You are on page 1of 19

PRIMER

Menopause
Susan R. Davis1, Irene Lambrinoudaki2, Maryann Lumsden3, Gita D. Mishra4,
Lubna Pal5, Margaret Rees6, Nanette Santoro7 and Tommaso Simoncini8
Abstract | Menopause is an inevitable component of ageing and encompasses the loss of ovarian
reproductive function, either occurring spontaneously or secondary to other conditions. It is not yet
possible to accurately predict the onset of menopause, especially early menopause, to give women
improved control of their fertility. The decline in ovarian oestrogen production at menopause can cause
physical symptoms that may be debilitating, including hot flushes and night sweats, urogenital atrophy,
sexual dysfunction, mood changes, bone loss, and metabolic changes that predispose to cardiovascular
disease and diabetes. The individual experience of the menopause transition varies widely. Important
influential factors include the age at which menopause occurs, personal health and wellbeing, and each
woman’s environment and culture. Management options range from lifestyle assessment and intervention
through to hormonal and non-hormonal pharmacotherapy, each of which has specific benefits and risks.
Decisions about therapy for perimenopausal and postmenopausal women depend on symptomatology,
health status, immediate and long-term health risks, personal life expectations, and the availability and
cost of therapies. More effective and safe therapies for the management of menopausal symptoms need
to be developed, particularly for women who have absolute contraindications to hormone therapy.
For an illustrated summary of this Primer, visit: http://go.nature.com/BjvJVX

Menopause is the permanent cessation of menstrual generally mild at this stage of the transition and most
cycles following the loss of ovarian follicular activity women will notice them but not require treatment.
(FIG. 1). This may be spontaneous (natural menopause) Clearly, the STRAW staging primarily applies to
or iatrogenic (secondary menopause). The latter includes women experiencing spontaneous menopause and not
removal of both ovaries (surgical menopause), as well as those with secondary menopause. It is also less useful
ovarian failure resulting from chemotherapy or radio- for women who are unable to observe a change in their
therapy. To facilitate research on menopause, investiga- menstrual bleeding patterns, owing to hysterectomy,
tors convened in 2001 and reported the first Staging of endometrial ablation, hormonal contraception with
Reproductive Aging Workshop (STRAW) recommenda- suppressed ovarian cycles or a progestin intrauterine
tions1. Staging is not only useful for research; it can also device, for example. For such women, the occurrence of
facilitate dialogue between a woman and her clinician, menopausal symptoms, due to the fall in ovarian oestro-
and between clinicians. A refined STRAW classification gen production, may provide the first indication of the
was published in 2012 and includes several data-driven menopause6. Although not all women experience sig-
adjustments to the original publication2–5. nificant symptoms, the fall in oestrogen at menopause
Briefly, the STRAW classification (summarized in results in changes throughout the body including bone
FIG. 2) separates a woman’s life into seven segments, loss, a tendency to increased abdominal fat and a more
with segments –2, –1 and 0 including the early meno- adverse cardiovascular risk profile. In this Primer article,
Correspondence to S.R.D. 
pausal transition, the late menopausal transition and we summarize the current understanding of the physiol-
e‑mail: susan.davis@
monash.edu
the final menstrual period, respectively. Age at natural ogy and clinical consequences of menopause, as well as
School of Public Health and menopause is used to indicate the timing of menopause the available treatment options.
Preventive Medicine, and is confirmed after 1 year of amenorrhoea. The early
Monash University, transition is defined as a departure from previously Epidemiology
99 Commercial Road,
Melbourne, Victoria 3004,
regular menstrual cycle lengths of ≥7 days, or a skipped The first reliable epidemiological estimates for the tim-
Australia. menstrual period. During this stage, oestrogen levels ing of menopause give a median age at natural meno-
are fluctuating but are sufficient overall, and cycles pause of 48–52  years among women in developed
Article number: 15004
doi:10.1038/nrdp.2015.4
are usually ovulatory. If oestrogen levels drop, they are nations7. In a more recent and broader meta-analysis8 of
Published online not maintained at a very low level for long, but will 36 studies spanning 35 countries, the overall mean age
23 April 2015 fluctuate until after menopause. Thus, symptoms are was 48.8 years (95% CI 48.3–49.2), with considerable

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 1

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Author addresses
variation by geographical region (TABLE 1). Although
studies in the United States and Asia tend to show results
1
School of Public Health and Preventive Medicine, Monash University, 99 Commercial close to this mean figure, age at menopause is gener-
Road, Melbourne, Victoria 3004, Australia. ally lower in Africa, Latin America and Middle-Eastern
2
Medical School, University of Athens, and Aretaieio University Hospital, Athens, countries (regional means: 47.2–48.4 years), and higher
Greece.
in Europe and Australia (50.5–51.2 years)8. The reasons
3
Reproductive & Maternal Medicine, Glasgow Royal Infirmary, Glasgow, UK.
4
School of Population Health, Faculty of Medicine and Biomedical Sciences, University
for such regional differences remain far from clear;
of Queensland, Australia. however, previous studies suggest a modest rise of the
5
Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University menopausal age for women in wealthy nations during
School of Medicine, New Haven, Connecticut, USA. the twentieth century 9–13. The biological and environ-
6
Women’s Centre, John Radcliffe Hospital, Oxford, UK. mental factors — and interactions between them — that
7
University of Colorado School of Medicine, Aurora, Colorado, USA. account for regional differences and historical trends in
8
University of Pisa, Pisa, Italy. the timing of menopause remain unclear.
Distinguishing the effects of menopausal transi-
tion on a woman’s health and quality of life (QoL) from
a Reproductive phase the effects of ageing is a major challenge for cross-­
GnRH
sectional, observational studies. Moreover, other major
life events can occur during midlife, such as needing
FSH to care for elderly parents and children leaving home
LH permanently. Findings from a study in the United
Oestradiol States indicate that the perimenopausal stage lasts on
Inhibin B average for almost 4 years, although for some women
Hormone levels

it can last much longer, with a longer perimenopause


being associated with a higher rate of medical consulta-
tions14. Vasomotor symptoms (hot flushes and cold or
night sweats) and vaginal dryness are well-established
symptoms during and after perimenopause15. Indeed,
75% of postmenopausal women <55 years of age report
vasomotor symptoms, and 28.5% of them have moder-
ate to severe symptoms16. Longitudinal analysis of data
0 14 28 from women in the Medical Research Council 1946
Time within cycle (days) British birth cohort has unravelled other effects on
b Menopause QoL. After adjusting for age, life events and a range of
other socio­economic and lifestyle factors, two aspects of
QoL declined with the menopausal transition: namely,
perceived physical health (including energy levels) and
Average hormone levels

psychosomatic status (such as nervous emotional state


and ability to concentrate). These changes were associ-
ated with a longer duration of perimenopause17, which
is consistent with an earlier study from the United States
that found longer perimenopause was associated with
higher rates of medical consultations14.
Vasomotor symptoms are among the most frequently
reported physiological symptoms during and after
meno­pause18,19, but their prevalence among women
in developed nations ranges from 30% to 75%16,20,21.
–5 0 +5 A systematic review of the prevalence of menopausal
Time from last period (years) symptoms in Asian countries found a predominance of
Figure 1 | Regulation of menstrual cycles.  a | In theNature
premenopausal
Reviews |years,
Diseasepulsatile
Primers other physical symptoms over vasomotor and psycho-
release of gonadotropin-releasing hormone (GnRH; indicated by arrows) stimulates the logical symptoms. Furthermore, vasomotor symptoms
synthesis and release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) occurred in premenopausal, perimenopausal and post-
from the anterior pituitary gland. FSH, in particular, stimulates oestradiol and inhibin B menopausal women22. However, most studies that were
production by ovarian follicles. Oestradiol and inhibin B exert feedback on the pituitary considered for this systematic review showed low exter-
gland and hypothalamus that, in turn, modifies the production of GnRH, LH and FSH. nal and internal validity when evaluated by a risk of bias
Co‑ordinated and timed production and release of pituitary FSH and LH result in the tool. Thus, further studies of representative samples and
development of ovarian follicles, ovulation and menstruation. Inhibin B is produced by
using validated questionnaires are needed to clarify the
the ovarian granulosa cells and inhibits FSH synthesis and secretion. b | After menopause,
the ovaries are depleted of follicles, oestradiol and inhibin B production falls, and prevalence of menopausal symptoms in Asian women.
ovulation and menstruation no longer occur. The loss of ovarian responsiveness to FSH Epidemiological studies have provided a detailed
and LH, and the loss of negative feedback of oestradiol and inhibin B on the picture of symptom trajectories experienced through
hypothalamic–pituitary unit, result in increased production and release of GnRH, FSH the menopausal transition and into postmenopause.
and LH. Increased FSH is particularly characteristic of postmenopause. For instance, findings from two studies15,23 have shown

2 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Regular menstrual cycle Subtle changes Menstrual cycle of Interval of amenorrhoea • Variable but • FSH stabilizes
in flow/length variable length, ≥60 days elevated FSH • Very low AMH
• Low FSH of menstrual persistent ≥7-day • Low AMH • Very low inhibin B
• Low AMH cycle difference in length • Elevated >25 IU/l FSH** • Low inhibin B • Very low antral
• Low antral follicle count of consecutive • Low AMH • Very low antral follicle count
• Variable FSH cycles • Low inhibin B follicle count
• Low AMH • Low antral follicle count
Regular menstrual cycle Increasing
• Low inhibin B • Variable but Vasomotor
• Low antral elevated FSH* Vasomotor symptoms likely symptoms most symptoms of
Variable to follicle count • Low AMH likely urogenital atrophy
regular menstrual • Low inhibin B
cycle • Low antral follicle
count
Stage
–5 –4 –3b –3a –2 –1 0 +1a +1b +1c +2

Early Peak Late Early Late Early Late


Reproductive Menopausal transition Postmenopause
Variable duration FMP (0)
Menarche Perimenopause
Principal criteria
Supportive criteria Variable Remaining
Descriptive characteristics duration 1–3 years 2 years (1+1) 3–4 years lifespan

Figure 2 | The stages of reproductive ageing in women.  The Stages of Reproductive Aging NatureWorkshop Disease defined
Reviews |(STRAW) Primers
seven stages ranging from the onset of menstrual cycles at menarche and the reproductive age to the perimenopausal
and postmenopausal phases. Principal (menstrual cycle), supportive (biochemical and imaging) and descriptive criteria
(symptoms) are used to characterize the phases. AMH, anti-Müllerian hormone; FMP, final menstrual period; FSH, follicle-
stimulating hormone. *Blood drawn on cycle days 2–5. **Approximate expected level based on assays using current
international pituitary standard. Figure reproduced from Climacteric (REF. 4) with permission from Informa Healthcare
(www.informahealthcare.com). Figure reprinted by permission from the American Society for Reproductive Medicine
(Fertility and Sterility, 2012, 97, 843–851). Figure republished with permission of Endocrine Press, from the Journal of
Clinical Endocrinology and Metabolism, Harlow, S. D. et al., 97, 1159–1168, 2012; permission conveyed through Copyright
Clearance Center, Inc. Figure reproduced from Harlow, S. D. et al. Executive summary of the Stages of Reproductive Aging
Workshop + 10: addressing the unfinished agenda of staging reproductive aging. Menopause 19 (4), 387–395 (2012).

that women who experienced strong vasomotor underlying biological mechanisms often remain unclear.
symptom peaks either before or after menopause had For instance, primary ovarian insufficiency (menopause
a quick decline in postmenopause. Such trajectories before the age of 40 years) is associated with reduced
for the severity of these symptoms were not evident risk of breast and ovarian cancers, but higher risk of
when analysed according to chronological age, but only cardiovascular disease and osteoporosis24–26. This does
when their timing was examined with respect to age at not necessarily imply any causal relationships, but could
menopause. Such results may help to guide the optimal result from common risk factors. This may be the case
treatment and management of symptoms. for cardio­vascular disease, for which recent findings
Age at menopause is also increasingly recognized suggest that pre-existing risk factors, such as raised total
as an indicator for health outcomes in later life, espe- serum cholesterol and blood pressure, are also associ-
cially those related to oestrogen exposure, although the ated with earlier menopause27. Overall, however, each
additional year of later menopause is linked with a 2%
reduction in all-cause mortality 28,29.
Table 1 | Geographical variation in age at menopause*
Region or n Number Mean age at Heterogeneity Mechanisms/pathophysiology
country of studies menopause (I‑squared; %) Factors influencing menopause
(95% CI) The functional lifespan of human ovaries is determined
Africa 1,175 3 48.4 (48.1–48.6) 0.0 by a complex and yet largely unidentified set of genetic,
Asia 39,158 8 48.8 (48.1–49.4) 98.9 hormonal and environmental factors (FIG. 3). Women
undergo menopause when follicles in their ovaries are
Australia 9,268 2 51.3 (49.8–52.8) 99.1
exhausted. However, the clinical manifestations of meno­
Europe 18,692 6 50.5 (50.0–51.1) 96.6 pause result from dynamic interactions between neuro­
Latin America 18,073 3 47.2 (45.9–48.6) 99.1 endocrine changes and alterations in the reproductive
Middle East 7,733 8 47.4 (46.9–47.8) 97.2 endocrine axis that governs the function of the ovaries.
It is unclear why ovaries start their function at
United States 15,690 6 49.1 (48.8–49.4) 94.6
puberty and stop it at menopause, and understanding
Total 109,789 36 48.8 (48.3–49.2) 99.6 this phenomenon would have far-reaching implications
*Data from REF. 8. for reproductive and health issues.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 3

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

• Genetic factors Hypothalamus Hypothalamic ageing identified a region on chromosome 8 that is associated
• Environment with age at menopause39. Interestingly, the gene encod-
• Lifestyle Desynchronized
GnRH GnRH secretion ing the gonadotropin-releasing hormone (GnRH) is
• Systemic diseases
near this region.
Brain Studies looking at associations between genes
Impaired timing
LH of LH surge encoding factors that are involved in reproductive
FSH pathophysiology and menopause have been disap-
FSH increase pointing. Most studies have failed to identify asso-
ciations or the results could not be replicated. Among
Pituitary gland the genes investigated, those encoding the oestrogen
receptor-α (ESR1), 17β-hydroxysteroid dehydrogenase
type 1 (HSD17B1), anti-Müllerian hormone (AMH)
Ovary
and AMH receptor type 2 (AMHR2) have been the
most promising 37. Women who are carriers of genetic
Uterus Follicle
loss Active mutations in early onset breast cancer 1 (BRCA1) or
follicles Functional
ovarian ageing BRCA2 are on average 3 years younger at menopause
than matched controls (50 years versus 53 years)40. A
AMH Decline in
inhibin B number of known genetic disorders result in earlier
Inhibin B
Oestradiol
menopause, including mutation of the gene encoding
Decline in AMH
forkhead box protein L2 (FOXL2), which also causes
Decrease in Oestradiol the blepharophimosis, ptosis and epicanthus inversus
ovarian follicle fluctuation (BPES) syndrome41. Mutations of the genes encoding
mass and function and decline
bone morphogenetic protein 15 (BMP15) and growth
and differentiation factor 9 (GDF9) result in premature
Anovulatory cycles and finally menopause42,43. Galactosaemia has been associated with
loss of menstrual cycles
menopause prior to the age of 40 years in one study 44,
Figure 3 | Menopausal loss of ovarian function.  Several processes
Nature Reviewscontribute
| DiseasetoPrimers
but no relationship was observed in a follow‑up study
declining ovarian function and the development of menopause including: hypothalamic of a similarly sized cohort 45. Variants of other genes that
and functional ovarian ageing; environmental, genetic and lifestyle factors; and systemic are involved in ovarian function, such as those encoding
diseases. Hypothalamic ageing leads to desynchronized gonadotropin-releasing follicle-stimulating hormone (FSH) and inhibin recep-
hormone (GnRH) production and an impaired surge of luteinizing hormone (LH) release tors, have been shown to be associated with early and
from the pituitary gland. These central nervous system changes, together with ovarian premature menopause43. Women who are carriers for
ageing, impair ovarian follicle maturation, hormone production (inhibin B, anti-Müllerian the fragile X mutation and have an intermediate num-
hormone (AMH) and oestradiol) and ovulation. This leads to cycle irregularities and ber of CGG repeats in the fragile X mental retardation 1
follicle-­stimulating hormone (FSH) upregulation.
(FMR1) gene have been observed to undergo early and
premature menopause46.
Recently, genome-wide association studies have
Genetic factors. The timing of menopause reflects found new genes that might influence ovarian ageing
a complex interplay of genetic, epigenetic, socio­ and menopause47–50. The most relevant are the BR serine/­
economic and lifestyle factors. Estimates of the herit- threonine kinase 1 gene (BRSK1), which encodes an
ability in meno­pausal age range from 30% to 85%30,31. AMP-activated protein kinase (AMPK)-related kinase,
Approximately 50% of the interindividual variability and the gene encoding the minichromosomal mainte-
in age at menopause is related to genetic effects 32. nance complex component 8 (MCM8), which is essential
Women whose mothers or other first-degree relatives for genome replication.
were known to have early menopause have been found
to be 6–12‑fold more likely to undergo early meno- Ovarian lifespan and follicle loss. The ovaries and the
pause themselves33,34. Furthermore, cross-sectional follicles are central to determining menopause timing 51.
and cohort studies have shown that a woman’s age at The number of follicular cells is determined before birth,
menopause is strongly associated with her mother’s age when oocytes expand to a maximum of 6–7 million at
at menopause30–36. However, genetic studies have so far mid-gestation. Afterwards, oocytes are rapidly lost due
failed to clearly identify the genetic traits that underlie to apoptosis, leading to a population of 700,000 at birth
this heritability 37. and 300,000 at puberty. Continuing apoptosis, along
Linkage analysis studies pinpoint areas on chromo- with the loss of oocytes during the 400–500 cycles of fol-
some X (region Xp21.3) that are associated with early licular recruitment in a normal reproductive life (some-
(<45 years) or premature (<40 years) menopause38. times involving multiple follicles per cycle), leads to
A region on chromosome 9 (9q21.3) is also associated final exhaustion of these cells at midlife and menopause
with age at menopause. This region contains multiple between 45 and 55 years of age51. These numbers high-
genes, including one that encodes a protein of the B cell light that the duration of ovarian functionality depends
lymphoma 2 (BCL‑2) family 38. BCL‑2 is involved in only little on ovulation and is mostly determined by the
apoptosis, and may thus influence menopause through extent and rapidity of oocyte apoptosis — what governs
follicular depletion. Other linkage analysis studies have this process is unknown.

4 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Although oocyte loss is fundamental for menopause, With time, the age-related hypothalamic modifica-
the specialized granulosa and theca steroid-secreting tions determine a decrease in oestrogen sensitivity and the
cells, and not the oocytes, determine the coordinated LH surge becomes more erratic. Follicles also become less
processes that drive the menstrual cycle. Follicular sensitive to gonadotropins, which leads to luteal-phase
cells are regulated by pituitary gonadotropins, as well defects and anovulatory cycles, and accordingly, to the
as by locally produced hormones. Loss of sensitivity to first menstrual irregularities. Hypothalamic insensitivity
stimulating factors by follicular cells is thought to have a to oestrogens also explains why menopausal symptoms —
key role in the decline of ovarian function52. Consistent such as hot flushes and night sweats — commonly occur
with this view, the most replicable and linear endocrine at this stage, when women have rather high levels of oes-
change throughout the menopausal transition is the pro- trogens, as well as why exogenous oestrogens are effective
gressive decline in inhibin B and AMH, which marks in reducing the symptoms.
the decrease in follicular mass and/or functionality and In summary, natural menopause is the consequence
explains why fertility is impaired in women well before of lost ovarian function. This is the final step in a long
any disruption of menstrual cyclicity 53. and irregular cascade of events taking place both in
the brain and in the ovaries. Genetic factors influence
Neuroendocrine events. The hypothalamic–pituitary the timing of this process, but the key molecular path-
reproductive axis undergoes considerable changes dur- ways involved are still unknown. Identifying such fac-
ing the menopausal transition. These modifications are tors would be invaluable to inform the development of
in part secondary to declining ovarian function, but new strategies to treat reproductive dysfunction and
several lines of evidence suggest that the brain under- menopause-­associated diseases.
goes independent functional modifications that are
important for reproductive ageing 54. According to this Symptoms and consequences
hypothesis, menopause may mirror puberty, a time at Menopausal symptoms. Most women entering meno-
which a set of hypothalamic processes also influences pause experience vasomotor symptoms. A hot flush
the reproductive axis. is a sudden episode of vasodilation in the face and
Increases in pituitary-derived FSH can be identified neck, which lasts 1–5 minutes and is accompanied by
in middle-aged women well before oestrogen declines profuse sweating. Women experiencing hot flushes
or cycle irregularities are noticed. Similarly, changes in are reported to have a narrower thermoneutral zone,
luteinizing hormone (LH) secretion patterns are found such that subtle changes of core temperature elicit
at this stage, with broader and less frequent pulses. thermoregulatory mechanisms such as vasodilation,
Experimental work in rodents55 suggests that an age- sweating or shivering. Declining levels of oestrogens
related desynchronization of the neurochemical signals and inhibin B, as well as increasing FSH levels, explain
involved in activating GnRH neurons takes place before only part of the disturbed thermoregulation, which is
changes in oestrous cyclicity. Several hypothalamic associated with changes in brain neurotransmitters
neuro­peptides and neurochemical agents (for example, and peripheral vascular reactivity 58.
glutamate, noradrenaline and vasoactive intestinal pep- Hot flushes usually occur in late perimenopause and
tide) that drive the oestrogen-mediated surge of GnRH the first postmenopausal years. Some women, however,
and LH seem to dampen with age or lack the precise may continue to experience vasomotor symptoms for
temporal coordination that is required for a specific many years after menopause59. Occasionally, hot flushes
pattern of GnRH secretion55. Disruption of this hypo- occur in the late reproductive years16,60, or several years
thalamic biological clock would lead to progressive after menopause60. The occurrence and intensity of
impairment in the timing of the preovulatory LH surge, menopausal symptoms vary widely between women
which would add to the poor ovarian responsiveness that and depend on genetic, environmental, racial, lifestyle
is encountered at this reproductive stage. and anthropometric factors. Black race, smoking and
overweight — in particular central obesity — increase
Endocrine changes during the transition. As discussed the prevalence and severity of vasomotor symptoms61,62.
above, the endocrine changes of the late reproduc- Sleep disturbances are also very common during the
tive years depend on the combined dysfunction of menopausal transition and they are mainly attributed
the ovaries and the hypothalamus56. A shortened fol- to frequent arousals due to night sweats and occurring
licular phase and the associated increase in FSH levels secondarily to psychological factors63,64. Mood disorders
are characteristic of early menopausal transition. This such as depression and anxiety are not caused by meno-
accounts for the shorter cycle intervals that are expe- pause; vulnerable women, however, may have their first
rienced by many women in this period of life. Cohort episode or a relapse during the transition65. Muscle and
studies have demonstrated that shortened follicular joint pain is also part of the menopausal symptomatol-
phases are associated with accelerated ovulation, which ogy 66 and it is highly correlated with hot flushes and
occurs at a smaller follicle size57. The prevailing expla- depressed mood67.
nation for this phenomenon is the loss of inhibin B
production, which leads to increased FSH release and, Urogenital atrophy. The anatomy and function of the
therefore, to an ‘overshoot’ of oestrogen production. female lower genital tract is oestrogen-dependent. Upon
This would facilitate (and accelerate) the achievement menopausal oestrogen decline, tissues lining the vagina,
of the LH surge57. vulva, bladder and urethra undergo atrophy, which

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 5

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Ageing Menopausal oestrogen decline tissue. Oestrogens regulate the coupling of the bone
resorption and formation processes. Postmenopausal
oestrogen decline leads to the uncoupling of bone
• Vitamin D deficiency • Reduced Osteoblasts remodelling, which results in excessive resorption.
• Decline in intestinal physical activity More specifically, oestrogen deficiency results in exces-
calcium absorption • Sarcopenia
• Impaired renal synthesis
sive production of the cytokine RANKL (receptor
• Increased RANKL activator of nuclear factor-κB ligand; also known as
of 1,25(OH)2 vitamin D3
• Decreased OPG
TNFSF11) by osteoblasts, which — upon binding to
its receptor RANK (also known as TNFRSF11A) on
Loss of mechanical stimulation of osteocytes
the surface of pre-osteoclasts and mature osteoclasts
— promotes osteoclasto­genesis and bone resorption72.
Osteoprotegerin (OPG; also known as TNFRSF11B)
Decreased bone formation Pre-osteoclasts
is a cytokine secreted by osteoblasts following oestro-
gen stimulation and is a natural inhibitor of RANKL72.
Secondary
Oestrogen deficiency is associated with decreased OPG
hyperparathyroidism Osteoclasts production, further augmenting RANKL activity73,74.
The age-associated decline in intestinal calcium absorp-
tion, vitamin D deficiency and impaired synthesis of
Increased bone resorption active 1,25‑dihydroxyvitamin D3 by the kidney lead to
secondary hyperparathyroidism, which further con-
tributes to accelerated bone resorption75. Finally, loss
Low bone mass and
loss of bone architecture of skeletal mechanical stimulation due to reduced daily
activity and loss of skeletal muscle mass may interact
with decreased bone formation due to the absence of
Low-trauma fracture oestrogen-mediated downregulation of sclerostin pro-
duction by osteocytes76. Both processes — increased
Figure 4 | Pathogenesis of postmenopausal osteoporosis.  Nature | Disease Primers
Reviewsoestrogen
Reduced
bone resorption and decreased bone formation — lead
production results in increased receptor activator of nuclear factor-κB ligand (RANKL)
levels, which leads to osteoclast activation and increased bone resorption. Furthermore,
to diminished bone strength and to fractures upon min-
production of osteoprotegerin (OPG), an osteoclast inhibitor, by osteoblasts is imal skeletal load. The earlier the age at menopause, the
decreased. These changes may be compounded by general age-related changes in bone higher is the risk of osteoporosis later in life77 (FIG. 4).
metabolism and remodelling, including disturbed vitamin D and calcium homeostasis,
secondary hyperparathyroidism and less mechanical stimulation of bone turnover. Metabolic consequences. The prevalence of obesity is
higher in postmenopausal women than in premenopau-
sal women78. This is a consequence of a multifactorial
causes a cluster of symptoms including vaginal dry- process that involves reduced energy expenditure due
ness, painful intercourse (dyspareunia), vulvar pruritus, to physical inactivity, which is sometimes compounded
burning and discomfort, as well as recurrent urogenital by depression, as well as due to muscle atrophy and
infections68. Vulvovaginal atrophy and urinary tract atro- a lower basal metabolic rate78. Whereas menopause
phy, due to oestrogen deficiency, are collectively called per se is not associated with weight gain, it leads to an
the genitourinary syndrome of menopause69. Unlike increase of total body fat and a redistribution of body
hot flushes and night sweats, which improve over time, fat from the periphery to the trunk, which results in
symptoms of urogenital atrophy persist throughout visceral adiposity78,79. Abdominal obesity and meno-
postmenopausal life and may have a serious impact on pausal oestrogen decline are associated with adverse
sexual health and QoL70. Pain during intercourse, sec- metabolic changes such as insulin resistance, a pro-
ondary to vulvovaginal atrophy, leads to diminished pensity to develop type 2 diabetes mellitus and dys-
sexual desire, arousal difficulties, relationship problems, lipidaemia characterized by high triglyceride levels,
and diminished physical and emotional sexual satisfac- low high-density lipoprotein (HDL) cholesterol levels
tion71. Although the majority of women have signs of and an increased frequency of small, dense low-­density
uro­genital atrophy upon physical examination, less than lipoprotein (LDL) particles 79,80. Altered adipokine
half of the postmenopausal population report bother- secretion, which leads to chronic inflammation, is a
some complaints68. Age, sexual activity, ethnicity and possible mechanism that links abdominal obesity to
attitudes towards menopause influence the occurrence its metabolic consequences79,81,82 (FIG. 5).
and the severity of urogenital symptoms70.
Cardiovascular disease. Beyond the beneficial effects
Osteoporosis. Bone is a dynamic tissue that undergoes on metabolic and immune parameters, oestrogens are
continuous remodelling throughout life. This process potent vasoactive hormones that promote vascular
begins with bone resorption, which is carried out by remodelling and elasticity, and regulate reactive dila-
multinucleated giant cells called osteoclasts. The lacuna tion and local inflammatory activity83. Oestrogen defi-
created by osteoclasts is subsequently filled with oste- ciency after menopause leads to the activation of the
oid — new non-mineralized bone synthesized by osteo­ renin–angiotensin system, the upregulation of the potent
blasts — which is later mineralized to form mature bone vasoconstrictor endothelin and the impairment of nitric

6 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Menopause Menopausal oestrogen decline


often bothersome for the affected women96. Although
and ageing menopause seems to be associated with changes in cog-
nition, it cannot be assumed that oestrogen therapy will
prevent cognitive decline. There is a general consensus
• Physical Indirect effects on Direct effects on the vasculature
inactivity cardiovascular risk factors • Activation of the renin– that oral oestrogen therapy should not be prescribed to
• Mood • Visceral adiposity angiotensin system prevent or treat cognitive decline97,98.
instability • Dyslipidaemia: • Increased angiotensin II
• Sarcopenia increased LDL-c and • Increased endothelin 1
decreased HDL-c • Decreased NO synthase Diagnosis, screening and prevention
• Increased triglycerides Risk factors for earlier menopause
• Insulin resistance Postnatal nutrition might influence the age at meno-
• Increased blood pressure • Oxidative stress
• Chronic inflammation • Vascular cell proliferation
pause. Women in the 1946 British birth cohort who had
• Vascular wall inflammation been breastfed experienced later menopause than those
• Arterial stiffness who had not 36,99, and women who had a low weight at
Obesity Atherosclerosis • Endothelial dysfunction 2 years of age had earlier menopause99. Similarly, Dutch
children who experienced severe caloric restriction as
Ischaemic heart disease a result of the 1944–1945 famine had an earlier meno-
and stroke pause than those who were not exposed to the famine100.
There is mixed evidence for relationships between age at
Nature Reviews
Figure 5 | Consequences of menopause on the cardiovascular | Disease
system.  Primers
The loss of menarche and age at menopause101.
oestrogen at menopause is associated with increased visceral fat, a more adverse lipid Emotional stress at a young age may affect repro-
profile and activation of the renin–angiotensin pathway, which leads to impaired
ductive ageing and there is evidence that women who
endothelial function. These changes, together with ageing-related changes, lead to an
increased risk of hypertension, obesity, type 2 diabetes mellitus, atherosclerosis, experience parental divorce early in life tend to have
ischaemic heart disease and stroke. HDL‑c, high-density lipoprotein cholesterol; LDL‑c, earlier menopause99,102. Some studies have also found
low-density lipoprotein cholesterol; NO, nitric oxide. that women with lower adult socioeconomic position,
which may indicate greater exposure to stress, tend to
experience menopause earlier than women of higher
oxide-mediated vasodilation. Oxidative stress, which is position, even after adjustment for smoking and par-
augmented by endothelin and angiotensin II, may fur- ity 103–107. A Latin-American study found that women
ther contribute to atherosclerotic processes84. Thus, soon living at altitudes above 2000 m were also more likely to
after menopause, women exhibit increases in blood pres- experience early menopause107.
sure, as well as subclinical vascular disease, which can be Of the various lifestyle and environmental factors in
observed as increased carotid and femoral artery intima- adulthood that have been investigated with respect to
media thickness, coronary artery calcium score and arte- the timing of menopause, only cigarette smoking 108–110
rial stiffness, and impaired flow-mediated dilation85–88. and nulliparity 34,111,112 are consistently associated with an
Clinical consequences of cardiovascular disease usually earlier menopause. It is controversial whether cessation
occur later in women than in men and ischaemic heart of smoking prior to menopause eliminates this effect,
disease usually manifests 10 years later in women than in with some studies implicating only current smoking
men89. The risk of stroke doubles during the first decade as a risk factor for earlier menopause113,114, and others
after menopause and ultimately exceeds that of men in implicating both past and current smoking 115. The lat-
older age90. These vascular events tend, however, to have ter association implies that smoking has an overall toxic
a more severe prognosis in women89. Early menopause effect on the follicle pool, presumably mediated by the
and primary ovarian insufficiency have consistently polycyclic aromatic hydrocarbons in cigarette smoke116.
been associated with increased risk of coronary heart Current, but not past, smoking has also been associated
disease, stroke and mortality91–93 (FIG. 5). with reduced levels of AMH, which is a marker for the
ovarian reserve117. Adverse life events, HIV infection and
Cognition. Oestrogens act in areas of the central nerv- street-drug use have also been related to earlier meno-
ous system that control learning, registering and retriev- pause113,118–120, but alcohol and coffee consumption gen-
ing information, judgement, evaluation processes and erally show no association115. Birth control pill use has
language skills. These areas mainly involve the prefron- been shown to be associated with a slightly later age at
tal cortex, hippocampus and striatum. Oestrogens act menopause in some but not all studies113,121.
through both genomic and non-genomic mechanisms. A recent systematic review found a modest asso-
They increase cellular proteins, thus promoting neuronal ciation between moderate to high physical activity
growth and survival, neural transmission and function, and earlier menopause in an unadjusted but not an
and also synaptogenesis94. Furthermore, oestrogens adjusted meta-analysis, whereas overweight had a
limit the inflammatory response in the central nervous modest association with later menopause8. Overweight
system, which helps to avoid repeated inflammatory and obesity have been linked with a later menopause
insults that might result in dystrophy and a propensity in several studies122,123. Premenopausal weight gain and
to dementia95. Most studies indicate that menopause premenopausal episodic weight loss of greater than 5 kg
affects cognitive function and, more specifically, aspects are each independently associated with a later meno-
related to verbal memory and verbal fluency. The effect pause124. Higher body mass index (BMI) has been asso-
size seems to be small; however, cognitive changes are ciated with a greater likelihood of transitioning from

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 7

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Table 2 | Common menopausal symptoms with timing of onset and duration


Symptom Typical age at onset Peak severity Average duration Refs
(years) (years)
Hot flushes 47 Late transition 6–10 129–131
Poor sleep 40–50 Late transition Variable 132
Adverse mood 49 Late transition 2–4 126,127,133
Vaginal dryness 49 Early to late transition Persists lifelong if untreated 128

premenopause to perimenopause but not from peri- oestradiol levels are expected to be consistently low
menopause to postmenopause123. Non-obese women (<20 pg ml–1); progesterone is no longer produced136.
tend to have a more rapid decline in oestradiol as they Of note, there is no acute change in testosterone levels
transit the menopause125. in relation to the menopause transition135,137.
AMH is a member of the transforming growth
Clinical diagnosis factor‑β (TGFβ) superfamily of proteins, and is found
The common symptoms of menopause, with their in ovarian granulosa cells and oocytes138. AMH might be
approximate onsets and durations126–133, are shown in useful for predicting the time of menopause. Serial AMH
TABLE 2. As women progress to the late menopausal measurements in a cohort study of 50 women indicated
transition (STRAW Stage –2 to –1)2–5, their menstrual that a drop of AMH below 0.05 ng ml–1 predicted meno-
cycle length increases to >60 days, anovulation becomes pause within the subsequent 5 years139. Overall, the sen-
more likely and periods of time with little or no oes- sitivity of AMH measurements in published studies to
trogen secretion occur. At this time, all of the com- date does not enable the clinician to predict a woman’s
monly observed menopausal symptoms worsen acutely. final menstrual period beyond simple historical and
Many symptoms peak in their intensity at this time, and clinical criteria, such as age and duration of amenor-
women may require treatment. rhoea140. Examination of AMH change over time holds
A woman is considered postmenopausal when she promise for refining our ability to prospectively deter-
is over the age of 45 and has gone at least 12 months mine when menopause will occur 141. Ultimately, with
without a spontaneous menstrual period. No specific the development of a sufficiently sensitive AMH assay,
diagnostic testing is required unless the clinical presen- it should be possible to help forecast the final menstrual
tation is atypical. Atypical presentations would include period with better accuracy.
substantial mood changes, new-onset anxiety or fatigue,
and arthralgia without flushes or sweats. Earlier ages of Diagnostic imaging
onset of prolonged amenorrhoea require consideration Other measurements have also been proposed for fore-
of other diagnoses such as polycystic ovary syndrome, casting the timing of permanent ovarian failure, includ-
secondary hypogonadotropic hypogonadism, hyper­ ing both antral follicle counts (assessed by transvaginal
prolactinaemia, pituitary tumours or uterine problems ultrasonography), recording of all follicles measuring
such as Asherman syndrome. A menopausal woman ≤7 mm in diameter (usually done in the early follicu-
who has undergone at least 12 months of amenorrhoea lar phase of the cycle), and functional ovarian reserve,
is unlikely to ever have another menstrual period, but it which is ascertained by dynamic testing of the ovary
can occur in about 10% of women134. with a stimulating agent (either clomiphene citrate
or FSH). However, these methods are better predic-
Biochemical assessment tors of the loss of fertility than they are of menopause
Although specific diagnostic testing for menopause per se142. Transvaginal ultrasonography is a very useful
is not recommended, clinical situations may arise in method for assessing fertility status and can provide
which it is beneficial to document primary gonadal information about ovarian ageing when appropriate.
failure. Usually, an increased FSH level will suffice to Measurement of the number of antral follicles can pro-
confirm this. Changes in FSH, oestradiol and inhi- vide useful information about the likelihood of preg-
bin B have been well documented in population-based nancy in women who are of advanced reproductive age
samples of women undergoing observational research. and wish to conceive. However, antral follicle counts
Measurement of oestradiol during the perimenopause have not been as useful in predicting menopause143.
is not clinically useful. Inhibin B is the first hormone Other ovarian measurements such as total ovarian vol-
to decline, and its drop precedes a rise in FSH135. The ume and stromal thickness have also been proposed,
compensatory rise in FSH causes follicles to continue but they currently lack the sensitivity and specificity to
to grow and leads to a shortening of the follicular phase be clinically useful.
of the menstrual cycle. Eventually, the follicle pool
becomes depleted and folliculogenesis no longer occurs Screening for osteoporosis
reliably. This juncture is when the late meno­pausal Operationally, osteoporosis is defined as reduced
transition begins. Follicle failure is intermittent in the bone mineral density measured by dual-energy X‑ray
late transition and is eventually superseded by perma- absorptio­metry (DXA). Although the estimated preva-
nent amenorrhoea. After menopause has happened, lence of osteoporosis in women aged 40–65 years is

8 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Table 3 | Differential diagnoses of menopausal symptoms


Type Conditions Investigation or laboratory finding
Other causes of low • Cyclical perimenstrual flushes Complete medical history
oestrogen • Post-partum changes
• Aromatase inhibitors
• Selective oestrogen receptor modulators
• GnRH agonists and antagonists
Chronic infections Various Body temperature measurement during flush
Dietary • Alcohol Careful medical history
• Spicy food
• Food additives (for example, monosodium
glutamate and sulphites)
Hormonal • Thyrotoxicosis Thyroid function tests
• Exogenous thyroid hormone excess
Medications • Some selective serotonin reuptake inhibitors Drug history
• Nicotinic acid
• Opiates
• Calcium channel blockers
Hormone-producing Carcinoid tumours leading to carcinoid Increased plasma chromogranin A and
tumours syndrome increased urine 5‑hydroxyindoleacetic acid
Phaeochromocytoma Increased plasma metanephrines
Other • Lymphoma Medical history, examination and tests as
• Medullary carcinoma of the thyroid appropriate
• Pancreatic islet-cell tumours
• Renal cell carcinoma
• Mastocytosis and mast cell disorders
GnRH, gonadotropin-releasing hormone.

low 144, there is a mean vertebral bone loss of 6.4% and Other causes of vasomotor symptoms should
neck of femur loss of 5% during the menopause transi- be excluded if the presentation is atypical (TABLE 3).
tion145. The available data do not support DXA screening Hormonal hot flushes do cause a significant rise in core
for postmenopausal women of ≤60 years of age who do temperature. If a woman records an increase in her oral
not have an identifiable medical condition, do not use temperature with flushing or night sweats, infective
medication that is associated with an increased risk of causes must be investigated. Thyrotoxicosis can mimic
osteoporosis or have no past history of a fracture due to menopause, with women presenting with agitation and
low trauma146. In line with this, the American College of anxiety, sleep disturbance, overheating, sweating and
Preventive Practice has recommended that DXA screen- palpitations. These symptoms may precede the classic
ing should be restricted to women aged 50–65 years with thyrotoxic symptoms of weight loss and tremor. A care-
one major risk factor (such as a history of menopause ful medical and medication history should identify other
prior to the age of 45 years or fragility fracture) or two possible causes. Serotonin-producing carcinoid tumours
minor risk factors (such as cigarette smoking or weight can present with nocturnal diarrhoea and episodic flush-
<57 kg)147. However, the debate is ongoing as to which ing without sweating. Phaeochromocytomas release the
women aged <65 years should be screened and currently, catecholamines adrenaline and noradrenaline, and are
no specific recommendations are universally accepted. characterized by persistent hypertension, flushing and
A recent study of women aged 40–65 years who under- profuse sweating 149.
went DXA screening has shown that the three strongest Some women may present with oligomenorrhoea
predictors of osteoporosis are being postmenopausal, and mood changes or depression. Perimenopausal
not using hormone therapy, and having a low BMI148. depression needs to be differentiated from major
depression and simple dysphoria. The diagnosis of
Differential diagnosis of menopause major depression requires at least 2 weeks of depressed
If a woman is >50 years of age, has stopped menstru- mood, or loss of interest or pleasure in nearly all activi-
ating and has classic oestrogen deficiency symptoms, a ties for most of the day, nearly every day, accompanied
diagnosis other than menopause is very unlikely. Other by at least four of the following symptoms: change in
causes of amenorrhoea should be considered in younger appetite or sleep, fatigue, psychomotor agitation or
women and in women aged >50 years who have atypical retardation, feelings of worthlessness and/or guilt,
symptoms. These causes include thyroid disease, pro- diminished concentration or indecisiveness, and sui-
lactinoma, severe depression or stress, and substantial cidal ideation150. By contrast, perimenopausal depres-
weight loss. Each of these may be associated with vaso- sion is usually accompanied by irritability, hostility or
motor symptoms and mood changes. Pregnancy should agitation, and anxiety 151. Clinically, it resembles the
always be considered in the setting of amenorrhoea. mood changes of premenstrual syndrome, with negative

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 9

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

mood, negative self-concept and less effective coping Prevention of menopause


abilities152. The lability of peri­menopausal depression is That menopause is inevitable is based on the concept
a distinguishing feature, in contrast to the pervasive low that human females have a fixed ovarian oocyte reserve
mood that is seen in major depression153. at birth and that oocyte numbers decline with age159.
In the later reproductive years, the quality of oocytes
Factors hindering diagnosis diminishes, such that fertilization and live birth rates fall,
Although one of the defining features of menopause is and the risk of birth defects and miscarriage increase.
the cessation of the menstrual period, it more broadly One group has identified oogonial stem cells in ovarian
encompasses the permanent cessation of ovarian repro- cortical tissue of young women160. This gives rise to the
ductive function. Thus, the traditional definition is not hypothesis that human ovarian germ cells are renewed
useful in the setting of iatrogenic causes of amenorrhoea, during the reproductive years, and even inspires the pos-
such as hysterectomy, endometrial ablation or progestin sibility that such cells could be used to extend a woman’s
intrauterine devices. Biochemical investigations may reproductive lifespan, and effectively delay or prevent
be warranted for women with iatrogenic amenorrhoea menopause. The finding of oogonial stem cells in human
(notably, measurement of FSH and oestradiol) and for ovaries is controversial161 and is yet to be replicated by
younger women (measurement of AMH). The latter is other groups162.
less useful in women >50 years of age, as most women
will have low AMH levels by this time. Some women may Management
report vasomotor symptoms when using combined oral Managing menopausal symptoms need not be complex.
contraception. In this instance, menopause can only be Bothersome vasomotor symptoms, disturbed noctur-
effectively diagnosed if the oral contraception is stopped nal sleep and an overall deterioration in QoL are by far
for several weeks, after which menopausal status can be the most common reasons for women to seek medical
assessed both clinically and biochemically. attention in midlife. Caregivers and patients can now
choose from an expanding repertoire of pharmacologi-
Screening for menopause cal options (both hormonal and non-hormonal)163–165, as
Population screening is not indicated for the diagnosis well as some complementary and alternative medications
of menopause. Nonetheless, women are increasingly to treat menopausal symptoms (TABLE 4). Here, we focus
delaying childbearing until their late reproductive years on standard therapies, as the evidence regarding com-
and many wish to know when their menopause is likely plementary and alternative medications is conflicting 166.
to occur. Taken together with age, serum AMH levels
provide a good indication. Tehrani et al.154 have reported Choosing the right therapy
that a combination of age and serum AMH levels can be Oestrogen deficiency is the principal pathophysiologi-
used to predict menopause using a mathematical equa- cal mechanism that underlies menopausal symptoms
tion. In their analyses, age alone had an 84% adequacy and various oestrogen formulations are prescribed as
in predicting age at menopause, and this rose to 92% menopausal hormone therapy, which remains the most
when AMH levels were added to their model. The use of effective therapeutic option available (TABLE 5). The addi-
AMH to predict menopause still has several limitations. tion of progesterone aims to protect against the con-
For example, AMH assays are not yet standardized155 sequences of systemic therapy with oestrogen only in
and are not sufficiently sensitive to predict menopause women with intact uteri160: namely, endometrial pathol-
with great accuracy. The decline in AMH can be accel- ogies, including hyperplasia and cancer. The risk-benefit
erated by factors such as cigarette smoking 141, and some ratios of all treatment options must be considered, tak-
clinical situations — such as hypogonadotropism156 and ing into account the nature and severity of symptoms,
obesity 21 — are associated with lower than normal AMH and individual treatment-related risks (TABLE 4).
levels that do not reflect a true functional ovarian reserve In the systemic circulation, oestradiol and oes-
deficit. Thus, the precision of using AMH to estimate trone are partly bound to sex hormone-binding glob-
time to the onset of menopause in individuals is yet to ulin (SHBG), as well as to albumin, as is testosterone.
be established. Increasing or decreasing SHBG levels will affect the
One group of women that may benefit from AMH amount of unbound oestrogen and testosterone in the
screening for diminished ovarian reserve, and hence circulation167. Oral oestrogen therapy increases SHBG
imminent menopause, is carriers of the fragile X gene synthesis in the liver through the first-pass effect; by
(see above). The FMR1 mutation affects approximately contrast, standard-dose transdermal oestrogen does
one in 200 women, and approximately 20% of affected not increase SHBG synthesis168. In some women, oral
women will experience premature ovarian failure157. oestrogen therapy results in very high SHBG levels, with
At all ages, carriers of the FMR1 mutation have lower a reduction in unbound hormone. This potentially leads
AMH levels than other women158. Although it has been to loss of efficacy of the administered oestrogen and/or
suggested that monitoring of ovarian function is appro- iatrogenic testosterone deficiency.
priate for women who carry the FMR1 mutation, the Upregulation of the hepatic synthesis of pro­
exact delineation of the onset of ovarian failure remains coagulants is another known effect of oral oestrogens.
challenging. AMH values for carriers of the FMR1 muta- Transdermal oestradiol does not seem to increase the
tion have been published158, but the clinical application risk of venous thromboembolic events 169,170. Thus,
of these is yet to be established. transdermal oestrogen therapy is the preferred mode of

10 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Table 4 | Therapeutic options for management of menopausal symptoms


Therapy Formulations Comments
Systemic hormone therapy
Oestrogen • Oral, transdermal (patch, gel or spray), vaginal Approved for management of menopausal symptoms
(ring) or implant
• Administered as oestradiol or synthetic
oestrogens
Progestogen • Oral, vaginal or intrauterine Approved for management of menopausal symptoms
• Administered as progesterone or progestin
(synthetic)
Oestrogen and progestogen • Combination Approved for management of menopausal symptoms
• Pill or patch
Tissue-selective oestrogen receptor Oral Recently approved in Europe and the United States for
complex (conjugated equine management of menopausal symptoms
oestrogen plus BZA (a SERM))
Ospemifene (SERM) Oral Approved for the treatment of women experiencing
moderate to severe dyspareunia (pain during sexual
intercourse)
Tibolone Oral • Approved for management of menopausal symptoms
in some countries
• Not approved or available in Canada or the United
States
Androgen therapy (testosterone) • Oral or vaginal • Prescribed for low desire and low arousal
• Topical, oral or implant • Off-label use of this class of drugs in most countries
Non-hormonal therapies
Selective serotonin reuptake Oral Low-dose paroxetine is the first and only non-hormonal
inhibitors drug to be approved by the FDA in the United States for
menopausal symptom relief
Selective noradrenaline reuptake Oral Use for menopausal symptom control represents
inhibitors off-label use of this class of drugs
Anticonvulsants (GABAergics) Oral Use for menopausal symptom control represents
off-label use of this class of drugs
α2-adrenergic receptor agonists Oral and transdermal Off-label use for the management of menopausal
vasomotor symptoms
Sedatives and hypnotics Oral Not recommended
Complementary or alternative therapies for vasomotor symptoms
Soy or isoflavones Oral Consistent benefit over placebo effects not
demonstrated
Black cohosh Oral Consistent benefit over placebo effects not
demonstrated
Vitamin E Oral Consistent benefit over placebo effects not
demonstrated
Acupuncture NA Consistent benefit over placebo effects not
demonstrated
Local vaginal therapy
Oestrogen Low-dose oestradiol, oestriol and conjugated Approved for the treatment of vaginal atrophy
oestrogen creams, rings and pessaries
Dehydroepiandrosterone Vaginal ovule Undergoing research trials; not approved
Testosterone Vaginal cream Undergoing research trials; not approved
Non-hormonal vaginal moisturizers Vaginal gels and creams Over-the-counter products
Lubricants Vaginal gels and creams Over-the-counter products
Other
Stellate ganglion blockade NA Requires specific expertise
Cognitive behavioural therapy NA Requires specific expertise
*BZA, bazedoxifene; GABA, γ-aminobutyric acid; NA, not applicable; SERM, selective oestrogen receptor modulator.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 11

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Table 5 | Prescription therapy options for management of menopausal symptoms


Treatment Benefits Risks Patient-specific considerations
option
Hormone therapy
Systemic • Symptom relief • VTE • Oestrogen-related risk of stroke, VTE and CVD is exaggerated with
oestrogen • Fracture risk • Stroke advancing age and increasing time since onset of menopause
alone* reduction • CVD • Comorbidities such as obesity, hypertension and diabetes all increase the
• Osteoporosis • Breast cancer risk of VTE, CVD and stroke
prevention (not seen in large • Risk reduction against VTE and gallstones achieved by using a transdermal
• Improved QoL clinical trials) route and reduced dose
• Gallstones • For women with natural menopause and bothersome vasomotor
symptoms who are aged <60 years or within 10 years of their
menopause, the benefits of hormone therapy outweigh the potential for
treatment-related harm
• For those with early or premature ovarian insufficiency, benefits outweigh
harm even in the absence of symptoms
Systemic • Symptom relief • VTE • Oestrogen-related risk of stroke, VTE and CVD is exaggerated with
oestrogen and • Fracture risk • Stroke advancing age and increasing time since onset of menopause
progestogen* reduction • CVD • Comorbidities such as obesity, hypertension and diabetes all increase the
• Osteoporosis • Breast cancer risk of VTE, CVD and stroke
prevention • Gallstones • Risk reduction against VTE may be achieved by using a transdermal route
• Improved QoL and reduced oestrogen dose
• Colon cancer risk • Observational data suggest that progesterone may have less impact on
reduction breast cancer risk than MPA in menopausal women, although ideally this
should be tested in an randomized trial
• Endometrial cancer risk is associated with oestrogen dose, as well as the
type, dose and duration of progestin
• Progesterone is less effective than synthetic progestins at negating the
proliferative effects of systemic oestrogen
• For women with natural menopause and bothersome vasomotor
symptoms who are aged <60 years or within 10 years of their
menopause, the benefits of hormone therapy outweigh the potential for
treatment-related harm
• For those with early or premature ovarian insufficiency, benefits outweigh
harm even in the absence of symptoms
Low-dose Effective against Negligible Significant systemic absorption can transiently occur with excessive use of
vaginal genitourinary vaginal oestrogen creams, particularly in the setting of an atrophic vaginal
oestrogen syndrome of epithelium
menopause
Ospemifene Effective against VTE risks are deemed Approved for management of moderate to severe dyspareunia due to
(SERM)* moderate to severe comparable to those of vulvovaginal atrophy in the United States and Europe
dyspareunia due low-dose oestrogen
to vulvovaginal
atrophy
TSEC Symptom relief Similar risk profile to • Approved in the United States and Europe
(combination oestrogen in terms of risk • Indicated for management of menopausal symptoms in women with intact
of conjugated for VTE, stroke, CVD and uteri
equine gallstones • Neutral effects on the uterus and on breast tissue (SERM effect)
oestrogen and • Potential for skeletal benefit as conjugated equine oestrogens and BZA are
BZA (SERM))* effective for fracture risk reduction
Tibolone • Symptom relief • Risk of stroke in women • Neutral effects on the endometrium; no need for concomitant
(synthetic • Fracture risk aged >60 years progestogen therapy
steroid with reduction • Risk of breast cancer may • Not available or approved in North America
oestrogenic, • Osteoporosis be lower than with other
progestogenic prevention therapies
and • Improved QoL
androgenic • Colon cancer risk
activity)* reduction
Options in women for whom hormone therapy is contraindicated
Selective • Relief of vasomotor • Relatively well tolerated • Less efficacious than hormone therapy
serotonin symptoms • Some agents may • Sexual dysfunction may worsen
reuptake • Anxiolytic effects impair chemoprotective
inhibitors efficacy of tamoxifen if
used concomitantly

12 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Table 5 (Cont.) | Prescription therapy options for management of menopausal symptoms


Treatment Benefits Risks Patient-specific considerations
option
Selective Relief of vasomotor • Relatively well tolerated Less efficacious than hormone therapy
noradrenaline symptoms • Slight rise in blood
reuptake pressure may be seen;
inhibitors caution advised in
the setting of known
hypertension
Clonidine (an Relief of vasomotor Postural hypotension Less efficacious than hormone therapy
α2-adrenergic symptoms
receptor
agonist)
GABAergics Relief of vasomotor • Relatively well tolerated • Less efficacious than hormone therapy
symptoms • Somnolence • Improved sleep, particularly in the presence of restless leg syndrome
*Caution advised when considering use in an older postmenopausal woman with existing comorbidities who is remote from onset of menopause. BZA, bazedoxifene;
CVD, cardiovascular disease; GABA, γ-aminobutyric acid; MPA, medroxyprogesterone acetate; QoL, quality of life; SERM, selective oestrogen receptor modulator;
TSEC, tissue-selective oestrogen complex; VTE, venous thromboembolism.

treatment in women with an increased thrombosis risk, Hormone therapy risk reduction
such as obese women and smokers. In addition, unlike In addition to an individualized risk-benefit assessment,
oral oestrogen, transdermal oestradiol does not increase mitigating the risk of hormone therapy is important.
the risk of gallbladder disease171. Several factors influence the treatment-related risks:
In the past 2 years, two new pharmaceutical prepa- oestrogen dose163,178 (lower doses reduce the thrombo­
rations were approved in the United States and Europe embolic risks of systemic oestrogen); administration
for the treatment of menopausal symptoms. An oral route179 (thromboembolic risk is suggested to be lower
selective oestrogen receptor modulator (SERM), for the use of transdermal oestrogens than oral oes-
ospemifene, has been approved for the treatment of trogens) and theoretically, systemic exposure can be
moderate to severe pain during intercourse associated diminished by intrauterine treatment with levonorg-
with vulvo­vaginal atrophy 172,173, and a tissue-specific estrel compared with oral formulations; progestogen
SERM–oestrogen complex (a combination of oral con- type179,180 (risk for postmenopausal breast cancer, as
jugated equine oestrogen and bazedoxifene (a SERM)) well as for thrombosis, may be lessened by the use of
has been approved for the management of moderate to pro­gesterone compared with synthetic progestins);
severe vasomotor symptoms in women with an intact and treatment schedule (infrequent progesterone dos-
uterus174. Tissue selectivity is achieved through the con- ing seems to reduce the risk of metabolic perturbations
current use of oestrogen and a SERM, which replaces and breast cancer compared with continuous combined
a progestogen and selectively blocks the undesirable oestrogen–progesterone regimens)6,180–183. Infrequent
actions of oestrogen. In the case of conjugated equine progestogen dosing schedules are increasingly used,
oestrogen–bazedoxifene, the proliferative effects of partly owing to patient preference, as they reduce the
oestrogen are blocked in the uterus and possibly also frequency of therapy-related bleeding episodes and, to
the breast, whereas the bone-sparing actions of oes- some extent, as a strategy to minimize breast exposure to
trogen are preserved. The role of testosterone for the progestogens. However, data on the endometrial safety
treatment of postmenopausal desire or arousal dis­ of long-cycle progestogen regimens are not only sparse,
orders and the long-term implications of such a therapy but even suggest a higher likelihood for endometrial
in postmenopausal women are unclear. This strategy hyperplasia and even cancer 184.
may benefit a certain subset of women, such as those Initiation of hormone therapy is usually contraindi-
with surgically induced menopause, who have persis- cated in women with a personal history of breast cancer
tent sexual symptoms despite optimization of meno- or venous thromboembolism, or those with a high risk
pausal hormone therapies175,176. Urogenital symptoms for breast cancer, thrombosis or stroke. Transdermal
are effectively treated with either local (vaginal) or sys- oestrogen therapy may be considered and preferred
temic oestrogen therapy 177. Oestrogen therapy restores when highly symptomatic women with type 2 diabetes
normal vaginal flora, lowers the pH, and thickens mellitus or obesity, or those at high risk of cardiovascu-
and revascularizes the vaginal lining. The number of lar disease, do not respond to non-hormonal therapies.
superficial epithelial cells is increased and symptoms In general, commencement of hormone therapy is not
of atrophy are alleviated. Importantly, low-dose vaginal recommended for women who are aged >60 years.
oestrogen improves vaginal atrophy without causing Recommendations regarding the duration of sys-
proliferation of the endometrium177. Given the docu- temic hormone therapy need to be individualized and
mented efficacy and proven safety, vaginal oestrogen depend on the end points of treatment. For women with
is the first-line approach to treat symptoms of vaginal premature ovarian failure or primary ovarian insuffi-
atrophy in the majority of women. ciency, or those with early menopause (before the age of

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 13

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

45 years), therapy can be continued until the average age may lessen vasomotor symptoms, as well as reduce the
of the natural menopause (early 50s), at which time the risks of cardiovascular disease; diabetes; urinary incon-
need for hormone therapy should be reassessed. There tinence; breast, pancreatic and endometrial cancer; and
are no arbitrary limits regarding the duration of meno- dementia78. Increasing physical activity has been recom-
pausal hormone therapy; it can be used for as long as mended to alleviate vasomotor symptoms; however, this
the woman feels the benefits outweigh the risks for her, may exacerbate symptoms in women with a low level of
but treatment should be re-evaluated frequently. When fitness193. Furthermore, a recent prospective study has
hormone therapy is initiated for vasomotor symptoms, shown no benefit of exercise in reducing these symp-
clinicians should strive to control symptoms with the toms194. Regular physical activity is to be encouraged to
lowest possible hormone dose, and continued therapy maintain muscle mass and balance, thereby reducing
for >5 years must be individualized on the basis of the risk of fall and fracture. A fairly high and consist-
symptom severity and the risk for breast cancer and ent degree of impact exercise seems to be required to
vascular events, and also with due regard to the patient’s improve bone density 195,196. It is unlikely that physical
preference and choice. activity alone will improve bone mineral density in
women who are underweight197. Yoga has been shown to
Non-hormonal therapies improve sleep, but not to reduce vasomotor symptoms198.
A number of non-hormonal therapies are efficacious
against menopausal vasomotor symptoms (TABLE 4) and Menopause management in perspective
should be considered for women who do not wish to Hormone therapy remains the most efficacious of the
take oestrogen or those with contraindications. available strategies for managing menopausal symp-
For vasomotor symptoms, many drugs have dem- toms. Safety and efficacy of low-dose topical oestrogen
onstrated efficacy in several studies: paroxetine, fluox- against local urogenital symptoms69,177 is well established
etine and citalopram (which are selective serotonin and now, oral ospemifene, if available, offers added flex-
reuptake inhibitors); venlafaxine and desvenlafaxine ibility for women experiencing isolated urogenital symp-
(selective noradrenaline reuptake inhibitors); clonidine toms199. In the majority of young and otherwise healthy
(α2-adrenergic receptor agonist); and anticonvulsants early menopausal women who are within 10 years of
(gabapentin and pregabalin)2,3. Paroxetine and fluoxe- onset of menopause, the benefits of systemic hormone
tine are potent cytochrome P450 2D6 (CYP2D6) inhibi- therapy will probably outweigh any potential for harm.
tors, and as they decrease the metabolism of tamoxifen By contrast, in older women or women with comor-
(a SERM used in the treatment of breast cancer) — which bidities that increase the risk for cardiovascular disease
may reduce its anticancer effects — these drugs should and stroke (such as hypertension, obesity, longstand-
be avoided in tamoxifen users185. However, consistency of ing smoking history, and/or a strong family history
treatment response and efficacy of the various alternative of stroke and premature atherosclerosis) the potential
options remain questionable186–189. Stellate ganglion block harm of systemic hormone therapy outweighs the ben-
achieved by injecting an anaesthetic such as bupivacaine efits. For these women, non-hormonal approaches are
under fluoroscopic guidance around the stellate ganglion the preferred first-line strategy to control menopausal
— which is the cervicothoracic ganglion of the sympa- symptoms. Treatment should always be individualized.
thetic system — has been shown to improve vasomotor Some women with severe symptoms that are refrac-
symptoms in a small randomized controlled trial190. tory to non-hormonal therapy may accept a substantial
Alternatives to vaginal therapies, such as vaginal degree of risk of hormone therapy in order to have an
lubricants and moisturizers, are less effective than vagi- acceptable QoL. To simplify the risk-benefit assessment
nal oestrogen191. Although vaginal moisturizers lower for the various treatment options, the North American
the vaginal pH and may normalize vaginal cell compo- Menopause Society has recently endorsed an easy-to-
sition, sustained symptom relief has not been seen in all follow algorithm182 (available as a mobile phone app).
studies191. Vaginal moisturizers do not reduce urinary It helps women’s healthcare providers from across the
tract symptoms or asymptomatic bacteriuria191. Vaginal globe to identify the optimal treatment strategy (hor-
lubricants offer transient relief from vaginal atrophy- monal or non-hormonal) on the basis of each patient’s
related discomfort when used during intercourse, but individualized risk profile.
they do not treat the underlying problem.
Quality of life
Physical activity, diet and lifestyle QoL has been shown to change during menopause,
All women at midlife should be encouraged to maintain often in relation to the presence or absence of symp-
or achieve a normal body weight, be physically active, toms200. The primary objective of the care of sympto-
adopt a healthy diet, limit alcohol consumption and not matic menopausal women is enhancement of QoL. Like
smoke. Anecdotally, some women find that avoidance pain, an individual’s perception of QoL is not easy to
of spicy food, hot drinks and alcohol lessens their vaso- determine and is purely subjective. There is no univer-
motor symptoms. Obesity is associated with a greater sal agreement on a QoL definition or how it should be
likelihood of vasomotor symptoms, although women measured, although it is becoming increasingly valued
who are overweight (BMI from 25 to <30 kg m–2), as as a therapeutic outcome (BOX 1). General QoL reflects
opposed to obese (BMI ≥30 kg m–2), are more likely to the individual’s beliefs about functioning and achieve-
have severe symptoms192. For obese women, weight loss ments in various aspects of life, and an overall sense

14 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

Box 1 | Assessing quality of life and menopausal symptoms


such as herbal preparations and cognitive behavioural
therapy 205–209. However, when HRQoL is assessed,
Several instruments can be used to measure quality of life (QoL) during menopause; there was no such improvement in the Women’s Health
the domains covered by each instrument are shown in brackets. Initiative study and the Heart and Oestrogen–Progestin-
Generic instruments Replacement Study, neither of which recruited women
• Short-form 36 health survey (SF‑36; physical and social functioning; bodily pain; with severe symptoms, as this would have led to unblind-
general and mental health; vitality; emotional and physical role) ing of the trials210–212. An improvement was observed in
• EuroQoL (mobility; self-care; usual activities; pain and discomfort; anxiety or the 2000 Finnish health survey, in which women had
depression) been prescribed hormone therapy for symptom relief 213.
Menopause-specific instruments This is perhaps not surprising as the purpose of the for-
• Greene climacteric scale (psychological, somatic and vasomotor symptoms) mer studies was to assess chronic disease prevention and
• Women’s health questionnaire (WHQ; depressed mood; somatic, vasomotor and symptom relief was of little importance.
menstrual symptoms; anxiety; sexual behaviour; sleep problems; memory and
concentration; attractiveness) Outlook
• Menopause-specific QoL questionnaire (MENQOL; vasomotor, psychosocial, physical Overall, our understanding from epidemiological stud-
and sexual domains) ies of the effects of menopausal transition on women
• Menopause rating scale (MRS; psychological, somato-vegetative and urogenital and the factors that influence its duration and timing
symptoms) remains unclear and incomplete. Basic questions, such
• Menopause QoL scale (MQOL; physical, vasomotor, psychosocial and sexual domains) as global statistics on the age of menopause and the
• Utian menopause QoL scale (QQOL; occupational, health-related, emotional and variation in the length of the perimenopause, remain
sexual QoL) unanswered. One way forward lies in recent steps that
have been taken to combine individual-level data from
Symptom-specific instruments
numerous international studies of women’s health to
• Kupperman index (menopausal symptoms)
provide a more comprehensive and detailed picture of
• Hot flush-related daily interference scale (vasomotor symptoms) the menopausal transition, its timing and long-term
• Sexual activity log (sexual activity) health implications, not just for women in developed
nations but also for those from developing regions and
from diverse populations.
of satisfaction and wellbeing. When QoL is related to With the increasing lifespan of women in less devel-
health and illness, it is usually known as health-related oped countries, the impact of the menopausal transition
QoL (HRQoL). HRQoL is the patient’s evaluation of the in these countries needs to be better understood22. Data
impact of a health condition and its treatment on life about the persistence of menopausal symptoms beyond
aspects that are most likely to be affected by a change in the age of 65 years and how such symptoms might
health status. This covers physical health and function, adversely affect older women are also sparse214. Studies
emotional function, role limitations and social func- investigating menopause in developing countries and in
tioning. All of these domains are relevant to a woman’s older women are sorely needed.
QoL and measuring their status goes beyond the mere We are getting closer to accurately forecasting the
counting of events. timing of menopause. This is important in the context
Assessment instruments are generic or disease-­ of women delaying childbearing and wanting to have
specific201; the former covering different aspects of daily better control of their fertility. Research to refine the
living. They can also be used in the evaluation of cost use of AMH as a predictor of menopause and studies
effectiveness in health economic analyses202. However, involving the isolation of oogonial stem cells to prevent
assessment of QoL needs to include not only the impact menopause are ongoing. However, we still lack a com-
of symptoms, but also personal and environmen- plete understanding of the basic mechanisms that are
tal factors. This has led to the development of several responsible for vasomotor symptoms, which in turn
menopause-­specific QoL instruments, which can be limits the development of novel non-hormonal treat-
used to assess the benefits of treatment 201. Often more ments for hot flushes and night sweats. More research
than one instrument must be used to cover different into the central thermal control mechanisms influ-
aspects of menopause and treatment in order to assess enced by oestrogen is needed, and our understanding
general QoL and specific symptoms. Also, it is difficult of the effects of oestrogen deficiency on this remains
to incorporate the impact of adverse effects of treatment in its infancy. We also need to better understand the
into the assessment of QoL as the instruments are not mechanisms by which oestrogen deficiency results in
designed to do this. sleep disturbance, as this has extensive adverse effects
The impact of menopausal hormone therapy on QoL on women that persist for years. For other symptoms
depends on whether women are symptomatic or not, as of oestrogen deficiency, the underlying mechanisms
might be expected203. Menopause-specific question- remain elusive, including arthralgia and mood changes,
naires show improvements in vasomotor symptoms, particularly anxiety. A greater understanding of the bio-
sexual functioning and sleep for hormone treatment chemical pathways by which oestrogen deficiency con-
compared with placebo, and this may vary with the tributes to central fat accumulation could lead to novel
type of therapy used204. This is also true of oestrogen– targeted approaches for preventing central weight gain
bazedoxifene, tibolone and alternative treatments, in women, and possibly in men.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 15

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

The pendulum has swung from the overt enthusiasm of There needs to be greater recognition that meno-
pharmaceutical companies to invest in menopausal thera- pause affects all women, that many of the symptoms
pies in the 1990s to the removal of several highly effective of oestrogen deficiency are not transient but persist for
hormone therapies from the market (such as intranasal decades, and that oestrogen loss adversely influences
oestradiol and transdermal testosterone patches) and bone, metabolic and cardiovascular health. Hence,
minimization of women’s health divisions in large compa- investment in the management of menopause trans-
nies that were previously world leaders in the provision of lates into investing in the health of what will equate to
midlife women’s health products. This damage has been a half of the life of young women today. Funding of basic
consequence of the fear of breast cancer, venous thrombosis research is needed to identify novel interventions that
and stroke, which was engendered by the very first publica- effectively alleviate the symptoms of oestrogen defi-
tions of the Women’s Health Initiative215. Thus, menopause, ciency — notably, vasomotor symptoms, sleep distur-
a condition that affects every woman, is now underfunded bance, mood changes and urogenital symptoms — and
and under researched. Furthermore, vast numbers of abrogate the adverse effects of oestrogen deficiency on
highly symptomatic women are not receiving therapies that body composition, bone density and the cardiovascular
are proven to be effective and many are resorting to alterna- system, as well as translational research to evaluate the
tives that are mostly ineffective and unregulated18,97,163,187,192. safety of such new therapies.

1. Soules, M. R. et al. Executive summary: Stages of a comprehensive review. Health Qual. Life Outcomes 36. Mishra, G., Hardy, R. & Kuh, D. Are the effects of risk
Reproductive Aging Workshop (STRAW). Fertil. Steril. 3, 47 (2005). factors for timing of menopause modified by age?
76, 874–878 (2001). 19. Thurston, R. C. & Joffe, H. Vasomotor symptoms and Results from a British birth cohort study. Menopause
2. Harlow, S. D. et al. Executive summary of the Stages menopause: findings from the Study of Women’s 14, 717–724 (2007).
of Reproductive Aging Workshop + 10: addressing Health across the Nation. Obstetr. Gynecol. Clin. 37. Voorhuis, M., Onland-Moret, N. C.,
the unfinished agenda of staging reproductive aging. North Amer. 38, 489–501 (2011). van der Schouw, Y. T., Fauser, B. C. & Broekmans, F. J.
Menopause 19, 387–395 (2012). 20. Gold, E. B. et al. Longitudinal analysis of the Human studies on genetics of the age at natural
3. Harlow, S. et al. Executive summary of the Stages of association between vasomotor symptoms and race/ menopause: a systematic review. Hum. Reprod.
Reproductive Aging Workshop + 10: addressing the ethnicity across the menopausal transition: study of Update 16, 364–377 (2010).
unfinished agenda of staging reproductive aging. women’s health across the nation. Am. J. Publ. Health 38. van Asselt, K. M. et al. Linkage analysis of extremely
Fertil. Steril. 97, 843–851 (2012). 96, 1226–1235 (2006). discordant and concordant sibling pairs identifies
4. Harlow, S. D. et al. Executive summary of the Stages 21. Freeman, E. W. et al. Association of anti-mullerian quantitative trait loci influencing variation in human
of Reproductive Aging Workshop +10: addressing hormone levels with obesity in late reproductive-age menopausal age. Am. J. Hum. Genet. 74, 444–453
the unfinished agenda of staging reproductive aging. women. Fertil. Steril. 87, 101–106 (2007). (2004).
Climacteric 15, 105–114 (2012). 22. Islam, M. R., Gartoulla, P., Bell, R. J., Fradkin, P. & 39. Murabito, J. M., Yang, Q., Fox, C. S. & Cupples, L. A.
5. Harlow, S. D. et al. Executive summary of the Stages Davis, S. R. Prevalence of menopausal symptoms in Genome-wide linkage analysis to age at natural
of Reproductive Aging Workshop + 10: addressing Asian midlife women: a systematic review. Climacteric menopause in a community-based sample: the
the unfinished agenda of staging reproductive aging. 1–42 (2014). Framingham Heart Study. Fertil. Steril. 84,
J. Clin. Endocrinol. Metabolism 97, 1159–1168 23. Mishra, G. D. & Kuh, D. Health symptoms during 1674–1679 (2005).
(2012). midlife in relation to menopausal transition: British 40. Lin, W. T. et al. Comparison of age at natural
6. Jane, F. M. & Davis, S. R. A practitioner’s toolkit for prospective cohort study. BMJ 344, e402 (2012). menopause in BRCA1/2 mutation carriers with a non-
managing the menopause. Climacteric 17, 1–16 This paper demonstrates the various trajectories of clinic based sample of women in northern California.
(2014). symptoms associated with menopause. Cancer 119, 1652–1659 (2013).
7. Tom, S. E. & Mishra, G. D. Current Topics in 24. Colditz, G. A. et al. Menopause and the risk of 41. Watkins, W. J. et al. An investigation into FOXE1
Menopause: a Life Course Approach to Reproductive coronary heart disease in women. N. Engl. J. Med. polyalanine tract length in premature ovarian failure.
Aging (Bentham Science Publishers Ltd., 2013). 316, 1105–1110 (1987). Mol. Hum. Reprod. 12, 145–149 (2006).
8. Schoenaker, D. A., Jackson, C. A., Rowlands, J. V. & 25. Anasti, J. N., Kalantaridou, S. N., Kimzey, L. M., 42. Chand, A. L., Ponnampalam, A. P., Harris, S. E.,
Mishra, G. D. Socioeconomic position, lifestyle factors Defensor, R. A. & Nelson, L. M. Bone loss in young Winship, I. M. & Shelling, A. N. Mutational analysis of
and age at natural menopause: a systematic review women with karyotypically normal spontaneous BMP15 and GDF9 as candidate genes for premature
and meta-analyses of studies across six continents. premature ovarian failure. Obstet. Gynecol. 91, ovarian failure. Fertil. Steril. 86, 1009–1012 (2006).
Int. J. Epidemiol. 43, 1542–1562 (2014). 12–15 (1998). 43. Ferrarini, E. et al. Clinical characteristics and genetic
This paper provides a global perspective of factors 26. Kalantaridou, S. N., Davis, S. R. & Nelson, L. M. analysis in women with premature ovarian
associated with age at menopause. Premature ovarian failure. Endocrinol. Metabolism insufficiency. Maturitas 74, 61–67 (2013).
9. Dratva, J. et al. Is age at menopause increasing Clin. North Amer. 27, 989–1006 (1998). This is a comprehensive summary of primary
across Europe? Results on age at menopause and 27. Kok, H. S. et al. Heart disease risk determines ovarian insufficiency.
determinants from two population-based studies. menopausal age rather than the reverse. J. Am. 44. Cramer, D. W., Harlow, B. L., Barbieri, R. L. &
Menopause 16, 385–394 (2009). College Cardiol. 47, 1976–1983 (2006). Ng, W. G. Galactose‑1‑phosphate uridyl transferase
10. Flint, M. P. Secular trends in menopause age. 28. Ossewaarde, M. E. et al. Age at menopause, cause- activity associated with age at menopause and
J. Psychosomat. Obstetr. Gynaecol. 18, 65–72 specific mortality and total life expectancy. reproductive history. Fertil. Steril. 51, 609–615
(1997). Epidemiology 16, 556–562 (2005). (1989).
11. Flint, M. Is there a secular trend in age of 29. Mondul, A. M., Rodriguez, C., Jacobs, E. J. & 45. Kaufman, F. R., Devgan, S. & Donnell, G. N. Results of
menopause? Maturitas 1, 133–139 (1978). Calle, E. E. Age at natural menopause and cause- a survey of carrier women for the galactosemia gene.
12. Nichols, H. B. et al. From menarche to menopause: specific mortality. Am. J. Epidemiol. 162, 1089–1097 Fertil. Steril. 60, 727–728 (1993).
trends among US Women born from 1912 to 1969. (2005). 46. Nelson, L. M. Clinical practice. Primary ovarian
Am. J. Epidemiol. 164, 1003–1011 (2006). 30. Kok, H. S., van, A. K. M., van, d. S. Y. T., Peeters, P. H. insufficiency. N. Engl. J. Med. 360, 606–614 (2009).
13. Rodstrom, K. et al. Evidence for a secular trend in & Wijmenga, C. Genetic studies to identify genes 47. He, C. et al. Genome-wide association studies identify
menopausal age: a population study of women in underlying menopausal age. Hum. Reprod. Update 11, loci associated with age at menarche and age at
Gothenburg. Menopause 10, 538–543 (2003). 483–493 (2005). natural menopause. Nat. Genet. 41, 724–728
14. McKinlay, S. M., Brambilla, D. J. & Posner, J. G. The 31. van Asselt, K. M. et al. Heritability of menopausal (2009).
normal menopause transition. Maturitas 61, 4–16 age in mothers and daughters. Fertil. Steril. 82, 48. Stolk, L. et al. Loci at chromosomes 13, 19 and 20
(2008). 1348–1351 (2004). influence age at natural menopause. Nat. Genet. 41,
15. Mishra, G. D. & Dobson, A. J. Using longitudinal 32. Murabito, J. M., Yang, O., Fox, C., Wilson, P. W. & 645–647 (2009).
profiles to characterize women’s symptoms through Cupples, L. A. Heritability of age at natural 49. Elks, C. E. et al. Thirty new loci for age at menarche
midlife: results from a large prospective study. menopause in the Framingham Heart Study. J. Clin. identified by a meta-analysis of genome-wide
Menopause 19, 549–555 (2012). Endocrinol. Metab. 90, 3427–3430 (2005). association studies. Nat. Genet. 42, 1077–1085
16. Gartoulla, P., Islam, M. R., Bell, R. J. & Davis, S. R. 33. Torgerson, D. J., Thomas, R. E. & Reid, D. M. Mothers (2010).
Prevalence of menopausal symptoms in Australian and daughters menopausal ages: is there a link? Eur. 50. Stolk, L. et al. Meta-analyses identify 13 loci
women at midlife: a systematic review. Climacteric J. Obstet. Gynecol. Reprod. Biol. 74, 63–66 (1997). associated with age at menopause and highlight DNA
17, 529–539 (2014). 34. Cramer, D. W., Xu, H. & Harlow, B. L. Family history as repair and immune pathways. Nat. Genet. 44,
17. Mishra, G. & Kuh, D. Perceived change in quality of a predictor of early menopause. Fertil. Steril. 64, 260–268 (2012).
life during the menopause. Social Sci. Med. 62, 740–745 (1995). 51. Nejat, E. J. & Chervenak, J. L. The continuum of
93–102 (2006). 35. de, B. J. P. et al. The role of genetic factors in age at ovarian aging and clinicopathologies associated with
18. Utian, W. H. Psychosocial and socioeconomic burden natural menopause. Hum. Reprod. 16, 2014–2018 the menopausal transition. Maturitas 66, 187–190
of vasomotor symptoms in menopause: (2001). (2010).

16 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

52. Broekmans, F. J., Soules, M. R. & Fauser, B. C. Ovarian or early menopause: long-term health consequences. 102. Hardy, R., Mishra, G. & Kuh, D. in Menopause,
aging: mechanisms and clinical consequences. Endocr. Maturitas 65, 161–166 (2010). Postmenopause and Aging. (eds Purdie, D. W. et al.)
Rev. 30, 465–493 (2009). 78. Davis, S. R. et al. Understanding weight gain at 11–19 (Royal Society of Medicine Press Ltd London,
53. Burger, H. G., Hale, G. E., Robertson, D. M. & menopause. Climacteric 15, 419–429 (2012). 2005).
Dennerstein, L. A review of hormonal changes during 79. Tchernof, A. & Despres, J. P. Pathophysiology of 103. Do, K. A. et al. Predictive factors of age at
the menopausal transition: focus on findings from the human visceral obesity: an update. Physiol. Rev. 93, menopause in a large Australian twin study. Hum.
Melbourne Women’s Midlife Health Project. Hum. 359–404 (2013). Biol. 70, 1073–1091 (1998).
Reprod. Update 13, 559–565 (2007). 80. Barton, M. Cholesterol and atherosclerosis: 104. Shinberg, D. S. An event history analysis of age at last
54. Wise, P. M. Neuroendocrine modulation of the modulation by oestrogen. Curr. Opin. Lipidol 24, menstrual period: correlates of natural and surgical
“menopause”: insights into the aging brain. Am. 214–220 (2013). menopause among midlife Wisconsin women. Social
J. Physiol. 277, E965–E970 (1999). 81. Chedraui, P. et al. Angiogenesis, inflammation and Sci. Med. 46, 1381–1396 (1998).
55. Downs, J. L. & Wise, P. M. The role of the brain in endothelial function in postmenopausal women 105. Torgerson, D. J., Avenell, A., Russell, I. T. & Reid, D. M.
female reproductive aging. Mol. Cell Endocrinol. 299, screened for the metabolic syndrome. Maturitas 77, Factors associated with onset of menopause in women
32–38 (2009). 370–374 (2014). aged 45–49. Maturitas 19, 83–92 (1994).
56. Butler, L. & Santoro, N. The reproductive endocrinology 82. Chedraui, P. et al. Circulating leptin, resistin, 106. Wise, L. A., Krieger, N., Zierler, S. & Harlow, B. L.
of the menopausal transition. Steroids 76, 627–635 adiponectin, visfatin, adipsin and ghrelin levels and Lifetime socioeconomic position in relation to onset of
(2011). insulin resistance in postmenopausal women with and perimenopause. J. Epidemiol. Commun. Health 56,
57. Santoro, N. et al. Impaired folliculogenesis and without the metabolic syndrome. Maturitas 79, 851–860 (2002).
ovulation in older reproductive aged women. J. Clin. 86–90 (2014). 107. Castelo-Branco, C. et al. Age at menopause in Latin
Endocrinol. Metab. 88, 5502–5509 (2003). 83. Hage, F. G. & Oparil, S. Ovarian hormones and America. Menopause 13, 706–712 (2006).
58. Archer, D. F. et al. Menopausal hot flushes and night vascular disease. Curr. Opin. Cardiol 28, 411–416 108. Bromberger, J. T. et al. Prospective study of the
sweats: where are we now? Climacteric 14, 515–528 (2013). determinants of age at menopause. Am. J. Epidemiol.
(2011). This paper describes the mechanisms of 145, 124–133 (1997).
59. Freeman, E. W., Sammel, M. D., Lin, H., Liu, Z. vasoprotection by endogenous oestrogens and the 109. Hardy, R., Kuh, D. & Wadsworth, M. Smoking, body
& Gracia, C. R. Duration of menopausal hot flushes development of cardiovascular disease in mass index, socioeconomic status and the menopausal
and associated risk factors. Obstet. Gynecol. 117, menopause. transition in a British national cohort. Int. J. Epidemiol.
1095–1104 (2011). 84. Abramson, B. L. & Melvin, R. G. Cardiovascular risk in 29, 845–851 (2000).
60. Gjelsvik, B., Rosvold, E. O., Straand, J., Dalen, I. & women: focus on hypertension. Can. J. Cardiol 30, 110. van Asselt, K. M. et al. Current smoking at menopause
Hunskaar, S. Symptom prevalence during menopause 553–559 (2014). rather than duration determines the onset of natural
and factors associated with symptoms and menopausal 85. El Khoudary, S. R. et al. Progression rates of carotid menopause. Epidemiology 15, 634–639 (2004).
age. Results from the Norwegian Hordaland Women’s intima-media thickness and adventitial diameter 111. Discigil, G., Gemalmaz, A., Tekin, N. & Basak, O.
Cohort study. Maturitas 70, 383–390 (2011). during the menopausal transition. Menopause 20, Profile of menopausal women in west Anatolian rural
61. Richard-Davis, G. & Wellons, M. Racial and ethnic 8–14 (2013). region sample. Maturitas 55, 247–254 (2006).
differences in the physiology and clinical symptoms of 86. Hildreth, K. L., Kohrt, W. M. & Moreau, K. L. 112. Hardy, R. & Kuh, D. Reproductive characteristics and
menopause. Semin. Reprod. Med. 31, 380–386 Oxidative stress contributes to large elastic arterial the age at inception of the perimenopause in a British
(2013). stiffening across the stages of the menopausal National Cohort. Am. J. Epidemiol. 149, 612–620
62. Duffy, O. K., Iversen, L., Aucott, L. & Hannaford, P. C. transition. Menopause 21, 624–632 (2014). (1999).
Factors associated with resilience or vulnerability to hot 87. Moreau, K. L., Hildreth, K. L., Meditz, A. L., 113. Gold, E. B. et al. Factors related to age at natural
flushes and night sweats during the menopausal Deane, K. D. & Kohrt, W. M. Endothelial function is menopause: longitudinal analyses. Am. J. Epidemiol.
transition. Menopause 20, 383–392 (2013). impaired across the stages of the menopause 178, 70–83 (2013).
63. Zervas, I. M. et al. Additive effect of depressed mood transition in healthy women. J. Clin. Endocrinol. 114. McKinlay, S. M., Bifano, N. L. & McKinlay, J. B.
and vasomotor symptoms on postmenopausal Metab. 97, 4692–4700 (2012). Smoking and age at menopause in women. Ann.
insomnia. Menopause 16, 837–842 (2009). 88. Stamatelopoulos, K. S. et al. Recently Intern. Med. 103, 350–356 (1985).
64. Al‑Safi, Z. A. & Santoro, N. Menopausal hormone postmenopausal women have the same prevalence of 115. Mikkelesen, T. F., Graff-Iverseon, S., Sundby, J. &
therapy and menopausal symptoms. Fertil. Steril. 101, subclinical carotid atherosclerosis as age and Bjertness, E. Early menopause, association with
905–915 (2014). traditional risk factor matched men. Atherosclerosis tobacco smoking, coffee consumption and other
65. Vivian-Taylor, J. & Hickey, M. Menopause and 221, 508–513 (2012). lifestyle factors: a cross-sectional study. BMC Public
depression: is there a link? Maturitas 79, 142–146 89. Lobo, R. A. et al. Prevention of diseases after Health 7, 149–167 (2007).
(2014). menopause. Climacteric 17, 540–556 (2014). 116. Mark-Kappeler, C. J., Hoyer, P. B. & Devine, P. J.
66. Bay-Jensen, A. C. et al. Role of hormones in cartilage 90. Lisabeth, L. & Bushnell, C. Stroke risk in women: the Xenobiotic effects on ovarian preantral follicles. Biol.
and joint metabolism: understanding an unhealthy role of menopause and hormone therapy. Lancet Reprod. 85, 871–883 (2011).
metabolic phenotype in osteoarthritis. Menopause 20, Neurol. 11, 82–91 (2012). 117. Plante, B. J., Cooper, G. S., Baird, D. D. &
578–586 (2013). 91. Rocca, W. A., Grossardt, B. R., Miller, V. M., Steiner, A. Z. The impact of smoking on antimullerian
67. Blumel, J. E. et al. Menopause could be involved in the Shuster, L. T. & Brown, R. D. Jr. Premature hormone levels in women aged 38 to 50 years.
pathogenesis of muscle and joint aches in mid-aged menopause or early menopause and risk of ischemic Maturitas 17, 571–576 (2010).
women. Maturitas 75, 94–100 (2013). stroke. Menopause 19, 272–277 (2012). 118. Luoto, R., Laprio, J. & Uutela, A. Age at natural
68. Nappi, R. E. & Kokot-Kierepa, M. Vaginal Health: 92. De Vos, M., Devroey, P. & Fauser, B. C. Primary menopause and sociodemographic status in Finland.
Insights, Views and Attitudes (VIVA) - results from an ovarian insufficiency. Lancet 376, 911–921 (2010). Am. J. Epidemiol. 139, 64–76 (1994).
international survey. Climacteric 15, 36–44 (2012). 93. Wellons, M., Ouyang, P., Schreiner, P. J., 119. Lawlor, D. A., Ebrahim, S. & Smith, G. D. The
69. Portman, D. J. & Gass, M. L. Genitourinary syndrome of Herrington, D. M. & Vaidya, D. Early menopause association of socio-economic position across the life
menopause: new terminology for vulvovaginal atrophy predicts future coronary heart disease and stroke: the course and age at menopause: teh British Women’s
from the International Society for the Study of Women’s Multi-Ethnic Study of Atherosclerosis. Menopause 19, Heart and Health Study. BJOG 110, 1078–1087
Sexual Health and The North American Menopause 1081–1087 (2012). (2003).
Society. Menopause 21, 1063–1068 (2014). 94. Luine, V. N. Estradiol and cognitive function: past, 120. Schoenbaum, E. E. et al. HIV infection, drug use, and
70. Nappi, R. E. & Palacios, S. Impact of vulvovaginal present and future. Horm. Behav. 66, 602–618 onset of natural menopause. Clin. Infect. Dis. 41,
atrophy on sexual health and quality of life at (2014). 1517–1524 (2005).
postmenopause. Climacteric 17, 3–9 (2014). 95. Silva, I. & Naftolin, F. Brain health and cognitive and 121. van Noord, P. A., Dubas, J. S., Dorland, M.,
71. Avis, N. E. et al. Longitudinal changes in sexual mood disorders in ageing women. Best Pract. Res. Boersma, H. & te Velde, E. Age at natural menopause
functioning as women transition through menopause: Clin. Obstet. Gynaecol. 27, 661–672 (2013). in a population-based screening cohort: the role of
results from the Study of Women’s Health Across the 96. Weber, M. T., Maki, P. M. & McDermott, M. P. menarche, fecundity, and lifestyle factors. Fertil. Steril.
Nation. Menopause 16, 442–452 (2009). Cognition and mood in perimenopause: a systematic 68, 95–102 (1997).
72. Boyle, W. J., Simonet, W. S. & Lacey, D. L. Osteoclast review and meta-analysis. J. Steroid Biochem. Mol. 122. Akahoshi, M. et al. The effects of body mass index on
differentiation and activation. Nature 423, 337–342 Biol. 142, 90–98 (2014). age at menopause. International journal of obesity
(2003). 97. de Villiers, T. J. et al. Updated 2013 International and related metabolic disorders. Int. J. Obes. Relat.
73. Tella, S. H. & Gallagher, J. C. Prevention and treatment Menopause Society recommendations on menopausal Metab. Disord. 26, 961–968 (2002).
of postmenopausal osteoporosis. J. Steroid Biochem. hormone therapy and preventive strategies for midlife 123. Sammel, M. D., Freeman, E. W., Liu, Z., Lin, H. &
Mol. Biol. 142, 155–170 (2014). health. Climacteric 16, 316–337 (2013). Guo, W. Factors that influence entry into stages of the
74. Khosla, S. Pathogenesis of age-related bone loss in 98. Henderson, V. W. Action of estrogens in the aging menopausal transition. Menopause 16, 1218–1227
humans. J. Gerontol. A Biol. Sci. Med. Sci. 68, brain: dementia and cognitive aging. Biochim. (2009).
1226–1235 (2013). Biophys. Acta 1800, 1077–1083 (2010). 124. Aydin, Z. D. Determinants of age at natural
This paper illustrates the mechanisms of 99. Hardy, R. & Kuh, D. Does early growth influence menopause in the Isparta Menopause and Health
postmenopausal bone loss. timing of the menopause? Evidence from a British Study: premenopausal body mass index gain rate and
75. Clarke, B. L. & Khosla, S. Physiology of bone loss. birth cohort. Hum. Reprod. 17, 2474–2479 (2002). episodic weight loss. Menopause 17, 494–505
Radiol Clin. North Am. 48, 483–495 (2010). 100. Elias, S. G., van Noord, P. A., Peeters, P. H., (2010).
76. Sapir-Koren, R. & Livshits, G. Is interaction between den Tonkelaar, T. I. & Grobbee, D. E. Childhood 125. Sowers, M. R. et al. Genetic polymorphisms and
age-dependent decline in mechanical stimulation and exposure to the 1944–1945 Dutch famine and obesity influence estradiol decline during the
osteocyte-estrogen receptor levels the culprit for subsequent female reproductive function. Hum. menopause. Clin. Endocrinol. (Oxf.) 74, 618–623
postmenopausal-impaired bone formation? Reprod. 20, 2483–2488 (2005). (2011).
Osteoporos. Int. 24, 1771–1789 (2013). 101. Mishra, G. D., Cooper, R., Tom, S. E. & Kuh, D. Early 126. Cohen, L. S., Soares, C. N., Vitonis, A. F., Otto, M. W.
77. Shuster, L. T., Rhodes, D. J., Gostout, B. S., life circumstances and their impact on menarche and & Harlow, B. L. Risk for new onset of depression
Grossardt, B. R. & Rocca, W. A. Premature menopause menopause. Women’s Health 5, 175–190 (2009). during the menopausal transition: the Harvard

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 17

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

study of moods and cycles. Arch. Gen. Psychiatry 63, 146. Hawker, G. et al. A clinical decision rule to enhance 169. Olie, V. et al. Hormone therapy and recurrence of
385–390 (2006). targeted bone mineral density testing in healthy mid- venous thromboembolism among postmenopausal
This is the first work to find that new-onset life women. Osteoporos Int. 23, 1931–1938 (2012). women. Menopause 18, 488–493 (2011).
depression is specifically associated with the late 147. Lim, L. S., Hoeksema, L. J. & Sherin, K. Screening for 170. Hoibraaten, E. et al. Increased risk of recurrent
menopause transition. osteoporosis in the adult U. S. population: ACPM venous thromboembolism during hormone
127. Bromberger, J. T. et al. Depressive symptoms during position statement on preventive practice. Am. replacement therapy—results of the randomized,
the menopausal transition: the Study of Women’s J. Prev. Med. 36, 366–375 (2009). double-blind, placebo-controlled estrogen in venous
Health Across the Nation (SWAN). J. Affect Disord. 148. Davis, S. R., Tan, A. & Bell, R. J. Targeted assessment thromboembolism trial (EVTET). Thromb. Haemost.
103, 267–272 (2007). of fracture risk in women at midlife. Osteoporos. Int. 84, 961–967 (2000).
128. Pastore, L. M., Carter, R. A., Hulka, B. S. & Wells, E. http://dx.doi.org/10.1007/s00198-015-3046-9 171. Liu, B. et al. Gallbladder disease and use of
Self-reported urogenital symptoms in (2015). transdermal versus oral hormone replacement therapy
postmenopausal women: Women’s Health Initiative. This paper offers a clinical approach to in postmenopausal women: prospective cohort study.
Maturitas 49, 292–303 (2004). pre-screening women at midlife using bone BMJ 337, a386 (2008).
129. Freeman, E. W., Sammel, M. D. & Sanders, R. J. Risk densitometry. 172. Constantine, G., Graham, S., Portman, D. J.,
of long-term hot flashes after natural menopause: 149. Lenders, J. W. et al. Pheochromocytoma and Rosen, R. C. & Kingsberg, S. A. Female sexual function
evidence from the Penn Ovarian Aging Study cohort. paraganglioma: an endocrine society clinical improved with ospemifene in postmenopausal women
Menopause 21, 924–932 (2014). practice guideline. J. Clin. Endocrinol. Metab. 99, with vulvar and vaginal atrophy: results of a
130. Politi, M. C., Schleinitz, M. D. & Col, N. F. Revisiting 1915–1942 (2014). randomized, placebo-controlled trial. Climacteric
the duration of vasomotor symptoms of menopause: a 150. American Psychiatric Association. Diagnostic and http://dx.doi.org/10.3109/13697137.2014.954996
meta-analysis. J. Gen. Intern. Med. 23, 1507–1513 Statistical Manual of Mental Disorders 5th edn (2014).
(2008). (American Psychiatric Association, 2013). 173. Wurz, G. T., Soe, L. H. & DeGregorio, M. W.
131. Col, N. F., Guthrie, J. R., Politi, M. & Dennerstein, L. 151. Bromberger, J. T. et al. Persistent mood symptoms in Ospemifene, vulvovaginal atrophy, and breast cancer.
Duration of vasomotor symptoms in middle-aged a multiethnic community cohort of pre- and Maturitas 74, 220–225 (2013).
women: a longitudinal study. Menopause 16, perimenopausal women. Am. J. Epidemiol. 158, 174. Pinkerton, J. V. et al. Evaluation of the efficacy and
453–457 (2009). 347–356 (2003). safety of bazedoxifene/conjugated estrogens for
132. Kravitz, H. M. & Joffe, H. Sleep during the 152. Morse, C. A., Dudley, E., Guthrie, J. & Dennerstein, L. secondary outcomes including vasomotor symptoms in
perimenopause: a SWAN story. Obstet. Gynecol. Clin. Relationships between premenstrual complaints and postmenopausal women by years since menopause in
North Am. 38, 567–586 (2011). perimenopausal experiences. J. Psychosom. Obstet. the Selective Estrogens, Menopause and Response to
133. Freeman, E. W., Sammel, M. D., Lin, H. & Gynaecol. 19, 182–191 (1998). Therapy (SMART) trials. J. Womens Health (Larchmt)
Nelson, D. B. Associations of hormones and 153. Holte, A. & Mikkelsen, A. The menopausal syndrome: 23, 18–28 (2014).
menopausal status with depressed mood in women a factor analytic replication. Maturitas 13, 193–203 175. Wierman, M. E. et al. Androgen therapy in women: a
with no history of depression. Arch. Gen. Psychiatry (1991). reappraisal: an endocrine society clinical practice
63, 375–382 (2006). 154. Tehrani, F. R., Solaymani-Dodaran, M., Tohidi, M., guideline. J. Clin. Endocrinol. Metab. 99, 3489–3510
134. Wallace, R. B., Sherman, B. M., Bean, J. A., Gohari, M. R. & Azizi, F. Modeling age at menopause (2014).
Treloar, A. E. & Schlabaugh, L. Probability of using serum concentration of anti-mullerian hormone. 176. Davis, S. R. Cardiovascular and cancer safety of
menopause with increasing duration of amenorrhea in J. Clin. Endocrinol. Metab. 98, 729–735 (2013). testosterone in women. Curr. Opin. Endocrinol.
middle-aged women. Am. J. Obstet. Gynecol. 135, 155. Dewailly, D. et al. The physiology and clinical utility of Diabetes Obes 18, 198–203 (2011).
1021–1024 (1979). anti-Mullerian hormone in women. Hum. Reprod. 177. Santen, R. J. Vaginal administration of estradiol:
135. Burger, H. G., Dudley, E. C., Cui, J., Dennerstein, L. & Update 20, 370–385 (2014). effects of dose, preparation and timing on plasma
Hopper, J. L. A prospective longitudinal study of 156. Tran, N. D., Cedars, M. I. & Rosen, M. P. The role of estradiol levels. Climacteric 17, 1–14 (2014).
serum testosterone, dehydroepiandrosterone sulfate, anti-mullerian hormone (AMH) in assessing ovarian 178. Grodstein, F., Manson, J. E., Stampfer, M. J. &
and sex hormone-binding globulin levels through the reserve. J. Clin. Endocrinol. Metab. 96, 3609–3614 Rexrode, K. Postmenopausal hormone therapy and
menopause transition. J. Clin. Endocrinol. Metab. 85, (2011). stroke: role of time since menopause and age at
2832–2838 (2000). 157. Sullivan, S. D., Welt, C. & Sherman, S. FMR1 and the initiation of hormone therapy. Arch. Intern. Med. 168,
136. Metcalf, M. G., D.Onald, R. A. & Livesey, J. H. continuum of primary ovarian insufficiency. Semin. 861–866 (2008).
Pituitary-ovarian function before, during and after the Reprod. Med. 29, 299–307 (2011). 179. American College of Obstetricians and Gynecologists.
menopause: a longitudinal study. Clin. Endocrinol. 158. Spath, M. A. et al. Intra-individual stability over time ACOG committee opinion no. 556: Postmenopausal
(Oxf.) 17, 489–494 (1982). of standardized anti-Mullerian hormone in FMR1 estrogen therapy: route of administration and risk of
137. Davison, S. L., Bell, R., Donath, S., Montalto, J. G. & premutation carriers. Hum. Reprod. 26, 2185–2191 venous thromboembolism. Obstet. Gynecol. 121,
Davis, S. R. Androgen levels in adult females: changes (2011). 887–890 (2013).
with age, menopause, and oophorectomy. J. Clin. 159. Richardson, S. J., Senikas, V. & Nelson, J. F. 180. Canonico, M. et al. Hormone therapy and venous
Endocrinol. Metab. 90, 3847–3853 (2005). Follicular depletion during the menopausal transition: thromboembolism among postmenopausal women:
138. Modi, D., Bhartiya, D. & Puri, C. Developmental evidence for accelerated loss and impact of the route of estrogen administration and
expressionand cellular distribution of Mullerian ultimate exhaustion. J. Clin. Endocrinol. Metab. 65, progestogens: the ESTHER study. Circulation 115,
inhibiting substance in the primate ovary. 1231–1237 (1987). 840–845 (2007).
Reproduction 132, 443–453 (2006). 160. Woods, D. C., White, Y. A. & Tilly, J. L. Purification of 181. Fournier, A. et al. Risks of endometrial cancer
139. Sowers, M. R. et al. Anti-mullerian hormone and oogonial stem cells from adult mouse and human associated with different hormone replacement
inhibin B in the defintion of ovarian aging and the ovaries: an assessment of the literature and a view therapies in the E3N cohort, 1992–2008. Am.
menopause transition. J. Clin. Endocrinol. Metab. 93, toward the future. Reprod. Sci. 20, 7–15 (2013). J. Epidemiol. 180, 508–517 (2014).
3478–3483 (2008). 161. Hanna, C. B. & Hennebold, J. D. Ovarian germline 182. Manson, J. E. et al. Algorithm and mobile app for
140. Santoro, N. et al. Helping midlife women predict the stem cells: an unlimited source of oocytes? Fertil. menopausal symptom management and hormonal/
onset of the final menses, SWAN, the Study of Steril. 101, 20–30 (2014). non-hormonal therapy decision making: a clinical
Women’s Health Across the Nation. Menopause 14, 162. Yuan, J. et al. No evidence for neo-oogenesis may link decision-support tool from The North American
415–424 (2007). to ovarian senescence in adult monkey. Stem Cells Menopause Society. Menopause 22, 247–253
141. Freeman, E. W., Sammel, M. D., Lin, H., 31, 2538–2550 (2013). (2014).
Boorman, D. W. & Gracia, C. R. Contribution of the 163. de Villiers, T. J. et al. Global Consensus Statement 183. Chin, J., Konje, J. C. & Hickey, M. Levonorgestrel
rate of change of antimullerian hormone in estimating on menopausal hormone therapy. Maturitas 74, intrauterine system for endometrial protection in
time to menopause for late reproductive-age women. 391–392 (2013). women with breast cancer on adjuvant tamoxifen.
Fertil. Steril. 98, 1254–1259 (2012). 164. Furness, S., Roberts, H., Marjoribanks, J. & Cochrane Database Syst. Rev. 4, CD007245 (2009).
This is the first work to examine the value of serial Lethaby, A. Hormone therapy in postmenopausal 184. Wan, Y. L. & Holland, C. The efficacy of levonorgestrel
AMH levels in predicting menopause. women and risk of endometrial hyperplasia. intrauterine systems for endometrial protection: a
142. La Marca, A. et al. Anti-Mullerian hormone-based Cochrane Database Syst. Rev. 8, CD000402 (2012). systematic review. Climacteric 14, 622–632 (2011).
prediction model for a live birth in assisted 165. Hall, E., Frey, B. N. & Soares, C. N. Non-hormonal 185. Stearns, V. et al. Active tamoxifen metabolite plasma
reproduction. Reprod. Biomed. Online 22, 341–349 treatment strategies for vasomotor symptoms: a concentrations after coadministration of tamoxifen
(2011). critical review. Drugs 71, 287–304 (2011). and the selective serotonin reuptake inhibitor
143. Hansen, K. R., Craig, L. B., Zavy, M. T., Klein, N. A. & 166. Borrelli, F. & Ernst, E. Alternative and complementary paroxetine. J. Natl Cancer Inst. 95, 1758–1764
Soules, M. R. Ovarian primordial and nongrowing therapies for the menopause. Maturitas 66, 333–343 (2003).
follicle counts according to the Stages of Reproductive (2010). 186. Lethaby, A. et al. Phytoestrogens for menopausal
Aging Workshop (STRAW) staging system. This is a comprehensive systematic review of vasomotor symptoms. Cochrane Database Syst. Rev.
Menopause 19, 164–171 (2012). alternative and complementary medicine for 12, CD001395 (2013).
144. Crandall, C. J. et al. Osteoporosis screening in menopausal symptoms. 187. Rada, G. et al. Non-hormonal interventions for hot
postmenopausal women 50 to 64 years old: 167. Dunn, J. F., Nisula, B. C. & Rodboard, D. Transport of flushes in women with a history of breast cancer.
comparison of US Preventive Services Task Force steroid hormones. Binding of 21 endogenous steroids Cochrane Database Syst. Rev. 9, CD004923 (2010).
strategy and two traditional strategies in the Women’s to both testosterone-binding globulin and cortico- 188. Leach, M. J. & Moore, V. Black cohosh (Cimicifuga spp.)
Health Initiative. J. Bone Miner. Res. 29, 1661–1666 steroid-binding globulin in human plasma. J. Clin. for menopausal symptoms. Cochrane Database Syst.
(2014). Endocrinol. Metab. 53, 58–68 (1981). Rev. 9, CD007244 (2012).
145. Sowers, M. R. et al. Amount of bone loss in relation to 168. Raisz, L. G. et al. Comparison of the effects of 189. Laakmann, E., Grajecki, D., Doege, K.,
time around the final menstrual period and follicle- estrogen alone and estrogen plus androgen on zu Eulenburg, C. & Buhling, K. J. Efficacy of Cimicifuga
stimulating hormone staging of the transmenopause. biochemical markers of bone form and resorption in racemosa, Hypericum perforatum and Agnus castus in
J. Clin. Endocrinol. Metabolism 95, 2155–2162 postmenopausal women. J. Clin. Endocrinol. Metab. the treatment of climacteric complaints: a systematic
(2010). 81, 37–43 (1995). review. Gynecol. Endocrinol. 28, 703–709 (2012).

18 | 2015 | VOLUME 1 www.nature.com/nrdp

© 2015 Macmillan Publishers Limited. All rights reserved


PRIMER

190. Walega, D. R., Rubin, L. H., Banuvar, S., 199. Portman, D. J., Bachmann, G. A. & Simon, J. A. quality of life and physiological parameters in
Shulman, L. P. & Maki, P. M. Effects of stellate Ospemifene, a novel selective estrogen receptor symptomatic postmenopausal women: a double-blind,
ganglion block on vasomotor symptoms: findings from modulator for treating dyspareunia associated with placebo-controlled trial. Swedish Alternative Med.
a randomized controlled clinical trial in postmenopausal vulvar and vaginal atrophy. Group. Int. J. Clin. Pharmacol. Res. 19, 89–99 (1999).
postmenopausal women. Menopause 21, 807–814 Menopause 20, 623–630 (2013). 210. Hays, J. et al. Effects of estrogen plus progestin on
(2014). 200. Avis, N. E. et al. Change in health-related quality of life health-related quality of life. N. Engl. J. Med. 348,
191. Sinha, A. & Ewies, A. A. Non-hormonal topical over the menopausal transition in a multiethnic cohort 1839–1854 (2003).
treatment of vulvovaginal atrophy: an up‑to‑date of middle-aged women: Study of Women’s Health 211. Brunner, R. L. et al. Effects of conjugated equine
overview. Climacteric 16, 305–312 (2013). Across the Nation. Menopause 16, 860–869 (2009). estrogen on health-related quality of life in
192. Gartoulla, P., Worsley, R., Bell, R. J. & Davis, S. R. This paper discusses the impact of menopause on postmenopausal women with hysterectomy: results
Moderate-severe vasomotor and sexual symptoms HRQoL. from the Women’s Health Initiative randomized clinical
remain problematic for 60–65 year old women. 201. Zollner, Y. F., Acquadro, C. & Schaefer, M. Literature trial. Arch. Internal Med. 165, 1976–1986 (2005).
Menopause http://dx.doi.org/10.1097/ review of instruments to assess health-related quality 212. Hlatky, M. A., Boothroyd, D., Vittinghoff, E., Sharp, P.
GME.0000000000000383 (2014). of life during and after menopause. Qual. Life Res. 14, & Whooley, M. A. Quality-of‑life and depressive
193. Elavsky, S., Gonzales, J. U., Proctor, D. N., Williams, N. 309–327 (2005). symptoms in postmenopausal women after receiving
& Henderson, V. W. Effects of physical activity on 202. Salpeter, S. R., Buckley, N. S., Liu, H. & Salpeter, E. E. hormone therapy: results from the Heart and Estrogen/
vasomotor symptoms: examination using objective and The cost-effectiveness of hormone therapy in younger Progestin Replacement Study (HERS) trial. JAMA 287,
subjective measures. Menopause 19, 1095–1103 and older postmenopausal women. Am. J. Med. 122, 591–597 (2002).
(2012). 42–52.e2 (2009). 213. Ylikangas, S., Sintonen, H. & Heikkinen, J. Decade-long
194. Daley, A. et al. The effectiveness of exercise as This paper reinforces the importance of QoL at a use of continuous combined hormone replacement
treatment for vasomotor menopausal symptoms: community level. therapy is associated with better health-related quality
randomised controlled trial. BJOG 122, 565–575 203. Utian, W. H. & Woods, N. F. Impact of hormone therapy of life in postmenopausal women, as measured by the
(2015). on quality of life after menopause. Menopause 20, generic 15D instrument. J. Br. Menopause Soc. 11,
This is a randomized trial in primary care that 1098–1105 (2013). 145–151 (2005).
examines the effects of exercise on symptoms of 204. Archer, D. F. et al. Long-term safety of drospirenone- 214. Zeleke, B. M., Davis, S. R., Fradkin, P. & Bell, R. J.
menopause. It shows no effect, thus questioning estradiol for hormone therapy: a randomized, double- Vasomotor symptoms and urogenital atrophy in older
the recommendation of exercise by healthcare blind, multicenter trial. Menopause 12, 716–727 women: a systematic review. Climacteric http://dx.doi.
providers as a first-line therapy. (2005). org/10.3109/13697137.2014.978754 (2014).
195. Babatunde, O. O., Forsyth, J. J. & Gidlow, C. J. A 205. Al‑Akoum, M. et al. Effects of Hypericum perforatum 215. Rossouw, J. et al. Risks and benefits of estrogen plus
meta-analysis of brief high-impact exercises for (St. John’s wort) on hot flashes and quality of life in progestin in healthy postmenopausal women: principal
enhancing bone health in premenopausal women. perimenopausal women: a randomized pilot trial. results from the Women’s Health Initiative Randomised
Osteoporos Int. 23, 109–119 (2012). Menopause 16, 307–314 (2009). Controlled Trial. JAMA 288, 321–333 (2002).
196. Martyn‑St James, M. & Carroll, S. A meta-analysis of 206. Kalay, A. E., Demir, B., Haberal, A., Kalay, M. &
impact exercise on postmenopausal bone loss: the Kandemir, O. Efficacy of citalopram on climacteric Author contributions
case for mixed loading exercise programmes. Br. symptoms. Menopause 14, 223–229 (2007). Introduction (S.R.D.); Epidemiology (G.D.M.); Mechanisms
J. Sports Med. 43, 898–908 (2009). 207. Somunkiran, A., Erel, C. T., Demirci, F. & Senturk, M. L. and pathophysiology (T.S. and I.L.); Diagnosis, screening and
197. Tucker, L. A., Fosson, E., Bailey, B. W. & The effect of tibolone versus 17β‑estradiol on prevention (N.S. and S.R.D.); Management (M.R., L.P. and
Lecheminant, J. D. Is the dose response relationship climacteric symptoms in women with surgical S.R.D.); Quality of life (M.L.); Outlook (S.R.D.); and overview
between body mass and hip bone mineral density in menopause: a randomized, cross-over study. Maturitas of Primer (S.R.D.).
women influenced by diet, physical activity, or 56, 61–68 (2007).
menopause? Am. J. Health Promot. 28, 325–327 208. Utian, W. et al. Bazedoxifene/conjugated estrogens Competing interests statement
(2013). and quality of life in postmenopausal women. S . R . D . h a s re c e i ve d a n h o n o ra r i u m f ro m A b b o t t
198. Newton, K. M. et al. Efficacy of yoga for vasomotor Maturitas 63, 329–335 (2009). Pharmaceuticals for one presentation and is an investigator
symptoms: a randomized controlled trial. Menopause 209. Wiklund, I. K., Mattsson, L. A., Lindgren, R. & for Trimel Pharmaceuticals and Lawley Pharmaceuticals; all
21, 339–346 (2014). Limoni, C. Effects of a standardized ginseng extract on other authors declare no competing interests.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 1 | 2015 | 19

© 2015 Macmillan Publishers Limited. All rights reserved

You might also like