known that vBMD by QCT
baseline compared with .% decrease under placebo; is more sensitive to treatment-induced bone changes
P 5 .), but no significant treatment effect for than aBMD by DXA. Accordingly, in sharp contrast
either hip trochanter, lumbar spine, or whole-body with the QCT data, there was an only small positive
BMD. Snyder et al. () observed in men (mean age, treatment effect for lumbar spine aBMD [.% (. to
 years) treated for  months daily with  mg of .); P 5 .] and no significant treatment effect at
transdermal T or placebo by scrotal patches no the femoral neck [.% (2. to .); P 5 .] and
treatment effect on aBMD at either the lumbar spine total hip [.% (2. to .); P 5 .] (). These
or the hip. A post hoc analysis revealed a negative findings for aBMD are in line with the earlier DXA-
interaction between baseline T levels and T treatment based studies and indicate that DXA underestimates
effect, with lumbar spine BMD increases above pla- the effects of T treatment on the bone. Therefore, the
cebo mainly restricted to the men with initial serum robustness of the reported bone effects of T treatment
T # ng/dL (. nmol/L). in the Bone Trial compared with earlier trials is due
Reported effects of T treatment on levels of bio- primarily to the use of QCT-based endpoints. How
chemical markers of bone turnover have been in- much the inclusion of men who are clearly hypo-
consistent and essentially limited to decreases in the gonadal in this study may have contributed to the
markers of bone resorption observed in only some robustness of the QCT data cannot be concluded.

944 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

In summary, there is a striking knowledge gap as to In an experimental setting in healthy older men (
the effects of T therapy for skeletal health in aging to  years of age) with manipulation of sex steroid
men: (i) there is no reliable data on T treatment in levels for a duration of  weeks, there were no dif-
older men with osteoporosis; (ii) except for a couple of ferences in cognitive function (memory, executive
studies, trials did not include older men with un- function, spatial cognition) when comparing men with
equivocally low serum T; and (iii) there are no data at induced hypogonadal T levels (GnRH agonist), men
all on the effect of T therapy on fracture risk, the most with T and E levels in the eugonadal range (GnRH
relevant clinical evaluation criterion. The data avail- agonist plus T-gel), men with T levels in the eugonadal
able suggest that T therapy in older men has a modest range and low E (GnRH agonist plus T-gel plus
suppressive effect on bone resorption, produces a aromatase inhibitor), or all placebo-treated men.
limited increase of aBMD at the lumbar spine, and These are negative findings, even though free T was
inconsistent, marginal, and overall nonsignificant in- positively related to spatial cognition and E was
creases of aBMD at the hip as assessed by DXA. negatively associated with working memory in post hoc
Treatment with T produces more substantial increases analyses of the combined treatment groups ().

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


of vBMD at the spine and significant increases of Emmelot-Vonk et al. () randomized healthy men
vBMD at the hip as assessed by QCT. Evidence from aged  to  years (mean,  years) with low to low-
several trials indicates that the increase of BMD during normal T (, ng/dL or . nmol/L) to oral
T therapy is related to the magnitude of the increase in treatment with  mg of T undecanoate twice daily for
serum T and E, which might explain why in older  months [n 5 ; baseline mean T,  ng/dL
studies significant aBMD increases were observed only ( nmol/L)] or matching placebo [n 5 ; baseline
in studies with T administration by intramuscular mean T,  ng/dL (. nmol/L)]. Although T
injections and not in studies with transdermal T ad- treatment resulted in an increase of lean body mass
ministration with patches. Finally, although there is and a decrease of fat mass, there was no treatment
some low-quality evidence suggesting a greater BMD effect on the results of a battery of eight memory and
response to T treatment in men with initially low cognition tests. However, interpretation is limited by
serum T, the limited data available do not allow the fact that it is not documented whether the oral T
conclusions on this point. administration substantially increased mean T levels.
In conclusion, although modest favorable effects on Huang et al. () in the TEAAM study randomized
“Osteoporosis is neither a
the maintenance of skeletal integrity may be an added men $ years of age with low to low-normal serum T specific nor sufficient
benefit of T treatment initiated for another indication in [ to  ng/dL (. to . nmol/L) or free indication for T therapy.”
older men with low serum T, there are presently clearly T , ng/dL ( pmol/L)] to treatment with . g of
insufficient data to support its use to treat osteoporosis. % gel daily or placebo. There was no effect of
Osteoporosis is neither a specific nor sufficient in- treatment of eight memory and cognition tests in
dication for T therapy. Specific osteoporosis treatment T-treated men [n 5 ; mean baseline T,  ng/dL
(e.g., with a bisphosphonate) should be initiated in (. nmol/L); mean follow-up,  months] compared
hypogonadal older men at high risk of fracture, re- with placebo-treated men [n 5 ; mean baseline T,
gardless of whether they are also treated with T to  ng/d/L (. nmol/L); mean follow-up,  months].
alleviate other symptoms of hypogonadism (, ). In the Cognition Function Trial, which is part of the
TTrials, Resnick et al. () assessed the effects on
Mental health cognition of treatment with T gel in men with low
Cognition. Studies on the effects of T therapy serum T (, ng/dL or . nmol/L) and impaired
on cognition in aging men have been heterogeneous sexual function, physical function, or vitality and with
in key aspects such as cognitive domains and as- age-associated memory impairment. From  men
sessment tools and baseline cognitive status and T included in the TTrials, a subgroup of  men [mean
levels. Not surprisingly, findings have been variable. age, . years; mean baseline T,  ng/dL (. nmol/
Although earlier studies suggested that T therapy L)] met criteria for age-associated memory impairment
might enhance cognitive function in older men based on baseline subjective memory complaints and
with low serum T, most of these studies included objective memory performance. There was no signifi-
only small number of subjects, and improvements cant mean change from baseline to  and  months in
were variable depending on cognitive domain and men treated with T (n 5 ) and placebo (n 5 ) for
methodology [see (, ) for reviews]. Similarly, scores for delayed paragraph recall, visual memory,
earlier studies reporting that T treatment might slow executive function, or spatial ability.
cognitive decline in men with mild cognitive im- Thus, the overall findings from randomized,
pairment or Alzheimer’s disease are also small in controlled studies do not indicate that T treatment has
scale and they included almost exclusively eugonadal significant beneficial (or adverse) effects on memory or
men (). Findings from more recent and lager other aspects of cognition in elderly men with low or
scale studies on the effects of T therapy on cognition low-to-normal serum T, regardless of whether they
in older men have been negative. have preexistent memory impairment.

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 945


REVIEW

Depression, mood, and well-being. Earlier Safety of T treatment


studies, although reporting variable results, provided The assessment of the risk/benefit balance of T therapy
some evidence that therapy can improve indices of in older men with low serum T and thus also eval-
mood in men with low to low-normal serum T (). uation of the desirability to initiate such treatment in
Nevertheless, in an earlier larger placebo-controlled an older individual is hampered by a knowledge gap as
study including  men with a median age of  to its long-term safety. Indeed, none of the performed
years (% $ years of age) and mean baseline RCTs in older men or in men in general had the
serum T ~ ng/dL ( nmo/L), T treatment (three T required power and duration to reliably establish
treatment groups, that is, two different doses of T-gel long-term CV safety and cancer risk of T therapy.
and a T patch) had no effect above placebo gel on Alternatively, findings from observational studies, in
categorical mood scores (), and in the above- particular those pertaining to CV safety, have gener-
discussed study by Emmelot-Vonk et al. () there ated controversy. Besides dosage and duration of T
was no treatment effect of oral T undecanoate on the therapy, the formulation and route of T administra-
SF-  questionnaire scores, including the scores tion might be relevant to safety aspects because of

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


related to mental health, emotional role, and social formulation-specific pharmacokinetics with marked
functioning, and no effect on the scores for the differences between formulations in the achieved
Questions on Life Satisfaction Modules. The results profiles of serum T levels as well as differences in the
of randomized placebo-controlled trials addressing production of the bioactive metabolites DHT and E.
the effects of T therapy in men with depression were Finally, RCTs on T therapy in older men have gen-
also variable (), with the only larger trial () erally excluded men at higher risk of adverse events so
reporting negative findings. A meta-analysis of  that the safety data do not apply to such patients.
controlled clinical trials using common depression Commonly applied exclusion criteria have been
rating scales, which included a total of  men, prostate abnormalities or estimated increased risk for
showed a significant positive impact of T therapy on prostate cancer, moderately severe lower urinary tract
mood. However, in subgroup analyses the effect size symptoms, (borderline) high Hct or Hb, recent CV
was larger in subclinical depression compared with events such as stroke and MI, markedly high blood
major depression and was not statistically significant pressure, and severe obstructive sleep apnea (OSA).
in eugonadal men and in men . years of age
(). Erythropoiesis
The Vitality trial, part of the TTrials, included Given the hematopoietic effects of androgen exposure,
men $ years of age with low T (, ng/dL or development of polycythemia (Hct .% to %;
. nmol/L) and self-reported low vitality and a score Hb .. to . g/dL) during T therapy is a concern.
of , on the Functional Assessment of Chronic Moreover, older men appear to be at higher risk. In an
Illness Therapy (FACIT)–Fatigue scale (range  to experimental setting in men with suppressed gonad-
; higher score indicates less fatigue). At  and otropins exposed to graded doses of T, increases in Hb
 months of treatment there was a significant dif- and Hct were dose-related and greater in older men
ference in favor of T treatment compared with ( to  years of age) than in younger men ( to 
placebo for the SF- vitality score, the Positive and years of age), also after adjusting for serum T or free T
Negative Affect Schedule scores, and the Patient Health levels (age effect P # .) (, ).
Questionnaire- depression score. However, the treat- Meta-analyses of interventional studies found that
ment effect was small, with all effect sizes ,.. The middle-aged and older men on T therapy had higher
primary efficacy criterion (i.e., $ point increase on the odds than did men on placebo to develop poly-
FACIT-Fatigue score) was not reached. When analyzing cythemia (in the literature interchangeably also re-
the FACIT-Fatigue score as a continuous variable, a ferred to as erythrocytosis) (, ), with an overall
positive treatment effect was present, which became incidence of .% among actively treated participants
significant only when considering the whole TTrials (). Similarly, other studies in elderly men report
study population (). polycythemia in % to % of participants (, ,
The whole of the available information suggests , –), although incidences up to % have
that T therapy can improve mood, depressive symp- been reported (). In a recent meta-analysis of RCTs
toms, and vitality in older men with low serum T. in middle-aged and older men with low serum T
However, these effects appear of small amplitude, and (, ng/dL or . nmol/L) and at least one or more
this conclusion is driven primarily by the recent symptom or sign of hypogonadism, the RR of poly-
findings from the TTrials. There is no convincing cythemia in T-treated men compared with placebo
documentation of an effect of T on mood or de- was . (% CI, . to .) (). In some studies,
pressive symptoms in older men with initially no low supraphysiologic T levels were found at the time of
serum T. There is also no convincing evidence to polycythemia (). The risk for polycythemia in
support the use of T therapy to help treat depression hypogonadal men under T therapy has been associated
in older men. with androgen sensitivity (as per CAG repeat length of

946 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

the AR gene) and with through levels of T between numerically higher in the subjects treated with injected
repeated injections of long-acting depot T preparation or transdermal T compared with placebo, but overall
(, ). Furthermore, polycythemia appears to there were only  prostate cancer events in both the
occur more frequently in men treated with (mainly short-term and long-term RCTs. In their meta-
short-acting) injectable T as compared with oral or analysis, Boyle et al. () found that in  RCTs
transdermal T (, ). there was no change in PSA (effect size, . ng/mL;
Taken together, clinically meaningful polycythemia % CI, 2. to .). For  trials reporting on
is a relatively frequent adverse effect of T therapy in prostate cancer cases, the summary OR for prostate
older men and carries a risk of severe complications cancer was . (. to .), which was based on 
(, ). This makes periodic screening of Hb or Hct prostate cancer cases. Kohn et al. () focused on
levels during treatment warranted, especially in men lower urinary tract symptoms in a meta-analysis of 
with other risk factors such as smoking, chronic RCTs of T treatment of LOH involving  men
pulmonary disease, or apnea syndrome. with a mean age of . years and an average follow-up
of . months. They found no significant difference in

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Prostate and lower urinary tract symptoms pooled IPSS change from baseline to follow-up between
Considering the important role of T in normal growth men treated with T [-. points (2. to .)] or with
and maintenance of the prostate gland and of AR placebo [. points (2. to .)], suggesting that T
signaling in stimulation of metastatic prostate cancer treatment in aging men with hypogonadism does not
growth, the potential for adverse effects of T therapy worsen lower urinary tract symptoms.
on prostate health has been a concern. Several meta- In the recent TTrials,  out of  T-treated men
analyses of RCTs have addressed this concern (, compared with  out of  placebo-treated men
, , ). Although their findings may seem increased their PSA levels by $ ng/mL. From the 
rather reassuring, they also show that the low number men with PSA above the trigger value during the year
of men enrolled in RCTs of longer duration and the of treatment or the year after treatment and referred
very low number of observed events preclude de- for urologic evaluation, only  had prostate biopsies; 
finitive conclusions as to longer-term prostate cancer man in the T group had a diagnosis of prostate cancer
risk. Calof et al. () performed a meta-analysis of  during the year of treatment, and  more men in the T
earlier RCTs of  months’ median duration ( days to group and  man in the placebo group had prostate “…clinically meaningful
 years) enrolling  T-treated and  placebo- cancer diagnosed in the year following treatment. In polycythemia is a relatively
treated men and found increased odds of the com- the TTrials the number of men with increased frequent adverse effect of T
bined occurrence of prostate adverse events (OR, .; IPSS . was not different between T-treated men therapy in older men….”
% CI, . to .), including a PSA . ng/mL or (n 5 ) and placebo-treated men (n 5 ) ().
. ng/mL increase during the study, an increase in Considering the modest number of enrolled
International Prostate Symptom Score (IPSS) by . subjects, the short duration of T exposure, and the
points, acute urinary retention, prostate biopsies, and small number of events, neither the individual RCTs
prostate cancer. For none of the individual events was reported as to date nor their meta-analyses provide
there a significant difference between T and placebo; reliable information on the longer-term prostate
prostate biopsies and PSA . ng/dL were numeri- cancer risk. Observational studies, which suffer from
cally higher in the T group. The meta-analysis by known limitations of high risk of bias and con-
Fernández-Balsells et al. () included low- to founding, nevertheless provide the only available
medium-quality randomized and nonrandomized information on longer T exposure in substantial
comparative trials (T vs placebo or no T) of at least numbers of men with hypogonadism. In a combined
 months’ study duration enrolling men with low or analysis of three parallel, prospective cumulative
low-normal baseline T. They found no significant registry studies including a total of  hypogonadal
differences between groups for individual prostate men treated with T undecanoate injections ( mg
outcomes or the composite outcome; prostate biopsies every  weeks) for a median duration of  years,
were numerically higher in the T-treated men (RR, prostate cancer was diagnosed in  patients: in the
.; % CI, . to .). Cui et al. () first cohort with a mean age of . years there were 
performed a meta-analysis of  RCTs of T treatment cases in  men (.%) with an incidence rate of .
involving a total of  men with hypogonadism, of per , patient-years; in the second cohort with a
which  were of duration shorter than  months and mean age of . years, there were  cases in  men
 were of  months to  years’ duration. Short-term (.%) with an incidence rate of . per ,
T treatment was more likely than placebo treatment to patients-years, whereas in the third cohort with a mean
increase PSA levels (P , .). For none of the age of  years, there were no cases in  men. It
formulations (injectable, transdermal, or oral) was was concluded that the incidence rate of prostate
there a significant increased risk for prostate biopsy, cancer in these T-treated men was not higher (and
prostate nodule, or prostate cancer. In the studies of even somewhat lower) than expected according to
longer duration, prostate biopsy and cancer were data from population screening studies (). A

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 947


REVIEW

multinational prospective registry study reported on ( mg/d; % gel), which was prematurely inter-
up to  months’ follow-up of  newly diagnosed rupted because of the higher rate of CV-related
hypogonadal men, of which  received T therapy adverse events in the T-treated men ( in 
and  remained untreated (mean age, . years;  men) compared with placebo-treated men ( in 
treated and  untreated men $ years of age). men). The reported events were diverse in nature
There were in total  cases of prostate cancer–positive and pathophysiology.
biopsies. Overall, the proportion of biopsies that were In their meta-analysis of randomized and non-
positive for cancer, the primary outcome of the study, randomized comparative intervention studies (T vs
did not differ between T-treated subjects (.%) and placebo/nonintervention), Fernández-Balsells et al.
untreated men (.%), and the prostate cancer rate in () found no significant differences in rates of death,
the T-treated men was not higher than in untreated MI, or revascularization procedures. In a meta-analysis
men or than expected in the general population. by Ruige et al. () of  double-blind RCTs of T vs
Treatment was associated with a limited PSA increase placebo, which included . men and reported on
during the first  months, which stabilized thereafter CV events, there was no significant difference in event

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


and with no increase in IPSS. The authors concluded rate between groups (estimated RR, .; % CI, .
that there was no evidence of increased prostate cancer to .). The power was low with only  highly di-
rate or progression of lower urinary tract symptoms in verse events. Xu et al. () performed a meta-analysis of
newly diagnosed hypogonadal men treated with T placebo-controlled RCTs of T therapy of at least
(). Note that in these observational studies a  weeks’ duration and reporting CV-related events.
substantial proportion of men were , years of age, The  selected trials included  mainly middle-
and the number of events was low. Moreover, in the aged and older men with low T and/or chronic dis-
latter registry the duration of observation is rather eases, who experienced a total of  CV-related
short and the reasons for no treatment vs T treatment events, defined as any event reported as such by the
are unknown (). authors or where the description fell within a corre-
In summary, overall the evidence available in- sponding category of the International Statistical
dicates that T treatment in aging men with low serum Classification of Diseases. The authors found that T
T may result in a small increase of PSA level occurring treatment increased the risk of CV-related events in a
within the first year of treatment. Treatment with T fixed effects model (OR, .; % CI, . to .)
does not worsen lower urinary tract symptoms as with a similar finding when the analysis was restricted
assessed with the IPSS. This applies only to men with to serious events (OR, .; % CI, . to .) and
an initial IPSS ,, as men with more severe urinary similar odds for CV-related death (OR, .; % CI,
tract symptoms have usually been excluded from . to .). The authors further reported that the risk
RCTs. The available data from RCTs and observa- of a CV event varies by the source of funding (P for
tional studies do not reveal any alarming signal as to interaction , .), with increased risk in  trials
prostate cancer risk and appear reassuring as to the ( men included) not funded by the pharmaceu-
short-term prostate safety of T treatment in aging men. tical industry (OR, .; % CI, . to .) but not
However, a critical appraisal of the data shows that in  trials ( men included) sponsored by industry
somewhat stronger evidence is in fact available for not (OR, .; % CI, . to .). The validity of the
much more than the first year of treatment. Owing to findings of this meta-analysis has been challenged by
the generally short duration of RCTs and their low some on the basis that as much as one third of the
power (i.e., a small to modest number of enrolled events in T-treated men in the  trials not sponsored
subjects and a small number of events) and to by industry are contributed by only  trials, whereas
the methodological and other limitations of obser- for the first of these  trials () a substantial proportion
vational studies, the longer-term risk of prostate cancer of events were not “classical” major adverse CV events,
associated with T treatment in older men remains and the second trial () pertains to treatment of men
unknown. with cirrhosis of the liver with very high doses of
an unapproved oral formulation of micronized T
Cardiovascular and venous thromboembolic events resulting in markedly high T levels and for which the
The CV effects of T in aging men and in particular CV events taken into account possibly included
the effects of exogenously administered T are the (esophageal) bleedings (). In a recent systematic
subject of ongoing debates, fueled by inconsistent review and meta-analysis of the CV risk of exogenous
findings from observational studies and by the lack T, Alexander et al. () included  RCT and 
of adequately powered RCTs with clinical CV events observational studies. They retained  placebo-
as the primary outcome (). Scrutiny of the CV controlled RCTs for meta-analysis, with death from
safety of T treatment has been intensified after all causes, MI, and stroke as primary outcomes, and
publication of the report () on an RCT in older men other CV endpoints such as arrhythmias, heart failure,
(mean age,  years) with limited mobility and low T coronary angiography, pulmonary embolism, or ve-
treated with a relatively high dose of transdermal T nous thrombosis as secondary endpoints. Compared

948 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

with placebo, T treatment did not show any significant within an integrated health care delivery system,
increase in the risk of MI ( RCTs; OR, .; % CI, comparing the hypogonadal men ever dispensed a T
. to .), stroke ( RCTs; OR, .; % CI, . to prescription (.%; n 5 ; mean age, . years;
.), or mortality ( RCTs; OR, .; % CI, . to .% prior CV disease; .-year median follow-up)
.). There was no significant increased risk for the with those never dispensed T (.%; n 5 ,;
composite outcome of MI, stroke, and death ( RCTs; mean age, . years; .% prior CV disease; .-year
OR, .; % CI, . to .). The strength of the median follow-up); % of the men were . years
evidence for the different outcomes was determined as of age. The primary outcome was a composite
low. CV endpoint including acute MI, coronary re-
In the TTrials in older men with low T, the oc- vascularization, unstable angina, stroke, transient is-
currence of CV events was similar in T-treated and chemic attack, and sudden cardiac death. The rates of
placebo-treated men with in both groups  out of  the composite CV endpoint were lower in the men
men with a major CV event during the -month ever dispensed T than in those never dispensed T (.
study; in both groups five out of the seven major CV and . per  person-years, respectively); the

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


events were stroke (). In a substudy of the TTrials, adjusted HR for the composite endpoint in the men
during the -year duration of the study, an increase in ever dispensed T was . (% CI, . to .). The
coronary artery noncalcified plaque volume, as mea- findings were similar for endpoints restricted to stroke
sured by coronary CT angiography, was significantly and transient ischemic attack (HR, .; % CI, .
greater (P 5 .) in T-treated men (n 5 ) to .) or restricted to the combined cardiac events
compared with placebo-treated men (n 5 ); there (HR, .; % CI, . to .). It is concluded that
was also an increase in total plaque volume (P 5 among men with androgen deficiency, dispensed T
.) but not in coronary artery calcification score prescriptions were associated with a lower risk of CV
(). In the TEAAM study men $ years of age outcomes during a median duration of . years ().
(mean, . years of age) with low to low-normal T Wallis et al. () reported a more nuanced picture in
( to  ng/dL or . to . nmol/L) were ran- their intention-to-treat observational cohort study.
domized to  mg of transdermal T (% gel) or placebo This was a population-based study using different
for  months. Between T-treated (n 5 ) and Canadian health care–related databases, comparing “…findings from RCTs and
placebo-treated (n 5 ) men there was no difference men $ years of age newly treated with T (n 5 observational studies do not
in the rate of change during  years in common carotid ,; .-year median follow-up) and controls allow us to draw conclusions
artery IMT (P 5 .) or in coronary calcium score matched for region of residence, comorbidity, diabetes as to the CV safety of T
(P 5 .), the primary outcomes of this study. Al- status, and index year (n 5 ,; .-year median treatment in aging men with
low T….”
though the authors reported that the small number of follow-up). Men treated with T had a lower mortality
unadjudicated CV events and major adverse CV than did controls (HR, .; % CI, . to .).
events did not differ between groups, the overall rate of However, when looking at cumulative exposure ef-
MI, coronary revascularization, stroke and death of fects, men in the lowest tertile of T exposure duration
CV event was nevertheless numerically higher in the (median of  months) had an increased risk of
T-treated men (n 5 ) compared with the placebo mortality (HR, .; % CI, . to .) and com-
group (n 5 ) (). posite CV events (HR, .; % CI, . to .),
Observational studies on CV risk and mortality of whereas in contrast the men in the highest tertile of T
exogenous T treatment as reviewed by Alexander et al. exposure had a lower risk of mortality (HR, .; %
() are characterized by substantial clinical and CI, . to .) and composite CV events (HR, .;
methodological heterogeneity, and the evidence was % CI, . to .) with a significant trend across
rated as of very low quality owing to the high risk of tertiles (P , .). A limitation of this latter ob-
bias, imprecision, and inconsistency. The findings have servational study compared with the former is the
been disparate, with some studies reporting that T absence of information on serum T with controls
treatment is associated with increased risk of matched for CV risk factors but not for hypogonadal
CV events, including MI, stroke, and/or mortality status. Therefore, the higher mortality and CV events
(–), others reporting no effect on the risk of MI in the shorter duration exposure might represent an
() and mortality (), and still others reporting effect of either treatment and/or of the (causes for the)
reduced risks in (subgroups of) T-treated men (, hypogonadal status itself. For both studies it is not
, ). A detailed discussion of their respective possible to exclude that the apparent longer-term
merits and limitations falls, however, beyond the scope favorable effects of T treatment on CV risk and
of this review. mortality represent at least in part a healthy user effect
Recent observational studies deserving mention and residual confounding. In a retrospective cohort
have once more delivered mixed messages. Cheetham study of older men identified with low T, Sharma et al.
et al. () reported on a retrospective cohort study of () reported that those treated with T and nor-
men with hypogonadism [T , ng/dL (. nmol/ malizing their T level (n 5 ,; median age, 
L) or coded diagnosis of hypogonadism] conducted years) had a lower risk of atrial fibrillation compared

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 949


REVIEW

with men treated with T without normalization of T with transdermal T was not significant, the point
(n 5 ,; median age,  years) (HR, .; % CI, estimate was higher than for T injections. This led the
. to .) and men who did not receive T therapy authors to postulate a detrimental effect resulting from
(n 5 ,; median age,  years) (HR, .; % CI, higher DHT serum levels under oral (and transdermal)
. to .), suggesting that normalization of T in treatment.
older men with low T is associated with a lower risk of In summary, the findings from RCTs and obser-
atrial fibrillation. vational studies do not allow us to draw conclusions as
Martinez et al. () performed a population-based to the CV safety of T treatment in aging men with low
case-control study involving , patients with T because the data are insufficient and inconsistent.
confirmed venous thromboembolism, comprising The data from RCTs and meta-analyses of RCTs
deep venous thrombosis and pulmonary embolism, might seem predominantly reassuring as to the ab-
and , age-matched controls and reported an sence of risk of MI and other major cardiac events,
increased risk of venous thromboembolism in the first stroke, and mortality, but there are also discordant
 months of T treatment (rate ratio, .; % CI, . findings. Moreover, the data on CV safety from RCTs

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


to .); after . months of therapy the rate ratio suffer from major limitations. They are mostly not
declines to . (% CI, . to .) and after ces- designed as studies with hard CV endpoints as primary
sation of treatment to . (% CI, . to .). or secondary outcomes, they have enrolled too few
Baillargeon et al. () performed a cohort study subjects and most were of short duration, and the
involving  cases of venous thromboembolism number of observed events was too low so that the
(% $ years) and , matched controls. Having power of RCTs and their meta-analysis was in-
filled a prescription for T therapy in the  days sufficient to make conclusions. Observational studies,
preceding the event was not associated with an in- both earlier and recent large observational cohort and
creased risk of venous thromboembolism:  cases case-control studies, have delivered even more con-
(.%) vs  controls (.%) with filled prescription flicting results, which is not surprising in view of the
in the  days preceding the corresponding index intrinsic limitations of observational data and their
dates; adjusted OR of . (% CI, . to .) with methodological heterogeneity. As to the risk of venous
similar results when the window of prescription is thromboembolism (deep venous thrombosis and
extended to  or  days preceding the event. None of pulmonary embolism), the more limited data have also
the examined specific routes of T administration was been conflicting, precluding definitive conclusions.
associated with an increased risk of venous throm- In conclusion, the CV risk profile and the risk of
boembolism. In their cohort study of men with low T, venous thromboembolism associated with T therapy
Sharma et al. () found no increased risk of venous in older men with low serum T remain to be further
thromboembolism in men with normalized serum T elucidated. This requires data from adequately pow-
under T treatment ( cases in , men) com- ered RCTs specifically designed to this end.
pared with untreated hypogonadal men ( cases in
, men) with an adjusted HR of . (% CI, . Sleep and OSA
to .) or compared with T-treated men with still low Men suffering from OSA not uncommonly have a low
T ( cases in , men) with an adjusted HR of . serum T, in particular when they are also obese. Serum
(% CI, . to .). T levels in men with OSA tend to be inversely related
As to possible differences in CV risk associated to the severity of hypoxemia. Severe (untreated) OSA
with T therapy among different modalities of T ad- is traditionally listed among contraindications for
ministration, available information is limited and initiation of T therapy (, ), which for a long time
inconsistent. Layton et al. () in a retrospective was based on no more than a few case reports of
cohort analysis of , men having initiated T worsening of OSA under T therapy. Liu et al. (),
therapy compared men treated with T injections, in a randomized sequence, double-blind, placebo-
T-gels, or T patches. They found that compared with controlled, crossover study in healthy, community-
treatment with T-gel, treatment by T injections was dwelling men . years of age (mean age, . years of
associated with a greater risk of death, hospitalization, age), showed by overnight polysomnography that
and CV events, but not venous thromboembolism, short-term injection of markedly supraphysiological
whereas the risk profile was similar for gels and doses of T resulted in reduced total time slept by about
patches. In contrast, in the observational study by  hour, increased duration of hypoxemia, and dis-
Cheetham et al. () there was a significant, albeit turbed breathing during sleep. Hoyos et al. ()
small, higher CV risk associated with topical T performed an -week placebo-controlled trial in men
treatment. Further blurring the picture, Borst et al. with obesity (BMI . kg/m) with OSA. All men
() from their meta-analysis of RCTs concluded that were put on a hypocaloric diet and were randomized
oral T treatment was associated with a significant CV to T injections ( mg T undecanoate at week , ,
risk, which was not the case for injected and trans- and ) (n 5 ; mean age,  years) or placebo
dermal T administration; although CV risk associated injections (n 5 ; mean age,  years). Therapy with

950 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

T compared with placebo worsened the oxygen osteoporosis treatments (see “Bone health” above).
desaturation index and nocturnal hypoxemia at the The findings from RCTs do not indicate that T therapy
first evaluation at week , but no longer at the final has a significant beneficial effect on cognition in older
evaluation at week  when the subjects had also lost men with low T, regardless of whether they have
weight. This effect was independent of baseline T. preexisting cognitive impairment (see “Cognition”
Overall, the limited data available thus tend to under “Mental health” above). Available data do in-
confirm that caution is to be recommended as to the dicate that T therapy in elderly men with low T can
initiation of T therapy in men with (untreated) severe improve mood, vitality, and depressive symptoms
OSA. with, however, only a small effect size. Such an effect
has not been shown in older men without low T, and T
Summary of risks/benefits and indications for therapy has not been shown to help treat depression
T treatment (see “Depression, mood, and well-being” under
As discussed in prior sections of this review, low “Mental health” above). Finally, T therapy can increase
sexual desire and function are probably the symptoms Hb in older men with low T and otherwise un-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


most consistently associated with low serum T in explained anemia ().
elderly men (see “Sexual function” and “Diagnosis of Thus, in terms of clinically relevant endpoints, the
Hypogonadism in Aging Men” above). In RCTs, global picture from available RCTs on the efficacy of T
sexual desire and function are also the clinically rel- therapy in older men with low T appears rather bleak.
evant endpoints most consistently improved by T As far as efficacy is concerned, amelioration of sexual
therapy in elderly men with low T. Nevertheless, the function in older men with low T and sexual symp-
observed improvements by T therapy are mostly of toms may be the only relatively well-defined clinical
modest amplitude, and the effect on ED, in particular, indication justifying consideration of the initiation of
appears less than that reported for the use of PDE T therapy, even though treatment effects are of
inhibitors and has not been convincingly documented moderate amplitude and of uncertain longer-term
to be additive to that of PDE inhibitors in combined durability. Because of the difficulties in defining
therapy (see “Sexual function” above). At the dosages low vitality and mood, and categorizing depressive
used in RCTs, T therapy in aging men improves body symptoms in clinical practice, and considering the only
composition with consistent but quantitatively small small effect size of T therapy, these conditions as such “…caution is to be
decreases of fat mass and increases in lean mass and are probably not sufficient to justify T treatment. recommended as to the
skeletal muscle mass. The latter translates variably in Nevertheless, when present in the former context of initiation of T therapy in men
either no gain or only limited gain in muscle strength sexual complaints in elderly men with low T, they may with (untreated) severe OSA.”
and power and thus, not surprisingly, in an overall be seen as strengthening the indication for T treat-
disappointing impact on physical function, with RCTs ment, as does the presence of otherwise unexplained
reporting small positive effects of uncertain clinical anemia. In the present state of the art, the effects on
significance or negative findings. Clearly, the data body composition, physical function, insulin sensi-
available as to date do not allow considering im- tivity, bone turnover, and BMD do not as such
provement of physical function, sarcopenia, or frailty constitute indications for T therapy and should
as a valid indication for T therapy in older men with or rather be regarded as fringe beneficial effects of a T
without low serum T (see “Body composition, muscle therapy initiated for another indication such as sexual
mass and strength, and physical performance” above). dysfunction.
The beneficial effects of T therapy on body compo- As to the risks linked to T therapy in aging men,
sition are consistently associated with small reductions clinically meaningful polycythemia is sufficiently
in fasting blood glucose and modest improvement in common to require regular monitoring of Hct and Hb
insulin sensitivity. However, in the results of RCTs this (see “Erythropoiesis” above). Treatment does not
again fails to translate into consistent and clinically worsen lower urinary tract symptoms, but this has not
relevant improvements of metabolic status. Therefore, been studied in men with initially severe symptoms.
to improve the metabolic state in older men with low The data appear reassuring as to the short-term (up to
or low-normal T is not a valid indication for T therapy,  year of T therapy) prostate safety, but longer-term
neither in general nor more specifically in men with safety has not been established (see “Prostate and lower
the MetS or TDM (see “Body composition and urinary tract symptoms” above). Even though the data
metabolic profile” above). Whereas T therapy in el- from RCTs may seem rather reassuring as to major CV
derly men tends to reduce bone turnover and results in events (MI, stroke, CV death), these data suffer from
small increases in aBMD as assessed by DXA and significant methodological limitations in this context
somewhat larger beneficial effects on vBMD by QCT, and there are also discordant findings, and the findings
low bone mass and osteoporosis are not valid in- from observational studies have been conflicting and
dications for T therapy in elderly men. Indeed, there is inconclusive. Whereas the longer-term safety is a
no data on the ability of T therapy to reduce fracture concern because of lack of controlled data, the risk of
risk or data comparing the effects of T with established thromboembolic events (DVT, pulmonary embolism)

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 951


REVIEW

might already be increased early after initiation of statements (, ) explicitly state that age is not a
treatment. From a critical appraisal it can only be contraindication.
concluded that the CV and thromboembolic safety What about older men who fulfill the above re-
profile of T therapy in older men with low T has yet to quirements for T therapy, that is, they have a con-
be fully established. Finally, T therapy may worsen firmed diagnosis of symptomatic hypogonadism, no
preexisting untreated sleep disorders, in particular unaddressed reversible causes of hypogonadism, and
obstructive sleep apnea (see “Sleep and OSA” above). no contraindications for T treatment? Whether they
The global safety profile of T therapy in elderly men can or should be proposed T therapy is an area of less
with low T is thus primarily characterized by un- consensus, but systematic initiation of treatment does
certainty on long-term (CV and prostate) safety not seem an option in view of the unconvincing
rather than by major concerns for short-term ( benefit/risk profile of therapy. On this point, recom-
year) safety. Nevertheless, adverse events such as mendations are not univocal and tend to be ambig-
fluid retention, polycythemia, DVT, and pulmonary uous. The FDA, concerned by the extensive use of T in
embolism can occur within the first months of an attempt to relieve symptoms in men with low T for

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


treatment. no apparent reason other than aging, took a clear and
Taken together, the benefit/risk profile of T therapy restrictive stance in a  Drug Safety Communi-
to aging men with low T is that of an only limited cation. They stated that T is FDA approved as re-
established efficacy in the face of an uncertain (longer- placement therapy only for men with low T due to
term) safety. This current state of the art clearly certain medical conditions, that is, disorders of the
demands caution and restraint in the clinical approach testis, pituitary, or brain that cause hypogonadism, and
to T therapy in elderly men. Whereas this is indeed that the benefits and safety of T have not been
reflected in warnings from health authorities () and established to treat men with low T due to aging, even
recently published clinical practice guidelines (, ), when they have symptoms that seem related to low T
the current widespread use and misuse of T world- (). The FDA required that manufacturers change the
wide, but in particular in the United States (), is labeling of approved T products to clarify the ap-
strikingly at odds with this appraisal. proved uses and further to include information on
Before considering T therapy for hypogonadism in possible increased risk for heart attacks and stroke in
elderly men, the first requirement is a firmly estab- patients taking T. Also in , there was a com-
lished diagnosis of hypogonadism (see “Diagnosis of munication by the European Medicine Agency on
Hypogonadism in Aging Men” above); that is, the T-containing medicines (). The Pharmacovigilance
patient should have unequivocally and persistently low Risk Assessment Committee of the European Medi-
(free) T and symptoms suggestive of hypogonadism. cine Agency conducted a review of T-containing
Although this may seem evident good clinical prac- medicines and concluded that they did not find
tice, it is disquieting that this is apparently the case consistent evidence that the use of T by men with
in only a minority of men prescribed T. Indeed, hypogonadism increases the risk of heart problems.
in many, a single low T was not confirmed by a Nevertheless, the Pharmacovigilance Risk Assessment
second measurement; moreover, many are treated Committee further recommended that the product
notwithstanding a normal serum T, and many are information should make it clear that T should only be
treated even without any prior assessment of serum T used when an abnormally low level of the hormone
(). Second, when there are potentially reversible has been confirmed by signs and symptoms and ap-
causes of hypogonadism involved, such as obesity, propriate laboratory tests. It is further stated that
some comorbidities, and medications, it seems ap- “Testosterone levels naturally fall somewhat with age,
propriate to first pursue resolution or mitigation of the but restoration of these levels in healthy older men is
impact of these causal factors whenever possible before not an authorized use of the medicine in the EU.” In
considering T therapy. Third, treatment should not be the recent update of the Endocrine Society clinical
initiated when there are contraindications. Commonly practice guideline on T therapy in men with hypo-
considered as (absolute or relative) contraindications gonadism (), the authors “suggest” against routinely
are (metastatic) prostate cancer, unevaluated prostate prescribing T therapy to all men $ years of age with
nodule or elevated PSA (. ng/mL or . ng/mL low T. They then pursue with the following non-
in men at higher risk of prostate carcinoma), severe committal recommendation: “in men . yr who
lower urinary tract symptoms (IPSS .) associated have symptoms or conditions suggestive of T de-
with benign prostate hypertrophy, breast cancer, ficiency (such as low libido or unexplained anemia)
polycythemia (Hct .% to %), thrombophilia, and consistently and unequivocally low morning T
recent history of MI or stroke (within the past concentrations, we suggest that clinicians offer T
 months), poorly controlled congestive heart failure, therapy on an individualized basis after explicit
and untreated severe OSA (, , ). Of note, older discussion of the potential risks and benefits.” A
age per se is not listed as a contraindication for ini- consensus statement by several andrological and
tiation of T therapy, and some position and consensus urological associations () formulated the following

952 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

similarly noncommittal summary recommendation: Endogenous levels of DHEA and DHEAS decline
“Age is not a contraindication to initiate testosterone substantially with aging (). As “replacement therapy,”
treatment. Individual assessment of co-morbidities (as DHEA is usually administered orally and it is partly
possible causes of symptoms) and potential risks converted into both T and E. In men, contribution to
versus benefits of testosterone treatment is particularly circulating T levels is at best marginal relative to
important in elderly men.” An Endocrine Society of prevailing T levels and unlikely to be of clinical sig-
Australia position statement on male hypogonadism nificance even in older men with low T, whereas the
() states that, “T replacement therapy is warranted in contribution to E levels is more significant. Whether
men with pathological hypogonadism, regardless of T and E concentrations in target tissues are signifi-
age” and that “Currently, there are limited data from cantly increased by local DHEA metabolism is un-
high-quality randomized controlled trials with clini- known. In a meta-analysis of placebo-controlled RCTs
cally meaningful outcomes to justify testosterone of DHEA supplementation in elderly men, there was
treatment in older men, usually with chronic disease, no significant clinical effect of treatment, except for a
who have low circulating testosterone levels but small decrease of fat mass (). DHT, the bioactive T

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


without hypothalamic, pituitary or testicular disease.” metabolite and major androgen in the prostate tissue,
In conclusion, there is apparent consensus that in is a strong AR agonist. There are scarce data from
men with a pathological hypogonadism, with an RCTs on transdermal therapy with DHT gel in aging
established (organic) testicular, pituitary, or hypo- men with low-to-normal or normal serum T. Treat-
thalamic disorder, age is not a contraindication for T ment results in an expected marked increase in serum
therapy. As to men with hypogonadism related to DHT levels while decreasing serum T levels. The
aging, often with contributions of other factors such as therapy appears to induce effects such as a modest
overweight and comorbidities, drug agencies have decrease in fat mass and an increase in lean mass, as
expressed clear restrictive views, whereas recom- well as some improvement in sexual function, with-
mendations of several scientific associations tend to be out major short-term prostate effects (, –).
more evasive and noncommittal, leaving the evalua- However, clinical data on DHT therapy are presently
tion of the benefit/risk balance with individual pre- too limited. Moreover, treatment is not equivalent to T
scribers and patients. therapy, as DHT is not aromatized to estrogens, which “Several hormonal alternatives
for T therapy in (older) men
may likely adversely affect long-term bone health [see
with hypogonadism have been
Alternatives to T treatment “Bone metabolism and skeletal integrity” as well as the subject of small-scale
In many instances hypogonadism in elderly men is “Bone health” (under “Efficacy of T therapy”) above] studies or are the subject of
potentially at least partly reversible, and there is limited and may also limit other treatment effects compared ongoing drug development
evidence that it can respond to lifestyle measures and with T therapy (). programs.”
optimization of comorbidities (). For several spe- In the absence of major testicular failure indicated
cific clinical signs and symptoms associated with by markedly elevated gonadotropins, serum T in-
hypogonadism, there are established nonhormonal creases in response to hCG injections. Whereas re-
treatments, for example, metformin, diet, exercise for peated hCG injections, whether in association with
insulin resistance and TDM, PDE inhibitors for ED, FSH injections, has a place in the treatment of (usually
and bisphosphonates for osteoporosis. A discussion of younger) men with hypogonadism with a desire for
their place in elderly men with low T falls beyond the fertility, such a treatment has not been studied and
scope of this review. would be impractical for longer-term therapy in older
Several hormonal alternatives for T therapy in men with low T. Moreover, T response to hCG is
(older) men with hypogonadism have been the subject diminished in older men with low T (, ). An
of small-scale studies or are the subject of ongoing alternative to hCG injections is stimulation of en-
drug development programs. However, for most, the dogenous gonadotropin secretion. Circulating E
data available are very limited and their use in this plays a major role in the negative feedback regulation
context is off-label or in clinical trials. As to date, of LH secretion, and in the absence of organic dis-
owing to insufficient data, none can be considered as orders of the gonadal axis, decreasing E hypothalamic
an established or valid alternative for T therapy in negative feedback action results in increased LH and T
aging men. We will therefore limit the discussion to serum levels. This can be achieved either by treatment
some general remarks. with SERMs, having (partial) antagonistic effects on
Potential hormonal alternatives for T therapy can hypothalamic ERs, for example, clomiphene citrate (or
be grouped in three categories: (i) T precursors (e.g., its trans isomer enclomiphene citrate), tamoxifen, and
DHEA) or metabolites (e.g., DHT); (ii) compounds raloxifene (–), or by decreasing circulating E
stimulating endogenous T production, that is, hCG, levels with an aromatase inhibitor, for example,
selective ER modulators (SERMs), and aromatase letrozole and anastrozole (–). These several
inhibitors; and (iii) synthetic compounds acting on drugs have been used off-label as treatment of
the AR, that is, anabolic steroids and selective AR hypogonadism, mostly in younger men with a desire
modulators. for fertility or for treatment of decreased fertility with

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 953


REVIEW

low sperm count (, ), but also in aging men with amount of new data became available, and the vast
obesity with secondary hypogonadism as an alterna- majority (i.e., %) of the work cited herein was
tive to T therapy (, , , –). Data on the published after the  review. Qualitative steps
benefits and risks of therapy with SERMs and aro- forward in the literature have mainly been in three
matase inhibitors in elderly men are presently very areas: substantially more prospective observational
limited, in particular in view of the complexity of their data on large cohorts; more clinical data based on
effects. Indeed, potential beneficial effects and risks of state-of-the-art sex steroid hormone assays; and
therapy with SERMs in elderly men are dependent on considerably more data from RCTs on T therapy in
the rise of T and E levels and the tissue-specific older men with documented low T. Our appraisals of
modulation of E effects. They can vary according to the literature on the clinical correlates of altered sex
the considered target tissue and factors such as sex steroid levels in aging men and on the effects of T
steroid levels both before and during treatment; for therapy are summarized in Table . Notwithstanding
example, the effects of SERMS on bone metabolism the wealth of new data, the conclusions that can be
differ according to pretreatment E levels (, ). drawn from the present review do not differ essentially

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


The effects of aromatase inhibitors, in turn, depend on from those made more than a decade ago. Although
the rise of T levels and the degree of suppression of E we now have improved documentation of the hor-
levels. The latter is usually only partial in men monal changes and their clinical correlates in older
responding to aromatase inhibition with increased T men, important areas of uncertainty persist, in par-
production, and the degree of E suppression may vary ticular concerning the direction and causality of the
according to treatment modalities. This is a relevant relationship between some hormonal and clinical
issue, as suppressed E levels may not only adversely changes, as well as pertaining to the long-term benefits
affect bone health (), but they also may have other and risks of T therapy.
treatment effects such as on fat mass and even on Overall, the data from prospective observational
sexual function (, ). studies confirm and extend prior findings from cross-
As possible alternatives for T therapy, there has sectional studies on reproductive hormone changes.
long been interest in compounds that may prefer- Aging is accompanied by a modest decline of total T,
entially exert wanted androgenic effects, in particular an increase of SHBG, and a more pronounced decrease
anabolic effects on muscle, with less risk of adverse of free T. This occurs with large between-subject
effects, such as on prostate and the CV system. Older variability, and observations in cohorts of community-
steroidal anabolic compounds, which are massively dwelling men have indicated the contribution of
abused in sports and body building, are not candidates comorbidities to these changes, with, in particular,
as alternatives to T therapy in older men with low T increased prevalence of overweight and obesity in
because of both lack of data and a pharmacological aging men being a significant contributing factor. As to
profile not documented to compare favorably with T the clinical implications of these hormonal changes, in
in this context. There are ongoing efforts of the phar- the  review, acknowledging the limitations of the
maceutical industry to develop more selective non- then mostly cross-sectional available data, it was
steroidal selective AR modulators, with a focus primarily concluded that whereas the age-related hormonal
on selective anabolic action on muscle and improved changes are likely to play at least in some men a
physical function (, ), which of course is only one contributory role in part of the clinical alterations that
aspect of health and well-being of aging men. accompany aging, it is also clear that for many signs
and symptoms in elderly men that are reminiscent of
the clinical picture in young men with hypogonadism,
Conclusions and Perspectives the data are inconclusive as to a role of an age-related
partial androgen deficiency. It was further stated that
In this review, we summarized the present state of “Overall, there is presently little if any conclusive
knowledge on the male reproductive system changes evidence for a role of ‘physiological’ age-related decline
observed during aging and their clinical implications. of sex steroid production on morbidity or deteriora-
Although changes affecting fertility are reviewed, the tion of quality of life in elderly men; nevertheless this
main focus is on the hormonal changes, being clini- does not mean that elderly men cannot suffer ‘path-
cally of greater potential significance in older men. ological’ hypogonadism with markedly subnormal
We reviewed possible underlying pathophysiological testosterone.” On the basis of the present updated
mechanisms as well as the clinical correlates of the review of the evidence, these conclusions can be largely
hormonal changes. We discussed androgen deficiency upheld. In fact, in several areas where cross-sectional
and the diagnosis of hypogonadism in older men and associations suggested a role of altered T levels in
where we stand in the appraisal of the risks and aging-related clinical changes, prospective studies ei-
benefits of, as well as indications for, T therapy in older ther did not confirm the association, gave variable
men. Since an extensive review of the topic was results, or suggested a reverse or bidirectional causal
published in this journal in  (), a considerable relationship.

954 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

Among the most consistent associations with low T safety of T therapy, which in any case demands re-
in elderly men, according to the more recent literature, strictive indications for T prescriptions to older men.
are the occurrences of sexual symptoms and lower Hct, In this regard, there are presently no data suggesting
and both seem at least partly responsive to T therapy. that age per se is a contraindication for T therapy in
Longitudinal data have confirmed the association older men with symptomatic hypogonadism due to
between low sex steroid hormone levels and altered hypothalamic, pituitary, or testicular disease.
bone metabolism with increased bone loss and higher There are many areas for further research, and we
fracture risk, but these associations are mainly with mention here only a few. In several observational
lower E and higher SHBG, with a less prominent role studies, free or non–SHBG-bound T levels were found
for T levels. As to the association of altered T levels to be more closely associated with clinical change than
with changes in body composition, metabolic health, was total T. Moreover, associations of SHBG with
and physical performance, the picture emerging from a clinical changes, independent of sex steroid levels, have
large amount of observational data is complex, with also been reported. The role of SHBG as a modulator
indications of bidirectional association and an im- of sex steroid action but also as an integrating marker

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


portant component of reverse causality. In RCTs, T of diverse metabolic and hormonal influences (i.e.,
therapy in elderly men with low-to-normal T con- insulin, adipokines, IGF-, sex steroids, thyroid hor-
sistently resulted in limited decreases of fat mass and mone) in elderly men deserves further exploration.
increases of lean mass, but the effects on metabolic There is also still much work to be done to standardize
health and physical function have been minimal, methodology to assess free T and to validate free T
unlikely to be of clinical significance, and they are reference ranges. Another area deserving further ex-
inconsistent across studies. The data from prospective ploration is the role of bioactive T metabolites, that is,
studies do not consistently show an association of low E and DHT. In particular, the meaning and possible
T in aging men with a decline of cognitive function or mechanisms of reported associations of DHT with
incident AD and, accordingly, findings from RCTs clinical events such as stroke and death need clarifi-
failed to reveal beneficial effects of T therapy on cation. Low T in elderly men has been associated with
cognition in elderly men. The findings on the asso- all-cause mortality, but whether low (free) T can be a “Among the most consistent
ciation of low T with depressive symptoms are mixed clinically useful biomarker of poor health status, re- associations with low T in
and inconclusive. Whereas markedly low T may sponsive to lifestyle measures and optimized man- elderly men, according to the
possibly be associated with the occurrence of de- agement of comorbidity, is an intriguing but yet more recent literature, are
pressive symptoms in elderly men, there is little evi- unexplored possibility. Also deserving exploration is the occurrences of sexual
dence for a role of moderate androgen decline. The the question of whether suppressed activity of the symptoms and lower Hct, and
both seem at least partly
whole of the information from RCTs suggests that T gonadal axis might somehow be a desirable adaptive responsive to T therapy.”
therapy has small-amplitude beneficial effects on phenomenon to ill health that might be undesirable to
mood, depressive symptoms, and vitality in elderly reverse with T therapy.
men with clearly low T. Findings from a large body of The setup of an adequately powered study of
observational data on the association of serum an- sufficient duration to assess long-term (CV and
drogens with CVD in aging men have been variable, prostate) safety of T therapy in aging men is still
most likely due to residual confounding and reverse needed and has been requested by the FDA. However,
causality related to general health status. A critical even if the outcome is reassuring as to the long-term
appraisal of the data does not indicate an association safety of T, this should not signal broad application of
between T levels and the occurrence of IHD or MI. An T replacement therapy in aging men with low T.
association has been found of lower T (and/or DHT) Indeed, as discussed earlier (see the introductory
with the occurrence of stroke and with CV and all- comments under “Treatment of Hypogonadism in
cause mortality. In particular, the relationship between Elderly Men” above), the concept of T replacement is
low T and increased mortality most likely reflects vague and outdated. If T therapy in older men will
reverse causality, with low T in elderly men as a marker acquire a place, beyond the treatment of a small
of poorer health status. The long-term CV effects of T number of older men with symptomatic hypo-
therapy in elderly men are yet to be determined. gonadism due to hypothalamic, pituitary, or testicular
Globally, neither the severity of documented ad- (organic) disorders, it is more likely to be as “phar-
verse consequences linked to low T, nor the nature and macotherapy” for well-defined indications in well-
magnitude of beneficial treatment effects justify the defined patient groups, for example, to improve
concept of a broadly applied T replacement therapy in specific aspects of physical function in older men with
older men with low T. Although there are areas in low T and chronic pulmonary disease or for those
need of further clarification concerning clinical con- recovering from surgery. It is conceivable that for some
sequences of low T and T treatment benefits, it seems indications clinical meaningful effects may require
unlikely that additional data will fundamentally alter supraphysiological T doses, which might be acceptable
this appraisal. This conclusion is, moreover, in- only when a favorable benefit/risk balance is dem-
dependent of the unresolved issue of the long-term onstrated in specifically tailored RCTs with defined

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 955


REVIEW

Table 7. Summary of Appraisal of the Literature on the Clinical Correlates of Altered Endogenous Sex Steroids Levels in Aging Men and of the Literature on
the Effects of T Therapy in Aging Men With Low Serum T
System/Function Associations With Endogenous Sex Steroids Effects of T Therapy

Bone health , (Free or bioavailable) E2 with , Serum markers bone resorption (small effect)

, BMD; . bone turnover; . bone loss; . nonvertebral fractures . Areal vertebral BMD by DXA (small effect size)

. SHBG (independent of T and E2) with 5 Areal BMD femoral neck by DXA (mostly negative findings)

. Bone loss; . nonvertebral fractures; . vertebral fractures . Volumetric vertebral BMD by QCT

Androgenic T action more marginal and indirect . Volumetric hip BMD by QCT
(i.e., on muscle, falls)

(? effect in osteoporosis and fracture risk)

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


(osteoporosis not indication for T therapy)

Body composition, , SHBG and total T with , Total body fat mass;
metabolic health, and
physical performance . Fat mass, obesity . Fat-free mass;

, SHBG and total T and free T with . (Prevented ,) skeletal muscle mass;

.. Fat mass, morbid obesity (quantitatively modest changes)

Reciprocal relationship , T and . fat mass with . Insulin sensitivity


predominant reverse causality

, Total T and/or SHBG cross-sectional with (modest effect size)

. Insulin resistance, MetS, T2DM (data do not support T treatment to improve metabolic status
in older men with low T in general or with MetS or T2DM)

, Total T and/or SHBG longitudinal with $ Muscle strength and power

. MetS and T2DM (variable: limited to no effect)

(inconsistent and not with free/bioavailable T) $ Physical function

. (Free/bioavailable) T with (no effect or small effects of uncertain clinical significance)

. Lean mass, , lean mass decline

(inconsistent findings)

. (Free/bioavailable) T with

. Indices muscle strength and /or physical


performance (findings inconsistent)

. Free/bioavailable T with

. Indices of frailty

Erythropoiesis , (Free/bioavailable) T and hypogonadism with . Risk of polycythemia

, Hemoglobin; . decline Hb . Hct, Hb in men with unexplained anemia

Cardiovascular disease Sex steroids with CVD: disparate, inconsistent ? MI, other major cardiac events, stroke
and mortality findings; residual confounding, reverse causality

Sex steroids not with

IHD, MI

, Or “not optimal” T (and/or DHT) with Thromboembolism (data not conclusive)

. Stroke (although they seem predominantly reassuring (i.e., 5) for short


term, data not conclusive and no reliable long-term data ?)

. CVD-specific and all-cause mortality


(Continued )

956 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

Table 7. Continued
System/Function Associations With Endogenous Sex Steroids Effects of T Therapy

Sexual function , (Free/bioavailable) T with . Sexual desire, . erectile function,

, Sexual desire; . Overall sexual activity

, indices of sexual function (moderate amplitude;

only if initially unequivocally low T and

prevalent sexual symptoms;

effect on erectile function less than

PDE5 inhibitors;

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


no potentiation of PDE5 inhibitors)

Mental health , Or suboptimal (free/bioavailable) T not consistently 5 Memory


with cognitive decline or incident Alzheimer’s disease

, (Free/bioavailable)T with 5 Other aspects of cognition

. Depressive symptoms (mixed findings; only limited . Mood, vitality


evidence for role of moderately low T)

, Depressive symptoms (small effect size; depression not


indication for T therapy)

See text for details.


Bold text indicates hormonal changes; clinical correlates accompany the changes listed.
Abbreviations: ., higher, more, increase; ,, lower, less, decrease; 5, unchanged, no effect; ?, not known.

hard clinical endpoints, in subjects representative of been shown to offer added value on top of non-
the target population for treatment, and for treatment pharmacologic measures (e.g., exercise, weight loss)
modalities and durations relevant for the intended and compared with, or on top of, existing pharma-
clinical use. Moreover, such T treatment should have cologic treatments for the same indication.

References
1. Kaufman JM, Vermeulen A. The decline of androgen increased risk of heart attack and stroke with 11. Ramasamy R, Chiba K, Butler P, Lamb DJ. Male
levels in elderly men and its clinical and therapeutic use. Available at: www.fda.gov/Drugs/DrugSafety/ biological clock: a critical analysis of advanced pa-
implications. Endocr Rev. 2005;26(6):833–876. ucm436259.htm. Accessed 14 May 2019. ternal age. Fertil Steril. 2015;103(6):1402–1406.
2. Jasuja GK, Bhasin S, Rose AJ. Patterns of testosterone 7. Basaria S, Coviello AD, Travison TG, Storer TW, 12. Johnson SL, Dunleavy J, Gemmell NJ, Nakagawa S.
prescription overuse. Curr Opin Endocrinol Diabetes Farwell WR, Jette AM, Eder R, Tennstedt S, Ulloor J, Consistent age-dependent declines in human se-
Obes. 2017;24(3):240–245. Zhang A, Choong K, Lakshman KM, Mazer NA, men quality: a systematic review and meta-analysis.
3. Baillargeon J, Kuo YF, Westra JR, Urban RJ, Goodwin JS. Miciek R, Krasnoff J, Elmi A, Knapp PE, Brooks B, Ageing Res Rev. 2015;19:22–33.
Testosterone Prescribing in the United States, Appleman E, Aggarwal S, Bhasin G, Hede-Brierley L, 13. Stone BA, Alex A, Werlin LB, Marrs RP. Age
2002–2016. JAMA. 2018;320(2):200–202. Bhatia A, Collins L, LeBrasseur N, Fiore LD, Bhasin S. thresholds for changes in semen parameters in men.
4. Yeap BB, Grossmann M, McLachlan RI, Handelsman Adverse events associated with testosterone ad- Fertil Steril. 2013;100(4):952–958.
DJ, Wittert GA, Conway AJ, Stuckey BG, Lording ministration. N Engl J Med. 2010;363(2):109–122. 14. Zhu QX, Meads C, Lu ML, Wu JQ, Zhou WJ, Gao ES.
DW, Allan CA, Zajac JD, Burger HG. Endocrine 8. Xu L, Freeman G, Cowling BJ, Schooling CM. Tes- Turning point of age for semen quality: a
Society of Australia position statement on male tosterone therapy and cardiovascular events among population-based study in Chinese men. Fertil Steril.
hypogonadism (part 1): assessment and indications men: a systematic review and meta-analysis of 2011;96(3):572–576.
for testosterone therapy. Med J Aust. 2016;205(4): placebo-controlled randomized trials. BMC Med. 15. Mahmoud AM, Goemaere S, El-Garem Y, Van
173–178. 2013;11(1):108. Pottelbergh I, Comhaire FH, Kaufman JM. Tes-
5. Bhasin S, Brito JP, Cunningham GR, Hayes FJ, Hodis 9. Mintziori G, Lambrinoudaki I, Kolibianakis EM, ticular volume in relation to hormonal indices
HN, Matsumoto AM, Snyder PJ, Swerdloff RS, Wu Ceausu I, Depypere H, Erel CT, Pérez-López FR, of gonadal function in community-dwelling el-
FC, Yialamas MA. Testosterone therapy in men with Schenck-Gustafsson K, van der Schouw Y, derly men. J Clin Endocrinol Metab. 2003;88(1):
hypogonadism: an Endocrine Society clinical prac- Simoncini T, Tremollieres F, Tarlatzis BC, Rees M, 179–184.
tice guideline. J Clin Endocrinol Metab. 2018;103(5): Goulis DG. EMAS position statement: late par- 16. Johnson L, Zane RS, Petty CS, Neaves WB. Quanti-
1715–1744. enthood. Maturitas. 2013;76(2):200–204. fication of the human Sertoli cell population: its
6. US Food and Drug Administration. Drug safety 10. Khandwala YS, Zhang CA, Lu Y, Eisenberg ML. The distribution, relation to germ cell numbers, and age-
communications: FDA cautions about using tes- age of fathers in the USA is rising: an analysis of 168 related decline. Biol Reprod. 1984;31(4):785–795.
tosterone products for low testosterone due to 867 480 births from 1972 to 2015. Hum Reprod. 2017; 17. Well D, Yang H, Houseni M, Iruvuri S, Alzeair S,
aging; requires labeling change to inform of possible 32(10):2110–2116. Sansovini M, Wintering N, Alavi A, Torigian DA.

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 957


REVIEW

Age-related structural and metabolic changes in the male germ line. Fertil Steril. 2017;107(2): results from the Massachusetts Male Aging Study.
the pelvic reproductive end organs. Semin Nucl 319–323. Eur J Endocrinol. 2006;155(3):443–452.
Med. 2007;37(3):173–184. 35. Levinger U, Gornish M, Gat Y, Bachar GN. Is vari- 52. Liu PY, Beilin J, Meier C, Nguyen TV, Center JR,
18. Paniagua R, Amat P, Nistal M, Martin A. Ultra- cocele prevalence increasing with age? Andrologia. Leedman PJ, Seibel MJ, Eisman JA, Handelsman DJ.
structural changes in Sertoli cells in ageing humans. 2007;39(3):77–80. Age-related changes in serum testosterone and sex
Int J Androl. 1985;8(4):295–312. 36. Avellino G, Theva D, Oates RD. Common urologic hormone binding globulin in Australian men:
19. Dakouane M, Bicchieray L, Bergere M, Albert M, diseases in older men and their treatment: how longitudinal analyses of two geographically separate
Vialard F, Selva J. A histomorphometric and cyto- they impact fertility. Fertil Steril. 2017;107(2): regional cohorts. J Clin Endocrinol Metab. 2007;92(9):
genetic study of testis from men 29–102 years old. 305–311. 3599–3603.
Fertil Steril. 2005;83(4):923–928. 37. Handelsman DJ, Staraj S. Testicular size: the effects 53. Travison TG, Araujo AB, O’Donnell AB, Kupelian V,
20. Neaves WB, Johnson L, Porter JC, Parker CR Jr, of aging, malnutrition, and illness. J Androl. 1985; McKinlay JB. A population-level decline in serum
Petty CS. Leydig cell numbers, daily sperm pro- 6(3):144–151. testosterone levels in American men. J Clin Endo-
duction, and serum gonadotropin levels in aging 38. Hurley EG, DeFranco EA. Influence of paternal age crinol Metab. 2007;92(1):196–202.
men. J Clin Endocrinol Metab. 1984;59(4): on perinatal outcomes. Am J Obstet Gynecol. 2017; 54. Lapauw B, Goemaere S, Zmierczak H, Van
756–763. 217(5):566.e1–566.e6. Pottelbergh I, Mahmoud A, Taes Y, De Bacquer
21. Meacham RB, Murray MJ. Reproductive function in 39. Malaspina D, Gilman C, Kranz TM. Paternal age and D, Vansteelandt S, Kaufman JM. The decline of
the aging male. Urol Clin North Am. 1994;21(3): mental health of offspring. Fertil Steril. 2015;103(6): serum testosterone levels in community-dwelling

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


549–556. 1392–1396. men over 70 years of age: descriptive data and
22. Sasano N, Ichijo S. Vascular patterns of the human 40. Conti SL, Eisenberg ML. Paternal aging and in- predictors of longitudinal changes. Eur J Endocrinol.
testis with special reference to its senile changes. creased risk of congenital disease, psychiatric dis- 2008;159(4):459–468.
Tohoku J Exp Med. 1969;99(3):269–280. orders, and cancer. Asian J Androl. 2016;18(3): 55. Camacho EM, Huhtaniemi IT, O’Neill TW, Finn JD,
23. Johnson L, Petty CS, Neaves WB. Influence of age on 420–424. Pye SR, Lee DM, Tajar A, Bartfai G, Boonen S,
sperm production and testicular weights in men. 41. Nybo Andersen AM, Urhoj SK. Is advanced paternal Casanueva FF, Forti G, Giwercman A, Han TS, Kula
J Reprod Fertil. 1984;70(1):211–218. age a health risk for the offspring? Fertil Steril. 2017; K, Keevil B, Lean ME, Pendleton N, Punab M,
24. Humm KC, Sakkas D. Role of increased male age 107(2):312–318. Vanderschueren D, Wu FC; EMAS Group. Age-
in IVF and egg donation: is sperm DNA frag- 42. Paul C, Robaire B. Ageing of the male germ line. Nat associated changes in hypothalamic–pituitary–
mentation responsible? Fertil Steril. 2013;99(1): Rev Urol. 2013;10(4):227–234. testicular function in middle-aged and older men
30–36. 43. Urhoj SK, Jespersen LN, Nissen M, Mortensen LH, are modified by weight change and lifestyle fac-
25. Ford WC, North K, Taylor H, Farrow A, Hull MG, Nybo Andersen AM. Advanced paternal age and tors: longitudinal results from the European Male
Golding J; The ALSPAC Study Team (Avon Lon- mortality of offspring under 5 years of age: a Ageing Study. Eur J Endocrinol. 2013;168(3):
gitudinal Study of Pregnancy and Childhood). In- register-based cohort study. Hum Reprod. 2014; 445–455.
creasing paternal age is associated with delayed 29(2):343–350. 56. Shi Z, Araujo AB, Martin S, O’Loughlin P,
conception in a large population of fertile couples: 44. Jennings MO, Owen RC, Keefe D, Kim ED. Man- Wittert GA. Longitudinal changes in testos-
evidence for declining fecundity in older men. Hum terone over five years in community-dwelling
agement and counseling of the male with advanced
Reprod. 2000;15(8):1703–1708. men. J Clin Endocrinol Metab. 2013;98(8):
paternal age. Fertil Steril. 2017;107(2):324–328.
26. Forster P, Hohoff C, Dunkelmann B, Schurenkamp 3289–3297.
45. Kroese AC, de Lange NM, Collins J, Evers JL.
M, Pfeiffer H, Neuhuber F, Brinkmann B. Correc- 57. Yeap BB, Manning L, Chubb SAP, Handelsman
Surgery or embolization for varicoceles in sub-
tion to “Elevated germline mutation rate in DJ, Almeida OP, Hankey GJ, Flicker L. Progressive
fertile men. Cochrane Database Syst Rev. 2012;10:
teenage fathers”. Proc Biol Sci. 2016;283(1837): impairment of testicular endocrine function
CD000479.t
20161723. in ageing men: testosterone and dihy-
46. Showell MG, Mackenzie-Proctor R, Brown J, Yazdani
27. Nybo Andersen AM, Hansen KD, Andersen PK, drotestosterone decrease, and luteinizing hor-
A, Stankiewicz MT, Hart RJ. Antioxidants for male
Davey Smith G. Advanced paternal age and risk of mone increases, in men transitioning from the
subfertility. Cochrane Database Syst Rev. 2014;
fetal death: a cohort study. Am J Epidemiol. 2004; 8th to 9th decades of life. Clin Endocrinol (Oxf).
12(12):CD007411.
160(12):1214–1222. 2018;88(1):88–95.
47. Wu FC, Tajar A, Pye SR, Silman AJ, Finn JD, O’Neill
28. Eisenberg ML, Meldrum D. Effects of age on fertility 58. Nieschlag E, Lammers U, Freischem CW, Langer K,
TW, Bartfai G, Casanueva F, Forti G, Giwercman A,
and sexual function. Fertil Steril. 2017;107(2): Wickings EJ. Reproductive functions in young fa-
Huhtaniemi IT, Kula K, Punab M, Boonen S,
301–304. thers and grandfathers. J Clin Endocrinol Metab.
29. Wu Y, Kang X, Zheng H, Liu H, Liu J. Effect of Vanderschueren D; European Male Aging Study 1982;55(4):676–681.
paternal age on reproductive outcomes of Group. Hypothalamic-pituitary-testicular axis dis- 59. Sartorius G, Spasevska S, Idan A, Turner L, Forbes E,
in vitro fertilization. PLoS One. 2015;10(9): ruptions in older men are differentially linked to age Zamojska A, Allan CA, Ly LP, Conway AJ, McLachlan
e0135734. and modifiable risk factors: the European Male RI, Handelsman DJ. Serum testosterone, dihy-
30. Ma NZ, Chen L, Hu LL, Dai W, Bu ZQ, Sun YP. The Aging Study. J Clin Endocrinol Metab. 2008;93(7): drotestosterone and estradiol concentrations in
influence of male age on treatment outcomes and 2737–2745. older men self-reporting very good health: the
neonatal birthweight following assisted re- 48. Morley JE, Kaiser FE, Perry HM III, Patrick P, Morley healthy man study. Clin Endocrinol (Oxf). 2012;77(5):
production technology involving intracytoplasmic PMK, Stauber PM, Vellas B, Baumgartner RN, Garry 755–763.
sperm injection (ICSI) cycles. Andrologia. 2018;50(1): PJ. Longitudinal changes in testosterone, luteinizing 60. Yeap BB, Alfonso H, Chubb SA, Handelsman DJ,
e12826. hormone, and follicle-stimulating hormone in Hankey GJ, Norman PE, Flicker L. Reference
31. Wu Y, Kang X, Zheng H, Liu H, Huang Q, Liu J. Effect healthy older men. Metabolism. 1997;46(4): ranges and determinants of testosterone, dihy-
of paternal age on reproductive outcomes of 410–413. drotestosterone, and estradiol levels measured
intracytoplasmic sperm injection. PLoS One. 2016; 49. Zmuda JM, Cauley JA, Kriska A, Glynn NW, Gutai using liquid chromatography-tandem mass spec-
11(2):e0149867. JP, Kuller LH. Longitudinal relation between en- trometry in a population-based cohort of older
32. Meijerink AM, Ramos L, Fleischer K, Veltman JA, dogenous testosterone and cardiovascular disease men. J Clin Endocrinol Metab. 2012;97(11):4030–
Hendriks JC, Braat DD. Influence of paternal age on risk factors in middle-aged men. A 13-year follow- 4039.
ongoing pregnancy rate at eight weeks’ gestation in up of former Multiple Risk Factor Intervention 61. Fukai S, Akishita M, Miyao M, Ishida K, Toba K,
assisted reproduction. Reprod Biomed Online. 2016; Trial participants. Am J Epidemiol. 1997;146(8): Ouchi Y. Age-related changes in plasma androgen
32(1):96–103. 609–617. levels and their association with cardiovascular risk
33. Park YS, Lee SH, Lim CK, Choi HW, An JH, Park CW, 50. Harman SM, Metter EJ, Tobin JD, Pearson J, factors in male Japanese office workers. Geriatr
Lee HS, Lee JS, Seo JT. Paternal age as an in- Blackman MR; Baltimore Longitudinal Study of Gerontol Int. 2010;10(1):32–39.
dependent factor does not affect embryo quality Aging. Longitudinal effects of aging on serum total 62. Kim YS, Hong D, Lee DJ, Joo NS, Kim KM. Total
and pregnancy outcomes of testicular sperm and free testosterone levels in healthy men. J Clin testosterone may not decline with ageing in Korean
extraction-intracytoplasmic sperm injection in Endocrinol Metab. 2001;86(2):724–731. men aged 40 years or older. Clin Endocrinol (Oxf).
azoospermia. Andrologia. 2018;50(2):e12864. 51. Mohr BA, Bhasin S, Link CL, O’Donnell AB, 2012;77(2):296–301.
34. Herati AS, Zhelyazkova BH, Butler PR, Lamb DJ. Age- McKinlay JB. The effect of changes in adiposity on 63. Liu Z, Liu J, Shi X, Wang L, Yang Y, Tao M. Dynamic
related alterations in the genetics and genomics of testosterone levels in older men: longitudinal alteration of serum testosterone with aging: a cross-

958 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

sectional study from Shanghai, China. Reprod Biol hormone agonist therapy than with surgical cas- 89. Crabbe P, Bogaert V, De Bacquer D, Goemaere S,
Endocrinol. 2015;13(1):111. tration: new insights attained by mass spectrom- Zmierczak H, Kaufman JM. Part of the in-
64. Xia F, Wang N, Han B, Li Q, Chen Y, Zhu C, Chen Y, etry. J Urol. 2012;187(5):1601–1606. terindividual variation in serum testosterone levels
Pu X, Cang Z, Zhu C, Lu M, Meng Y, Guo H, Chen C, 76. Eisenhofer G, Peitzsch M, Kaden D, Langton K, in healthy men reflects differences in androgen
Lin D, Zheng J, Kuang L, Tu W, Li B, Hu L, Shen Z, Lu Pamporaki C, Masjkur J, Tsatsaronis G, Mangelis A, sensitivity and feedback set point: contribution of
Y. Hypothalamic-pituitary-gonadal axis in aging Williams TA, Reincke M, Lenders JWM, Bornstein the androgen receptor polyglutamine tract poly-
men and women: increasing total testosterone in SR. Reference intervals for plasma concentrations of morphism. J Clin Endocrinol Metab. 2007;92(9):
aging men. Neuroendocrinology. 2017;104(3):291– adrenal steroids measured by LC-MS/MS: impact of 3604–3610.
301. gender, age, oral contraceptives, body mass index 90. Rajender S, Singh L, Thangaraj K. Phenotypic het-
65. Feldman HA, Longcope C, Derby CA, Johannes CB, and blood pressure status. Clin Chim Acta. 2017;470: erogeneity of mutations in androgen receptor gene.
Araujo AB, Coviello AD, Bremner WJ, McKinlay JB. 115–124. Asian J Androl. 2007;9(2):147–179.
Age trends in the level of serum testosterone and 77. Misra A, Khurana L. Obesity-related non-communicable 91. Huhtaniemi IT, Pye SR, Limer KL, Thomson W,
other hormones in middle-aged men: longitudinal diseases: South Asians vs white Caucasians. Int J Obes. O’Neill TW, Platt H, Payne D, John SL, Jiang M,
results from the Massachusetts male aging study. 2011;35(2):167–187. Boonen S, Borghs H, Vanderschueren D, Adams JE,
J Clin Endocrinol Metab. 2002;87(2):589–598. 78. Heald AH, Ivison F, Anderson SG, Cruickshank K, Ward KA, Bartfai G, Casanueva F, Finn JD, Forti G,
66. Ohlsson C, Nilsson ME, Tivesten A, Ryberg H, Laing I, Gibson JM. Significant ethnic variation in Giwercman A, Han TS, Kula K, Lean ME, Pendleton
Mellström D, Karlsson MK, Ljunggren Ö, Labrie F, total and free testosterone concentration. Clin N, Punab M, Silman AJ, Wu FC; European Male

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Orwoll ES, Lee DM, Pye SR, O’Neill TW, Finn JD, Endocrinol (Oxf). 2003;58(3):262–266. Ageing Study Group. Increased estrogen rather
Adams JE, Ward KA, Boonen S, Bartfai G, Casanueva 79. Rajan TV, Kerstetter J, Feinn R, Kenny A. Evidence than decreased androgen action is associated with
FF, Forti G, Giwercman A, Han TS, Huhtaniemi IT, for low androgenicity among Indian (South Asian) longer androgen receptor CAG repeats. J Clin
Kula K, Lean ME, Pendleton N, Punab M, men. Aging Male. 2014;17(1):30–34. Endocrinol Metab. 2009;94(1):277–284.
Vanderschueren D, Wu FC, Vandenput L; EMAS 80. Kupelian V, Hayes FJ, Link CL, Rosen R, McKinlay JB. 92. Bogaert V, Vanbillemont G, Taes Y, De Bacquer D,
Study Group. Comparisons of immunoassay and Inverse association of testosterone and the meta- Deschepper E, Van Steen K, Kaufman JM. Small
mass spectrometry measurements of serum es- bolic syndrome in men is consistent across race and effect of the androgen receptor gene GGN repeat
tradiol levels and their influence on clinical asso- ethnic groups. J Clin Endocrinol Metab. 2008;93(9): polymorphism on serum testosterone levels in
ciation studies in men. J Clin Endocrinol Metab. 2013; 3403–3410. healthy men. Eur J Endocrinol. 2009;161(1):171–177.
98(6):E1097–E1102. 81. Giton F, Fiet J, Cornu JN, Cussenot O, Bélanger A, 93. Edwards A, Hammond HA, Jin L, Caskey CT,
67. Swerdloff RS, Dudley RE, Page ST, Wang C, Salameh Urien S, Oliva A, Blanchet P, Multigner L. Serum sex Chakraborty R. Genetic variation at five trimeric
WA. Dihydrotestosterone: biochemistry, physiol- steroids measured in middle-aged European and and tetrameric tandem repeat loci in four human
ogy, and clinical implications of elevated blood African–Caribbean men by gas chromatography- population groups. Genomics. 1992;12(2):241–253.
levels. Endocr Rev. 2017;38(3):220–254. mass spectrometry. Eur J Endocrinol. 2011;165(6): 94. Ackerman CM, Lowe LP, Lee H, Hayes MG, Dyer AR,
68. Shiraishi S, Lee PW, Leung A, Goh VH, Swerdloff RS, 917–924. Metzger BE, Lowe WL, Urbanek M; Hapo Study
Wang C. Simultaneous measurement of serum 82. Richard A, Rohrmann S, Zhang L, Eichholzer M, Cooperative Research Group. Ethnic variation in
testosterone and dihydrotestosterone by liquid Basaria S, Selvin E, Dobs AS, Kanarek N, Menke A, allele distribution of the androgen receptor (AR)
chromatography-tandem mass spectrometry. Clin Nelson WG, Platz EA. Racial variation in sex steroid (CAG)n repeat. J Androl. 2012;33(2):210–215.
Chem. 2008;54(11):1855–1863. hormone concentration in black and white men: a 95. Krithivas K, Yurgalevitch SM, Mohr BA, Wilcox CJ,
69. Handelsman DJ, Yeap B, Flicker L, Martin S, Wittert meta-analysis. Andrology. 2014;2(3):428–435. Batter SJ, Brown M, Longcope C, McKinlay JB,
GA, Ly LP. Age-specific population centiles for 83. Rohrmann S, Nelson WG, Rifai N, Brown TR, Dobs Kantoff PW. Evidence that the CAG repeat in the
androgen status in men. Eur J Endocrinol. 2015; A, Kanarek N, Yager JD, Platz EA. Serum estrogen, androgen receptor gene is associated with the age-
173(6):809–817. but not testosterone, levels differ between black related decline in serum androgen levels in men.
70. Jasuja GK, Travison TG, Davda M, Murabito JM, and white men in a nationally representative J Endocrinol. 1999;162(1):137–142.
Basaria S, Zhang A, Kushnir MM, Rockwood AL, sample of Americans. J Clin Endocrinol Metab. 2007; 96. Walsh S, Zmuda JM, Cauley JA, Shea PR, Metter EJ,
Meikle W, Pencina MJ, Coviello A, Rose AJ, 92(7):2519–2525. Hurley BF, Ferrell RE, Roth SM. Androgen receptor
D’Agostino R, Vasan RS, Bhasin S. Age trends in 84. Orwoll ES, Nielson CM, Labrie F, Barrett-Connor E, CAG repeat polymorphism is associated with fat-
estradiol and estrone levels measured using liquid Cauley JA, Cummings SR, Ensrud K, Karlsson M, Lau free mass in men. J Appl Physiol (1985). 2005;98(1):
chromatography tandem mass spectrometry in E, Leung PC, Lunggren O, Mellström D, Patrick AL, 132–137.
community-dwelling men of the Framingham Stefanick ML, Nakamura K, Yoshimura N, Zmuda J, 97. Van Pottelbergh I, Lumbroso S, Goemaere S, Sultan
Heart Study. J Gerontol A Biol Sci Med Sci. 2013;68(6): Vandenput L, Ohlsson C; Osteoporotic Fractures in C, Kaufman JM. Lack of influence of the androgen
733–740. Men (MrOS) Research Group. Evidence for geo- receptor gene CAG-repeat polymorphism on sex
71. Hsu B, Cumming RG, Waite LM, Blyth FM, graphical and racial variation in serum sex steroid steroid status and bone metabolism in elderly men.
Naganathan V, Le Couteur DG, Seibel MJ, levels in older men. J Clin Endocrinol Metab. 2010; Clin Endocrinol (Oxf). 2001;55(5):659–666.
Handelsman DJ. Longitudinal relationships between 95(10):E151–E160. 98. Eriksson AL, Lorentzon M, Mellström D, Vandenput
reproductive hormones and cognitive decline in 85. Eendebak RJ, Swiecicka A, Gromski PS, Pye SR, L, Swanson C, Andersson N, Hammond GL,
older men: the Concord Health and Ageing in Men O’Neill TW, Marshall A, Keevil BG, Tampubolon G, Jakobsson J, Rane A, Orwoll ES, Ljunggren O, Johnell
Project. J Clin Endocrinol Metab. 2015;100(6): Goodacre R, Wu FC, Rutter MK. Ethnic differences O, Labrie F, Windahl SH, Ohlsson C. SHBG gene
2223–2230. in male reproductive hormones and relationships promoter polymorphisms in men are associated
72. Orwoll E, Lambert LC, Marshall LM, Phipps K, Blank with adiposity and insulin resistance in older men. with serum sex hormone-binding globulin, an-
J, Barrett-Connor E, Cauley J, Ensrud K, Cummings S. Clin Endocrinol (Oxf). 2017;86(5):660–668. drogen and androgen metabolite levels, and hip
Testosterone and estradiol among older men. J Clin 86. Meikle AW, Stringham JD, Bishop DT, West DW. bone mineral density. J Clin Endocrinol Metab. 2006;
Endocrinol Metab. 2006;91(4):1336–1344. Quantitating genetic and nongenetic factors 91(12):5029–5037.
73. Tannenbaum C, Barrett-Connor E, Laughlin GA, influencing androgen production and clearance 99. Vanbillemont G, Bogaert V, De Bacquer D, Lapauw
Platt RW. A longitudinal study of dehydroepian- rates in men. J Clin Endocrinol Metab. 1988;67(1): B, Goemaere S, Toye K, Van Steen K, Taes Y,
drosterone sulphate (DHEAS) change in older men 104–109. Kaufman JM. Polymorphisms of the SHBG gene
and women: the Rancho Bernardo Study. Eur J 87. Ring HZ, Lessov CN, Reed T, Marcus R, Holloway L, contribute to the interindividual variation of sex
Endocrinol. 2004;151(6):717–725. Swan GE, Carmelli D. Heritability of plasma sex steroid hormone blood levels in young, middle-
74. Labrie F, Bélanger A, Cusan L, Gomez JL, Candas B. hormones and hormone binding globulin in adult aged and elderly men. Clin Endocrinol (Oxf). 2009;
Marked decline in serum concentrations of adrenal male twins. J Clin Endocrinol Metab. 2005;90(6): 70(2):303–310.
C19 sex steroid precursors and conjugated an- 3653–3658. 100. Huhtaniemi IT, Pye SR, Holliday KL, Thomson W,
drogen metabolites during aging. J Clin Endocrinol 88. Bogaert V, Taes Y, Konings P, Van Steen K, De O’Neill TW, Platt H, Payne D, John SL, Jiang M,
Metab. 1997;82(8):2396–2402. Bacquer D, Goemaere S, Zmierczak H, Crabbe P, Bartfai G, Boonen S, Casanueva FF, Finn JD, Forti G,
75. van der Sluis TM, Bui HN, Meuleman EJH, Heijboer Kaufman JM. Heritability of blood concentrations Giwercman A, Han TS, Kula K, Lean ME, Pendleton
AC, Hartman JF, van Adrichem N, Boevé E, de of sex-steroids in relation to body composition in N, Punab M, Silman AJ, Vanderschueren D, Labrie F,
Ronde W, van Moorselaar RJ, Vis AN. Lower tes- young adult male siblings. Clin Endocrinol (Oxf). Wu FC; European Male Aging Study Group. Effect
tosterone levels with luteinizing hormone-releasing 2008;69(1):129–135. of polymorphisms in selected genes involved in

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 959


REVIEW

pituitary-testicular function on reproductive hor- receptor alpha and beta gene polymorphisms with 124. Zumoff B, Strain GW, Miller LK, Rosner W, Senie R,
mones and phenotype in aging men. J Clin Endo- sex steroid levels and body fat content in men. Exp Seres DS, Rosenfeld RS. Plasma free and non-sex-
crinol Metab. 2010;95(4):1898–1908. Clin Endocrinol Diabetes. 2012;120(3):154–159. hormone-binding-globulin-bound testosterone are
101. Ohlsson C, Wallaschofski H, Lunetta KL, Stolk L, 111. Meyer TE, Chu LW, Li Q, Yu K, Rosenberg PS, decreased in obese men in proportion to their
Perry JR, Koster A, Petersen AK, Eriksson J, Lehtimäki Menashe I, Chokkalingam AP, Quraishi SM, Huang degree of obesity. J Clin Endocrinol Metab. 1990;
T, Huhtaniemi IT, Hammond GL, Maggio M, WY, Weiss JM, Kaaks R, Hayes RB, Chanock SJ, Hsing 71(4):929–931.
Coviello AD, Ferrucci L, Heier M, Hofman A, AW. The association between inflammation-related 125. Giagulli VA, Kaufman JM, Vermeulen A. Patho-
Holliday KL, Jansson JO, Kähönen M, Karasik D, genes and serum androgen levels in men: the genesis of the decreased androgen levels in obese
Karlsson MK, Kiel DP, Liu Y, Ljunggren O, Lorentzon prostate, lung, colorectal, and ovarian study. Pros- men. J Clin Endocrinol Metab. 1994;79(4):997–1000.
M, Lyytikäinen LP, Meitinger T, Mellström D, Melzer tate. 2012;72(1):65–71. 126. Brodsky IG, Balagopal P, Nair KS. Effects of testos-
D, Miljkovic I, Nauck M, Nilsson M, Penninx B, Pye 112. Saadat M, Bahaoddini S, Saadat I. Alteration of terone replacement on muscle mass and muscle
SR, Vasan RS, Reincke M, Rivadeneira F, Tajar A, serum sex hormonal profile in male gasoline filling protein synthesis in hypogonadal men—a clinical
Teumer A, Uitterlinden AG, Ulloor J, Viikari J, Völker station workers in respect to their polymorphism of research center study. J Clin Endocrinol Metab. 1996;
U, Völzke H, Wichmann HE, Wu TS, Zhuang WV, glutathione S-transferase M1. Environ Toxicol 81(10):3469–3475.
Ziv E, Wu FC, Raitakari O, Eriksson A, Bidlingmaier Pharmacol. 2013;35(2):265–269. 127. Allan CA, Strauss BJ, Burger HG, Forbes EA,
M, Harris TB, Murray A, de Jong FH, Murabito JM, 113. Lindgren I, Giwercman A, Axelsson J, Lundberg McLachlan RI. Testosterone therapy prevents gain
Bhasin S, Vandenput L, Haring R; EMAS Study Giwercman Y. Association between follicle-stimulating in visceral adipose tissue and loss of skeletal muscle

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Group. Genetic determinants of serum testoster- hormone receptor polymorphisms and reproduc- in nonobese aging men. J Clin Endocrinol Metab.
one concentrations in men. PLoS Genet. 2011;7(10): tive parameters in young men from the general 2008;93(1):139–146.
e1002313. population. Pharmacogenet Genomics. 2012;22(9): 128. Hamilton EJ, Gianatti E, Strauss BJ, Wentworth J,
102. Svartberg J, Schirmer H, Wilsgaard T, Mathiesen EB, 667–672. Lim-Joon D, Bolton D, Zajac JD, Grossmann M.
Njølstad I, Løchen ML, Jorde R. Single-nucleotide 114. Grigorova M, Punab M, Poolamets O, Sõber S, Increase in visceral and subcutaneous abdominal fat
polymorphism, rs1799941 in the sex hormone- Vihljajev V, Žilaitienė B, Erenpreiss J, Matulevičius V, in men with prostate cancer treated with androgen
binding globulin (SHBG) gene, related to both se- Tsarev I, Laan M. Study in 1790 Baltic men: FSHR deprivation therapy. Clin Endocrinol (Oxf). 2011;
rum testosterone and SHBG levels and the risk of Asn680Ser polymorphism affects total testes vol- 74(3):377–383.
myocardial infarction, type 2 diabetes, cancer and ume. Andrology. 2013;1(2):293–300. 129. Grossmann M. Low testosterone in men with type
mortality in men: the Tromsø Study. Andrology. 115. Ahmetov II, Donnikov AE, Trofimov DY. ACTN3 2 diabetes: significance and treatment. J Clin
2014;2(2):212–218. genotype is associated with testosterone levels of Endocrinol Metab. 2011;96(8):2341–2353.
103. Zmuda JM, Cauley JA, Kuller LH, Ferrell RE. A athletes. Biol Sport. 2014;31(2):105–108. 130. Cheung AS, Hoermann R, Dupuis P, Joon DL, Zajac
common promotor variant in the cytochrome 116. Laczmanski L, Lwow F, Mossakowska M, Puzianowska- JD, Grossmann M. Relationships between insulin
P450c17a (CYP17) gene is associated with bio- Kuznicka M, Szwed M, Kolackov K, Krzyzanowska-
resistance and frailty with body composition and
availability testosterone levels and bone size in men. Swiniarska B, Bar-Andziak E, Chudek J, Sloka N,
testosterone in men undergoing androgen depri-
J Bone Miner Res. 2001;16(5):911–917. Milewicz A. Association between vitamin D con-
vation therapy for prostate cancer. Eur J Endocrinol.
104. Sheu YT, Zmuda JM, Cauley JA, Moffett SP, Rosen centration and levels of sex hormones in an elderly
2016;175(3):229–237.
CJ, Ishwad C, Ferrell RE. Nuclear receptor Polish population with different genotypes of VDR
131. Corona G, Rastrelli G, Monami M, Saad F, Luconi M,
coactivator-3 alleles are associated with serum polymorphisms (rs10735810, rs1544410, rs7975232,
Lucchese M, Facchiano E, Sforza A, Forti G,
bioavailable testosterone, insulin-like growth factor- rs731236). Gene. 2015;559(1):73–76.
Mannucci E, Maggi M. Body weight loss reverts
1, and vertebral bone mass in men. J Clin Endocrinol 117. Shen O, Ding X, Nie J, Xia Y, Wang X, Tong J, Zhang J.
obesity-associated hypogonadotropic hypogonad-
Metab. 2006;91(1):307–312. Variants of the CLOCK gene affect the risk of id-
ism: a systematic review and meta-analysis. Eur J
105. Eriksson AL, Lorentzon M, Vandenput L, Labrie F, iopathic male infertility in the Han-Chinese pop-
Endocrinol. 2013;168(6):829–843.
Lindersson M, Syvänen AC, Orwoll ES, Cummings ulation. Chronobiol Int. 2015;32(7):959–965.
132. Rastrelli G, Carter EL, Ahern T, Finn JD, Antonio L,
SR, Zmuda JM, Ljunggren O, Karlsson MK, 118. De Toni L, Di Nisio A, Speltra E, Rocca MS, Ghezzi M,
O’Neill TW, Bartfai G, Casanueva FF, Forti G, Keevil
Mellström D, Ohlsson C. Genetic variations in sex Zuccarello D, Turiaco N, Ferlin A, Foresta C. Poly-
steroid-related genes as predictors of serum es- morphism rs2274911 of GPRC6A as a novel risk B, Maggi M, Giwercman A, Han TS, Huhtaniemi IT,
trogen levels in men. J Clin Endocrinol Metab. 2009; factor for testis failure. J Clin Endocrinol Metab. 2016; Kula K, Lean ME, Pendleton N, Punab M,
94(3):1033–1041. 101(3):953–961. Vanderschueren D, Wu FC; EMAS Study Group.
106. Jakobsson J, Palonek E, Lorentzon M, Ohlsson C, 119. Ratan SK, Sharma A, Kapoor S, Polipalli SK, Dubey D, Development of and recovery from secondary
Rane A, Ekström L. A novel polymorphism in the Mishra TK, Sinha SK, Agarwal SK. Polymorphism of hypogonadism in aging men: prospective results
17b-hydroxysteroid dehydrogenase type 5 (aldo- 39 UTR of MAMLD1 gene is also associated with from the EMAS. J Clin Endocrinol Metab. 2015;
keto reductase 1C3) gene is associated with lower increased risk of isolated hypospadias in Indian 100(8):3172–3182.
serum testosterone levels in caucasian men. Phar- children: a preliminary report. Pediatr Surg Int. 2016; 133. Tena-Sempere M. Interaction between energy
macogenomics J. 2007;7(4):282–289. 32(5):515–524. homeostasis and reproduction: central effects of
107. Swanson C, Mellström D, Lorentzon M, Vandenput 120. Shiota M, Fujimoto N, Yokomizo A, Takeuchi A, leptin and ghrelin on the reproductive axis. Horm
L, Jakobsson J, Rane A, Karlsson M, Ljunggren O, Kashiwagi E, Dejima T, Kiyoshima K, Inokuchi J, Metab Res. 2013;45(13):919–927.
Smith U, Eriksson AL, Bélanger A, Labrie F, Ohlsson Tatsugami K, Eto M. The prognostic impact of 134. Luukkaa V, Pesonen U, Huhtaniemi I, Lehtonen A,
C. The uridine diphosphate glucuronosyltransferase serum testosterone during androgen-deprivation Tilvis R, Tuomilehto J, Koulu M, Huupponen R.
2B15 D85Y and 2B17 deletion polymorphisms therapy in patients with metastatic prostate can- Inverse correlation between serum testosterone
predict the glucuronidation pattern of androgens cer and the SRD5A2 polymorphism. Prostate Cancer and leptin in men. J Clin Endocrinol Metab. 1998;
and fat mass in men. J Clin Endocrinol Metab. 2007; Prostatic Dis. 2016;19(2):191–196. 83(9):3243–3246.
92(12):4878–4882. 121. Shiota M, Fujimoto N, Takeuchi A, Kashiwagi E, 135. Costanzo PR, Knoblovits P. Male gonadal axis
108. Grigorova M, Punab M, Zilaitienė B, Erenpreiss J, Dejima T, Inokuchi J, Tatsugami K, Yokomizo A, function in patients with type 2 diabetes. Horm Mol
Ausmees K, Matuleviĉius V, Tsarev I, Jørgensen N, Kajioka S, Uchiumi T, Eto M. The association Biol Clin Investig. 2016;26(2):129–134.
Laan M. Genetically determined dosage of follicle- of polymorphisms in the gene encoding gonadotropin- 136. Pittas AG, Joseph NA, Greenberg AS. Adipocyto-
stimulating hormone (FSH) affects male repro- releasing hormone with serum testosterone level kines and insulin resistance. J Clin Endocrinol Metab.
ductive parameters. J Clin Endocrinol Metab. 2011; during androgen deprivation therapy and prognosis 2004;89(2):447–452.
96(9):E1534–E1541. of metastatic prostate cancer. J Urol. 2018;199(3): 137. Veldhuis J, Yang R, Roelfsema F, Takahashi P.
109. Jin G, Sun J, Kim ST, Feng J, Wang Z, Tao S, Chen Z, 734–740. Proinflammatory cytokine infusion attenuates LH’s
Purcell L, Smith S, Isaacs WB, Rittmaster RS, Zheng 122. Allan CA, McLachlan RI. Androgens and obesity. feedforward on testosterone secretion: modulation
SL, Condreay LD, Xu J. Genome-wide association Curr Opin Endocrinol Diabetes Obes. 2010;17(3): by age. J Clin Endocrinol Metab. 2016;101(2):
study identifies a new locus JMJD1C at 10q21 that 224–232. 539–549.
may influence serum androgen levels in men. Hum 123. Fui MN, Dupuis P, Grossmann M. Lowered tes- 138. Pagotto U, Marsicano G, Cota D, Lutz B, Pasquali R.
Mol Genet. 2012;21(23):5222–5228. tosterone in male obesity: mechanisms, morbidity The emerging role of the endocannabinoid system
110. Sakka C, Efstathiadou ZA, Polyzos SA, Goutou M, and management. Asian J Androl. 2014;16(2): in endocrine regulation and energy balance. Endocr
Stakias N, Koukoulis GN. Associations of estrogen 223–231. Rev. 2006;27(1):73–100.

960 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

139. Porte D Jr, Baskin DG, Schwartz MW. Insulin sig- and sexual desire on men in the month of Ram- insulin-like growth factor-1 levels, mental reaction
naling in the central nervous system: a critical role in adan. Urol J. 2015;12(2):2099–2102. time, and maximal aerobic exercise in sedentary and
metabolic homeostasis and disease from C. elegans 154. Derouiche A, Jafri A, Driouch I, El Khasmi M, long-term physically trained elderly males. Int J
to humans. Diabetes. 2005;54(5):1264–1276. Adlouni A, Benajiba N, Bamou Y, Saile R, Neurosci. 2004;114(5):623–637.
140. George JT, Millar RP, Anderson RA. Hypothesis: Benouhoud M. Effect of argan and olive oil con- 170. Lovell DI, Cuneo R, Wallace J, McLellan C. The
kisspeptin mediates male hypogonadism in obesity sumption on the hormonal profile of androgens hormonal response of older men to sub-maximum
and type 2 diabetes. Neuroendocrinology. 2010;91(4): among healthy adult Moroccan men. Nat Prod aerobic exercise: the effect of training and
302–307. Commun. 2013;8(1):51–53. detraining. Steroids. 2012;77(5):413–418.
141. Tajar A, Forti G, O’Neill TW, Lee DM, Silman AJ, Finn 155. Gardner-Thorpe D, O’Hagen C, Young I, Lewis SJ. 171. Arazi H, Damirchi A, Asadi A. Age-related hormonal
JD, Bartfai G, Boonen S, Casanueva FF, Giwercman A, Dietary supplements of soya flour lower serum adaptations, muscle circumference and strength
Han TS, Kula K, Labrie F, Lean ME, Pendleton N, testosterone concentrations and improve markers development with 8 weeks moderate intensity
Punab M, Vanderschueren D, Huhtaniemi IT, Wu of oxidative stress in men. Eur J Clin Nutr. 2003;57(1): resistance training. Ann Endocrinol (Paris). 2013;
FC; EMAS Group. Characteristics of secondary, 100–106. 74(1):30–35.
primary, and compensated hypogonadism in aging 156. Nagata C, Takatsuka N, Shimizu H, Hayashi H, 172. Hayes LD, Sculthorpe N, Herbert P, Baker JS, Hullin
men: evidence from the European Male Ageing Akamatsu T, Murase K. Effect of soymilk con- DA, Kilduff LP, Grace FM. Resting steroid hormone
Study. J Clin Endocrinol Metab. 2010;95(4):1810– sumption on serum estrogen and androgen con- concentrations in lifetime exercisers and lifetime
1818. centrations in Japanese men. Cancer Epidemiol sedentary males. Aging Male. 2015;18(1):22–26.

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


142. Dhindsa S, Furlanetto R, Vora M, Ghanim H, Biomarkers Prev. 2001;10(3):179–184. 173. Sato K, Iemitsu M, Matsutani K, Kurihara T,
Chaudhuri A, Dandona P. Low estradiol concen- 157. Janevic T, Kahn LG, Landsbergis P, Cirillo PM, Cohn Hamaoka T, Fujita S. Resistance training restores
trations in men with subnormal testosterone BA, Liu X, Factor-Litvak P. Effects of work and life muscle sex steroid hormone steroidogenesis in
concentrations and type 2 diabetes. Diabetes Care. stress on semen quality. Fertil Steril. 2014;102(2): older men. FASEB J. 2014;28(4):1891–1897.
2011;34(8):1854–1859. 530–538. 174. Hayes LD, Herbert P, Sculthorpe NF, Grace FM.
143. Dhindsa S, Batra M, Kuhadiya N, Dandona P. 158. Nordkap L, Jensen TK, Hansen ÅM, Lassen TH, Bang Exercise training improves free testosterone in
Oestradiol concentrations are not elevated in AK, Joensen UN, Blomberg Jensen M, Skakkebæk lifelong sedentary aging men. Endocr Connect. 2017;
obesity-associated hypogonadotrophic hypo- NE, Jørgensen N. Psychological stress and testicular 6(5):306–310.
gonadism. Clin Endocrinol (Oxf). 2014;80(3):464. function: a cross-sectional study of 1,215 Danish 175. Tissandier O, Péres G, Fiet J, Piette F. Testosterone,
144. Pellitero S, Olaizola I, Alastrue A, Martı́nez E, men. Fertil Steril. 2016;105(1):174–187.e1–2. dehydroepiandrosterone, insulin-like growth factor
Granada ML, Balibrea JM, Moreno P, Serra A, 159. Fabre B, Machulsky NF, Grosman H, Gonzalez D, 1, and insulin in sedentary and physically trained
Navarro-Dı́az M, Romero R, Puig-Domingo M. Oneto A, Repetto EM, Mesch V, Nolazco C, Mazza aged men. Eur J Appl Physiol. 2001;85(1–2):177–184.
O, Gidron Y, Berg G. Life events are positively as- 176. Kostka T, Patricot MC, Mathian B, Lacour JR,
Hypogonadotropic hypogonadism in morbidly
sociated with luteinizing hormone in middle age Bonnefoy M. Anabolic and catabolic hormonal
obese males is reversed after bariatric surgery. Obes
adult men: role of cortisol as a third variable. Stress.
Surg. 2012;22(12):1835–1842. responses to experimental two-set low-volume
2014;17(4):328–333.
145. Aarts E, van Wageningen B, Loves S, Janssen I, resistance exercise in sedentary and active elderly
160. Lennartsson AK, Kushnir MM, Bergquist J, Billig H,
Berends F, Sweep F, de Boer H. Gonadal status and people. Aging Clin Exp Res. 2003;15(2):123–130.
Jonsdottir IH. Sex steroid levels temporarily increase
outcome of bariatric surgery in obese men. Clin 177. Khoo J, Tian HH, Tan B, Chew K, Ng CS, Leong D,
in response to acute psychosocial stress in healthy
Endocrinol (Oxf). 2014;81(3):378–386. Teo RC, Chen RY. Comparing effects of low- and
men and women. Int J Psychophysiol. 2012;84(3):
146. Chosich J, Bradford AP, Allshouse AA, Reusch JE, high-volume moderate-intensity exercise on sexual
246–253.
Santoro N, Schauer IE. Acute recapitulation of the function and testosterone in obese men. J Sex Med.
161. Hardy MP, Gao H-B, Dong Q, Ge R, Wang Q, Chai
hyperinsulinemia and hyperlipidemia characteristic 2013;10(7):1823–1832.
WR, Feng X, Sottas C. Stress hormone and male
of metabolic syndrome suppresses gonadotropins. 178. Hayes LD, Sculthorpe N, Herbert P, Baker JS, Spagna
reproductive function. Cell Tissue Res. 2005;322(1):
Obesity (Silver Spring). 2017;25(3):553–560. R, Grace FM. Six weeks of conditioning exercise
147–153.
147. Hämäläinen E, Adlercreutz H, Puska P, Pietinen P. increases total, but not free testosterone in lifelong
162. Deslypere JP, Vermeulen A. Leydig cell function in
Diet and serum sex hormones in healthy men. normal men: effect of age, life-style, residence, diet, sedentary aging men. Aging Male. 2015;18(3):
J Steroid Biochem. 1984;20(1):459–464. and activity. J Clin Endocrinol Metab. 1984;59(5): 195–200.
148. Dorgan JF, Judd JT, Longcope C, Brown C, Schatzkin 955–962. 179. Yeap BB, Almeida OP, Hyde Z, Norman PE, Chubb
A, Clevidence BA, Campbell WS, Nair PP, Franz C, 163. Cumming DC, Wheeler GD, McColl EM. The effects SA, Jamrozik K, Hankey GJ, Flicker L. Healthier
Kahle L, Taylor PR. Effects of dietary fat and fiber on of exercise on reproductive function in men. Sports lifestyle predicts higher circulating testosterone in
plasma and urine androgens and estrogens in men: Med. 1989;7(1):1–17. older men: the Health in Men Study. Clin Endocrinol
a controlled feeding study. Am J Clin Nutr. 1996; 164. Wheeler GD, Singh M, Pierce WD, Epling WF, (Oxf). 2009;70(3):455–463.
64(6):850–855. Cumming DC. Endurance training decreases serum 180. Zhao J, Leung JY, Lin SL, Schooling CM. Cigarette
149. Wang C, Catlin DH, Starcevic B, Heber D, Ambler C, testosterone levels in men without change in smoking and testosterone in men and women: a
Berman N, Lucas G, Leung A, Schramm K, Lee PW, luteinizing hormone pulsatile release. J Clin Endo- systematic review and meta-analysis of observa-
Hull L, Swerdloff RS. Low-fat high-fiber diet de- crinol Metab. 1991;72(2):422–425. tional studies. Prev Med. 2016;85(1):1–10.
creased serum and urine androgens in men. J Clin 165. Kupchak BR, Kraemer WJ, Hoffman MD, Phinney 181. Field AE, Colditz GA, Willett WC, Longcope C,
Endocrinol Metab. 2005;90(6):3550–3559. SD, Volek JS. The impact of an ultramarathon on McKinlay JB. The relation of smoking, age, relative
150. Sallinen J, Pakarinen A, Fogelholm M, Alen M, Volek hormonal and biochemical parameters in men. weight, and dietary intake to serum adrenal steroids,
JS, Kraemer WJ, Häkkinen K. Dietary intake, serum Wilderness Environ Med. 2014;25(3):278–288. sex hormones, and sex hormone-binding globulin in
hormones, muscle mass and strength during 166. Häkkinen K, Pakarinen A. Acute hormonal re- middle-aged men. J Clin Endocrinol Metab. 1994;
strength training in 49 - 73-year-old men. Int J Sports sponses to heavy resistance exercise in men and 79(5):1310–1316.
Med. 2007;28(12):1070–1076. women at different ages. Int J Sports Med. 1995; 182. English KM, Pugh PJ, Parry H, Scutt NE, Channer KS,
151. Moran LJ, Brinkworth GD, Martin S, Wycherley TP, 16(8):507–513. Jones TH. Effect of cigarette smoking on levels of
Stuckey B, Lutze J, Clifton PM, Wittert GA, Noakes 167. Häkkinen K, Pakarinen A, Newton RU, Kraemer WJ. bioavailable testosterone in healthy men. Clin Sci
M. Long-term effects of a randomised controlled Acute hormone responses to heavy resistance (Lond). 2001;100(6):661–665.
trial comparing high protein or high carbohydrate lower and upper extremity exercise in young versus 183. Schooling CM. Smoking, sex, risk factors and ab-
weight loss diets on testosterone, SHBG, erectile old men. Eur J Appl Physiol Occup Physiol. 1998; dominal aortic aneurysm: is it all down to testos-
and urinary function in overweight and obese men. 77(4):312–319. terone? J Epidemiol Community Health. 2015;69(5):
PLoS One. 2016;11(9):e0161297. 168. Cooper CS, Taaffe DR, Guido D, Packer E, Holloway 495.
152. Armamento-Villareal R, Aguirre LE, Qualls C, L, Marcus R. Relationship of chronic endurance 184. Välimäki MJ, Härkönen M, Eriksson CJ, Ylikahri RH.
Villareal DT. Effect of lifestyle intervention on the exercise to the somatotropic and sex hormone Sex hormones and adrenocortical steroids in men
hormonal profile of frail, obese older men. J Nutr status of older men. Eur J Endocrinol. 1998;138(5): acutely intoxicated with ethanol. Alcohol. 1984;1(1):
Health Aging. 2016;20(3):334–340. 517–523. 89–93.
153. Talib RA, Canguven O, Al-Rumaihi K, Al Ansari A, 169. Ari Z, Kutlu N, Uyanik BS, Taneli F, Buyukyazi G, 185. Sparrow D, Bosse R, Rowe JW. The influence of age,
Alani M. The effect of fasting on erectile function Tavli T. Serum testosterone, growth hormone, and alcohol consumption, and body build on gonadal

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 961


REVIEW

function in men. J Clin Endocrinol Metab. 1980;51(3): abnormalities and implications. J Gastroenterol TT; Multicenter AIDS Cohort Study. Longitudinal
508–512. Hepatol. 2015;30(2):244–251. changes over 10 years in free testosterone among
186. Shiels MS, Rohrmann S, Menke A, Selvin E, Crespo 201. Grossmann M, Hoermann R, Gani L, Chan I, Cheung HIV-infected and HIV-uninfected men. J Acquir
CJ, Rifai N, Dobs A, Feinleib M, Guallar E, Platz EA. A, Gow PJ, Li A, Zajac JD, Angus P. Low testosterone Immune Defic Syndr. 2016;71(1):57–64.
Association of cigarette smoking, alcohol con- levels as an independent predictor of mortality in 217. Zirkin BR, Santulli R, Strandberg JD, Wright WW,
sumption, and physical activity with sex steroid men with chronic liver disease. Clin Endocrinol (Oxf). Ewing LL. Testicular steroidogenesis in the aging
hormone levels in US men. Cancer Causes Control. 2012;77(2):323–328. brown Norway rat. J Androl. 1993;14(2):118–123.
2009;20(6):877–886. 202. Kim S, Kwon H, Park JH, Cho B, Kim D, Oh SW, Lee 218. Wang Y, Chen F, Ye L, Zirkin B, Chen H. Ste-
187. Venkat KK, Arora MM, Singh P, Desai M, CM, Choi HC. A low level of serum total testos- roidogenesis in Leydig cells: effects of aging and
Khatkhatay I. Effect of alcohol consumption on terone is independently associated with non- environmental factors. Reproduction. 2017;154(4):
bone mineral density and hormonal parameters in alcoholic fatty liver disease. BMC Gastroenterol. R111–R122.
physically active male soldiers. Bone. 2009;45(3): 2012;12(1):69. 219. Chen H, Hardy MP, Huhtaniemi I, Zirkin BR. Age-
449–454. 203. Iglesias P, Carrero JJ, Dı́ez JJ. Gonadal dysfunction in related decreased Leydig cell testosterone pro-
188. Jensen TK, Swan S, Jørgensen N, Toppari J, Redmon men with chronic kidney disease: clinical features, duction in the brown Norway rat. J Androl. 1994;
B, Punab M, Drobnis EZ, Haugen TB, Zilaitiene B, prognostic implications and therapeutic options. 15(6):551–557.
Sparks AE, Irvine DS, Wang C, Jouannet P, Brazil C, J Nephrol. 2012;25(1):31–42. 220. Chen H, Hardy MP, Zirkin BR. Age-related decreases
Paasch U, Salzbrunn A, Skakkebæk NE, Andersson 204. Wu HC, Lee LC, Wang WJ. The association between in Leydig cell testosterone production are not re-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


AM. Alcohol and male reproductive health: a cross- serum testosterone and mortality among elderly stored by exposure to LH in vitro. Endocrinology.
sectional study of 8344 healthy men from Europe men on hemodialysis. J Clin Lab Anal. 2018;32(5): 2002;143(5):1637–1642.
and the USA. Hum Reprod. 2014;29(8):1801–1809. e22394. 221. Sokanovic SJ, Janjic MM, Stojkov NJ, Baburski AZ,
189. Sarkola T, Eriksson CJ. Testosterone increases in men 205. Nakashima A, Ohkido I, Yokoyama K, Mafune A, Bjelic MM, Andric SA, Kostic TS. Age related
after a low dose of alcohol. Alcohol Clin Exp Res. Urashima M, Yokoo T. Associations between low changes of cAMP and MAPK signaling in Leydig
2003;27(4):682–685. serum testosterone and all-cause mortality and cells of Wistar rats. Exp Gerontol. 2014;58(1):19–29.
190. Castilla-Garcı́a A, Santolaria-Fernández FJ, González- infection-related hospitalization in male hemodi- 222. Liao C, Reaven E, Azhar S. Age-related decline in the
Reimers CE, Batista-López N, González-Garcı́a C, alysis patients: a prospective cohort study. Kidney steroidogenic capacity of isolated rat Leydig cells: a
Jorge-Hernández JA, Hernández-Nieto L. Alcohol- Int Rep. 2017;2(6):1160–1168. defect in cholesterol mobilization and processing.
induced hypogonadism: reversal after ethanol 206. Kamischke A, Kemper DE, Castel MA, Lüthke M, J Steroid Biochem Mol Biol. 1993;46(1):39–47.
withdrawal. Drug Alcohol Depend. 1987;20(3): Rolf C, Behre HM, Magnussen H, Nieschlag E. 223. Hales DB. Testicular macrophage modulation of
255–260. Testosterone levels in men with chronic obstructive Leydig cell steroidogenesis. J Reprod Immunol. 2002;
191. Schooling CM, Au Yeung SL, Freeman G, Cowling BJ. pulmonary disease with or without glucocorticoid 57(1-2):3–18.
The effect of statins on testosterone in men and therapy. Eur Respir J. 1998;11(1):41–45. 224. Rebrin I, Kamzalov S, Sohal RS. Effects of age and
women, a systematic review and meta-analysis of 207. Van Vliet M, Spruit MA, Verleden G, Kasran A, Van caloric restriction on glutathione redox state in
randomized controlled trials. BMC Med. 2013;11(1): Herck E, Pitta F, Bouillon R, Decramer M. Hypo- mice. Free Radic Biol Med. 2003;35(6):626–635.
57. gonadism, quadriceps weakness, and exercise in- 225. Hanukoglu I. Antioxidant protective mechanisms
192. Cepeda MS, Zhu V, Vorsanger G, Eichenbaum G. tolerance in chronic obstructive pulmonary disease. against reactive oxygen species (ROS) generated by
Effect of opioids on testosterone levels: cross- Am J Respir Crit Care Med. 2005;172(9):1105–1111. mitochondrial P450 systems in steroidogenic cells.
sectional study using NHANES. Pain Med. 2015; 208. Gordon D, Beastall GH, Thomson JA, Sturrock RD. Drug Metab Rev. 2006;38(1–2):171–196.
16(12):2235–2242. Androgenic status and sexual function in males 226. Luo L, Chen H, Trush MA, Show MD, Anway MD,
193. Nakagawa K, Obara T, Matsubara M, Kubo M. with rheumatoid arthritis and ankylosing spondy- Zirkin BR. Aging and the brown Norway rat Leydig
Relationship of changes in serum concentrations of litis. Q J Med. 1986;60(231):671–679. cell antioxidant defense system. J Androl. 2006;27(2):
prolactin and testosterone during dopaminergic 209. Cutolo M, Balleari E, Giusti M, Monachesi M, 240–247.
modulation in males. Clin Endocrinol (Oxf). 1982; Accardo S. Sex hormone status of male patients 227. Paniagua R, Nistal M, Sáez FJ, Fraile B. Ultrastructure
17(4):345–352. with rheumatoid arthritis: evidence of low serum of the aging human testis. J Electron Microsc Tech.
194. MacAdams MR, White RH, Chipps BE. Reduction of concentrations of testosterone at baseline and after 1991;19(2):241–260.
serum testosterone levels during chronic gluco- human chorionic gonadotropin stimulation. Ar- 228. Paniagua R, Amat P, Nistal M, Martin A. Ultra-
corticoid therapy. Ann Intern Med. 1986;104(5): thritis Rheum. 1988;31(10):1314–1317. structure of Leydig cells in human ageing testes.
648–651. 210. Siminoski K, D’Costa M, Walfish PG. Hypo- J Anat. 1986;146:173–183.
195. Kristensen DM, Desdoits-Lethimonier C, Mackey gonadotropic hypogonadism in idiopathic hemo- 229. Wu FC, Tajar A, Beynon JM, Pye SR, Silman AJ, Finn
AL, Dalgaard MD, De Masi F, Munkbøl CH, chromatosis: evidence for combined hypothalamic JD, O’Neill TW, Bartfai G, Casanueva FF, Forti G,
Styrishave B, Antignac JP, Le Bizec B, Platel C, Hay- and pituitary involvement. J Endocrinol Invest. 1990; Giwercman A, Han TS, Kula K, Lean ME, Pendleton
Schmidt A, Jensen TK, Lesné L, Mazaud-Guittot S, 13(10):849–853. N, Punab M, Boonen S, Vanderschueren D, Labrie F,
Kristiansen K, Brunak S, Kjaer M, Juul A, Jégou B. 211. Dobs AS, Few WL III, Blackman MR, Harman SM, Huhtaniemi IT; EMAS Group. Identification of late-
Ibuprofen alters human testicular physiology to Hoover DR, Graham NM. Serum hormones in men onset hypogonadism in middle-aged and elderly
produce a state of compensated hypogonadism with human immunodeficiency virus-associated men. N Engl J Med. 2010;363(2):123–135.
(published correction appears inProc Natl Acad Sci wasting. J Clin Endocrinol Metab. 1996;81(11): 230. Longcope C. The effect of human chorionic go-
USA.2018;115(17):E4143). Proc Natl Acad Sci USA. 4108–4112. nadotropin on plasma steroid levels in young and
2018;115(4):E715–E724. 212. Monroe AK, Dobs AS, Palella FJ, Kingsley LA, Witt old men. Steroids. 1973;21(4):583–592.
196. Kalyani RR, Gavini S, Dobs AS. Male hypogonadism MD, Brown TT. Morning free and total testosterone 231. Winters SJ, Troen P. Episodic luteinizing hormone
in systemic disease. Endocrinol Metab Clin North in HIV-infected men: implications for the assess- (LH) secretion and the response of LH and follicle-
Am. 2007;36(2):333–348. ment of hypogonadism. AIDS Res Ther. 2014;11(1):6. stimulating hormone to LH-releasing hormone in
197. Grossmann M, Matsumoto AM. A perspective on 213. Schürmeyer TH, Müller V, von zur Mühlen A, aged men: evidence for coexistent primary testic-
middle-aged and older men with functional Schmidt RE. Endocrine testicular function in HIV- ular insufficiency and an impairment in gonado-
hypogonadism: focus on holistic management. J Clin infected outpatients. Eur J Med Res. 1997;2(7): tropin secretion. J Clin Endocrinol Metab. 1982;55(3):
Endocrinol Metab. 2017;102(3):1067–1075. 275–281. 560–565.
198. Völzke H, Aumann N, Krebs A, Nauck M, Steveling 214. Arver S, Sinha-Hikim I, Beall G, Guerrero M, Shen R, 232. Veldhuis JD, Veldhuis NJ, Keenan DM, Iranmanesh
A, Lerch MM, Rosskopf D, Wallaschofski H. Hepatic Bhasin S. Serum dihydrotestosterone and testos- A. Age diminishes the testicular steroidogenic re-
steatosis is associated with low serum testosterone terone concentrations in human immunodefi- sponse to repeated intravenous pulses of recombinant
and high serum DHEAS levels in men. Int J Androl. ciency virus-infected men with and without weight human LH during acute GnRH-receptor blockade in
2010;33(1):45–53. loss. J Androl. 1999;20(5):611–618. healthy men. Am J Physiol Endocrinol Metab. 2005;
199. Longcope C, Pratt JH, Schneider S, Fineberg E. Es- 215. Rochira V, Guaraldi G. Hypogonadism in the HIV- 288(4):E775–E781.
trogen and androgen dynamics in liver disease. infected man. Endocrinol Metab Clin North Am. 233. Veldhuis JD, Liu PY, Keenan DM, Takahashi PY.
J Endocrinol Invest. 1984;7(6):629–634. 2014;43(3):709–730. Older men exhibit reduced efficacy of and
200. Sinclair M, Grossmann M, Gow PJ, Angus PW. 216. Slama L, Jacobson LP, Li X, Palella FJ Jr, Margolick JB, heightened potency downregulation by in-
Testosterone in men with advanced liver disease: Kingsley LA, Wiley DJ, Pialoux G, Dobs AS, Brown travenous pulses of recombinant human LH: a

962 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

study in 92 healthy men. Am J Physiol Endocrinol humans. J Gerontol A Biol Sci Med Sci. 2008;63(5): of estrogen receptors with E2 downregulates per-
Metab. 2012;302(1):E117–E122. 447–453. oxisome proliferator-activated receptor g in he-
234. Kaufman JM, Giri M, Deslypere JM, Thomas G, 246. Simó R, Sáez-López C, Barbosa-Desongles A, patocellular carcinoma. Oncol Rep. 2013;30(6):
Vermeulen A. Influence of age on the re- Hernández C, Selva DM. Novel insights in SHBG 3027–3031.
sponsiveness of the gonadotrophs to luteinizing regulation and clinical implications. Trends Endo- 261. Plaisance EP, Grandjean PW, Judd RL, Jones KW,
hormone-releasing hormone in males. J Clin Endo- crinol Metab. 2015;26(7):376–383. Taylor JK. The influence of sex, body composition,
crinol Metab. 1991;72(6):1255–1260. 247. Lewerin C, Johansson H, Lerner UH, Karlsson MK, and nonesterified fatty acids on serum adipokine
235. Iranmanesh A, Mulligan T, Veldhuis JD. Age in men Lorentzon M, Barrett-Connor E, Smith U, Ohlsson concentrations. Metabolism. 2009;58(11):1557–
does not determine gonadotropin-releasing hor- C, Mellström D. High serum adiponectin is asso- 1563.
mone’s dose-dependent stimulation of luteinizing ciated with low blood haemoglobin in elderly men: 262. Sáez-López C, Soriguer F, Hernandez C, Rojo-
hormone secretion under an exogenous testos- the Swedish MrOS study. J Intern Med. 2015;278(1): Martinez G, Rubio-Martı́n E, Simó R, Selva DM.
terone clamp. J Clin Endocrinol Metab. 2010;95(6): 68–76. Oleic acid increases hepatic sex hormone binding
2877–2884. 248. Lanfranco F, Zitzmann M, Simoni M, Nieschlag E. globulin production in men. Mol Nutr Food Res.
236. Deslypere JP, Kaufman JM, Vermeulen T, Vogelaers Serum adiponectin levels in hypogonadal males: 2014;58(4):760–767.
D, Vandalem JL, Vermeulen A. Influence of age on influence of testosterone replacement therapy. Clin 263. Takahashi J, Higashi Y, LaNasa JA, Yoshida K,
pulsatile luteinizing hormone release and re- Endocrinol (Oxf). 2004;60(4):500–507. Winters SJ, Oshima H, Troen P. Studies of the
sponsiveness of the gonadotrophs to sex hormone 249. Vermeulen A, Kaufman JM, Giagulli VA. Influence of human testis. XVIII. Simultaneous measurement of

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


feedback in men. J Clin Endocrinol Metab. 1987; some biological indexes on sex hormone-binding nine intratesticular steroids: evidence for reduced
64(1):68–73. globulin and androgen levels in aging or obese mitochondrial function in testis of elderly men.
237. Veldhuis JD, Keenan DM, Liu PY, Iranmanesh A, males. J Clin Endocrinol Metab. 1996;81(5): J Clin Endocrinol Metab. 1983;56(6):1178–1187.
Takahashi PY, Nehra AX. The aging male 1821–1826. 264. Hammar M, Ahlstrand C, Berg AA, Läckgren G. In
hypothalamic–pituitary–gonadal axis: pulsatility 250. Lecomte P, Lecureuil N, Lecureuil M, Lemonnier Y, vitro conversion of 3H-pregnenolone by human
and feedback. Mol Cell Endocrinol. 2009;299(1): Mariotte N, Valat C, Garrigue MA. Sex differences in testicular tissue from fetal age to senescence. Arch
14–22. the control of sex-hormone-binding globulin in the Androl. 1984;13(2–3):203–212.
238. Liu PY, Takahashi PY, Roebuck PD, Bailey JN, Keenan elderly: role of insulin-like growth factor-I and in- 265. Hammar M, Petersson F, Berg AA. In vitro con-
DM, Veldhuis JD. Testosterone’s short-term positive sulin. Eur J Endocrinol. 1998;139(2):178–183. version of progesterone in the human testis at
effect on luteinizing-hormone secretory-burst mass 251. Vanbillemont G, Lapauw B, De Naeyer H, Roef G, different ages, pathophysiological conditions, and
and its negative effect on secretory-burst frequency Kaufman JM, Taes YE. Sex hormone-binding during treatment with estrogens or gonadotrophic
are attenuated in middle-aged men. J Clin Endo- globulin at the crossroad of body composition, hormones. Arch Androl. 1985;14(2–3):143–149.
crinol Metab. 2009;94(10):3978–3986. somatotropic axis and insulin/glucose homeostasis 266. Pirke KM, Sintermann R, Vogt HJ. Testosterone and
239. Eendebak RJAH, Ahern T, Swiecicka A, Pye SR, in young healthy men. Clin Endocrinol (Oxf). 2012; testosterone precursors in the spermatic vein and in
O’Neill TW, Bartfai G, Casanueva FF, Maggi M, Forti 76(1):111–118. the testicular tissue of old men. Reduced oxygen
252. Kalme T, Koistinen H, Loukovaara M, Koistinen R, supply may explain the relative increase of testicular
G, Giwercman A, Han TS, Słowikowska-Hilczer J,
Leinonen P. Comparative studies on the regulation progesterone and 17a-hydroxyprogesterone con-
Lean MEJ, Punab M, Pendleton N, Keevil BG,
of insulin-like growth factor-binding protein-1 tent and production in old age. Gerontology. 1980;
Vanderschueren D, Rutter MK, Tampubolon G,
(IGFBP-1) and sex hormone-binding globulin 26(4):221–230.
Goodacre R, Huhtaniemi IT, Wu FCW; EMAS
(SHBG) production by insulin and insulin-like 267. Vermeulen A, Deslypere JP. Intratesticular un-
Group. Elevated luteinizing hormone despite nor-
growth factors in human hepatoma cells. J Steroid conjugated steroids in elderly men. J Steroid Bio-
mal testosterone levels in older men-natural history,
Biochem Mol Biol. 2003;86(2):197–200. chem. 1986;24(5):1079–1083.
risk factors and clinical features. Clin Endocrinol
253. Selva DM, Hogeveen KN, Innis SM, Hammond GL. 268. Bélanger A, Candas B, Dupont A, Cusan L, Diamond
(Oxf). 2018;88(3):479–490.
Monosaccharide-induced lipogenesis regulates the P, Gomez JL, Labrie F. Changes in serum concen-
240. Molnár CS, Vida B, Sipos MT, Ciofi P, Borsay BÁ, Rácz
human hepatic sex hormone-binding globulin gene. trations of conjugated and unconjugated steroids in
K, Herczeg L, Bloom SR, Ghatei MA, Dhillo WS,
J Clin Invest. 2007;117(12):3979–3987. 40- to 80-year-old men. J Clin Endocrinol Metab.
Liposits Z, Hrabovszky E. Morphological evidence 254. Ding EL, Song Y, Manson JE, Hunter DJ, Lee CC, Rifai 1994;79(4):1086–1090.
for enhanced kisspeptin and neurokinin B signaling N, Buring JE, Gaziano JM, Liu S. Sex hormone- 269. Hardy MP, Schlegel PN. Testosterone production in
in the infundibular nucleus of the aging man. En- binding globulin and risk of type 2 diabetes in the aging male: where does the slowdown occur?
docrinology. 2012;153(11):5428–5439. women and men. N Engl J Med. 2009;361(12): Endocrinology. 2004;145(10):4439–4440.
241. Sánchez-Garrido MA, Ruiz-Pino F, Manfredi-Lozano 1152–1163. 270. Mulligan T, Iranmanesh A, Veldhuis JD. Pulsatile iv
M, Leon S, Garcia-Galiano D, Castaño JP, Luque RM, 255. Simó R, Barbosa-Desongles A, Lecube A, Hernandez infusion of recombinant human LH in leuprolide-
Romero-Ruiz A, Castellano JM, Diéguez C, Pinilla L, C, Selva DM. Potential role of tumor necrosis factor- suppressed men unmasks impoverished Leydig-cell
Tena-Sempere M. Obesity-induced hypogonadism a in downregulating sex hormone-binding globulin. secretory responsiveness to midphysiological LH
in the male: premature reproductive neuroendo- Diabetes. 2012;61(2):372–382. drive in the aging male. J Clin Endocrinol Metab.
crine senescence and contribution of Kiss1- 256. Simó R, Barbosa-Desongles A, Hernandez C, Selva 2001;86(11):5547–5553.
mediated mechanisms. Endocrinology. 2014;155(3): DM. IL1b down-regulation of sex hormone-binding 271. Vanderschueren D, Laurent MR, Claessens F, Gielen
1067–1079. globulin production by decreasing HNF-4a via E, Lagerquist MK, Vandenput L, Börjesson AE,
242. Kunimura Y, Iwata K, Ishigami A, Ozawa H. Age- MEK-1/2 and JNK MAPK pathways. Mol Endocrinol. Ohlsson C. Sex steroid actions in male bone. Endocr
related alterations in hypothalamic kisspeptin, 2012;26(11):1917–1927. Rev. 2014;35(6):906–960.
neurokinin B, and dynorphin neurons and in pul- 257. Weyer C, Funahashi T, Tanaka S, Hotta K, 272. Stoch SA, Parker RA, Chen L, Bubley G, Ko YJ,
satile LH release in female and male rats. Neurobiol Matsuzawa Y, Pratley RE, Tataranni PA. Hypo- Vincelette A, Greenspan SL. Bone loss in men with
Aging. 2017;50(1):30–38. adiponectinemia in obesity and type 2 diabetes: prostate cancer treated with gonadotropin-releasing
243. Eghlidi DH, Garyfallou VT, Kohama SG, Urbanski HF. close association with insulin resistance and hormone agonists. J Clin Endocrinol Metab. 2001;
Age-associated gene expression changes in the hyperinsulinemia. J Clin Endocrinol Metab. 2001; 86(6):2787–2791.
arcuate nucleus of male rhesus macaques. J Mol 86(5):1930–1935. 273. Mittan D, Lee S, Miller E, Perez RC, Basler JW, Bruder
Endocrinol. 2017;59(2):141–149. 258. Hammond GL. Plasma steroid-binding proteins: JM. Bone loss following hypogonadism in men with
244. Cnop M, Havel PJ, Utzschneider KM, Carr DB, Sinha primary gatekeepers of steroid hormone action. prostate cancer treated with GnRH analogs. J Clin
MK, Boyko EJ, Retzlaff BM, Knopp RH, Brunzell JD, J Endocrinol. 2016;230(1):R13–R25. Endocrinol Metab. 2002;87(8):3656–3661.
Kahn SE. Relationship of adiponectin to body fat 259. Liu CC, Huang SP, Cheng KH, Hsieh TJ, Huang CN, 274. Smith MR, Fallon MA, Lee H, Finkelstein JS.
distribution, insulin sensitivity and plasma lipo- Wang CJ, Yeh HC, Tsai CC, Bao BY, Wu WJ, Lee YC. Raloxifene to prevent gonadotropin-releasing hor-
proteins: evidence for independent roles of age and Lower SHBG level is associated with higher leptin mone agonist-induced bone loss in men with
sex. Diabetologia. 2003;46(4):459–469. and lower adiponectin levels as well as metabolic prostate cancer: a randomized controlled trial. J Clin
245. Atzmon G, Pollin TI, Crandall J, Tanner K, Schechter syndrome, independent of testosterone. Sci Rep. Endocrinol Metab. 2004;89(8):3841–3846.
CB, Scherer PE, Rincon M, Siegel G, Katz M, Lipton 2017;7(1):2727. 275. Smith MR, Morton RA, Barnette KG, Sieber PR,
RB, Shuldiner AR, Barzilai N. Adiponectin levels and 260. Lin YM, Velmurugan BK, Yeh YL, Tu CC, Ho TJ, Lai Malkowicz SB, Rodriguez D, Hancock ML, Steiner
genotype: a potential regulator of life span in TY, Tsai CH, Tsai FJ, Tsai CH, Huang CY. Activation MS. Toremifene to reduce fracture risk in men

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 963


REVIEW

receiving androgen deprivation therapy for prostate 288. Cauley JA, Ewing SK, Taylor BC, Fink HA, Ensrud KE, Older men with low serum estradiol and high se-
cancer. J Urol. 2010;184(4):1316–1321. Bauer DC, Barrett-Connor E, Marshall L, Orwoll ES; rum SHBG have an increased risk of fractures. J Bone
276. Smith MR, Goode M, Zietman AL, McGovern FJ, Osteoporotic Fractures in Men Study (MrOS) Re- Miner Res. 2008;23(10):1552–1560.
Lee H, Finkelstein JS. Bicalutamide monotherapy search Group. Sex steroid hormones in older men: 302. LeBlanc ES, Nielson CM, Marshall LM, Lapidus JA,
versus leuprolide monotherapy for prostate cancer: longitudinal associations with 4.5-year change in hip Barrett-Connor E, Ensrud KE, Hoffman AR, Laughlin
effects on bone mineral density and body com- bone mineral density—the osteoporotic fractures G, Ohlsson C, Orwoll ES; Osteoporotic Fractures in
position. J Clin Oncol. 2004;22(13):2546–2553. in men study. J Clin Endocrinol Metab. 2010;95(9): Men Study Group. The effects of serum testos-
277. Wadhwa VK, Weston R, Parr NJ. Bicalutamide 4314–4323. terone, estradiol, and sex hormone binding globulin
monotherapy preserves bone mineral density, 289. Woo J, Kwok T, Leung JC, Ohlsson C, Vandenput L, levels on fracture risk in older men. J Clin Endocrinol
muscle strength and has significant health-related Leung PC. Sex steroids and bone health in older Metab. 2009;94(9):3337–3346.
quality of life benefits for osteoporotic men with Chinese men. Osteoporos Int. 2012;23(5):1553–1562. 303. Vandenput L, Mellström D, Kindmark A, Johansson
prostate cancer. BJU Int. 2011;107(12):1923–1929. 290. Finkelstein JS, Lee H, Leder BZ, Burnett-Bowie SA, H, Lorentzon M, Leung J, Redlund-Johnell I,
278. Matsumoto AM, Tenover L, McClung M, Mobley D, Goldstein DW, Hahn CW, Hirsch SC, Linker A, Rosengren BE, Karlsson MK, Wang YX, Kwok T,
Geller J, Sullivan M, Grayhack J, Wessells H, Kadmon Perros N, Servais AB, Taylor AP, Webb ML, Ohlsson C. High serum SHBG predicts incident
D, Flanagan M, Zhang GK, Schmidt J, Taylor AM, Lee Youngner JM, Yu EW. Gonadal steroid-dependent vertebral fractures in elderly men. J Bone Miner Res.
M, Waldstreicher J; Pless Study Group. The long- effects on bone turnover and bone mineral density 2016;31(3):683–689.
term effect of specific type II 5a-reductase in- in men. J Clin Invest. 2016;126(3):1114–1125. 304. Orwoll E, Lambert LC, Marshall LM, Blank J, Barrett-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


hibition with finasteride on bone mineral density in 291. Lormeau C, Soudan B, d’Herbomez M, Pigny P, Connor E, Cauley J, Ensrud K, Cummings SR; Os-
men: results of a 4-year placebo controlled triala.. Duquesnoy B, Cortet B. Sex hormone-binding teoporotic Fractures in Men Study Group.
J Urol. 2002;167(5):2105–2108. globulin, estradiol, and bone turnover markers in Endogenous testosterone levels, physical perfor-
279. Amory JK, Watts NB, Easley KA, Sutton PR, Anawalt male osteoporosis. Bone. 2004;34(6):933–939. mance, and fall risk in older men. Arch Intern Med.
BD, Matsumoto AM, Bremner WJ, Tenover JL. Ex- 292. Bjørnerem A, Emaus N, Berntsen GK, Joakimsen RM, 2006;166(19):2124–2131.
ogenous testosterone or testosterone with finas- Fønnebø V, Wilsgaard T, Oian P, Seeman E, Straume 305. Vandenput L, Mellström D, Laughlin GA, Cawthon
teride increases bone mineral density in older men B. Circulating sex steroids, sex hormone-binding PM, Cauley JA, Hoffman AR, Karlsson MK, Rosengren
with low serum testosterone. J Clin Endocrinol globulin, and longitudinal changes in forearm BE, Ljunggren Ö, Nethander M, Eriksson AL,
Metab. 2004;89(2):503–510. bone mineral density in postmenopausal women Lorentzon M, Leung J, Kwok T, Orwoll ES, Ohlsson C.
280. Borst SE, Yarrow JF, Conover CF, Nseyo U, and men: the Tromsø study. Calcif Tissue Int. 2007; Low testosterone, but not estradiol, is associated
Meuleman JR, Lipinska JA, Braith RW, Beck DT, 81(2):65–72. with incident falls in older men: the International
Martin JS, Morrow M, Roessner S, Beggs LA, McCoy 293. Paller CJ, Shiels MS, Rohrmann S, Basaria S, Rifai N, MrOS Study. J Bone Miner Res. 2017;32(6):1174–
SC, Cannady DF II, Shuster JJ. Musculoskeletal and Nelson W, Platz EA, Dobs A. Relationship of sex 1181.
steroid hormones with bone mineral density (BMD) 306. Vanbillemont G, Lapauw B, Bogaert V, Goemaere S,
prostate effects of combined testosterone and
in a nationally representative sample of men. Clin
finasteride administration in older hypogonadal Zmierczak HG, Taes Y, Kaufman JM. Sex hormone-
Endocrinol (Oxf). 2009;70(1):26–34.
men: a randomized, controlled trial. Am J Physiol binding globulin as an independent determinant of
294. Zha X-Y, Hu Y, Pang X-N, Zhu J-H, Chang G-L, Li L.
Endocrinol Metab. 2014;306(4):E433–E442. cortical bone status in men at the age of peak
Sex hormone-binding globulin (SHBG) as an in-
281. Yeap BB, Alfonso H, Chubb SAP, Gauci R, Byrnes E, bone mass. J Clin Endocrinol Metab. 2010;95(4):
dependent determinant of bone mineral density
Beilby JP, Ebeling PR, Handelsman DJ, Allan CA, 1579–1586.
(BMD) among Chinese middle-aged and elderly
Grossmann M, Norman PE, Flicker L. Higher serum 307. Orwoll ES, Lapidus J, Wang PY, Vandenput L,
men. Endocrine. 2014;47(2):590–597.
undercarboxylated osteocalcin and other bone Hoffman A, Fink HA, Laughlin GA, Nethander M,
295. Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk
turnover markers are associated with reduced di- Ljunggren Ö, Kindmark A, Lorentzon M, Karlsson
of fracture after androgen deprivation for prostate
abetes risk and lower estradiol concentrations in MK, Mellström D, Kwok A, Khosla S, Kwok T,
cancer. N Engl J Med. 2005;352(2):154–164.
older men. J Clin Endocrinol Metab. 2015;100(1): Ohlsson C; Osteoporotic Fractures in Men (MrOS)
296. Bjørnerem A, Ahmed LA, Joakimsen RM, Berntsen
63–71. Study Research Group. The limited clinical utility of
GK, Fønnebø V, Jørgensen L, Øian P, Seeman E,
282. Khosla S, Melton LJ III, Robb RA, Camp JJ, Atkinson testosterone, estradiol and sex hormone binding
Straume B. A prospective study of sex steroids, sex
EJ, Oberg AL, Rouleau PA, Riggs BL. Relationship of hormone-binding globulin, and non-vertebral globulin measurements in the prediction of fracture
volumetric BMD and structural parameters at fractures in women and men: the Tromso Study. risk and bone loss in older men. J Bone Miner Res.
different skeletal sites to sex steroid levels in men. Eur J Endocrinol. 2007;157(1):119–125. 2017;32(3):633–640.
J Bone Miner Res. 2005;20(5):730–740. 297. Meier C, Nguyen TV, Handelsman DJ, Schindler C, 308. Jackson AS, Janssen I, Sui X, Church TS, Blair SN.
283. Khosla S, Melton LJ III, Achenbach SJ, Oberg AL, Kushnir MM, Rockwood AL, Meikle AW, Center JR, Longitudinal changes in body composition asso-
Riggs BL. Hormonal and biochemical determinants Eisman JA, Seibel MJ. Endogenous sex hormones ciated with healthy ageing: men, aged 20–96 years.
of trabecular microstructure at the ultradistal radius and incident fracture risk in older men: the Dubbo Br J Nutr. 2012;107(7):1085–1091.
in women and men. J Clin Endocrinol Metab. 2006; Osteoporosis Epidemiology Study. Arch Intern Med. 309. LeBlanc ES, Wang PY, Lee CG, Barrett-Connor E,
91(3):885–891. 2008;168(1):47–54. Cauley JA, Hoffman AR, Laughlin GA, Marshall LM,
284. Vandenput L, Lorentzon M, Sundh D, Nilsson ME, 298. Cawthon PM, Schousboe JT, Harrison SL, Ensrud KE, Orwoll ES. Higher testosterone levels are associated
Karlsson MK, Mellström D, Ohlsson C. Serum es- Black D, Cauley JA, Cummings SR, LeBlanc ES, with less loss of lean body mass in older men. J Clin
tradiol levels are inversely associated with cortical Laughlin GA, Nielson CM, Broughton A, Kado DM, Endocrinol Metab. 2011;96(12):3855–3863.
porosity in older men. J Clin Endocrinol Metab. 2014; Hoffman AR, Jamal SA, Barrett-Connor E, Orwoll ES; 310. Genton L, Karsegard VL, Chevalley T, Kossovsky MP,
99(7):E1322–E1326. Osteoporotic Fractures in Men (MrOS) Study Re- Darmon P, Pichard C. Body composition changes
285. Khosla S, Melton LJ III, Atkinson EJ, O’Fallon WM. search Group. Sex hormones, sex hormone binding over 9 years in healthy elderly subjects and impact
Relationship of serum sex steroid levels to longi- globulin, and vertebral fractures in older men. Bone. of physical activity. Clin Nutr. 2011;30(4):436–442.
tudinal changes in bone density in young versus 2016;84:271–278. 311. Han TS, Correa E, Lean MEJ, Lee DM, O’Neill TW,
elderly men. J Clin Endocrinol Metab. 2001;86(8): 299. Amin S, Zhang Y, Felson DT, Sawin CT, Hannan MT, Bartfai G, Forti G, Giwercman A, Kula K, Pendleton
3555–3561. Wilson PW, Kiel DP. Estradiol, testosterone, and the N, Punab M, Rutter MK, Vanderschueren D,
286. Gennari L, Merlotti D, Martini G, Gonnelli S, Franci risk for hip fractures in elderly men from the Fra- Huhtaniemi IT, Wu FC, Casanueva FF; and the
B, Campagna S, Lucani B, Dal Canto N, Valenti R, mingham Study. Am J Med. 2006;119(5):426–433. EMAS Study Group. Changes in prevalence of
Gennari C, Nuti R. Longitudinal association be- 300. Roddam AW, Appleby P, Neale R, Dowsett M, obesity and high waist circumference over four
tween sex hormone levels, bone loss, and bone Folkerd E, Tipper S, Allen NE, Key TJ. Association years across European regions: the European male
turnover in elderly men. J Clin Endocrinol Metab. between endogenous plasma hormone concen- ageing study (EMAS). Endocrine. 2017;55(2):456–
2003;88(11):5327–5333. trations and fracture risk in men and women: the 469.
287. Van Pottelbergh I, Goemaere S, Kaufman JM. Bio- EPIC-Oxford prospective cohort study. J Bone Miner 312. Dey DK, Bosaeus I, Lissner L, Steen B. Changes in
available estradiol and an aromatase gene poly- Metab. 2009;27(4):485–493. body composition and its relation to muscle
morphism are determinants of bone mineral 301. Mellström D, Vandenput L, Mallmin H, Holmberg strength in 75-year-old men and women: a 5-year
density changes in men over 70 years of age. J Clin AH, Lorentzon M, Odén A, Johansson H, Orwoll ES, prospective follow-up study of the NORA cohort in
Endocrinol Metab. 2003;88(7):3075–3081. Labrie F, Karlsson MK, Ljunggren O, Ohlsson C. Göteborg, Sweden. Nutrition. 2009;25(6):613–619.

964 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

313. Derby CA, Zilber S, Brambilla D, Morales KH, Metter E, Valenti G, Guralnik JM, Ferrucci L. As- 338. Maffei L, Murata Y, Rochira V, Tubert G, Aranda C,
McKinlay JB. Body mass index, waist circumference sociation between hormones and metabolic syn- Vazquez M, Clyne CD, Davis S, Simpson ER, Carani
and waist to hip ratio and change in sex steroid drome in older Italian men. J Am Geriatr Soc. 2006; C. Dysmetabolic syndrome in a man with a novel
hormones: the Massachusetts Male Ageing Study. 54(12):1832–1838. mutation of the aromatase gene: effects of tes-
Clin Endocrinol (Oxf). 2006;65(1):125–131. 327. Oh JY, Barrett-Connor E, Wedick NM, Wingard DL; tosterone, alendronate, and estradiol treatment.
314. Araujo AB, Travison TG, Bhasin S, Esche GR, Rancho Bernardo Study. Endogenous sex hormones J Clin Endocrinol Metab. 2004;89(1):61–70.
Williams RE, Clark RV, McKinlay JB. Association and the development of type 2 diabetes in older 339. Herrmann BL, Saller B, Janssen OE, Gocke P,
between testosterone and estradiol and age-related men and women. Diabetes Care. 2002;25(1):55–60. Bockisch A, Sperling H, Mann K, Broecker M. Impact
decline in physical function in a diverse sample of 328. Hall SA, Esche GR, Araujo AB, Travison TG, Clark RV, of estrogen replacement therapy in a male with
men. J Am Geriatr Soc. 2008;56(11):2000–2008. Williams RE, McKinlay JB. Correlates of low tes- congenital aromatase deficiency caused by a novel
315. Schaap LA, Pluijm SM, Deeg DJ, Penninx BW, Nicklas tosterone and symptomatic androgen deficiency mutation in the CYP19 gene. J Clin Endocrinol
BJ, Lips P, Harris TB, Newman AB, Kritchevsky SB, in a population-based sample. J Clin Endocrinol Metab. 2002;87(12):5476–5484.
Cauley JA, Goodpaster BH, Tylavsky FA, Yaffe K, Metab. 2008;93(10):3870–3877. 340. Finkelstein JS, Lee H, Burnett-Bowie S-AM, Pallais JC,
Visser M; Health ABC study. Low testosterone levels 329. van den Beld AW, de Jong FH, Grobbee DE, Pols HA, Yu EW, Borges LF, Jones BF, Barry CV, Wulczyn KE,
and decline in physical performance and muscle Lamberts SW. Measures of bioavailable serum Thomas BJ, Leder BZ. Gonadal steroids and body
strength in older men: findings from two pro- testosterone and estradiol and their relationships composition, strength, and sexual function in men.
spective cohort studies. Clin Endocrinol (Oxf). 2008; with muscle strength, bone density, and body N Engl J Med. 2013;369(11):1011–1022.

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


68(1):42–50. composition in elderly men. J Clin Endocrinol Metab. 341. Li C, Ford ES, Li B, Giles WH, Liu S. Association of
316. De Buyser SL, Petrovic M, Taes YE, Toye KRC, 2000;85(9):3276–3282. testosterone and sex hormone-binding globulin
Kaufman JM, Lapauw B, Goemaere S. Validation of 330. Tsai EC, Matsumoto AM, Fujimoto WY, Boyko EJ. with metabolic syndrome and insulin resistance in
the FNIH sarcopenia criteria and SOF frailty index as Association of bioavailable, free, and total testos- men. Diabetes Care. 2010;33(7):1618–1624.
predictors of long-term mortality in ambulatory terone with insulin resistance: influence of sex 342. Barrett-Connor E, Khaw KT, Yen SS. Endogenous sex
older men. Age Ageing. 2016;45(5):602–608. hormone-binding globulin and body fat. Diabetes hormone levels in older adult men with diabetes
317. Yeap BB, McCaul KA, Flicker L, Hankey GJ, Almeida Care. 2004;27(4):861–868. mellitus. Am J Epidemiol. 1990;132(5):895–901.
OP, Golledge J, Norman PE. Diabetes, myocardial 331. Nielsen TL, Hagen C, Wraae K, Brixen K, Petersen 343. Colangelo LA, Ouyang P, Liu K, Kopp P, Golden SH,
infarction and stroke are distinct and duration- PH, Haug E, Larsen R, Andersen M. Visceral and Dobs AS, Szklo M, Vaidya D, Cushman M, Gapstur
dependent predictors of subsequent cardiovascu- subcutaneous adipose tissue assessed by magnetic SM. Association of endogenous sex hormones with
lar events and all-cause mortality in older men. J Clin resonance imaging in relation to circulating an- diabetes and impaired fasting glucose in men:
Endocrinol Metab. 2015;100(3):1038–1047. drogens, sex hormone-binding globulin, and lutei- multi-ethnic study of atherosclerosis. Diabetes Care.
318. Shamliyan T, Talley KM, Ramakrishnan R, Kane RL. nizing hormone in young men. J Clin Endocrinol 2009;32(6):1049–1051.
Association of frailty with survival: a systematic Metab. 2007;92(7):2696–2705. 344. Selvin E, Feinleib M, Zhang L, Rohrmann S, Rifai N,
literature review. Ageing Res Rev. 2013;12(2): 332. Vikan T, Schirmer H, Njølstad I, Svartberg J. Low Nelson WG, Dobs A, Basaria S, Golden SH, Platz EA.
719–736. Androgens and diabetes in men: results from the
testosterone and sex hormone-binding globulin
319. Nguyen PL, Alibhai SMH, Basaria S, D’Amico AV, Third National Health and Nutrition Examination
levels and high estradiol levels are independent
Kantoff PW, Keating NL, Penson DF, Rosario DJ, Survey (NHANES III) (published correction appears
predictors of type 2 diabetes in men. Eur J Endo-
Tombal B, Smith MR. Adverse effects of androgen in Diabetes Care. 2007;30(6):1683). Diabetes Care.
crinol. 2010;162(4):747–754.
deprivation therapy and strategies to mitigate 2007;30(2):234–238.
333. Antonio L, Wu FC, O’Neill TW, Pye SR, Ahern TB,
them. Eur Urol. 2015;67(5):825–836. 345. Laaksonen DE, Niskanen L, Punnonen K, Nyyssönen
Laurent MR, Huhtaniemi IT, Lean ME, Keevil BG,
320. Vandenput L, Mellström D, Karlsson MK, Orwoll E, K, Tuomainen TP, Valkonen VP, Salonen R, Salonen
Rastrelli G, Forti G, Bartfai G, Casanueva FF, Kula K,
Labrie F, Ljunggren O, Ohlsson C. Serum estradiol is JT. Testosterone and sex hormone-binding globulin
Punab M, Giwercman A, Claessens F, Decallonne B,
associated with lean mass in elderly Swedish men. predict the metabolic syndrome and diabetes in
Vanderschueren D; European Male Ageing Study
Eur J Endocrinol. 2010;162(4):737–745. middle-aged men. Diabetes Care. 2004;27(5):
Study Group. Low free testosterone is associated
321. Cawthon PM, Ensrud KE, Laughlin GA, Cauley JA, 1036–1041.
with hypogonadal signs and symptoms in men with
Dam TTL, Barrett-Connor E, Fink HA, Hoffman AR, 346. Kupelian V, Page ST, Araujo AB, Travison TG,
Lau E, Lane NE, Stefanick ML, Cummings SR, Orwoll normal total testosterone. J Clin Endocrinol Metab. Bremner WJ, McKinlay JB. Low sex hormone-
ES; Osteoporotic Fractures in Men (MrOS) Research 2016;101(7):2647–2657. binding globulin, total testosterone, and symp-
Group. Sex hormones and frailty in older men: the 334. Antonio L, Wu FCW, O’Neill TW, Pye SR, Carter EL, tomatic androgen deficiency are associated with
Osteoporotic Fractures in Men (MrOS) study. J Clin Finn JD, Rutter MK, Laurent MR, Huhtaniemi IT, development of the metabolic syndrome in non-
Endocrinol Metab. 2009;94(10):3806–3815. Han TS, Lean MEJ, Keevil BG, Pendleton N, Rastrelli obese men. J Clin Endocrinol Metab. 2006;91(3):
322. Bann D, Wu FC, Keevil B, Lashen H, Adams J, Hardy G, Forti G, Bartfai G, Casanueva FF, Kula K, Punab M, 843–850.
R, Muniz G, Kuh D, Ben-Shlomo Y, Ong KK. Giwercman A, Claessens F, Decallonne B, 347. Lakshman KM, Bhasin S, Araujo AB. Sex hormone-
Changes in testosterone related to body compo- Vanderschueren D; EMAS Study Group. Associa- binding globulin as an independent predictor of
sition in late midlife: findings from the 1946 British tions between sex steroids and the development of incident type 2 diabetes mellitus in men. J Gerontol
birth cohort study. Obesity (Silver Spring). 2015; metabolic syndrome: a longitudinal study in Eu- A Biol Sci Med Sci. 2010;65(5):503–509.
23(7):1486–1492. ropean men. J Clin Endocrinol Metab. 2015;100(4): 348. Rodriguez A, Muller DC, Metter EJ, Maggio M,
323. Yeap BB, Knuiman MW, Divitini ML, Handelsman 1396–1404. Harman SM, Blackman MR, Andres R. Aging, an-
DJ, Beilby JP, Beilin J, McQuillan B, Hung J. Differential 335. Haring R, Völzke H, Spielhagen C, Nauck M, drogens, and the metabolic syndrome in a longi-
associations of testosterone, dihydrotestosterone Wallaschofski H. The role of sex hormone-binding tudinal study of aging. J Clin Endocrinol Metab. 2007;
and oestradiol with physical, metabolic and health- globulin and testosterone in the risk of incident 92(9):3568–3572.
related factors in community-dwelling men aged metabolic syndrome. Eur J Prev Cardiol. 2013;20(6): 349. Holmboe SA, Jensen TK, Linneberg A, Scheike T,
17–97 years from the Busselton Health Survey. Clin 1061–1068. Thuesen BH, Skakkebaek NE, Juul A, Andersson AM.
Endocrinol (Oxf). 2014;81(1):100–108. 336. Travison TG, Araujo AB, Kupelian V, O’Donnell AB, Low testosterone: a risk marker rather than a risk
324. Trabert B, Graubard BI, Nyante SJ, Rifai N, Bradwin G, McKinlay JB. The relative contributions of aging, factor for type 2 diabetes. J Clin Endocrinol Metab.
Platz EA, McQuillan GM, McGlynn KA. Relationship health, and lifestyle factors to serum testosterone 2016;101(8):3180–3190.
of sex steroid hormones with body size and with decline in men. J Clin Endocrinol Metab. 2007;92(2): 350. Brand JS, Rovers MM, Yeap BB, Schneider HJ,
body composition measured by dual-energy X-ray 549–555. Tuomainen TP, Haring R, Corona G, Onat A,
absorptiometry in US men. Cancer Causes Control. 337. Eriksson J, Haring R, Grarup N, Vandenput L, Maggio M, Bouchard C, Tong PC, Chen RY, Akishita
2012;23(12):1881–1891. Wallaschofski H, Lorentzen E, Hansen T, Mellström M, Gietema JA, Gannagé-Yared MH, Undén AL,
325. Muller M, Grobbee DE, den Tonkelaar I, Lamberts D, Pedersen O, Nauck M, Lorentzon M, Nystrup Hautanen A, Goncharov NP, Kumanov P, Chubb
SW, van der Schouw YT. Endogenous sex hormones Husemoen LL, Völzke H, Karlsson M, Baumeister SE, SA, Almeida OP, Wittchen HU, Klotsche J,
and metabolic syndrome in aging men. J Clin Linneberg A, Ohlsson C. Causal relationship be- Wallaschofski H, Völzke H, Kauhanen J, Salonen JT,
Endocrinol Metab. 2005;90(5):2618–2623. tween obesity and serum testosterone status in Ferrucci L, van der Schouw YT. Testosterone, sex
326. Maggio M, Lauretani F, Ceda GP, Bandinelli S, men: a bidirectional mendelian randomization hormone-binding globulin and the metabolic
Basaria S, Ble A, Egan J, Paolisso G, Najjar S, Jeffrey analysis. PLoS One. 2017;12(4):1–15. syndrome in men: an individual participant data

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 965


REVIEW

meta-analysis of observational studies. PLoS One. 364. Bhasin S, Travison TG, Storer TW, Lakshman K, with frailty in older men. Clin Endocrinol (Oxf). 2013;
2014;9(7):e100409. Kaushik M, Mazer NA, Ngyuen A-H, Davda MN, Jara 78(5):752–759.
351. Hsu B, Cumming RG, Naganathan V, Blyth FM, Le H, Aakil A, Anderson S, Knapp PE, Hanka S, 376. Swiecicka A, Lunt M, Ahern T, O’Neill TW, Bartfai G,
Couteur DG, Seibel MJ, Waite LM, Handelsman DJ. Mohammed N, Daou P, Miciek R, Ulloor J, Zhang A, Casanueva FF, Forti G, Giwercman A, Han TS, Lean
Associations between circulating reproductive Brooks B, Orwoll K, Hede-Brierley L, Eder R, Elmi A, ME, Pendleton N, Punab M, Slowikowska-Hilczer J,
hormones and SHBG and prevalent and incident Bhasin G, Collins L, Singh R, Basaria S. Effect of Vanderschueren D, Huhtaniemi IT, Wu FC, Rutter
metabolic syndrome in community-dwelling older testosterone supplementation with and without a MK; EMAS Study Group.Nonandrogenic anabolic
men: the Concord Health and Ageing in Men dual 5a-reductase inhibitor on fat-free mass in men hormones predict risk of frailty: European Male
Project. J Clin Endocrinol Metab. 2014;99(12): with suppressed testosterone production: a ran- Ageing Study prospective data. J Clin Endocrinol
E2686–E2691. domized controlled trial. JAMA. 2012;307(9): Metab. 2017;102(8):2798–2806.
352. Bhasin S, Jasjua GK, Pencina M, D’Agostino R Sr, 931–939. 377. Bann D, Holly JMP, Lashen H, Hardy R, Adams J, Kuh
Coviello AD, Vasan RS, Travison TG. Sex hormone- 365. Bhasin S, Woodhouse L, Casaburi R, Singh AB, Mac D, Ong KK, Ben-Shlomo Y. Changes in insulin-like
binding globulin, but not testosterone, is associated RP, Lee M, Yarasheski KE, Sinha-Hikim I, Dzekov C, growth factor-I and -II associated with fat but not
prospectively and independently with incident Dzekov J, Magliano L, Storer TW. Older men are as lean mass in early old age. Obesity (Silver Spring).
metabolic syndrome in men: the framingham heart responsive as young men to the anabolic effects of 2015;23(3):692–698.
study. Diabetes Care. 2011;34(11):2464–2470. graded doses of testosterone on the skeletal muscle. 378. Ohlander SJ, Varghese B, Pastuszak AW. Eryth-
353. Schipf S, Haring R, Friedrich N, Nauck M, Lau K, Alte J Clin Endocrinol Metab. 2005;90(2):678–688. rocytosis following testosterone therapy. Sex Med

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


D, Stang A, Völzke H, Wallaschofski H. Low total 366. Schaap LA, Pluijm SMF, Smit JH, van Schoor NM, Rev. 2018;6(1):77–85.
testosterone is associated with increased risk of Visser M, Gooren LJG, Lips P. The association of sex 379. Guralnik JM, Eisenstaedt RS, Ferrucci L, Klein HG,
incident type 2 diabetes mellitus in men: results hormone levels with poor mobility, low muscle Woodman RC. Prevalence of anemia in persons 65
from the Study of Health in Pomerania (SHIP). Aging strength and incidence of falls among older men years and older in the United States: evidence for a
Male. 2011;14(3):168–175. and women. Clin Endocrinol (Oxf). 2005;63(2): high rate of unexplained anemia. Blood. 2004;104(8):
354. Jasuja GK, Travison TG, Davda M, Rose AJ, Zhang A, 152–160. 2263–2268.
Kushnir MM, Rockwood AL, Meikle W, Coviello AD, 367. Krasnoff JB, Basaria S, Pencina MJ, Jasuja GK, Vasan 380. Andrès E, Serraj K, Federici L, Vogel T, Kaltenbach G.
D’Agostino R, Vasan RS, Bhasin S. Circulating es- RS, Ulloor J, Zhang A, Coviello A, Kelly-Hayes M, Anemia in elderly patients: new insight into an old
trone levels are associated prospectively with di- D’Agostino RB, Wolf PA, Bhasin S, Murabito JM. Free disorder. Geriatr Gerontol Int. 2013;13(3):519–527.
abetes risk in men of the Framingham Heart Study. testosterone levels are associated with mobility 381. Lewerin C, Nilsson-Ehle H, Jacobsson S, Johansson H,
Diabetes Care. 2013;36(9):2591–2596. limitation and physical performance in community- Sundh V, Karlsson MK, Lorentzon M, Barrett-
355. Mather KJ, Kim C, Christophi CA, Aroda VR, dwelling men: the Framingham Offspring Study. Connor E, Vandenput L, Ohlsson C, Mellström D.
Knowler WC, Edelstein SE, Florez JC, Labrie F, Kahn J Clin Endocrinol Metab. 2010;95(6):2790–2799. Serum estradiol associates with blood hemoglobin
SE, Goldberg RB, Barrett-Connor E; Diabetes Pre- 368. O’Donnell AB, Travison TG, Harris SS, Tenover JL, in elderly men: the MrOS Sweden study. J Clin
vention Program. Steroid sex hormones, sex McKinlay JB. Testosterone, dehydroepiandroster- Endocrinol Metab. 2014;99(7):2549–2556.
hormone-binding globulin, and diabetes incidence one, and physical performance in older men: results 382. Bhatia V, Chaudhuri A, Tomar R, Dhindsa S, Ghanim
in the diabetes prevention program. J Clin Endo- from the Massachusetts Male Aging Study. J Clin H, Dandona P. Low testosterone and high C-re-
crinol Metab. 2015;100(10):3778–3786. Endocrinol Metab. 2006;91(2):425–431. active protein concentrations predict low hemat-
356. Salminen M, Vahlberg T, Räihä I, Niskanen L, Kivelä 369. Jasuja GK, Travison TG, Murabito JM, Davda MN, ocrit in type 2 diabetes. Diabetes Care. 2006;29(10):
S-L, Irjala K. Sex hormones and the risk of type 2 Rose AJ, Basaria S, Coviello A, Vasan RS, D’Agostino 2289–2294.
diabetes mellitus: a 9-year follow up among elderly R, Bhasin S. Circulating estrogen levels and self- 383. Grossmann M, Panagiotopolous S, Sharpe K,
men in Finland. Geriatr Gerontol Int. 2015;15(5): reported health and mobility limitation in MacIsaac RJ, Clarke S, Zajac JD, Jerums G, Thomas
559–564. community-dwelling men of the Framingham MC. Low testosterone and anaemia in men with
357. Evans NA. Current concepts in anabolic-androgenic Heart Study. J Gerontol A Biol Sci Med Sci. 2017;72(8): type 2 diabetes. Clin Endocrinol (Oxf). 2009;70(4):
steroids. Am J Sports Med. 2004;32(2):534–542. 1137–1142. 547–553.
358. Szulc P, Duboeuf F, Marchand F, Delmas PD. 370. Yuki A, Otsuka R, Kozakai R, Kitamura I, Okura T, 384. Ferrucci L, Maggio M, Bandinelli S, Basaria S,
Hormonal and lifestyle determinants of appen- Ando F, Shimokata H. Relationship between low Lauretani F, Ble A, Valenti G, Ershler WB, Guralnik
dicular skeletal muscle mass in men: the MINOS free testosterone levels and loss of muscle mass. Sci JM, Longo DL. Low testosterone levels and the risk
study. Am J Clin Nutr. 2004;80(2):496–503. Rep. 2013;3(1):1818. of anemia in older men and women. Arch Intern
359. Auyeung TW, Lee JS, Kwok T, Leung J, Ohlsson C, 371. Mohr BA, Bhasin S, Kupelian V, Araujo AB, Med. 2006;166(13):1380–1388.
Vandenput L, Leung PC, Woo J. Testosterone but O’Donnell AB, McKinlay JB. Testosterone, sex 385. Paller CJ, Shiels MS, Rohrmann S, Menke A, Rifai N,
not estradiol level is positively related to muscle hormone-binding globulin, and frailty in older men. Nelson WG, Platz EA, Dobs AS. Association be-
strength and physical performance independent of J Am Geriatr Soc. 2007;55(4):548–555. tween sex steroid hormones and hematocrit in a
muscle mass: a cross-sectional study in 1489 older 372. Hyde Z, Flicker L, Almeida OP, Hankey GJ, McCaul nationally representative sample of men. J Androl.
men. Eur J Endocrinol. 2011;164(5):811–817. KA, Chubb SAP, Yeap BB. Low free testosterone 2012;33(6):1332–1341.
360. Lapauw B, Goemaere S, Crabbe P, Kaufman JM, predicts frailty in older men: the Health in Men 386. Tajar A, Huhtaniemi IT, O’Neill TW, Finn JD, Pye SR,
Ruige JB. Is the effect of testosterone on body Study. J Clin Endocrinol Metab. 2010;95(7): Lee DM, Bartfai G, Boonen S, Casanueva FF, Forti G,
composition modulated by the androgen receptor 3165–3172. Giwercman A, Han TS, Kula K, Labrie F, Lean ME,
gene CAG repeat polymorphism in elderly men? Eur 373. Travison TG, Nguyen AH, Naganathan V, Stanaway Pendleton N, Punab M, Vanderschueren D, Wu FC;
J Endocrinol. 2007;156(3):395–401. FF, Blyth FM, Cumming RG, Le Couteur DG, EMAS Group. Characteristics of androgen de-
361. Hsu B, Cumming RG, Naganathan V, Blyth FM, Le Sambrook PN, Handelsman DJ. Changes in re- ficiency in late-onset hypogonadism: results from
Couteur DG, Seibel MJ, Waite LM, Handelsman DJ. productive hormone concentrations predict the the European Male Aging Study (EMAS). J Clin
Longitudinal relationships of circulating re- prevalence and progression of the frailty syndrome Endocrinol Metab. 2012;97(5):1508–1516.
productive hormone with functional disability, in older men: the Concord Health and Ageing in 387. Ahern T, Swiecicka A, Eendebak RJ, Carter EL, Finn
muscle mass, and strength in community-dwelling Men Project. J Clin Endocrinol Metab. 2011;96(8): JD, Pye SR, O’Neill TW, Antonio L, Keevil B, Bartfai G,
older men: the Concord Health and Ageing in Men 2464–2474. Casanueva FF, Forti G, Giwercman A, Han TS, Kula
project. J Clin Endocrinol Metab. 2014;99(9): 374. Swiecicka A, Eendebak RJ, Lunt M, O’Neill TW, K, Lean ME, Pendleton N, Punab M, Rastrelli G,
3310–3318. Bartfai G, Casanueva FF, Forti G, Giwercman A, Han Rutter MK, Vanderschueren D, Huhtaniemi IT, Wu
362. Renoud A, Ecochard R, Marchand F, Chapurlat R, TS, Slowikowska-Hilczer J, Lean MEJ, Pendleton N, FC; EMAS study group. Natural history, risk factors
Szulc P. Predictive parameters of accelerated muscle Punab M, Vanderschueren D, Huhtaniemi IT, Wu and clinical features of primary hypogonadism in
loss in men-MINOS study. Am J Med. 2014;127(6): FCW. Reproductive hormone levels predict changes ageing men: longitudinal data from the European
554–561. in frailty status in community-dwelling older men: Male Ageing Study. Clin Endocrinol (Oxf). 2016;85(6):
363. Hsu B, Cumming RG, Hirani V, Blyth FM, European Male Ageing Study prospective data. 891–901.
Naganathan V, Le Couteur DG, Seibel MJ, Waite LM, J Clin Endocrinol Metab. 2018;103(2):701–709. 388. Bachman E, Travison TG, Basaria S, Davda MN, Guo
Handelsman DJ. Temporal trend in androgen status 375. Yeap BB, Paul Chubb SA, Lopez D, Ho KK, Hankey W, Li M, Connor Westfall J, Bae H, Gordeuk V,
and androgen-sensitive outcomes in older men. GJ, Flicker L. Associations of insulin-like growth Bhasin S. Testosterone induces erythrocytosis via
J Clin Endocrinol Metab. 2016;101(4):1836–1846. factor-I and its binding proteins and testosterone increased erythropoietin and suppressed hepcidin:

966 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

evidence for a new erythropoietin/hemoglobin set 402. van den Beld AW, Bots ML, Janssen JA, Pols HA, 416. Shores MM, Biggs ML, Arnold AM, Smith NL,
point. J Gerontol A Biol Sci Med Sci. 2014;69(6): Lamberts SW, Grobbee DE. Endogenous hormones Longstreth WT Jr, Kizer JR, Hirsch CH, Cappola AR,
725–735. and carotid atherosclerosis in elderly men. Am J Matsumoto AM. Testosterone, dihydrotestoster-
389. Dhindsa S, Ghanim H, Batra M, Kuhadiya ND, Epidemiol. 2003;157(1):25–31. one, and incident cardiovascular disease and
Abuaysheh S, Green K, Makdissi A, Chaudhuri A, 403. Mäkinen J, Järvisalo MJ, Pöllänen P, Perheentupa A, mortality in the cardiovascular health study. J Clin
Dandona P. Effect of testosterone on hepcidin, Irjala K, Koskenvuo M, Mäkinen J, Huhtaniemi I, Endocrinol Metab. 2014;99(6):2061–2068.
ferroportin, ferritin and iron binding capacity in Raitakari OT. Increased carotid atherosclerosis in 417. Ärnlöv J, Pencina MJ, Amin S, Nam B-H, Benjamin EJ,
patients with hypogonadotropic hypogonadism andropausal middle-aged men. J Am Coll Cardiol. Murabito JM, Wang TJ, Knapp PE, D’Agostino RB Sr,
and type 2 diabetes. Clin Endocrinol (Oxf). 2016; 2005;45(10):1603–1608. Bhasin S, Vasan RS. Endogenous sex hormones and
85(5):772–780. 404. Svartberg J, von Mühlen D, Mathiesen E, Joakimsen cardiovascular disease incidence in men. Ann Intern
390. Ip FF, di Pierro I, Brown R, Cunningham I, O, Bønaa KH, Stensland-Bugge E. Low testosterone Med. 2006;145(3):176–184.
Handelsman DJ, Liu PY. Trough serum testosterone levels are associated with carotid atherosclerosis in 418. Haring R, Teng Z, Xanthakis V, Coviello A, Sullivan L,
predicts the development of polycythemia in men. J Intern Med. 2006;259(6):576–582. Bhasin S, Murabito JM, Wallaschofski H, Vasan RS.
hypogonadal men treated for up to 21 years with 405. Kische H, Gross S, Wallaschofski H, Völzke H, Dörr Association of sex steroids, gonadotrophins, and
subcutaneous testosterone pellets. Eur J Endocrinol. M, Nauck M, Felix SB, Haring R. Serum androgen their trajectories with clinical cardiovascular disease
2010;162(2):385–390. concentrations and subclinical measures of car- and all-cause mortality in elderly men from the
391. Moriyama Y, Fisher JW. Effects of testosterone and diovascular disease in men and women. Athero- Framingham Heart Study. Clin Endocrinol (Oxf).

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


erythropoietin on erythroid colony formation in sclerosis. 2016;247:193–200. 2013;78(4):629–634.
human bone marrow cultures. Blood. 1975;45(5): 406. Park BJ, Shim JY, Lee YJ, Lee JH, Lee HR. Inverse 419. Vandenplas G, De Bacquer D, Calders P, Fiers T,
665–670. relationship between bioavailable testosterone and Kaufman JM, Ouwens DM, Ruige JB. Endogenous
392. Kim SW, Hwang JH, Cheon JM, Park NS, Park SE, subclinical coronary artery calcification in non- oestradiol and cardiovascular disease in healthy
Park SJ, Yun HJ, Kim S, Jo DY. Direct and indirect obese Korean men. Asian J Androl. 2012;14(4): men: a systematic review and meta-analysis of
effects of androgens on survival of hematopoietic 612–615. prospective studies. Heart. 2012;98(20):1478–1482.
progenitor cells in vitro. J Korean Med Sci. 2005; 407. Travison TG, O’Donnell CJ, Bhasin S, Massaro JM, 420. Araujo AB, Dixon JM, Suarez EA, Murad MH, Guey
20(3):409–416. Hoffmann U, Vasan RS, D’Agostino RB Sr, Basaria S. LT, Wittert GA. Clinical review: endogenous tes-
393. Bachman E, Feng R, Travison T, Li M, Olbina G, Circulationg sex steroids and vascular calcification tosterone and mortality in men: a systematic review
Ostland V, Ulloor J, Zhang A, Basaria S, Ganz T, in community-dwelling men: the Framingham and meta-analysis. J Clin Endocrinol Metab. 2011;
Westerman M, Bhasin S. Testosterone suppresses Heart Study. J Clin Endocrinol Metab. 2016;101(5): 96(10):3007–3019.
hepcidin in men: a potential mechanism for 2160–2167. 421. Menke A, Guallar E, Rohrmann S, Nelson WG, Rifai
testosterone-induced erythrocytosis. J Clin Endo- 408. Yeap BB, Hyde Z, Norman PE, Chubb SAP, Golledge N, Kanarek N, Feinleib M, Michos ED, Dobs A, Platz
J. Associations of total testosterone, sex hormone-
crinol Metab. 2010;95(10):4743–4747. EA. Sex steroid hormone concentrations and risk of
binding globulin, calculated free testosterone, and
394. Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh death in US men. Am J Epidemiol. 2010;171(5):
luteinizing hormone with prevalence of abdominal
AB, Bhasin S. Effects of graded doses of testosterone 583–592.
aortic aneurysm in older men. J Clin Endocrinol
on erythropoiesis in healthy young and older men. 422. Haring R, Völzke H, Steveling A, Krebs A, Felix SB,
Metab. 2010;95(3):1123–1130.
J Clin Endocrinol Metab. 2008;93(3):914–919. Schöfl C, Dörr M, Nauck M, Wallaschofski H. Low
409. Yeap BB. Testosterone and cardiovascular disease
395. Maggio M, Snyder PJ, Ceda GP, Milaneschi Y, Luci serum testosterone levels are associated with in-
risk. Curr Opin Endocrinol Diabetes Obes. 2015;22(3):
M, Cattabiani C, Masoni S, Vignali A, Volpi R, creased risk of mortality in a population-based
193–202.
Lauretani F, Peachey H, Valenti G, Cappola AR, cohort of men aged 20–79. Eur Heart J. 2010;
410. Yeap BB, Alfonso H, Chubb SAP, Hankey GJ,
Longo D, Ferrucci L. Is the haematopoietic effect of 31(12):1494–1501.
Handelsman DJ, Golledge J, Almeida OP, Flicker L,
testosterone mediated by erythropoietin? The re- 423. Haring R, Teumer A, Völker U, Dörr M, Nauck M,
Norman PE. In older men, higher plasma testos-
sults of a clinical trial in older men. Andrology. 2013; Biffar R, Völzke H, Baumeister SE, Wallaschofski H.
terone or dihydrotestosterone is an independent
1(1):24–28. Mendelian randomization suggests non-causal as-
predictor for reduced incidence of stroke but not
396. Zitzmann M, Nieschlag E. Androgen receptor gene sociations of testosterone with cardiometabolic risk
myocardial infarction. J Clin Endocrinol Metab. 2014;
CAG repeat length and body mass index modulate 99(12):4565–4573. factors and mortality. Andrology. 2013;1(1):17–23.
the safety of long-term intramuscular testosterone 411. Abbott RD, Launer LJ, Rodriguez BL, Ross GW, 424. Hyde Z, Norman PE, Flicker L, Hankey GJ, Almeida
undecanoate therapy in hypogonadal men. J Clin Wilson PW, Masaki KH, Strozyk D, Curb JD, Yano K, OP, McCaul KA, Chubb SA, Yeap BB. Low free
Endocrinol Metab. 2007;92(10):3844–3853. Popper JS, Petrovitch H. Serum estradiol and risk of testosterone predicts mortality from cardiovascular
397. Beggs LA, Yarrow JF, Conover CF, Meuleman JR, stroke in elderly men. Neurology. 2007;68(8): disease but not other causes: the Health in Men
Beck DT, Morrow M, Zou B, Shuster JJ, Borst SE. 563–568. Study. J Clin Endocrinol Metab. 2012;97(1):179–189.
Testosterone alters iron metabolism and stimulates 412. Yeap BB, Hyde Z, Almeida OP, Norman PE, Chubb 425. Yeap BB, Alfonso H, Chubb SAP, Handelsman DJ,
red blood cell production independently of dihy- SA, Jamrozik K, Flicker L, Hankey GJ. Lower tes- Hankey GJ, Almeida OP, Golledge J, Norman PE,
drotestosterone. Am J Physiol Endocrinol Metab. tosterone levels predict incident stroke and tran- Flicker L. In older men an optimal plasma testos-
2014;307(5):E456–E461. sient ischemic attack in older men. J Clin Endocrinol terone is associated with reduced all-cause mor-
398. Aghazadeh M, Pastuszak AW, Johnson WG, Metab. 2009;94(7):2353–2359. tality and higher dihydrotestosterone with reduced
McIntyre MG, Hsieh TM, Lipshultz LI. Elevated 413. Shores MM, Arnold AM, Biggs ML, Longstreth WT ischemic heart disease mortality, while estradiol
dihydrotestosterone is associated with testosterone Jr, Smith NL, Kizer JR, Cappola AR, Hirsch CH, Marck levels do not predict mortality. J Clin Endocrinol
induced erythrocytosis. J Urol. 2015;194(1):160–165. BT, Matsumoto AM. Testosterone and dihy- Metab. 2014;99(1):E9–E18.
399. Hara N, Nishiyama T, Takizawa I, Saito T, Kitamura drotestosterone and incident ischaemic stroke in 426. Holmboe SA, Skakkebæk NE, Juul A, Scheike T,
Y, Takahashi K. Decline of the red blood cell count men in the Cardiovascular Health Study. Clin Jensen TK, Linneberg A, Thuesen BH, Andersson
in patients receiving androgen deprivation therapy Endocrinol (Oxf). 2014;81(5):746–753. AM. Individual testosterone decline and future
for localized prostate cancer: impact of ADT on 414. Ohlsson C, Barrett-Connor E, Bhasin S, Orwoll E, mortality risk in men. Eur J Endocrinol. 2018;178(1):
insulin-like growth factor-1 and erythropoiesis. Labrie F, Karlsson MK, Ljunggren O, Vandenput L, 123–130.
Urology. 2010;75(6):1441–1445. Mellström D, Tivesten A. High serum testosterone 427. Pye SR, Huhtaniemi IT, Finn JD, Lee DM, O’Neill TW,
400. Ruige JB, Ouwens DM, Kaufman JM. Beneficial and is associated with reduced risk of cardiovascular Tajar A, Bartfai G, Boonen S, Casanueva FF, Forti G,
adverse effects of testosterone on the cardiovas- events in elderly men. The MrOS (Osteoporotic Giwercman A, Han TS, Kula K, Lean ME, Pendleton
cular system in men. J Clin Endocrinol Metab. 2013; Fractures in Men) study in Sweden. J Am Coll N, Punab M, Rutter MK, Vanderschueren D, Wu FC;
98(11):4300–4310. Cardiol. 2011;58(16):1674–1681. EMAS Study Group. Late-onset hypogonadism and
401. Srinath R, Hill Golden S, Carson KA, Dobs A. En- 415. Soisson V, Brailly-Tabard S, Helmer C, Rouaud O, mortality in aging men. J Clin Endocrinol Metab.
dogenous testosterone and its relationship to Ancelin ML, Zerhouni C, Guiochon-Mantel A, 2014;99(4):1357–1366.
preclinical and clinical measures of cardiovascular Scarabin PY. A J-shaped association between 428. Nguyen PL, Je Y, Schutz FA, Hoffman KE, Hu JC,
disease in the atherosclerosis risk in communities plasma testosterone and risk of ischemic arterial Parekh A, Beckman JA, Choueiri TK. Association of
study. J Clin Endocrinol Metab. 2015;100(4): event in elderly men: the French 3C cohort study. androgen deprivation therapy with cardiovascular
1602–1608. Maturitas. 2013;75(3):282–288. death in patients with prostate cancer: a meta-

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 967


REVIEW

analysis of randomized trials. JAMA. 2011;306(21): testosterone levels at baseline in the Testosterone 458. Joshi D, van Schoor NM, de Ronde W, Schaap LA,
2359–2366. Trials. J Clin Endocrinol Metab. 2015;100(3): Comijs HC, Beekman AT, Lips P. Low free testos-
429. Mäkinen JI, Perheentupa A, Raitakari OT, Koskenvuo 1146–1155. terone levels are associated with prevalence and
M, Pöllänen P, Mäkinen J, Huhtaniemi I. Sexual 443. Hsu B, Cumming RG, Blyth FM, Naganathan V, Le incidence of depressive symptoms in older men.
symptoms in aging men indicate poor life satisfaction Couteur DG, Seibel MJ, Waite LM, Handelsman DJ. Clin Endocrinol (Oxf). 2010;72(2):232–240.
and increased health service consumption. Urology. The longitudinal relationship of sexual function and 459. Ford AH, Yeap BB, Flicker L, Hankey GJ, Chubb SAP,
2007;70(6):1194–1199. androgen status in older men: the Concord Health Handelsman DJ, Golledge J, Almeida OP. Pro-
430. Meuleman EJ, van Lankveld JJ. Hypoactive sexual and Ageing in Men Project. J Clin Endocrinol Metab. spective longitudinal study of testosterone and
desire disorder: an underestimated condition in 2015;100(4):1350–1358. incident depression in older men: The Health In
men. BJU Int. 2005;95(3):291–296. 444. Yeap BB. Hormonal changes and their impact on Men Study. Psychoneuroendocrinology. 2016;64:
431. Hyde Z, Flicker L, Hankey GJ, Almeida OP, McCaul cognition and mental health of ageing men. 57–65.
KA, Chubb SAP, Yeap BB. Prevalence of sexual Maturitas. 2014;79(2):227–235. 460. Matsumoto AM, Bremner WJ. Testicular disorders.
activity and associated factors in men aged 75 to 95 445. Hua JT, Hildreth KL, Pelak VS. Effects of testosterone In: Melmed S, Polonsky KS, Larsen PR, Kronenberg
years: a cohort study. Ann Intern Med. 2010;153(11): therapy on cognitive function in aging: a systematic HM, eds. Williams Textbook of Endocrinology. 13th
693–702. review. Cogn Behav Neurol. 2016;29(3):122–138. ed. Philadelphia, PA: Elsevier; 2016:688–777.
432. Hyde Z, Flicker L, Hankey GJ, Almeida OP, McCaul 446. Moffat SD, Zonderman AB, Metter EJ, Blackman 461. Huhtaniemi I, Forti G. Male late-onset hypo-
KA, Chubb SA, Yeap BB. Prevalence and predictors MR, Harman SM, Resnick SM. Longitudinal as- gonadism: pathogenesis, diagnosis and treatment.

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


of sexual problems in men aged 75–95 years: a sessment of serum free testosterone concentration Nat Rev Urol. 2011;8(6):335–344.
population-based study. J Sex Med. 2012;9(2): predicts memory performance and cognitive status 462. Corona G, Rastrelli G, Vignozzi L, Mannucci E, Maggi
442–453. in elderly men. J Clin Endocrinol Metab. 2002;87(11): M. How to recognize late-onset hypogonadism in
433. Araujo AB, Mohr BA, McKinlay JB. Changes in sexual 5001–5007. men with sexual dysfunction. Asian J Androl. 2012;
function in middle-aged and older men: longitu- 447. Moffat SD, Zonderman AB, Metter EJ, Kawas C, 14(2):251–259.
dinal data from the Massachusetts Male Aging Blackman MR, Harman SM, Resnick SM. Free tes- 463. Araujo AB, Esche GR, Kupelian V, O’Donnell AB,
Study. J Am Geriatr Soc. 2004;52(9):1502–1509. tosterone and risk for Alzheimer disease in older Travison TG, Williams RE, Clark RV, McKinlay JB.
434. Corona G, Maggi M. The role of testosterone in men. Neurology. 2004;62(2):188–193. Prevalence of symptomatic androgen deficiency in
erectile dysfunction. Nat Rev Urol. 2010;7(1):46–56. 448. Geerlings MI, Strozyk D, Masaki K, Remaley AT, men. J Clin Endocrinol Metab. 2007;92(11):
435. Rosen RC, Araujo AB, Connor MK, Gerstenberger Petrovitch H, Ross GW, White LR, Launer LJ. En- 4241–4247.
EP, Morgentaler A, Seftel AD, Miner MM, Shabsigh dogenous sex hormones, cognitive decline, and 464. Morley JE, Charlton E, Patrick P, Kaiser FE, Cadeau P,
R. The NERI hypogonadism screener: psychometric future dementia in old men. Ann Neurol. 2006;60(3): McCready D, Perry HM III. Validation of a screening
validation in male patients and controls. Clin 346–355. questionnaire for androgen deficiency in aging
Endocrinol (Oxf). 2011;74(2):248–256. 449. Yaffe K, Barnes D, Lindquist K, Cauley J, Simonsick
males. Metabolism. 2000;49(9):1239–1242.
436. Handelsman DJ. Testosterone and male ageing: EM, Penninx B, Satterfield S, Harris T, Cummings SR;
465. Heinemann LA, Saad F, Heinemann K, Thai DM.
spinning the wheels. Med J Aust. 2010;193(7): Health ABC Investigators. Endogenous sex hor-
Can results of the Aging Males’ Symptoms (AMS)
mone levels and risk of cognitive decline in an older
379–380. scale predict those of screening scales for androgen
biracial cohort. Neurobiol Aging. 2007;28(2):171–178.
437. Corona G, Mannucci E, Ricca V, Lotti F, Boddi V, deficiency? Aging Male. 2004;7(3):211–218.
450. Hogervorst E, Matthews FE, Brayne C. Are optimal
Bandini E, Balercia G, Forti G, Maggi M. The age- 466. Smith KW, Feldman HA, McKinlay JB. Construction
levels of testosterone associated with better cog-
related decline of testosterone is associated with and field validation of a self-administered screener
nitive function in healthy older women and men?
different specific symptoms and signs in patients for testosterone deficiency (hypogonadism) in
Biochim Biophys Acta. 2010;1800(10):1145–1152.
with sexual dysfunction. Int J Androl. 2009;32(6): ageing men. Clin Endocrinol (Oxf). 2000;53(6):
451. LeBlanc ES, Wang PY, Janowsky JS, Neiss MB, Fink
720–728. 703–711.
HA, Yaffe K, Marshall LM, Lapidus JA, Stefanick ML,
438. Travison TG, Morley JE, Araujo AB, O’Donnell AB, 467. Corona G, Mannucci E, Petrone L, Balercia G, Fisher
Orwoll ES; Osteoporotic Fractures in Men (MrOS)
McKinlay JB. The relationship between libido and AD, Chiarini V, Forti G, Maggi M. ANDROTEST: a
Research Group. Association between sex steroids
testosterone levels in aging men. J Clin Endocrinol structured interview for the screening of hypo-
and cognition in elderly men. Clin Endocrinol (Oxf).
Metab. 2006;91(7):2509–2513. gonadism in patients with sexual dysfunction. J Sex
2010;72(3):393–403.
439. Millar AC, Lau AN, Tomlinson G, Kraguljac A, Simel Med. 2006;3(4):706–715.
452. Kische H, Gross S, Wallaschofski H, Grabe HJ, Völzke
DL, Detsky AS, Lipscombe LL. Predicting low tes- H, Nauck M, Haring R. Associations of androgens 468. Wang C, Nieschlag E, Swerdloff R, Behre HM,
tosterone in aging men: a systematic review. CMAJ. with depressive symptoms and cognitive status in Hellstrom WJ, Gooren LJ, Kaufman JM, Legros JJ,
2016;188(13):E321–E330. the general population. PLoS One. 2017;12(5): Lunenfeld B, Morales A, Morley JE, Schulman C,
440. Amory JK, Anawalt BD, Matsumoto AM, Page ST, e0177272. Thompson IM, Weidner W, Wu FC. Investigation,
Bremner WJ, Wang C, Swerdloff RS, Clark RV. The 453. McGinty HL, Phillips KM, Jim HS, Cessna JM, Asvat treatment, and monitoring of late-onset hypo-
effect of 5alpha-reductase inhibition with dutas- Y, Cases MG, Small BJ, Jacobsen PB. Cognitive gonadism in males: ISA, ISSAM, EAU, EAA, and ASA
teride and finasteride on bone mineral density, functioning in men receiving androgen deprivation recommendations. Eur J Endocrinol. 2008;159(5):
serum lipoproteins, hemoglobin, prostate specific therapy for prostate cancer: a systematic review and 507–514.
antigen and sexual function in healthy young men. meta-analysis. Support Care Cancer. 2014;22(8): 469. Lunenfeld B, Mskhalaya G, Zitzmann M, Arver S,
J Urol. 2008;179(6):2333–2338. 2271–2280. Kalinchenko S, Tishova Y, Morgentaler A. Recom-
441. O’Connor DB, Lee DM, Corona G, Forti G, Tajar A, 454. Khosrow-Khavar F, Rej S, Yin H, Aprikian A, Azoulay mendations on the diagnosis, treatment and
O’Neill TW, Pendleton N, Bartfai G, Boonen S, L. Adrogen deprivation therapy and risk of de- monitoring of hypogonadism in men. Aging Male.
Casanueva FF, Finn JD, Giwercman A, Han TS, mentia in patients with prostate cancer. J Clin Oncol. 2015;18(1):5–15.
Huhtaniemi IT, Kula K, Labrie F, Lean MEJ, Punab M, 2017;35(2):201–207. 470. Travison TG, Vesper HW, Orwoll E, Wu F, Kaufman
Silman AJ, Vanderschueren D, Wu FC; European 455. Shores MM, Sloan KL, Matsumoto AM, Moceri VM, JM, Wang Y, Lapauw B, Fiers T, Matsumoto AM,
Male Ageing Study Group. The relationships be- Felker B, Kivlahan DR. Increased incidence of di- Bhasin S. Harmonized reference ranges for circu-
tween sex hormones and sexual function in middle- agnosed depressive illness in hypogonadal older ating testosterone levels in men of four cohort
aged and older European men. J Clin Endocrinol men. Arch Gen Psychiatry. 2004;61(2):162–167. studies in the United States and Europe. J Clin
Metab. 2011;96(10):E1577–E1587. 456. Shores MM, Moceri VM, Sloan KL, Matsumoto AM, Endocrinol Metab. 2017;102(4):1161–1173.
442. Cunningham GR, Stephens-Shields AJ, Rosen RC, Kivlahan DR. Low testosterone levels predict in- 471. Handelsman DJ, Wartofsky L. Requirement for mass
Wang C, Ellenberg SS, Matsumoto AM, Bhasin S, cident depressive illness in older men: effects of age spectrometry sex steroid assays in the Journal of
Molitch ME, Farrar JT, Cella D, Barrett-Connor E, and medical morbidity. J Clin Psychiatry. 2005;66(1): Clinical Endocrinology and Metabolism. J Clin
Cauley JA, Cifelli D, Crandall JP, Ensrud KE, Fluharty 7–14. Endocrinol Metab. 2013;98(10):3971–3973.
L, Gill TM, Lewis CE, Pahor M, Resnick SM, Storer 457. T’Sjoen GG, De Vos S, Goemaere S, Van Pottelbergh 472. Závada M, Safarcik K, Topolcan O. Some problems
TW, Swerdloff RS, Anton S, Basaria S, Diem S, I, Dierick M, Van Heeringen C, Kaufman JM. Sex of radioimmunoassay control. J Radioanal Chem.
Tabatabaie V, Hou X, Snyder PJ. Association of steroid level, androgen receptor polymorphism, and 1978;46(1):57–66.
sex hormones with sexual function, vitality, and depressive symptoms in healthy elderly men. J Am 473. Monaghan PJ, Keevil BG, Stewart PM, Trainer PJ.
physical function of older men with low Geriatr Soc. 2005;53(4):636–642. Case for the wider adoption of mass spectrometry-

968 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

based adrenal steroid testing, and beyond. J Clin dialysis. J Clin Endocrinol Metab. 2018;103(6): 501. Rosen RC, Allen KR, Ni X, Araujo AB. Minimal
Endocrinol Metab. 2014;99(12):4434–4437. 2167–2174. clinically important differences in the erectile
474. Wierman ME, Auchus RJ, Haisenleder DJ, Hall JE, 488. Fiers T, Delanghe J, T’Sjoen G, Van Caenegem E, function domain of the International Index of
Handelsman D, Hankinson S, Rosner W, Singh RJ, Wierckx K, Kaufman JM. A critical evaluation of Erectile Function scale. Eur Urol. 2011;60(5):
Sluss PM, Stanczyk FZ. Editorial: the new in- salivary testosterone as a method for assessment of 1010–1016.
structions to authors for the reporting of steroid serum testosterone. Steroids. 2014;86:5–9. 502. Snyder PJ, Bhasin S, Cunningham GR, Matsumoto
hormone measurements. J Clin Endocrinol Metab. 489. Blom T, Ojanotko-Harri A, Laine M, Huhtaniemi I. AM, Stephens-Shields AJ, Cauley JA, Gill TM,
2014;99(12):4375. Metabolism of progesterone and testosterone in Barrett-Connor E, Swerdloff RS, Wang C, Ensrud KE,
475. Taylor AE, Keevil B, Huhtaniemi IT. Mass spec- human parotid and submandibular salivary glands Lewis CE, Farrar JT, Cella D, Rosen RC, Pahor M,
trometry and immunoassay: how to measure ste- in vitro. J Steroid Biochem Mol Biol. 1993;44(1):69–76. Crandall JP, Molitch ME, Cifelli D, Dougar D,
roid hormones today and tomorrow. Eur J 490. Liu Z, Liu J, Shi X, Wang L, Yang Y, Tao M, Fu Q. Fluharty L, Resnick SM, Storer TW, Anton S, Basaria
Endocrinol. 2015;173(2):D1–D12. Comparing calculated free testosterone with total S, Diem SJ, Hou X, Mohler ER III, Parsons JK, Wenger
476. Handelsman DJ. Mass spectrometry, immunoassay testosterone for screening and diagnosing late- NK, Zeldow B, Landis JR, Ellenberg SS; Testosterone
and valid steroid measurements in reproductive onset hypogonadism in aged males: a cross- Trials Investigators. Effects of testosterone treat-
medicine and science. Hum Reprod. 2017;32(6): sectional study. J Clin Lab Anal. 2017;31(5):e22073. ment in older men. N Engl J Med. 2016;374(7):
1147–1150. 491. Rastrelli G, O’Neill TW, Ahern T, Bártfai G, 611–624.
477. Rosner W, Auchus RJ, Azziz R, Sluss PM, Raff H. Casanueva FF, Forti G, Keevil B, Giwercman A, Han 503. Cunningham GR, Stephens-Shields AJ, Rosen RC,

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Utility, limitations, and pitfalls in measuring tes- TS, Slowikowska-Hilczer J, Lean ME, Pendleton N, Wang C, Bhasin S, Matsumoto AM, Parsons JK, Gill
tosterone: an Endocrine Society position statement. Punab M, Antonio L, Tournoy J, Vanderschueren D, TM, Molitch ME, Farrar JT, Cella D, Barrett-Connor
J Clin Endocrinol Metab. 2007;92(2):405–413. Maggi M, Huhtaniemi IT, Wu FC; EMAS study E, Cauley JA, Cifelli D, Crandall JP, Ensrud KE,
478. Huhtaniemi IT, Tajar A, Lee DM, O’Neill TW, Finn group. Symptomatic androgen deficiency develops Gallagher L, Zeldow B, Lewis CE, Pahor M, Swerdloff
JD, Bartfai G, Boonen S, Casanueva FF, Giwercman A, only when both total and free testosterone decline RS, Hou X, Anton S, Basaria S, Diem SJ, Tabatabaie V,
Han TS, Kula K, Labrie F, Lean ME, Pendleton N, in obese men who may have incident biochemical Ellenberg SS, Snyder PJ. Testosterone treatment and
Punab M, Silman AJ, Vanderschueren D, Forti G, Wu secondary hypogonadism: prospective results from sexual function in older men with low testosterone
FC; EMAS Group. Comparison of serum testos- the EMAS. Clin Endocrinol (Oxf). 2018;89(4): levels. J Clin Endocrinol Metab. 2016;101(8):
terone and estradiol measurements in 3174 Euro- 459–469. 3096–3104.
pean men using platform immunoassay and mass 492. Buvat J, Maggi M, Guay A, Torres LO. Testosterone 504. Müller A, Smith L, Parker M, Mulhall JP. Analysis of
spectrometry; relevance for the diagnostics in aging deficiency in men: systematic review and standard the efficacy and safety of sildenafil citrate in the
men. Eur J Endocrinol. 2012;166(6):983–991. operating procedures for diagnosis and treatment. geriatric population. BJU Int. 2007;100(1):117–121.
479. Haring R, Baumeister SE, Nauck M, Völzke H, Keevil J Sex Med. 2013;10(1):245–284. 505. Brock G, Heiselman D, Maggi M, Kim SW, Rodrı́guez
493. Morales A, Bebb RA, Manjoo P, Assimakopoulos P,
BG, Brabant G, Wallaschofski H. Testosterone and Vallejo JM, Behre HM, McGettigan J, Dowsett SA,
Axler J, Collier C, Elliott S, Goldenberg L, Gottesman
cardiometabolic risk in the general population—the Hayes RP, Knorr J, Ni X, Kinchen K. Effect of tes-
I, Grober ED, Guyatt GH, Holmes DT, Lee JC; Ca-
impact of measurement method on risk associations: tosterone solution 2% on testosterone concen-
nadian Men’s Health Foundation Multidisciplinary
a comparative study between immunoassay and tration, sex drive and energy in hypogonadal men:
Guidelines Task Force on Testosterone Deficiency.
mass spectrometry. Eur J Endocrinol. 2013;169(4): results of a placebo-controlled study. J Urol. 2016;
Diagnosis and management of testosterone de-
463–470. 195(3):699–705.
ficiency syndrome in men: clinical practice guide-
480. Dittadi R, Matteucci M, Meneghetti E, Ndreu R. 506. Algeffari M, Jayasena CN, MacKeith P, Thapar A,
line. CMAJ. 2015;187(18):1369–1377.
Reassessment of the Access Testosterone chem- Dhillo WS, Oliver N. Testosterone therapy for sexual
494. Iwamoto T, Yanase T, Horie H, Namiki M, Okuyama
iluminescence assay and comparison with LC-MS dysfunction in men with type 2 diabetes: a sys-
A. Late-onset hypogonadism (LOH) and androgens:
method. J Clin Lab Anal. 2018;32(3):e22286. tematic review and meta-analysis of randomized
validity of the measurement of free testosterone
481. Goldman AL, Bhasin S, Wu FCW, Krishna M, controlled trials. Diabet Med. 2018;35(2):195–202.
levels in the diagnostic criteria in Japan. Int J Urol.
Matsumoto AM, Jasuja R. A reappraisal of testos- 507. Spitzer M, Basaria S, Travison TG, Davda MN, Paley
2009;16(2):168–174.
terone’s binding in circulation: physiological and A, Cohen B, Mazer NA, Knapp PE, Hanka S,
495. Vesper HW, Wang Y, Vidal M, Botelho JC, Caudill SP.
clinical implications. Endocr Rev. 2017;38(4):302– Serum total testosterone concentrations in the US Lakshman KM, Ulloor J, Zhang A, Orwoll K, Eder R,
324. household population from the NHANES 2011–2012 Collins L, Mohammed N, Rosen RC, DeRogatis L,
482. Södergård R, Bäckström T, Shanbhag V, Carstensen study population. Clin Chem. 2015;61(12):1495– Bhasin S. Effect of testosterone replacement on
H. Calculation of free and bound fractions of tes- 1504. response to sildenafil citrate in men with erectile
tosterone and estradiol-17b to human plasma 496. Litman HJ, Bhasin S, Link CL, Araujo AB, McKinlay JB. dysfunction: a parallel, randomized trial. Ann Intern
proteins at body temperature. J Steroid Biochem. Serum androgen levels in black, Hispanic, and white Med. 2012;157(10):681–691.
1982;16(6):801–810. men. J Clin Endocrinol Metab. 2006;91(11):4326– 508. Buvat J, Montorsi F, Maggi M, Porst H, Kaipia A,
483. Vermeulen A, Verdonck L, Kaufman JM. A critical 4334. Colson MH, Cuzin B, Moncada I, Martin-Morales A,
evaluation of simple methods for the estimation of 497. Coviello AD, Lakshman K, Mazer NA, Bhasin S. Yassin A, Meuleman E, Eardley I, Dean JD, Shabsigh
free testosterone in serum. J Clin Endocrinol Metab. Differences in the apparent metabolic clearance R. Hypogonadal men nonresponders to the PDE5
1999;84(10):3666–3672. rate of testosterone in young and older men with inhibitor tadalafil benefit from normalization of
484. Mazer NA. A novel spreadsheet method for cal- gonadotropin suppression receiving graded doses testosterone levels with a 1% hydroalcoholic tes-
culating the free serum concentrations of testos- of testosterone. J Clin Endocrinol Metab. 2006;91(11): tosterone gel in the treatment of erectile dys-
terone, dihydrotestosterone, estradiol, estrone and 4669–4675. function (TADTEST study). J Sex Med. 2011;8(1):
cortisol: with illustrative examples from male and 498. Boloña ER, Uraga MV, Haddad RM, Tracz MJ, Sideras 284–293.
female populations. Steroids. 2009;74(6):512–519. K, Kennedy CC, Caples SM, Erwin PJ, Montori VM. 509. Spitzer M, Basaria S, Travison TG, Davda MN,
485. Ly LP, Handelsman DJ. Empirical estimation of free Testosterone use in men with sexual dysfunction: a DeRogatis L, Bhasin S. The effect of testosterone on
testosterone from testosterone and sex hormone- systematic review and meta-analysis of randomized mood and well-being in men with erectile dys-
binding globulin immunoassays. Eur J Endocrinol. placebo-controlled trials. Mayo Clin Proc. 2007; function in a randomized, placebo-controlled trial.
2005;152(3):471–478. 82(1):20–28. Andrology. 2013;1(3):475–482.
486. Zakharov MN, Bhasin S, Travison TG, Xue R, Ulloor J, 499. Corona G, Isidori AM, Buvat J, Aversa A, Rastrelli G, 510. Isidori AM, Giannetta E, Greco EA, Gianfrilli D,
Vasan RS, Carter E, Wu F, Jasuja R. A multi-step, Hackett G, Rochira V, Sforza A, Lenzi A, Mannucci E, Bonifacio V, Isidori A, Lenzi A, Fabbri A. Effects of
dynamic allosteric model of testosterone’s binding Maggi M. Testosterone supplementation and sex- testosterone on body composition, bone meta-
to sex hormone binding globulin (published cor- ual function: a meta-analysis study. J Sex Med. 2014; bolism and serum lipid profile in middle-aged men:
rection appears in. Mol Cell Endocrinol. 2017;454: 11(6):1577–1592. a meta-analysis. Clin Endocrinol (Oxf). 2005;63(3):
167). Mol Cell Endocrinol. 2015;399(C):190–200. 500. Corona G, Rastrelli G, Morgentaler A, Sforza A, 280–293.
487. Fiers T, Wu F, Moghetti P, Vanderschueren D, Mannucci E, Maggi M. Meta-analysis of results of 511. Ottenbacher KJ, Ottenbacher ME, Ottenbacher AJ,
Lapauw B, Kaufman JM. Reassessing free- testosterone therapy on sexual function based on Acha AA, Ostir GV. Androgen treatment and
testosterone calculation by liquid chromatogra- International Index of Erectile Function scores? Eur muscle strength in elderly men: a meta-analysis.
phy–tandem mass spectrometry direct equilibrium Urol. 2017;72(6):1000–1011. J Am Geriatr Soc. 2006;54(11):1666–1673.

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 969


REVIEW

512. Kenny AM, Kleppinger A, Annis K, Rathier M, JP, Cunningham GR, Ensrud KE, Gill TM, testosterone: a controlled clinical trial. JAMA Intern
Browner B, Judge JO, McGee D. Effects of trans- Matsumoto AM, Molitch ME, Pahor M, Preston PE, Med. 2017;177(4):471–479.
dermal testosterone on bone and muscle in older Hou X, Cifelli D, Snyder PJ. The effect of testosterone 535. Kaufman JM, Lapauw B, Goemaere S. Current and
men with low bioavailable testosterone levels, low on cardiovascular biomarkers in the testosterone future treatments of osteoporosis in men. Best Pract
bone mass, and physical frailty. J Am Geriatr Soc. trials. J Clin Endocrinol Metab. 2018;103(2):681–688. Res Clin Endocrinol Metab. 2014;28(6):871–884.
2010;58(6):1134–1143. 524. Huang G, Pencina KM, Li Z, Basaria S, Bhasin S, 536. Rochira V, Antonio L, Vanderschueren D. EAA
513. Srinivas-Shankar U, Roberts SA, Connolly MJ, Travison TG, Storer TW, Harman SM, Tsitouras P. clinical guideline on management of bone health in
O’Connell MD, Adams JE, Oldham JA, Wu FC. Ef- Long-term testosterone administration on insulin the andrological outpatient clinic. Andrology. 2018;
fects of testosterone on muscle strength, physical sensitivity in older men with low or low-normal 6(2):272–285.
function, body composition, and quality of life in testosterone levels. J Clin Endocrinol Metab. 2018; 537. Young LA, Neiss MB, Samuels MH, Roselli CE,
intermediate-frail and frail elderly men: a ran- 103(4):1678–1685. Janowsky JS. Cognition is not modified by large but
domized, double-blind, placebo-controlled study. 525. Ng Tang Fui M, Prendergast LA, Dupuis P, Raval M, temporary changes in sex hormones in men. J Clin
J Clin Endocrinol Metab. 2010;95(2):639–650. Strauss BJ, Zajac JD, Grossmann M. Effects of tes- Endocrinol Metab. 2010;95(1):280–288.
514. O’Connel MDL, Roberts SA, Srinivas-Shankar U, tosterone treatment on body fat and lean mass in 538. Emmelot-Vonk MH, Verhaar HJ, Nakhai Pour HR,
Tajar A, Connolly MJ, Adams JE, Oldham JA, Wu FC. obese men on a hypocaloric diet: a randomized Aleman A, Lock TM, Bosch JL, Grobbee DE, van der
Do the effects of testosterone on muscle strength, controlled trial. BMC Medicine. 2016;14(1):153. Schouw YT. Effect of testosterone supplementation
physical function, body composition, and quality of 526. Aversa A, Bruzziches R, Francomano D, Rosano G, on functional mobility, cognition, and other pa-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


life persist six months after treatment in Isidori AM, Lenzi A, Spera G. Effects of testosterone rameters in older men: a randomized controlled
intermediae-frail and frail elderly men? J Clin undecanoate on cardiovascular risk factors and trial. JAMA. 2008;299(1):39–52.
Endocrinol Metab. 2011;96(2):454–458. atherosclerosis in middle-aged men with late-onset 539. Huang G, Wharton W, Bhasin S, Harman SM,
515. Caminiti G, Volterrani M, Iellamo F, Marazzi G, hypogonadism and metabolic syndrome: results Pencina KM, Tsitouras P, Li Z, Hally KA, Asthana S,
Massaro R, Miceli M, Mammi C, Piepoli M, Fini M, from a 24-month, randomized, double-blind, Storer TW, Basaria S. Effects of long-term testos-
Rosano GMC. Effect of long-acting testosterone placebo-controlled study. J Sex Med. 2010;7(10): terone administration on cognition in older men
treatment on functional exercise capacity, skeletal 3495–3503. with low or low-to-normal testosterone concen-
muscle performance, insulin resistance, and baror- 527. Hackett G, Cole N, Bhartia M, Kennedy D, Raju J, trations: a prespecified secondary analysis of data
eflex sensitivity in elderly patients with chronic Wilkinson P; BLAST Study Group. Testosterone from the randomised, double-blind, placebo-
heart failure a double-blind, placebo-controlled, replacement therapy improves metabolic param- controlled TEAAM trial. Lancet Diabetes Endo-
randomized study. J Am Coll Cardiol. 2009;54(10): eters in hypogonadal men with type 2 diabetes but crinol. 2016;4(8):657–665.
919–927. not in men with coexisting depression: the BLAST 540. Resnick SM, Matsumoto AM, Stephens-Shields AJ,
516. Toma M, McAlister FA, Coglianese EE, Vidi V, study. J Sex Med. 2014;11(3):840–856. Ellenberg SS, Gill TM, Shumaker SA, Pleasants DD,
Vasaiwala S, Bakal JA, Armstrong PW, Ezekowitz JA. 528. Jones TH, Arver S, Behre HM, Buvat J, Meuleman E, Barrett-Connor E, Bhasin S, Cauley JA, Cella D,
Testosterone supplementation in heart failure: a Moncada I, Morales AM, Volterrani M, Yellowlees A, Crandall JP, Cunningham GR, Ensrud KE, Farrar JT,
meta-analysis. Circ Heart Fail. 2012;5(3):315–321. Howell JD, Channer KS; TIMES2 Investigators. Lewis CE, Molitch ME, Pahor M, Swerdloff RS, Cifelli
517. Storer TW, Bhasin S, Travison TG, Pencina K, Miciek Testosterone replacement in hypogonadal men D, Anton S, Basaria S, Diem SJ, Wang C, Hou X,
R, McKinnon J, Basaria S. Testosterone attenuates with type 2 diabetes and/or metabolic syndrome Snyder PJ. Testosterone treatment and cognitive
age-related fall in aerobic function in mobility (the TIMES2 study). Diabetes Care. 2011;34(4): function in older men with low testosterone and
limited older men with low testosterone. J Clin 828–837. age-associated memory impairment. JAMA. 2017;
Endocrinol Metab. 2016;101(6):2562–2569. 529. Gianatti EJ, Dupuis P, Hoermann R, Strauss BJ, 317(7):717–727.
518. Storer TW, Basaria S, Traustadottir T, Harman SM, Wentworth JM, Zajac JD, Grossmann M. Effect of 541. Steidle C, Schwartz S, Jacoby K, Sebree T, Smith T,
Pencina K, Li Z, Travison TG, Miciek R, Tsitouras P, testosterone treatment on glucose metabolism in Bachand R; North American AA2500 T Gel Study
Hally K, Huang G, Bhasin S. Effects of testosterone men with type 2 diabetes: a randomized controlled Group. AA2500 testosterone gel normalizes an-
supplementation for 3 years on muscle perfor- trial. Diabetes Care. 2014;37(8):2098–2107. drogen levels in aging males with improvements in
mance and physical function in older men. J Clin 530. Grossmann M, Hoermann R, Wittert G, Yeap BB. body composition and sexual function. J Clin
Endocrinol Metab. 2017;102(2):583–593. Effects of testosterone treatment on glucose Endocrinol Metab. 2003;88(6):2673–2681.
519. Hildreth KL, Barry DW, Moreau KL, Vande Griend J, metabolism and symptoms in men with type 2 542. Pope HG Jr, Amiaz R, Brennan BP, Orr G, Weiser M,
Meacham RB, Nakamura T, Wolfe P, Kohrt WM, diabetes and the metabolic syndrome: a systematic Kelly JF, Kanayama G, Siegel A, Hudson JI, Seidman
Ruscin JM, Kittelson J, Cress ME, Ballard R, Schwartz review and meta-analysis of randomized controlled SN. Parallel-group placebo-controlled trial of tes-
RS. Effects of testosterone and progressive re- clinical trials. Clin Endocrinol (Oxf). 2015;83(3): tosterone gel in men with major depressive disorder
sistance exercise in healthy, highly functioning older 344–351. displaying an incomplete response to standard
men with low-normal testosterone levels. J Clin 531. Tracz MJ, Sideras K, Boloña ER, Haddad RM, antidepressant treatment. J Clin Psychopharmacol.
Endocrinol Metab. 2013;98(5):1891–1900. Kennedy CC, Uraga MV, Caples SM, Erwin PJ, 2010;30(2):126–134.
520. Sattler F, Bhasin S, He J, Chou C-P, Castaneda- Montori VM. Testosterone use in men and its 543. Amanatkar HR, Chibnall JT, Seo BW, Manepalli JN,
Sceppa C, Yarasheski K, Binder E, Schroeder ET, effects on bone health. A systematic review and Grossberg GT. Impact of exogenous testosterone
Kawakubo M, Zhang A, Roubenoff R, Azen S. meta-analysis of randomized placebo-controlled on mood: a systematic review and meta-analysis of
Testosterone threshold levels and lean tissue mass trials. J Clin Endocrinol Metab. 2006;91(6):2011–2016. randomized placebo-controlled trials. Ann Clin
targets needed to enhance skeletal muscle strength 532. Kenny AM, Prestwood KM, Gruman CA, Marcello Psychiatry. 2014;26(1):19–32.
and function: the HORMA trial. J Gerontol A Biol Sci KM, Raisz LG. Effects of transdermal testosterone 544. Calof OM, Singh AB, Lee ML, Kenny AM, Urban RJ,
Med Sci. 2011;66(1):122–129. on bone and muscle in older men with low bio- Tenover JL, Bhasin S. Adverse events associated with
521. Corona G, Giagulli VA, Maseroli E, Vignozzi L, Aversa available testosterone levels. J Gerontol A Biol Sci testosterone replacement in middle-aged and older
A, Zitzmann M, Saad F, Mannucci E, Maggi M. Med Sci. 2001;56(5):M266–M272. men: a meta-analysis of randomized, placebo-
Therapy of endocrine disease: testosterone sup- 533. Snyder PJ, Peachey H, Hannoush P, Berlin JA, Loh L, controlled trials. J Gerontol A Biol Sci Med Sci.
plementation and body composition: results from a Holmes JH, Dlewati A, Staley J, Santanna J, Kapoor 2005;60(11):1451–1457.
meta-analysis study. Eur J Endocrinol. 2016;174(3): SC, Attie MF, Haddad JG Jr, Strom BL. Effect of 545. Fernández-Balsells MM, Murad MH, Lane M,
R99–R116. testosterone treatment on bone mineral density in Lampropulos JF, Albuquerque F, Mullan RJ, Agrwal
522. Basu R, Dalla Man C, Campioni M, Basu A, Nair KS, men over 65 years of age. J Clin Endocrinol Metab. N, Elamin MB, Gallegos-Orozco JF, Wang AT, Erwin
Jensen MD, Khosla S, Klee G, Toffolo G, Cobelli C, 1999;84(6):1966–1972. PJ, Bhasin S, Montori VM. Clinical review 1: adverse
Rizza RA. Effect of 2 years of testosterone re- 534. Snyder PJ, Kopperdahl DL, Stephens-Shields AJ, effects of testosterone therapy in adult men: a
placement on insulin secretion, insulin action, Ellenberg SS, Cauley JA, Ensrud KE, Lewis CE, Barrett- systematic review and meta-analysis. J Clin Endo-
glucose effectiveness, hepatic insulin clearance, and Connor E, Schwartz AV, Lee DC, Bhasin S, crinol Metab. 2010;95(6):2560–2575.
postprandial glucose turnover in elderly men. Di- Cunningham GR, Gill TM, Matsumoto AM, 546. Tan WS, Low WY, Ng CJ, Tan WK, Tong SF, Ho C,
abetes Care. 2007;30(8):1972–1978. Swerdloff RS, Basaria S, Diem SJ, Wang C, Hou X, Khoo EM, Lee G, Lee BC, Lee V, Tan HM. Efficacy
523. Mohler ER III, Ellenberg SS, Lewis CE, Wenger NK, Cifelli D, Dougar D, Zeldow B, Bauer DC, Keaveny and safety of long-acting intramuscular testoster-
Budoff MJ, Lewis MR, Barrett-Connor E, Swerdloff TM. Effect of testosterone treatment on volumetric one undecanoate in aging men: a randomised
RS, Stephens-Shields A, Bhasin S, Cauley JA, Crandall bone density and strength in older men with low controlled study. BJU Int. 2013;111(7):1130–1140.

970 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972
REVIEW

547. Jick SS, Hagberg KW. The risk of adverse outcomes RS, Stephens-Shields A, Cauley JA, Crandall JP, 573. Sharma R, Oni OA, Chen G, Sharma M, Dawn B,
in association with use of testosterone products: a Cunningham GR, Ensrud KE, Gill TM, Matsumoto Sharma R, Parashara D, Savin VJ, Barua RS, Gupta K.
cohort study using the UK-based general practice AM, Molitch ME, Nakanishi R, Nezarat N, Association between testosterone replacement ther-
research database. Br J Clin Pharmacol. 2013;75(1): Matsumoto S, Hou X, Basaria S, Diem SJ, Wang C, apy and the incidence of DVT and pulmonary
260–270. Cifelli D, Snyder PJ. Testosterone treatment and embolism: a retrospective cohort study of the Veterans
548. Roy CN, Snyder PJ, Stephens-Shields AJ, Artz AS, coronary artery plaque volume in older men with Administration Database. Chest. 2016;150(3):563–571.
Bhasin S, Cohen HJ, Farrar JT, Gill TM, Zeldow B, low testosterone. JAMA. 2017;317(7):708–716. 574. Layton JB, Meier CR, Sharpless JL, Stürmer T, Jick SS,
Cella D, Barrett-Connor E, Cauley JA, Crandall JP, 560. Basaria S, Harman SM, Travison TG, Hodis H, Brookhart MA. Comparative safety of testosterone
Cunningham GR, Ensrud KE, Lewis CE, Matsumoto Tsitouras P, Budoff M, Pencina KM, Vita J, Dzekov C, dosage forms. JAMA Intern Med. 2015;175(7):
AM, Molitch ME, Pahor M, Swerdloff RS, Cifelli D, Mazer NA, Coviello AD, Knapp PE, Hally K, Pinjic E, 1187–1196.
Hou X, Resnick SM, Walston JD, Anton S, Basaria S, Yan M, Storer TW, Bhasin S. Effects of testosterone 575. Borst SE, Shuster JJ, Zou B, Ye F, Jia H, Wokhlu A,
Diem SJ, Wang C, Schrier SL, Ellenberg SS. Associ- administration for 3 years on subclinical athero- Yarrow JF. Cardiovascular risks and elevation of
ation of testosterone levels with anemia in older sclerosis progression in older men with low or low- serum DHT vary by route of testosterone admin-
men a controlled clinical trial. JAMA Intern Med. normal testosterone levels: a randomized clinical istration: a systematic review and meta-analysis.
2017;177(4):480–490. trial. JAMA. 2015;314(6):570–581. BMC Med. 2014;12(1):211.
549. Ponce OJ, Spencer-Bonilla G, Alvarez-Villalobos N, 561. Vigen R, O’Donnell CI, Barón AE, Grunwald GK, 576. Liu PY, Yee B, Wishart SM, Jimenez M, Jung DG,
Serrano V, Singh-Ospina N, Rodriguez-Gutierrez R, Maddox TM, Bradley SM, Barqawi A, Woning G, Grunstein RR, Handelsman DJ. The short-term ef-

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Salcido-Montenegro A, Benkhadra R, Prokop LJ, Wierman ME, Plomondon ME, Rumsfeld JS, Ho PM. fects of high-dose testosterone on sleep, breathing,
Bhasin S, Brito JP. The efficacy and adverse events of Association of testosterone therapy with mortality, and function in older men. J Clin Endocrinol Metab.
testosterone replacement therapy in hypogonadal myocardial infarction, and stroke in men with low 2003;88(8):3605–3613.
men: a systematic review and meta-analysis of testosterone levels. JAMA. 2013;310(17):1829–1836. 577. Hoyos CM, Killick R, Yee BJ, Grunstein RR, Liu PY.
randomized, placebo-controlled trials. J Clin Endo- 562. Finkle WD, Greenland S, Ridgeway GK, Adams JL, Effects of testosterone therapy on sleep and
crinol Metab. 2018;103(5):1745–1754. Frasco MA, Cook MB, Fraumeni JF Jr, Hoover RN. breathing in obese men with severe obstructive
550. Cui Y, Zong H, Yan H, Zhang Y. The effect of Increased risk of non-fatal myocardial infarction sleep apnoea: a randomized placebo-controlled
testosterone replacement therapy on prostate following testosterone therapy prescription in men. trial. Clin Endocrinol (Oxf). 2012;77(4):599–607.
cancer: a systematic review and meta-analysis. PLoS One. 2014;9(1):e85805. 578. European Medecine Agency. PRAC review does
Prostate Cancer Prostatic Dis. 2014;17(2):132–143. 563. Etminan M, Skeldon SC, Goldenberg SL, Carleton B, not confirm increase in heart problems with
551. Boyle P, Koechlin A, Bota M, d’Onofrio A, Zaridze Brophy JM. Testosterone therapy and risk of testosterone medicines. Available at: www.ema.europa.eu/
DG, Perrin P, Fitzpatrick J, Burnett AL, Boniol M. myocardial infarction: a pharmacoepidemiologic ema/index.jsp%3Fcurl%3Dpages/medicines/human/
Endogenous and exogenous testosterone and the study. Pharmacotherapy. 2015;35(1):72–78. referrals/Testosterone-containing_medicines/human_
risk of prostate cancer and increased prostate- 564. Baillargeon J, Urban RJ, Kuo YF, Ottenbacher KJ, Raji
referral_prac_000037.jsp%26mid%3DWC0b01ac05805c516f.
specific antigen (PSA) level: a meta-analysis. BJU MA, Du F, Lin Y-L, Goodwin JS. Risk of myocardial
Accessed 1 July 2018.
Int. 2016;118(5):731–741. infarction in older men receiving testosterone
579. Corona G, Rastrelli G, Giagulli VA, Sila A, Sforza A,
552. Kohn TP, Mata DA, Ramasamy R, Lipshultz LI. Ef- therapy. Ann Pharmacother. 2014;48(9):1138–1144.
Forti G, Mannucci E, Maggi M. Dehydroepian-
fects of testosterone replacement therapy on lower 565. Eisenberg ML, Li S, Herder D, Lamb DJ, Lipshultz LI.
drosterone supplementation in elderly men: a
urinary tract symptoms: a systematic review and Testosterone therapy and mortality risk. Int J Impot
meta-analysis study of placebo-controlled trials.
meta-analysis. Eur Urol. 2016;69(6):1083–1090. Res. 2015;27(2):46–48.
J Clin Endocrinol Metab. 2013;98(9):3615–3626.
553. Snyder PJ, Bhasin S, Cunningham GR, Matsumoto 566. Shores MM, Smith NL, Forsberg CW, Anawalt BD,
580. Ly LP, Jimenez M, Zhuang TN, Celermajer DS,
AM, Stephens-Shields AJ, Cauley JA, Gill TM, Matsumoto AM. Testosterone treatment and
Conway AJ, Handelsman DJ. A double-blind,
Barrett-Connor E, Swerdloff RS, Wang C, Ensrud KE, mortality in men with low testosterone levels. J Clin
placebo-controlled, randomized clinical trial of
Lewis CE, Farrar JT, Cella D, Rosen RC, Pahor M, Endocrinol Metab. 2012;97(6):2050–2058.
transdermal dihydrotestosterone gel on muscular
Crandall JP, Molitch ME, Resnick SM, Budoff M, 567. Sharma R, Oni OA, Gupta K, Chen G, Sharma M,
strength, mobility, and quality of life in older men
Mohler ER III, Wenger NK, Cohen HJ, Schrier S, Dawn B, Sharma R, Parashara D, Savin VJ, Ambrose
Keaveny TM, Kopperdahl D, Lee D, Cifelli D, JA, Barua RS. Normalization of testosterone level is with partial androgen deficiency. J Clin Endocrinol
Ellenberg SS. Lessons from the Testosterone Trials. associated with reduced incidence of myocardial Metab. 2001;86(9):4078–4088.
Endocr Rev. 2018;39(3):369–386. infarction and mortality in men. Eur Heart J. 2015; 581. Kunelius P, Lukkarinen O, Hannuksela ML, Itkonen
554. Haider A, Zitzmann M, Doros G, Isbarn H, 36(40):2706–2715. O, Tapanainen JS. The effects of transdermal
Hammerer P, Yassin A. Incidence of prostate cancer 568. Cheetham TC, An J, Jacobsen SJ, Niu F, Sidney S, dihydrotestosterone in the aging male: a pro-
in hypogonadal men receiving testosterone ther- Quesenberry CP, VanDenEeden SK. Association of spective, randomized, double blind study. J Clin
apy: observations from 5-year median followup of 3 testosterone replacement with cardiovascular Endocrinol Metab. 2002;87(4):1467–1472.
registries. J Urol. 2015;193(1):80–86. outcomes among men with androgen deficiency. 582. Sartorius GA, Ly LP, Handelsman DJ. Male sexual
555. Debruyne FM, Behre HM, Roehrborn CG, Maggi M, JAMA Intern Med. 2017;177(4):491–499. function can be maintained without aromatization:
Wu FCW, Schröder FH, Jones TH, Porst H, Hackett 569. Wallis CJ, Lo K, Lee Y, Krakowsky Y, Garbens A, randomized placebo-controlled trial of dihy-
G, Wheaton OA, Martin-Morales A, Meuleman E, Satkunasivam R, Herschorn S, Kodama RT, Cheung drotestosterone (DHT) in healthy, older men for
Cunningham GR, Divan HA, Rosen RC; RHYME P, Narod SA, Nam RK. Survival and cardiovascular 24 months. J Sex Med. 2014;11(10):2562–2570.
Investigators. Testosterone treatment is not asso- events in men treated with testosterone replace- 583. Duschek EJJ, Gooren LJ, Netelenbos C. Effects of
ciated with increased risk of prostate cancer or ment therapy: an intention-to-treat observational raloxifene on gonadotrophins, sex hormones, bone
worsening of lower urinary tract symptoms: pros- cohort study. Lancet Diabetes Endocrinol. 2016;4(6): turnover and lipids in healthy elderly men. Eur J
tate health outcomes in the Registry of Hypo- 498–506. Endocrinol. 2004;150(4):539–546.
gonadism in Men. BJU Int. 2017;119(2):216–224. 570. Sharma R, Oni OA, Gupta K, Sharma M, Sharma R, 584. Uebelhart B, Herrmann F, Pavo I, Draper MW, Rizzoli
556. The Copenhagen Study Group for Liver Diseases. Singh V, Parashara D, Kamalakar S, Dawn B, Chen G, R. Raloxifene treatment is associated with increased
Testosterone treatment of men with alcoholic Ambrose JA, Barua RS. Normalization of testos- serum estradiol and decreased bone remodeling in
cirrhosis: a double-blind study. Hepatology. 1986; terone levels after testosterone replacement ther- healthy middle-aged men with low sex hormone
6(5):807–813. apy is associated with decreased incidence of atrial levels. J Bone Miner Res. 2004;19(9):1518–1524.
557. Morgentaler A, Miner MM, Caliber M, Guay AT, fibrillation. J Am Heart Assoc. 2017;6(5):e004880. 585. Chua ME, Escusa KG, Luna S, Tapia LC, Dofitas B,
Khera M, Traish AM. Testosterone therapy and 571. Martinez C, Suissa S, Rietbrock S, Katholing A, Morales M. Revisiting oestrogen antagonists (clo-
cardiovascular risk: advances and controversies. Freedman B, Cohen AT, Handelsman DJ. Testos- miphene or tamoxifen) as medical empiric therapy
Mayo Clin Proc. 2015;90(2):224–251. terone treatment and risk of venous thrombo- for idiopathic male infertility: a meta-analysis.
558. Alexander GC, Iyer G, Lucas E, Lin D, Singh S. embolism: population based case-control study. Andrology. 2013;1(5):749–757.
Cardiovascular risks of exogenous testosterone use BMJ. 2016;355:i5968. 586. Pelusi C, Giagulli VA, Baccini M, Fanelli F, Mezzullo
among men: a systematic review and meta-analysis. 572. Baillargeon J, Urban RJ, Morgentaler A, Glueck CJ, M, Fazzini A, Bianchi N, Carbone MD, De Pergola G,
Am J Med. 2017;130(3):293–305. Baillargeon G, Sharma G, Kuo YF. Risk of venous Mastroroberto M, Morselli Labate AM, Pasquali R.
559. Budoff MJ, Ellenberg SS, Lewis CE, Mohler ER III, thromboembolism in men receiving testosterone Clomiphene citrate effect in obese men with low
Wenger NK, Bhasin S, Barrett-Connor E, Swerdloff therapy. Mayo Clin Proc. 2015;90(8):1038–1045. serum testosterone treated with metformin due to

doi: 10.1210/er.2018-00178 https://academic.oup.com/edrv 971


REVIEW

dysmetabolic disorders: a randomized, double-blind, 593. Bhasin S, Calof OM, Storer TW, Lee ML, Mazer NA, heart disease; CV, cardiovascular; CVD, CV disease; DHEA,
placebo-controlled study. PLoS One. 2017;12(9):e0183369. Jasuja R, Montori VM, Gao W, Dalton JT. Drug insight: dehydroepiandrosterone; DHEAS, DHEA sulfate; DISF-SDD,
587. Rodriguez KM, Pastuszak AW, Lipshultz LI. Enclo- testosterone and selective androgen receptor modu- Sexual Desire Domain of the Derogatis Interview for Sexual
miphene citrate for the treatment of secondary lators as anabolic therapies for chronic illness and aging. Function; DXA, dual-energy X-ray absorptiometry; E2, es-
male hypogonadism. Expert Opin Pharmacother. Nat Clin Pract Endocrinol Metab. 2006;2(3):146–159. tradiol; ED, erectile dysfunction; EMAS, European Male
2016;17(11):1561–1567. 594. Dalton JT. The long and winding road for selective Aging Study; ER, estrogen receptor; FACIT, Functional
588. T’Sjoen GG, Giagulli VA, Delva H, Crabbe P, De androgen receptor modulators. Br J Clin Pharmacol. Assessment of Chronic Illness Therapy; FDA, Food and
2017;83(10):2131–2133. Drug Administration; GDS, Geriatric Depression Scale; hCG,
Bacquer D, Kaufman JM. Comparative assessment
human chorionic gonadotropin; Hct, hematocrit; HDL,
in young and elderly men of the gonadotropin
high-density lipoprotein; HIMS, Health in Men Study; HNF,
response to aromatase inhibition. J Clin Endocrinol hepatocyte nuclear factor; HOMA, homeostatic model
Metab. 2005;90(10):5717–5722. Acknowledgments
assessment; HOMA-IR, HOMA of insulin resistance; HPT,
589. Loves S, Ruinemans-Koerts J, de Boer H. Letrozole Current Affiliation: I. T. Huhtaniemi’s current affiliation is
hypothalamic–pituitary–testicular; HR, hazard ratio; ICSI,
once a week normalizes serum testosterone in the Research Centre for Integrative Physiology and Phar-
intracytoplasmic sperm injection; IHD, ischemic heart
macology (formerly the Department of Physiology), Institute
obesity-related male hypogonadism. Eur J Endo- disease; IIEF, International Index of Erectile Function; IMT,
of Biomedicine, University of Turku, 20520 Turku, Finland.
crinol. 2008;158(5):741–747. intima–media thicknes; IPSS, International Prostate
Correspondence and Reprint Requests: Jean-Marc
590. Burnett-Bowie SA, McKay EA, Lee H, Leder BZ. Symptom Score; IVF, in vitro fertilization; LC-MS/MS, liquid
Kaufman, MD, PhD, Department of Endocrinology, Ghent

Downloaded from https://academic.oup.com/edrv/article/40/4/906/5381905 by guest on 11 February 2022


Effects of aromatase inhibition on bone mineral chromatography–tandem MS; LOH, late-onset hypo-
University Hospital, Corneel Heymanslaan 10, 9000 Ghent,
density and bone turnover in older men with low gonadism; MetS, metabolic syndrome; MI, myocardial in-
Belgium. E-mail: jean.kaufman@ugent.be.
testosterone levels. J Clin Endocrinol Metab. 2009; farction; MMAS, Massachusetts Male Aging Study; MMSE,
Disclosure Summary: G.T. has received scientific grants
94(12):4785–4792. as a prinicipal investigator from Ipsen, Bayer Shering, and
Mini-Mental State Examination; MONICA, Monitoring
591. Loves S, de Jong J, van Sorge A, Telting D, Tack CJ, Trends and Determinants of Cardiovascular Disease; MS,
Sandoz; consulting fees as an advisory board member from
Hermus A, Westerterp K, de Boer H. Somatic and mass spectrometry; OSA, obstructive sleep apnea; PDE5,
Ipsen and Novartis; and lecturer fees as a speaker from Ferring
phosphodiesterase 5; PPARg, peroxisome proliferator-
psychological effects of low-dose aromatase in- and Novartis. The remaining authors have nothing to
activated receptor g; PPT, Physical Performance Test;
hibition in men with obesity-related hypogonado- disclose.
PRT, progressive resistance training; PSA, prostate-specific
tropic hypotestosteronemia. Eur J Endocrinol. 2013; antigen; QCT, quantitative CT; RCT, randomized controlled
169(5):705–714. Abbreviations trial; RR, relative risk; SERM, selective ER modulator; SHIP,
592. Dias JP, Melvin D, Simonsick EM, Carlson O, Shardell 3MS, modified MMSE; 5-diol, 5-androstene-3b,17b-diol; Study of Health in Pomerania; SNP, single-nucleotide
MD, Ferrucci L, Chia CW, Basaria S, Egan JM. Effects aBMD, area BMD; AD, Alzheimer’s dementia; ADT, an- polymorphism; T, testosterone; T2DM, type 2 diabetes
of aromatase inhibition vs. testosterone in older drogen deprivation therapy; AR, androgen receptor; BMD, mellitus; TEAAM, Testosterone’s Effects on Atherosclerosis
men with low testosterone: randomized-controlled bone mineral density; BMI, body mass index; CHAMP, Progression in Aging Men; TTrials, Testosterone Trials;
trial. Andrology. 2016;4(1):33–40. Concord Health and Ageing in Men Project; CHD, coronary vBMD, volumetric BMD..

972 Kaufman et al Aging and the Male Reproductive System Endocrine Reviews, August 2019, 40(4):906–972

You might also like