Polycystic ovary syndrome: pathophysiology and therapeutic opportunities


Abstract

Polycystic ovary syndrome is characterised by excessive levels of androgens and ovulatory dysfunction, and is a common endocrine disorder in women of reproductive age. Polycystic ovary syndrome arises as a result of polygenic susceptibility in combination with environmental influences that might include epigenetic alterations and in utero programming. In addition to the well recognised clinical manifestations of hyperandrogenism and ovulatory dysfunction, women with polycystic ovary syndrome have an increased risk of adverse mental health outcomes, pregnancy complications, and cardiometabolic disease. Unlicensed treatments have limited efficacy, mostly because drug development has been hampered by an incomplete understanding of the underlying pathophysiological processes. Advances in genetics, metabolomics, and adipocyte biology have improved our understanding of key changes in neuroendocrine, enteroendocrine, and steroidogenic pathways, including increased gonadotrophin releasing hormone pulsatility, androgen excess, insulin resistance, and changes in the gut microbiome. Many patients with polycystic ovary syndrome have high levels of 11-oxygenated androgens, with high androgenic potency, that might mediate metabolic risk. These advances have prompted the development of new treatments, including those that target the neurokinin-kisspeptin axis upstream of gonadotrophin releasing hormone, with the potential to lessen adverse clinical sequelae and improve patient outcomes.

http://creativecommons.org/licenses/by-nc/4.0/

This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/.

https://doi.org/10.1136/bmjmed-2023-000548

Statistics from Altmetric.com

See more details

Posted by 1 X users

11 readers on Mendeley

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Request permissions

Introduction

Polycystic ovary syndrome is a common metabolic and reproductive disorder characterised variably by high levels of androgens, insulin resistance, and ovulatory dysfunction, with not all patients affected by these three parameters. These changes manifest as hyperandrogenism (hirsutism, acne, or scalp hair loss, or a combination of these), oligomenorrhoea or amenorrhoea, and morphological features of polycystic ovaries on ultrasound. Long recognised as a reproductive disorder, polycystic ovary syndrome is now also established as a metabolic condition associated with long term health risks, including type 2 diabetes and cardiovascular disease.1 Adverse mental health outcomes and reduced quality of life have also been reported.1 Polycystic ovary syndrome is therefore associated with substantial healthcare costs and resource utilisation. International surveys report a high level of dissatisfaction with care,2 not least because current treatments are often only modestly effective in alleviating symptoms and minimising long term risks. International guidelines recognise the low quality of evidence and the critical need for better research.3

An improved understanding of the pathogenesis of the disease might result in the development of new treatments and better patient outcomes. Recent studies have advanced our understanding of these pathophysiological processes. Neuroendocrine dysregulation leads to abnormal high frequency pulsatile secretion of gonadotrophin releasing hormone, and hypothalamic kisspeptin, neurokinin B, and dynorphin A neurons (so-called KNDy neurons) are integral regulators of this process.4 Although increased production of ovarian and adrenal androgens contribute to hyperandrogenism, peripherally generated 11-oxygenated androgens are emerging as important predictors of metabolic risk.5 6 Together with advances in our understanding of adipocyte biology, insulin resistance, the gut microbiome, and insights from genome-wide association studies, these studies could improve our understanding of the pathogenesis of this disease. New treatments based on these observations are now in various stages of preclinical or clinical development.

This review outlines our current understanding of the key pathophysiological processes in polycystic ovary syndrome. We discuss the significance of new research into neuroendocrine dysfunction, disrupted steroidogenesis, and changes in adipocyte biology, and the potential implications for the diagnosis and management of polycystic ovary syndrome. Finally, we consider the benefits and limitations of current drug treatments, along with a review of the evidence for emerging drug treatments.

Epidemiology

Polycystic ovary syndrome is a common endocrine disorder in women of reproductive age,7 with a prevalence of 4-21% depending on the diagnostic criteria used.8 A systematic review of 13 studies found a slightly higher estimate of the prevalence in black and Middle-Eastern than in Chinese and white populations,9 although inconsistencies in diagnostic criteria and recruitment methods make comparisons between ethnic groups challenging.10 The global disease burden seems to be increasing at a high rate. In 2019, an age standardised point prevalence of 1677.8 per 100 000 and an annual incidence of 59.8 per 100 000 population were reported based on data from 204 countries, representing increases of 30.4% and 29.5%, respectively, since 1990.11 The rising incidence, and accompanying morbidity,11 emphasises the importance of recognising polycystic ovary syndrome as an international public health priority.

Sources and selection criteria

We searched PubMed, Medline, and Embase from 1 January 2010 to 28 February 2023 for articles in the English language, with the search terms: “polycystic ovary syndrome,” “polycystic ovarian syndrome,” “PCOS,” “aetiology,” “etiology,” “cause,” “pathogenesis,” and “pathophysiology.” We excluded articles on endocrine conditions that might lead to secondary polycystic ovary syndrome, including acromegaly, androgen secreting tumours, congenital adrenal hyperplasia, and Cushing’s syndrome. To identify registered clinical trials, we searched ClinicalTrials.gov, the Cochrane Central Register of Controlled Trials (CENTRAL), and the International Standard Randomised Controlled Trial Number (ISRCTN) registry with the search terms “PCOS,” “polycystic ovary syndrome,” and “polycystic ovarian syndrome.” We prioritised large scale, randomised controlled trials and systematic reviews. We also included relevant articles identified from reference lists of retrieved articles.

Pathogenesis

Neuroendocrine disruption

Polycystic ovary syndrome is characterised by increased pulse frequency of gonadotrophin releasing hormone and reduced negative feedback from sex steroids at the level of the hypothalamus.4 12 Gonadotrophin releasing hormone is released from neurons in the hypothalamic infundibular nucleus in a pulsatile manner, resulting in increased secretion of luteinising hormone and follicle stimulating hormone. The pulse frequency of gonadotrophin releasing hormone is controlled by multiple upstream endocrine and neural factors, with a higher frequency favouring secretion of luteinising hormone and a lower frequency favouring secretion of follicle stimulating hormone. In women with polycystic ovary syndrome, raised levels of luteinising hormone cause excess production of ovarian thecal androgens, whereas relative deficiency of follicle stimulating hormone causes follicular arrest, polycystic ovarian morphology, and oligo-ovulation.4 The reduction in sex steroid feedback on release of gonadotrophin releasing hormone is thought to occur upstream of the hormone itself because gonadotrophin releasing hormone neurons do not have receptors for oestrogens or progesterone13 (figure 1). KNDy neurons have an important role in this regard (figure 1).

Kisspeptins are a family of peptides encoded by the KISS1 gene which act on the neuronal G protein coupled receptor KISS1R. KISS1 encodes prepro-kisspeptin, which is cleaved to produce the biologically active peptides KP54, KP14, KP13, and KP10.14 Two discrete neuronal populations exist: KNDy neurons in the infundibular nucleus function as the gonadotrophin releasing hormone pulse generator15 and mediate negative feedback from oestradiol,16 whereas a separate kisspeptin population located in the preoptic area mediates oestradiol positive feedback to produce the mid-cycle surge in luteinising hormone.16 17 Kisspeptin neurons express sex steroid receptors (progesterone and oestrogen receptors) required for negative feedback on gonadotrophin releasing hormone pulsatility.17 18 KISS1 is also expressed in adipose tissue where it is regulated independently of hypothalamic KISS1.19 Circulating levels of kisspeptin are higher in patients with polycystic ovary syndrome than in controls20 and although the origin of this excess is not entirely clear, a raised pulse frequency of kisspeptin in women with oligomenorrhoea and polycystic ovary syndrome suggests a hypothalamic source.21 Moreover, physiological coupling of kisspeptin and luteinising hormone pulsatility is lost in these women.21 The exact mechanisms for these effects are unclear, with inconsistent data from preclinical models on the existence and direction of dysregulated gonadotrophin releasing hormone pulsatility mediated by kisspeptin.22

Figure 1

Figure 1

Pathophysiology and neuroendocrine disruption of the hypothalamo-pituitary-gonadal axis in polycystic ovary syndrome. (Left) Increased pulsatility of gonadotrophin releasing hormone (GnRH) causes increased secretion of luteinising hormone, consequent disrupted folliculogenesis, and increased production of ovarian androgens. Adrenal androgens are also increased, including 11-oxygenated androgens which are activated peripherally by renal 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2) and aldo-keto reductase 1C3 (AKR1C3) in adipocytes. Steroid-5α-reductase (SRD5A) converts 11-ketotestosterone to 11-ketodihydrotestosterone. Excess levels of androgens stimulate deposition of abdominal adipose tissue which subsequently increases insulin resistance and hyperinsulinism. Hyperinsulinism stimulates AKR1C3 activity, increases androgen production from the ovaries (by its action as a co-gonadotrophin) and adrenal cortex, reduces production of hepatic sex hormone binding globulin, and inhibits progesterone mediated negative feedback onto GnRH neurons, worsening androgen excess in a vicious cycle. (Right) Kisspeptin, neurokinin B, and dynorphin A neurons (KNDy neurons) act in a paracrine and autocrine way to regulate release of kisspeptin onto GnRH neurons and consequent GnRH pulsatility. Neurokinin B binds to neurokinin 3 receptors (NK3R) to stimulate release of kisspeptin whereas dynorphin binds to kappa opioid receptors to inhibit kisspeptin release. γ-aminobutyric acid (GABA) and anti-müllerian hormone (AMH) bind to GABAA receptors (GABAAR) and AMH receptor type 2 (AMHR2), respectively, to stimulate GnRH pulsatility. Impaired negative feedback from oestradiol and progesterone is seen at the level of the hypothalamus. Neuroendocrine abnormalities in the control of these components are shown in red. OR=oestrogen receptor; PR=progesterone receptor

Neurokinin B and dynorphin are expressed by KNDy neurons and act in an autocrine and paracrine way to control release of kisspeptin (figure 1). Neurokinin B preferentially binds to the neurokinin 3 receptor (encoded by TACR3) to stimulate gonadotrophin releasing hormone pulsatility.4 23 Unlike KISS1 null mice, mice deficient in components of neurokinin B signalling can still generate surges in luteinising hormone and conceive, suggesting that compensatory pathways exist which contribute to the generation of kisspeptin and gonadotrophin releasing hormone pulses.17 24 25 This milder effect of neurokinin B blockade might avoid excessive reduction in gonadotrophin releasing hormone pulsatility, making it an attractive target for treatment.4 Dynorphin, which activates kappa opioid receptors on KNDy neurons to inhibit secretion of gonadotrophin releasing hormone,22 26 has been shown to mediate progesterone negative feedback on gonadotrophin releasing hormone neurons in sheep27 and humans.22 28

Neuronal activity of gonadotrophin releasing hormone is also regulated by other substances, including γ-aminobutyric acid (GABA) and anti-müllerian hormone, both of which stimulate gonadotrophin releasing hormone neurons directly. GABA exerts an excitatory effect on gonadotrophin releasing hormone neurons through GABAA receptors, and GABA levels in cerebrospinal fluid can be raised in patients with polycystic ovary syndrome.29 Anti-müllerian hormone is secreted by ovarian granulosa cells, where raised levels in women with polycystic ovary syndrome disrupt folliculogenesis and ovulation.30 Anti-müllerian hormone might also have neuroendocrine effects: 50% of gonadotrophin releasing hormone neurons in mice and humans express anti-müllerian hormone receptor type 2,31 with studies implicating anti-müllerian hormone in neuronal migration of gonadotrophin releasing hormone,32 gonadotrophin releasing hormone pulsatility, and secretion of luteinising hormone.30

Classical pathway of androgen synthesis

High levels of androgens is a primary defect in polycystic ovary syndrome. Cholesterol is converted to androgens by a cascade of enzymes common to all steroid producing organs, with tissue specific variations resulting in different steroid hormone profiles.33 In polycystic ovary syndrome, increased production of ovarian androgens by the classical pathway is driven by increased secretion of pituitary luteinising hormone, the action of insulin as a co-gonadotrophin, and increased thecal cell hypersensitivity to luteinising hormone.34–36 Figure 2 summarises the classical pathway of steroidogenesis. Through a sequence of reactions, cholesterol is converted to dehydroepiandrosterone, which is then converted to androstenedione by 3β-hydroxysteroid dehydrogenase type II and subsequently to testosterone by aldo-keto reductase type 1C3 (AKR1C3).35

Figure 2

Figure 2

Classical pathway of androgen synthesis. Luteinising hormone stimulates the classical pathway of androgen synthesis in ovarian theca cells. Cholesterol is transported to the inner mitochrondrial membrane by steroidogenic acute regulatory protein (StAR). A cleavage system of the cytochrome P450 enzyme, CYP11A1, ferrodoxin, and ferrodoxin reductase converts cholesterol to pregnenolone. Expression of CYP11A1 is stimulated by activation of the luteinising hormone receptor. Pregnenolone is transported to smooth endoplasmic reticulum where it is converted to 17-hydroxypregnenolone and subsequently to dehydroepiandrosterone by the 17-hydroxylase and 17,20-lyase subunit of the CYP17A1 enzyme, respectively. Dehydroepiandrosterone is then converted to androstenedione or androstenediol and subsequently to testosterone by a combination of 3β-hydroxysteroid dehydrogenase type II (HSD3B2) and aldo-keto reductase type 1C3 (AKR1C3). 17β-hydroxysteroid dehydrogenase 1 (HSD17B1) also catalyses the conversion of dehydroepiandrosterone to androstenediol. HSD3B2 converts pregnenolone and 17-hydroxypregnenolone to progesterone and 17-hydroxyprogesterone, respectively, which are substrates for a back door alternative pathway of androgen synthesis. Androstenedione and testosterone diffuse into granulosa cells where they are converted to oestrogens by the action of aromatase (CYP19A1), under the control of follicle stimulating hormone receptor activation. Testosterone can be converted to dihydrotestosterone by steroid 5α-reductase (SRD5A) in peripheral tissues

Increased activity of ovarian 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which converts inactive cortisone to active cortisol, might also have a role in the pathogenesis of polycystic ovary syndrome.37 Overexpression of ovarian 11β-HSD1 in rats caused polycystic ovarian morphology, oestrous cycle, and reproductive hormone abnormalities.37 Although 11β-HSD1 is widely expressed, dysregulation seems to be tissue specific, because hepatic 11β-HSD1 activity is impaired and expression of 11β-HSD1 in subcutaneous adipose tissue is increased in patients with polycystic ovary syndrome.38 Raised circulating levels and ovarian expression of vascular endothelial growth factor also contribute to the hypervascular, hyperplastic appearance of the ovarian stroma and theca interna in polycystic ovary syndrome, and might contribute to increased ovarian androgen synthesis.39

Androgen synthesis in adrenal glands and peripheral tissues

Polycystic ovary syndrome was previously thought to be primarily a disease of excess production of androgens in the ovaries, but the adrenal glands and peripheral tissues are now considered important sources of androgens in patients with polycystic ovary syndrome. Increased concentrations of dehydroepiandrosterone sulphate, an almost exclusive product of the adrenal cortex,40 are apparent in 20-30% of patients with polycystic ovary syndrome.41 This finding seems to be the result of increased secretory activity of the adrenal cortex because no change in pituitary responsiveness to corticotrophin releasing hormone or reduction in the minimal stimulatory dose of adrenocorticotropic hormone required for adrenal hormone production is seen.42 Changes in steroidogenesis, such as increased enzymatic activity of the 17-hydroxylase subunit of the cytochrome P450 enzyme, CYP17A1, might account for this hyper-responsiveness.43

Other adrenal androgens are also secreted in excess, including 11β-hydroxyandrostenedione and 11β-hydroxytestosterone.5 44 The adrenal androgen 11β-hydroxyandrostenedione is abundant and was previously thought to have little physiological importance because of its weak androgenic activity. Recent studies, however, have shown that 11β-hydroxyandrostenedione can be metabolised to 11-ketotestosterone and 11-ketodihydrotestosterone, termed 11-oxygenated androgens, because of the presence of an oxygen atom on carbon 11.45 Both 11-ketotestosterone and 11-ketodihydrotestosterone bind to androgen receptors with similar affinity and potency to testosterone and dihydrotestosterone.46 47 Mass spectrometry analyses have shown that 11-oxygenated androgens are the dominant circulating androgens in women with polycystic ovary syndrome and correlate substantially with markers of metabolic risk.5 The synthesis of 11-oxygenated androgens is reliant on the peripheral activation of adrenal derived androgens (figure 3). 11β-hydroxysteroid dehydrogenase type 2 is an enzyme expressed by the kidney that converts 11β-hydroxyandrostenedione to 11-ketoandrostenedione, and 11β-hydroxytestosterone to 11-ketotestosterone.45 Adipose tissue also has enzymes responsible for potent androgen formation, however, and might represent the dominant source of circulating 11-oxygenated androgens.45 48

Expression of the androgen activating enzyme, AKR1C3, in subcutaneous adipose tissue is increased in women with polycystic ovary syndrome compared with matched controls.6 49 Thus concentrations of androgens in adipose tissue are increased in women with polycystic ovary syndrome, accompanied by inhibition of lipolysis and increased de novo lipogenesis.6 These observations suggest that inhibition of AKR1C3 might be an attractive therapeutic target in patients with polycystic ovary syndrome.

Figure 3

Figure 3

Pathway for 11-oxygenated androgen synthesis, which begins in the adrenal cortex. Androstenedione and testosterone are produced by the classical pathway (figure 2). Dehydroepiandrosterone is diverted to downstream androgens or sulphonated to dehydroepiandrosterone sulphate by the sulphotransferase, SULT2A1. Androstenedione and testosterone are hydroxylated by 11β-hydroxylase (CYP11B) to produce abundant 11β-hydroxyandrostenedione (11OHA4) and smaller amounts of 11β-hydroxytestosterone (11OHT). Renal 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2) converts 11OHT to 11-ketotestosterone (11KT) and 11OHA4 to 11-ketoandrostenedione (11KA4). In adipose tissue, 11KA4 is metabolised to 11KT and 11-ketodihydrotestosterone (11DHKT) by aldo-keto reductase type 1C3 (AKR1C3) and steroid-5α-reductase (SRD5A), respectively. 11OHA4 is metabolised to 11OHT and 11β-hydroxydihydrotestosterone (11OHDHT) by 17β-hydroxysteroid dehydrogenase 2 (HSD17B2) and SRD5A, respectively. 11KT and 11KDHT are potent agonists of the androgen receptor whereas 11OHT and 11OHDHT have milder potency. StAR=steroidogenic acute regulatory protein; HSD3B2=3β-hydroxysteroid dehydrogenase type II; CYP11A1, CYP17A1, CYP11B1=cytochrome P450 enzymes

Hyperinsulinism

Insulin resistance, and the consequent hyperinsulinism, have an important role in driving androgen synthesis in many endocrine tissues. Insulin acts as a co-gonadotrophin in the ovaries,36 impairs progesterone mediated inhibition of the gonadotrophin releasing hormone pulse generator,50 and facilitates synthesis of androgens in the adrenal glands by increasing adrenocorticotropic hormone stimulated steroidogenesis.51 AKR1C3 expression and activity in adipocytes is increased by insulin, contributing to increased synthesis of androgens in adipocytes in polycystic ovary syndrome.52 Insulin also inhibits sex hormone binding globulin, facilitating hyperandrogenism by increasing the percentage of free biologically active androgens.53 Excess production of androgens then stimulates hyperinsulinism, leading to a vicious cycle between androgen and insulin excess.7 54 Several studies have also implicated hyperandrogenism in the accumulation of abdominal and visceral adipose tissue in polycystic ovary syndrome55 56; this hyperandrogenism further drives insulin resistance and consequent production of androgens (figure 1).

In common with hyperandrogenism, insulin resistance is not a universal feature of polycystic ovary syndrome, although a systematic review of hyperinsulinaemic-euglycaemic clamp studies of 1224 women with polycystic ovary syndrome and 741 controls showed that insulin sensitivity was lower in women with polycystic ovary syndrome than in controls (mean effect size −27%, 99% confidence interval −21 to −33).57 Studies exploring steroid metabolomics in patients with polycystic ovary syndrome might give more information. One such cross sectional study (n=488) combining machine learning with mass spectrometry multisteroid profiling has identified three distinct groups of patients based on the predominant source of androgens.58 These subgroups have distinct steroid metabolomes and risk of metabolic complications: a gonadal derived classical androgen excess group, an adrenal derived androgen excess group (comprising 11-oxygenated androgens), and a group with comparably mild androgen excess.58 The adrenal derived androgen group had the highest rates of hirsutism, insulin resistance, and type 2 diabetes. These insights challenge our understanding of polycystic ovary syndrome as one entity and might prompt a reconsideration of the classification of the disease based on the metabolomic signature.

Changes in adipocyte structure and function

Changes in white adipose tissue morphology and function is seen in women with polycystic ovary syndrome, including enlarged adipocytes, reduced lipoprotein lipase activity,59 and increased secretion of proinflammatory cytokines.60 The function of brown adipose tissue might also be disrupted because women with polycystic ovary syndrome showed reduced postprandial thermogenesis compared with controls matched for body mass index.61 This defect could be driven by androgen excess, because prenatally androgenised sheep have reduced postprandial thermogenesis in adulthood,62 accompanied by reduced adipose expression of thermogenic uncoupling proteins and sympathetic activity. Adolescent prenatally androgenised sheep also showed reduced hepatic expression and circulating levels of fibroblast growth factor 21,63 a hormone that regulates adipocyte function, insulin sensitivity, and energy balance. Targeting expression of fibroblast growth factor 21 during an appropriate period in development might be a therapeutic option.

Gut microbiota and bile acid metabolism

Recent studies have implicated changes in the gut microbiome in the pathogenesis of polycystic ovary syndrome. Women with polycystic ovary syndrome have higher intestinal levels of Bacteroides vulgatus and lower levels of glycodeoxycholic acid and tauroursodeoxycholic acid.64 Oral gavage of wild-type mice with faecal microbiota from individuals with polycystic ovary syndrome or pure B vulgatus caused insulin resistance, changes in bile acid metabolism, reduced secretion of interleukin 22, and disrupted oestrous cycle and ovarian morphology.64 Administration of interleukin 22 or glycodeoxycholic acid to mice treated with B vulgatus improved insulin sensitivity, testosterone levels, and oestrous cycles. Hence modifying the gut microbiota or bile acid metabolism, increasing levels of interleukin 22, or a combination of these actions, might be therapeutically valuable in polycystic ovary syndrome.64

Insights from genome-wide association studies

Genome-wide association studies have identified numerous susceptibility loci for polycystic ovary syndrome, including 11 in Han Chinese populations,65 66 eight in European populations,67 68 and eight in a Korean population.69 Robust candidate susceptibility loci are near genes belonging to metabolic (insulin receptor (INSR), insulin gene-variable number of tandem repeats (INS-VNTR), and DENN domain containing protein 1A (DENND1A))70 and neuroendocrine (follicle stimulating hormone receptor, luteinising hormone receptor, and thyroid adenoma associated (THADA)) pathways.70 Meta-analyses of genome-wide association studies have shown that the genetic architecture of polycystic ovary syndrome is consistent across different diagnostic criteria and ethnic groups.71 72 These observations indicate a shared ancestry for polycystic ovary syndrome and reinforce the importance of neuroendocrine and metabolic pathways in the pathogenesis of the disease.

Developmental programming

Genetic loci identified by genome-wide association studies currently account for only 10% of the known heritability (about 70%) of polycystic ovary syndrome,73 74 suggesting other influences on the pathogenesis of the disease. Emerging evidence indicates that polycystic ovary syndrome might have its origins in utero, and thus could be subject to developmental programming and epigenetic modifications. Prenatal exposure to androgens in several preclinical models caused a permanent polycystic ovary syndrome-like phenotype postnatally.75–77 A programming effect might also persist transgenerationally, because pregnant mice treated with dihydrotestosterone produced female offspring with polycystic ovary syndrome-like phenotypes from the first to the third generations of offspring.78 Cautious interpretation is needed, however, because these models might not accurately reflect the human phenotype. Anti-müllerian hormone might also be involved in in utero programming: levels of anti-müllerian hormone increased significantly in pregnant women with polycystic ovary syndrome (P<0.001), and use of this hormone caused gonadotrophin releasing hormone neuronal hyperactivity and androgen excess in pregnant mice.79 Epigenetic mechanisms might also be involved in mediating susceptibility to polycystic ovary syndrome, with differential methylation patterns and microRNA expression detected in adipose tissue and ovarian tissue of patients with polycystic ovary syndrome compared with controls.80

Health risks

Polycystic ovary syndrome is well established as a reproductive disorder associated with hyperandrogenism, and is the leading cause of oligomenorrhoea and amenorrhoea.81 Patients with polycystic ovary syndrome are at increased risk of mental health disorders,82 83 endometrial cancer,84 and ovarian hyperstimulation syndrome after induction of ovulation.85 Consistent with our understanding of the pathogenesis, however, polycystic ovary syndrome is also recognised as a metabolic disorder, with long term health risks, including hypertension, type 2 diabetes, dyslipidaemia, insulin resistance, and obesity.1 These health risks could be associated with an increased risk of cardiovascular events86 and several adverse pregnancy outcomes.87 Although the reproductive aspects might diminish with age, metabolic features typically persist or can worsen.88

Therapeutic goals

Difficulty in losing weight, irregular menses, infertility, and excessive hair growth were the most important health problems reported by patients with polycystic ovary syndrome in an international survey.2 These problems should therefore represent the main targets for therapeutic intervention, although priority setting partnerships are still needed to help focus research priorities. Existing drug treatments have not been licensed specifically for polycystic ovary syndrome and are used off-label to target symptoms. Also, previous studies have not emphasised health related quality of life measures when evaluating response to treatment. An ideal treatment for polycystic ovary syndrome should look at the health risks, reduce key processes in the pathogenesis of the disease, and be responsive to the symptom profile and needs of the individual. Where relevant, treatments should reduce clinical and biochemical hyperandrogenism, restore ovulatory cycles and fertility, normalise the length of the menstrual cycle, improve insulin sensitivity, reduce weight and cardiometabolic risk, and improve condition specific quality of life.

Existing treatments

Non-pharmacological interventions

International guidelines highlight the importance of modifications to lifestyle in the management of the disease.3 Changes in lifestyle can improve fasting insulin levels and anthropometric outcomes, although benefits on hyperandrogenism are modest89 and adherence is often difficult to sustain in clinical practice. Data on reproductive benefits are limited,90 although a recent small randomised controlled trial of 68 women with polycystic ovary syndrome showed that a behavioural modification programme improved menstrual regularity compared with a minimal intervention group.91 Laser treatment might have a role in the treatment of facial hirsutism, although further trials are needed to confirm the benefits on quality of life and cost effectiveness.3

Contraceptive pill

In women not attempting to conceive, combined contraceptive pills are first line treatments for menstrual irregularity and hyperandrogenism.3 The oestrogen component increases sex hormone binding globulin, thus reducing free testosterone and improving hyperandrogenism. Because this stimulatory effect on hepatic production of proteins also causes hypercoagulability, ethinyloestradiol based contraceptive pills containing the lowest effective dose of oestrogen (eg, 20-30 μg of ethinyloestradiol) are recommended.3 Combined contraceptive pills containing newer, more physiological, oestrogenic compounds have recently been developed, and might have a lower risk of venous thromboembolism than ethinyloestradiol.92 The progestogen component reduces ovarian androgen production by inhibiting secretion of luteinising hormone and protects the endometrium from hyperplasia.93 Combined contraceptive pills containing androgenic progestogens, such as norethisterone, should be avoided because of the potential to aggravate hyperandrogenic symptoms. Furthermore, ethinyloestradiol based contraceptive pills containing cyproterone acetate, the most potent anti-androgenic progestogen, are not currently recommended as first line treatment because of the increased risk of venous thromboembolism.3 A recent systematic review of 19 randomised controlled trials, however, concluded that the ethinyloestradiol-cyproterone acetate combination improved serum testosterone (mean difference 0.38 nmol/L, 95% confidence interval 0.33 to 0.43) and hirsutism compared with conventional combined contraceptive pills.94 Thus combinations of cyproterone acetate and newer oestrogenic compounds might have the potential to improve hyperandrogenism in patients with polycystic ovary syndrome without the added risk of venous thromboembolism.

Anti-androgen agents

Currently available anti-androgen agents act by blocking androgen receptors (cyproterone acetate, spironolactone, and flutamide) or reducing production of androgens (finasteride and dutasteride). Guidance on specific preparations or doses in polycystic ovary syndrome is necessarily vague, because studies on these agents are few in number and small scale.3 Furthermore, although targeting excess production of androgens might be crucial to improved patient outcomes, the use of currently available anti-androgen drugs is limited by side effects. All anti-androgen drugs carry a risk of feminisation of a male fetus and therefore use must be restricted to patients with adequate contraception in place.3

Insulin sensitisers

Metformin modulates hepatic insulin sensitivity and glucose production by activating AMP activated protein kinase and AMP activated protein kinase independent pathways. More recently, metformin has also been shown to mediate its antiglycaemic effects by actions on the gastrointestinal tract and the gut microbiome.95 Metformin is used to manage weight and metabolic outcomes in adult women with polycystic ovary syndrome with a body mass index ≥25.3 Metformin might also improve ovulation and live birth rates but is less effective than clomifene citrate or letrozole.3 96 Nevertheless, because of its wide availability and low cost, metformin could still be valuable in improving reproductive outcomes in women with polycystic ovary syndrome, especially in healthcare economies where access to assisted reproduction is limited.

Thiazolidinediones improve insulin sensitivity by activating nuclear peroxisome proliferator activated receptor γ. A meta-analysis of eight randomised controled trials concluded that thiazolidinediones reduce insulin and fasting glucose levels in polycystic ovary syndrome, but do not seem to affect hirsutism scores or serum levels of androgens.97 Existing data on thiazolidinediones in polycystic ovary syndrome are limited. Thiazolidinediones are associated with intrauterine growth restriction in animal studies and weight gain in humans.98 Thiazolidinediones are thus not recommended for use in polycystic ovary syndrome outside of the licensed indication in type 2 diabetes. Preliminary studies suggest that the insulin sensitiser inositol might improve glycaemic control99 and new international guidelines recommend shared decision making on using inositol for its potential metabolic benefits in polycystic ovary syndrome. Specific doses, forms, or combinations of the substance, however, cannot be recommended because of a lack of high quality evidence.3

New therapeutic targets

Kisspeptin based treatment

Kisspeptin has a major role as a regulator of the hypothalamic-pituitary-gonadal axis, and therefore extensive efforts have been made in investigating the effects of kisspeptin based treatment in women with polycystic ovary syndrome and in other disorders of reproduction. KP54 and KP10 are the most studied native kisspeptins in humans and have been investigated for their potential role in optimising oocyte maturation in patients undergoing in vitro fertilisation.100 101 Although the two compounds bind to KISS1R with similar affinity, KP54 has a longer serum half-life than KP10 and a more profound effect on secretion of luteinising hormone.102 A bolus dose of KP54 induces oocyte maturation in patients with polycystic ovary syndrome without causing clinically significant ovarian hyperstimulation syndrome.100 101 Administration of a subcutaneous bolus injection of native kisspeptin is safe and well tolerated103 and also results in higher expression of gonadotrophin receptors (follicle stimulating hormone receptor and luteinising hormone receptor) and steroidogenic enzymes (including aromatase CYP19A1, steroidogenic acute regulatory protein, and 3β-hydroxysteroid dehydrogenase type II) in ovarian granulosa cells, potentially promoting an ovarian environment favouring progesterone synthesis and ovarian implantation.104

Use of KP54 as an ovulation induction agent, however, might be limited by tachyphylaxis.105 Because KP54 preferentially stimulates secretion of luteinising hormone over follicle stimulating hormone,106 concerns also exist that long term administration of kisspeptin might exacerbate pre-existing deficiency of follicle stimulating hormone in polycystic ovary syndrome.4 Nevertheless, KP54 effectively induced ovulation in neonatally androgenised rats but not in prenatal androgenisation or post-weaning androgenisation models.107 Simultaneous increases in luteinising hormone and follicle stimulating hormone were seen after KP54 use in the neonatal androgenisation model, suggesting that ovulation induced by kisspeptin is linked to its ability to stimulate increases in both luteinising hormone and follicle stimulating hormone.107 In a first-in-woman pilot study of 12 patients with polycystic ovary syndrome, twice daily use of KP54 over three weeks significantly increased levels of luteinising hormone (P=0.04) and oestradiol (P=0.03) but not follicle stimulating hormone or inhibin B.107 Two of the 12 women developed a dominant follicle with subsequent ovulation. Hence long term administration of KP54 might be suitable for follicular maturation in a subset of patients with polycystic ovary syndrome, but more studies are needed to identify the patient characteristics that predict response to treatment.

KISS1R agonists are currently in development with modifications that increase potency and are resistant to proteolytic degradation. KISS1R agonists might have a lower risk of tachyphylaxis because of their longer duration of action, allowing less frequent dosing. The KISS1R agonist, MVT-602, showed greater potency than KP54, increasing release of luteinising hormone in healthy women with a longer duration of gonadotrophin releasing hormone neuronal activation in vitro.108 When tested in patients with polycystic ovary syndrome, MVT-602 increased luteinising hormone with a similar amplitude but greater duration than KP54 in healthy women (area under the curve of luteinising hormone exposure 171.30 v 38.5 IU×h/L), similar to the natural mid-cycle surge in luteinising hormone.108 These findings warrant further investigation in polycystic ovary syndrome and in other female reproductive disorders.

Paradoxically, kisspeptin receptor antagonists have also been suggested as therapeutic agents in polycystic ovary syndrome based on their potential to normalise hypersecretion of luteinising hormone, restore folliculogenesis and ovulation, and improve ovarian hyperandrogenism.109 Existing KISS1R antagonists, such as P234 and P271, have inconsistent effects on kisspeptin induced stimulation of gonadotrophin releasing hormone-luteinising hormone across species.110 111 Compound 15 a is a small molecular KISS1R antagonist with antagonistic activity at the receptor and good permeability of the blood-brain barrier in rats.112 KISS1R antagonists have yet to be tested in humans, however, and concerns exist that these agents will overly suppress secretion of luteinising hormone and stop ovulation.4

Neurokinin 3 receptor antagonists

Inhibition of the neurokinin 3 receptor is believed to cause a tempered inhibition of gonadotrophin releasing hormone pulsatility without excessive reduction because of the presence of compensatory pathways.4 25 MLE4901 (also named AZD4901) had promising effects on levels of reproductive hormones in 65 women with polycystic ovary syndrome in a phase 2 randomised controlled trial.113 After seven days of treatment with MLE4901 80 mg/day, the area under the luteinising hormone curve was reduced by 52.0% (95% confidence interval 29.6% to 67.3%), total testosterone concentration was reduced by 28.7% (13.9% to 40.9%), and luteinising hormone pulses were reduced by 3.55 pulses/8 hours (2.0 to 5.1).113 MLE4901 was discontinued, however, because of increased levels of transaminases in some patients.114 115 Hepatotoxicity is believed to be specific to MLE4901 and has not been reported with other neurokinin 3 receptor antagonists.115

In a phase 2 randomised controlled trial in 64 women with polycystic ovary syndrome, treatment with the neurokinin 3 receptor antagonist, fezolinetant, for 12 weeks at 60 mg or 180 mg, reduced levels of testosterone by 17% (95% confidence interval −28.7% to −4.6%) and 33% (−45.91% to −20.4%), respectively, compared with 1% (−8.8% to 11.7%) with placebo.116 Levels of luteinising hormone but not follicle stimulating hormone were significantly reduced (P<0.001) in a dose dependent manner, reducing the ratio of luteinising hormone to follicle stimulating hormone.

Preclinical studies have also highlighted the potential metabolic benefits of neurokinin 3 receptor antagonists. In a dihydrotestosterone induced mouse model of polycystic ovary syndrome, treatment with neurokinin 3 receptor antagonists decreased body weight and adiposity.117 No changes in food intake or energy expenditure were seen, although an increased respiratory exchange ratio suggested that neurokinin 3 receptor antagonists cause a shift to a carbohydrate predominant utilisation of fuel.117 These promising observations suggest that neurokinin 3 receptor antagonists fulfil many of the properties of an ideal treatment for polycystic ovary syndrome, and further clinical trials are awaited with interest.

Dynorphin, γ-aminobutyric acid, and anti-müllerian hormone based treatment

When dynorphin binds to kappa opioid receptors, release of kisspeptin onto gonadotrophin releasing hormone neurons is inhibited, and therefore selective kappa receptor agonists with a central action might reduce gonadotrophin releasing hormone pulsatility.4 A new generation of peripherally selective kappa receptor agonists have been developed that might access brain regions, including the infundibular nucleus, by fenestrated capillaries in the median eminence.118 The kappa receptor agonist, difelikefalin, does not seem to cause the centrally mediated side effects of dysphoria and sedation of previous kappa receptor agonists, and has recently been approved in the US for the treatment of moderate-to-severe pruritus in adults undergoing haemodialysis.119 120 In a prenatally androgenised mouse model of polycystic ovary syndrome, difelikefalin reduced serum levels of luteinising hormone and testosterone, restored oestrous cyclicity and ovulation, and reduced overexpression of KISS1 mRNA in the hypothalamic preoptic area.121

Centrally acting GABAA antagonists might also benefit patients with polycystic ovary syndrome. Although weight gain could in part explain the increased incidence of polycystic ovary syndrome in women receiving sodium valproate, this drug also increases levels of GABAA in the central nervous system.122 In contrast, peripheral levels of GABA are reduced in patients with polycystic ovary syndrome,123 and enteral administration of GABAA reduced body mass index and levels of testosterone in a letrozole induced polycystic ovary syndrome model.124 Antagonism of the anti-müllerian hormone pathway might also be therapeutically useful; recent insights into the structural basis for binding of anti-müllerian hormone to anti-müllerian hormone receptor type 2 could facilitate the rational design of anti-müllerian hormone antagonists.125

Targeting key enzymes in steroidogenesis

AKR1C3 functions as the gatekeeper in classical and 11-oxygenated androgen synthesis by mediating enzymatic conversion of androstenedione to testosterone and 11-ketoandrostenedione to 11-ketotestosterone.6 Various AKR1C3 inhibitors have been developed, with mixed results for their antineoplastic effects and ability to inhibit prostaglandin F synthase activity in castration resistant prostate cancer, acute myeloid leukaemia, and oestrogen receptor positive breast cancers.126–128 Although steroidal based inhibitors of AKR1C3 are in development, the therapeutic potential in preclinical models of polycystic ovary syndrome has not been examined.129 Selective inhibition of 11β-HSD1 with BVT.2733 in a rodent model of polycystic ovary syndrome improved insulin resistance, reproductive hormone dysfunction, and polycystic ovarian morphology.37 These observations are encouraging but further preclinical work is needed before the potential therapeutic benefits of AKR1C3 and 11β-HSD1 inhibitors can be tested in patients.

Glucagon-like peptide 1 inhibitors

Glucagon-like peptide 1 (GLP-1) receptor agonists increase glucose dependent insulin secretion, suppress secretion of glucagon, and increase peripheral insulin sensitivity by weight loss (by stimulation of satiety) and suppression of inflammation in adipose tissue.130 Although these properties might be therapeutically attractive in patients with polycystic ovary syndrome, previous randomised controlled trials were largely small, single centre, and of limited duration.131 Nevertheless, in a network meta-analysis of 941 women with polycystic ovary syndrome and overweight or obesity, liraglutide was superior to metformin (mean difference −3.82, 95% confidence interval −4.44 to −3.20) and orlistat (−1.95, −3.74 to −0.16) in reducing body weight.132 Some studies showed that GLP-1 receptor agonists improved menstrual regularity or frequency, and that these improvements in menstrual frequency correlated with reduction in body weight.133 134 GLP-1 receptor agonists have also been shown to lower levels of androgens133 135 and improve markers of cardiovascular risk.136 137 A meta-analysis of eight randomised controlled trials concluded that GLP-1 agonists were more effective than metformin in improving homeostasis model assessment-insulin resistance (standard mean difference −0.40, 95% confidence interval −0.74 to −0.06), abdominal circumference (−0.45, −0.89 to −0.00), and body mass index (−1.02, −1.85 to −0.19), but not in improving menstrual frequency (0.15, –0.24 to 0.54) or serum levels of testosterone (0.64, –0.08 to 1.35).138 Newer longer acting GLP-1 analogues, such as semaglutide or dulaglutide,139 140 or dual GLP-1-glucose dependent insulinotropic polypeptide agonists, such as tirzepatide,141 could provide more therapeutic opportunities, with the potential benefits of greater effects on weight loss, longer duration of action, and improved adherence. Semaglutide, the only GLP-1 agonist currently available in an oral formulation, is being investigated in a clinical trial in adolescent girls with polycystic ovary syndrome and obesity (Treating PCOS With Semaglutide vs Active Lifestyle Intervention (TEAL), NCT03919929). More adequately powered trials with a focus on core outcomes of polycystic ovary syndrome142 are needed to establish whether these new drugs have a role in clinical management.

Sodium-glucose co-transporter inhibitors

Sodium-glucose co-transporter 2 inhibitors reduce reabsorption of glucose in the proximal convoluted tubules of the kidney, promoting excretion of urinary glucose, and also reduce weight and cardiovascular events in other populations.143 Current data in patients with polycystic ovary syndrome are limited to four small randomised controlled trials.144–148 Canagliflozin showed greater improvements in body mass index (P=0.006), basal metabolic rate (P=0.02), and fat mass (P=0.02) than metformin in women with polycystic ovary syndrome, but not in hormonal or metabolic parameters.144 In overweight and obese patients with polycystic ovary syndrome, combined canagliflozin-metformin treatment for three months produced greater reductions than metformin monotherapy in total testosterone (−0.33 v −0.18 ng/mL, P=0.02), area under the curve for glucose (−158.00 v 2.63 mmol/L×min, P=0.02), and the ratio of the area under the curve for insulin and glucose (−2.86 v 0.51, P=0.02),146 but no significant differences were found in menstrual frequency, body mass index, or homeostasis model assessment-insulin resistance between the treatment groups.146

Licogliflozin is a dual inhibitor of sodium-glucose co-transporter 1 (SGLT1) and 2 (SGLT2). Simultaneous SGLT1 and SGLT2 inhibition could provide more effective weight loss because SGLT1 inhibition alone stimulates intestinal secretion of GLP-1.131 In a phase 2 randomised controlled trial of 29 patients, licogliflozin reduced levels of androstenedione by 19%, dehydroepiandrosterone sulphate by 24%, and hyperinsulinaemia by 70% in women with polycystic ovary syndrome.148 The outcome of a recently completed randomised controlled trial of dapagliflozin on insulin resistance and serum levels of androgens in patients with polycystic ovary syndrome (Dapagliflozin Efficacy and Action in PCOS (DEAP), NCT04213677) is awaited with interest. Table 1 summarises the emerging treatments for polycystic ovary syndrome described in this review.

Table 1

Emerging drug treatments for polycystic ovary syndrome

Guidelines

Guidelines on polycystic ovary syndrome vary in their methodological quality, approach to diagnosis, approach to screening for health risks, and recommendations for the use of drug treatments.149 The 2023 update to the international polycystic ovary syndrome guidelines, which uses consensus methodology and clear grading systems for clinical recommendations, has now been released.3 These evidence based guidelines were developed after consultation with international multidisciplinary and consumer bodies to support clinicians and patients in the diagnosis and management of polycystic ovary syndrome and reduce variation in care.

Conclusions

Polycystic ovary syndrome is a common reproductive and metabolic disorder resulting from polygenic and environmental influences. Key pathological changes include neuroendocrine dysregulation, excess production of androgens, insulin resistance, and changes in adipose tissue biology, with variation in dysfunction of these pathways contributing to differences in phenotypic expression and severity of the disease. Advances in genetic understanding, together with new techniques to assess the steroid metabolome, have identified new biological targets, challenged the perception of polycystic ovary syndrome as one entity, and could facilitate an individualised approach to long term cardiometabolic surveillance based on the metabolomic signature. These advances could, for the first time, enable the development of specific drug treatments for the disorder based on an improved understanding of the underlying pathophysiology. Well designed, multicentre, patient centred clinical trials of neurokinin receptor antagonists, kisspeptin based treatments, and repurposed antidiabetic drugs are now needed to investigate new therapeutic options for polycystic ovary syndrome.

Questions for future research

  • Should polycystic ovary syndrome be classified based on the steroid metabolomic signature, and specific treatments developed accordingly?
  • Does the steroid metabolome predict which patients are at increased risk of cardiometabolic disease?
  • Can reduction of 11-oxygenated androgens (eg, by inhibition of aldo-keto reductase type 1C3 (AKR1C3)) improve metabolic risk in patients with polycystic ovary syndrome?
  • Can later phase clinical trials of neurokinin 3 receptor antagonists show improvements in clinical hyperandrogenism and reproductive outcomes?

Polycystic ovary syndrome


Summary

Polycystic ovary syndrome (PCOS) affects 5–18% of women, and is a reproductive, metabolic, and psychological condition with impacts across the lifespan. The cause is complex, and includes genetic and epigenetic susceptibility, hypothalamic and ovarian dysfunction, excess androgen exposure, insulin resistance, and adiposity-related mechanisms. Diagnosis is recommended based on the 2003 Rotterdam criteria and confirmed with two of three criteria: hyperandrogenism (clinical or biochemical), irregular cycles, and polycystic ovary morphology. In adolescents, both the criteria of hyperandrogenism and irregular cycles are needed, and ovarian morphology is not included due to poor specificity. The diagnostic criteria generates four phenotypes, and clinical features are heterogeneous, with manifestations typically arising in childhood and then evolving across adolescent and adult life. Treatment involves a combination of lifestyle alterations and medical management. Lifestyle optimisation includes a healthy balanced diet and regular exercise to prevent excess weight gain, limit PCOS complications and target weight reduction when needed. Medical management options include metformin to improve insulin resistance and metabolic features, combined oral contraceptive pill for menstrual cycle regulation and hyperandrogenism, and if needed, anti-androgens for refractory hyperandrogenism. In this Review, we provide an update on the pathophysiology, diagnosis, and clinical features of PCOS, and discuss the needs and priorities of thoseL with PCOS, including lifestyle, and medical and infertility treatment. Further we discuss the status of international evidence-based guidelines (EBG) and translation, to support patient self management, healthcare provision, and to set research priorities.

Source: Lancet

No rush to label teens as having PCOS, says expert


Dr Veronique Celine Viardot-Foucault.

Diagnosis of polycystic ovary syndrome (PCOS) is challenging, and there should be no rush to label an adolescent as having the condition before a thorough evaluation of symptoms, according to a leading endocrinologist who was speaking at the RCOG World Congress 2018 in Singapore.

“Common features of PCOS such as hirsutism, acne, and obesity are often present in otherwise ‘normal’ adolescents,” said Dr Veronique Celine Viardot-Foucault from the KK Women’s and Children’s Hospital, Singapore, adding that these features may not necessarily be indicative of PCOS.

Appropriate diagnosis of PCOS in adolescents should involve careful evaluation of symptoms such as menstrual irregularities, hyperandrogenism, and polycystic ovarian morphology, she said. Menstrual irregularities—including secondary amenorrhoea and oligomenorrhoea in girls beyond 2 years after menarche, or primary amenorrhoea in those who have completed puberty—may be indicative of androgen excess. [Horm Res Paediatr 2017;88:371-395]

As symptom such as acne is common in adolescence and usually transient, it may not be indicative of hyperandrogenism, said Viardot-Foucault. Also, isolated cases of acne and/or alopecia should not be considered as diagnostic criteria for PCOS in adolescence, but moderate or severe inflammatory acne that is unresponsive to topical therapy may require investigation of androgen excess. [Horm Res Paediatr 2017;88:371-395]

Another feature commonly seen with PCOS is hirsutism, which can be evaluated using the modified Ferriman–Gallwey (FG) scoring system. “However, the FG scoring system is not applicable to younger, perimenarchal patients [younger than 15 years old],” she advised, pointing out that biochemical evidence of hyperandrogenism is preferred in this group.

As there is no clear cut-off of testosterone levels for adolescents, biochemical hyperandrogenism should be defined based on the methodology used, informed Viardot-Foucault. “Ideally, to establish the existence of androgen excess, assaying for free testosterone levels is the gold standard as it is more sensitive than measuring the total testosterone levels,” she said. “But a downside of this is that it requires equilibrium dialysis techniques which are costly and not widely available.”

However, most commercial laboratories use direct analogue radio-immunoassay, which is notoriously inaccurate for measuring free testosterone, cautioned Viardot-Foucault. “If uncertain regarding the quality of the free testosterone assay, it is preferable to rely on calculated free testosterone, which has a good concordance and correlation with free testosterone levels measured by equilibrium dialysis methods,” she suggested. [J Clin Endocrinol Metab 1999;84:3666-3672]

Also, the value of measuring other androgens besides free testosterone in patients with PCOS is relatively low, although increased levels of dehydroepiandrosterone sulphate (DHEAS) have been observed in 30–35 percent of PCOS patients. [Ann N Y Acad Sci 2006;1092:130-137]

“Transabdominal pelvic ultrasound has a lower diagnostic accuracy,” said Viardot-Foucault. “The presence of polycystic ovarian morphology [on ultrasound] in an adolescent who does not have hyperandrogenism or oligo-anovulation does not indicate a diagnosis of PCOS.”

When menstrual irregularities are concerned, the first-line treatment should be cyclical progestogens when contraception is not required and there are no signs of hyperandrogenism, according to Viardot-Foucault. If there is clinical hyperandrogenism or a need for contraception in those sexually active, third-generation oral contraceptives such as ethinyl estradiol 30 µg can be considered.

“There is room for local treatment of hirsutism such as laser [hair removal, but only for patients beyond] 16 years old and [who are] at least 2 years post-menarche,” she said. “If there are metabolic complications, [patients should be referred] to the endocrinologist.”

Links Between Fatty Liver Disease and Polycystic Ovary Syndrome


The possible link between these two conditions may have a number of implications for the diagnosis and treatment of various diseases among women.

A growing body of research indicates that non-alcoholic fatty liver disease (NAFLD) and polycystic ovary syndrome (PCOS) may be related and, due to shared mechanisms, may increase the risk of type 2 diabetes (T2D) and other cardiometabolic complications. The findings may have a number of implications for the diagnosis and treatment of various diseases.

Although the etiology of PCOS, one of the most common endocrine disorders in women of reproductive age, is uncertain, obesity and insulin resistance are frequently present in affected individuals, and they play a role in its development. The condition affects 5% to 18% of this population depending on the diagnostic criteria used, and clinical features consist of menstrual dysfunction, infertility, hirsutism, acne, and alopecia.

1,2

Similarly, obesity and insulin resistance appear to contribute to the pathogenesis of NAFLD, which is characterized by increased accumulation of fat in the liver in the absence of significant alcohol consumption. NAFLD includes a range of diseases, from simple steatosis to non-alcoholic steatohepatitis to cirrhosis, and it has an estimated worldwide prevalence of 6.3% to 33.0%; however, if obesity or T2D is involved, the prevalence of NAFLD rises to approximately 75%.

2

A connection, but few reasons as to why

NAFLD and PCOS occur together more frequently than expected, in many cases simply by chance alone. Studies have consistently shown that NAFLD rates are elevated in young women with PCOS, independent of weight and metabolic syndrome features, and limited data suggest that these women have better odds of experiencing more severe forms of NAFLD.

3

In one recent study, by Evangeline Vassilatou, MD, PhD, an endocrinologist at the Attikon University General Hospital, in Athens, Greece, NAFLD was detected in 71 of 110 (64.5%) overweight and obese (yet otherwise apparently healthy) premenopausal women, and women with NAFLD were more often diagnosed with PCOS than women without NAFLD (43.7% versus 23.1%, respectively).

4

It’s currently unclear how the two conditions may influence each other: Are they a consequence of shared risk factors? Or does one condition contribute to the other independently of these factors? Accumulating evidence indicates that NAFLD may exacerbate insulin resistance and release multiple pro-inflammatory, pro-coagulant, and pro-fibrogenic mediators that could contribute to the pathophysiology of PCOS.

3

On the other hand, insulin resistance and androgen excess are the main characteristics of PCOS that could increase the risk of developing NAFLD.

1

To examine the most relevant factors associated with NAFLD in women with PCOS, investigators recently conducted a cross-sectional study including 600 Caucasian women diagnosed with PCOS between May 2008 and May 2013 and 125 women matched for body mass index.

5

NAFLD, which was diagnosed by an NAFLD liver fat score, was identified in 50.6% of women with PCOS, compared with 34.0% of controls. Women with PCOS had higher readings for waist circumference, lipid accumulation product (a combination of waist circumference and fasting triglyceride levels), insulin resistance, total cholesterol, and triglycerides than controls. Upon further analysis, insulin resistance and lipid accumulation product were independently associated with NAFLD.

5

“This study provided further evidence that PCOS women are more prone to develop NAFLD compared with non-PCOS premenopausal women, and that features of the metabolic dysfunction that characterize PCOS are the main factors implicated in the development of NAFLD in these patients,” says Dr. Vassilatou, who wasn’t involved with the study. “Some research also suggests that androgen excess, which is a key feature of PCOS and is interrelated to insulin resistance, may be an additional contributing factor for the development of NAFLD in PCOS.”

Just scratching the surface

Dr. Vassilatou says that more research is needed to understand the role of androgens, if any, in the pathophysiology of NAFLD in women with PCOS. Also, long-term follow-up studies are necessary to reveal the range of liver-related outcomes in women with PCOS and to determine the natural history of NAFLD in these women—for example, how often it progresses from its early stages to severe liver disease. It will also be important to investigate whether obese patients with PCOS and NAFLD present more frequently with an advanced stage of liver disease, as reported in a few studies.

Although additional studies are needed to clarify the association between PCOS and NAFLD, accumulating data over the past decade indicate that clinicians should at least be aware of this connection. “Thus, these women should be screened for NAFLD, particularly obese patients with features of the metabolic syndrome. Conversely, premenopausal women with NAFLD should be screened for PCOS,” suggests Dr. Vassilatou.

Despite the need for greater screening, more work is necessary to identify the most appropriate methods, which could include ultrasound, liver function tests, and algorithms such as the NAFLD liver fat score. Furthermore, because there’s no medical therapy of proven benefit for treating NAFLD, well-designed interventional studies—with lifestyle modifications and/or the use of medical therapy targeting insulin resistance, impaired glucose tolerance, and dyslipidemia—are needed to determine the optimal management of affected patients.

To get a sense of where we are now, diet, weight loss, and exercise are currently the cornerstone of therapy and are often combined with insulin sensitizers, hypolipidemic drugs, and hepatoprotective agents, like antioxidants.

1

Published: March 17, 2017

7 Subtle Signs You Could Have PCOS


Experts believe that more than half of women with polycystic ovary syndrome don’t even realize they have it.
PCOS_Feature

If you’ve skipped a period or two (and know you’re not pregnant) and have been breaking out like you’re a teenager again, it’s easy to chalk it all up to stress. But something more serious may be going on, such as polycystic ovary syndrome (PCOS), a stealth health issue caused by a hormonal imbalance and marked by a series of small cysts on the ovaries.

Five to 10 percent of women of childbearing age are affected by the condition, but less than half of women are diagnosed, according to the PCOS Foundation. That means millions of women have PCOS and don’t even know it. To shed some light on this silent disease, here are the most common not-so-obvious signs of the hormonal disorder. If you’re experiencing any of these symptoms, bring them up with your gynecologist or general practitioner and get them evaluated.

1. Your cycle is all over the place.

Unpredictable menstrual cycles or skipping several periods are one of the hallmarks of PCOS. “Our menstrual cycle is like a vital sign,” says Maryam Siddiqui, MD, assistant professor of obstetrics-gynecology at the University of Chicago Medicine. “It tells us if our metabolism is in a good state; if you’re too thin, overweight, or stressed, that can throw your cycles off. Having irregular periods or more likely, skipping multiple periods could be a sign of a hormonal imbalance like PCOS.” Menstrual irregularities like these should raise a red flag and warrant a doctor’s attention.

2. You’re growing hair in unexpected places.

With PCOS, the ovaries produce excessive amounts of a type of hormones called androgens, which stimulate hair growth. We’re not talking about the hairs on your head. “You’ll get hair growth in funny places—around the nipples, on your chest, the inside of your thighs, and your belly,” says Siddiqui. “Places were women don’t typically have a lot of hair growth.”

3. You’re breaking out.

Those same high levels of androgens also trigger acne. The hormones boost sebum production, and the combo of excess oil and old skin tissue plugs pores. To add insult to injury, bacteria that flourish on sebum increase, triggering inflammation.

4. There’s a dark “ring” around your neck.

You might blame it on a cheap necklace leaving a ring of residue on your skin at first, but PCOS can cause a stubborn darkening of the skin around the back of your neck. “It’s a velvety, dark discoloration that doesn’t wash off,” explains Siddiqui. The pigmentation and skin texture changes can also appear under your arms and around the vulva.

5. Your belly is getting bigger and you don’t know why.

Unexplained, persistent weight gain, particularly around the abdomen, is a sign of the hormonal disorder. Although it’s not fully understood why weight gain is a symptom, insulin resistance appears to play a role. “With PCOS, you can have trouble metabolizing blood sugar, known as insulin resistance,” explains Siddiqui. “When you have insulin resistance, your pancreas has to work really hard and make a lot of insulin just to lower your blood sugar. That is linked to weight gain and central obesity.” (Women with PCOS are at higher risk for developing diabetes.)

6. Those annoying skin tags keep popping up.

Although it’s not fully understood why, those flesh-colored nubs of excess skin tend to crop up around the neck area and under the arms of women with PCOS, according to the U.S. Department of Health and Human Services. It’s worth noting, though, that skin tags, which are benign and can be triggered by friction, are also common in people who don’t have PCOS, so don’t automatically freak out if you have them.

7. You’re having trouble getting pregnant.

The hormonal imbalance interferes with the body’s ability to ovulate normally, which is essential for pregnancy to occur. So it’s no surprise that PCOS is one of the most common causes of infertility. In fact, it’s responsible for 70 percent of infertility problems in women who have trouble ovulating, according to the PCOS Foundation.

Polycystic Ovary Syndrome Might Start in the Brain, Not the Ovaries


Finally, some answers.

A new study has found evidence that the common and debilitating reproductive condition, polycystic ovary syndrome, could start in the brain, not the ovaries, as researchers have long assumed.

If verified, the research could change the way we think about the painful and severely misunderstood condition, which affects at least one in 10 women worldwide.

Anyone who has polycystic ovary syndrome (PCOS) – or knows someone with the condition – will be aware of how incredibly frustrating it can be.

Thanks to the variety of symptoms it can cause – from weight gain, large ovarian cysts, difficulty ovulating, acne, facial hair, depression, and agonising and heavy periods – it can take women years to get diagnosed.

Even then, there’s very little in the way of treatment options. Most women are simply told to go on the pill or take other hormonal medications to manage their individual symptoms, but not the underlying cause.

In the long-term, PCOS can lead to metabolic disorders, such as type 2 diabetes, cardiovascular disease, and hormonal dysfunction, including infertility. In fact, PCOS is the cause of more than 75 percent of anovulatory infertility, which is infertility caused by a woman not ovulating.

And yet, despite the severity of the condition, researchers still don’t understand how PCOS arises and how we can treat it.

Now, researchers led by the University of New South Wales in Australia have shown that mice without receptors for androgens – a group of steroid hormones commonly associated with males, such as testosterone – in their brains can’t develop PCOS. But if the androgen receptors in the ovaries are removed, the condition can still arise

 Seeing as mouse and human reproductive systems share many similarities, it’s compelling early evidence that doctors and scientists might have been focussing on the wrong piece of the puzzle all along.

“For the first time we have a new direction of where we should be looking to try and develop treatments that will treat the cause of PCOS, the androgen excess in the ovary but also in the brain,” said lead researcher Kirsty Walters in an emailed press release.

Before this, researchers knew that an increase in androgens, known as hyperandrogenism, was linked to the onset of PCOS. But exactly how and where these androgens act in the body was poorly understood.

“Hyperandrogenism is the most consistent PCOS characteristic; however, it is unclear whether androgen excess, which is treatable, is a cause or a consequence of PCOS,” the researchers write in their paper.

To get a better idea, the researchers took four groups of mice:

  • a control group of normal mice
  • a group of mice genetically engineered to have no androgen receptors (ARs) anywhere in their bodies
  • a group that had been engineered to have no ARs in just their brains
  • a final group that only had ARs missing from their ovaries.

The team then used a high dose of androgen to attempt to trigger PCOS in all four groups of mice.

While the control group developed PCOS as they expected, the mice missing ARs entirely, or just missing them from their brains, didn’t get the condition.

Interestingly, the mice that were only missing ARs from their ovaries still went on to develop PCOS, although at a lower rate than the control group. That means androgens acting on the ovaries can’t be the sole cause of PCOS.

The result suggests two important things: researchers were right about an excess of androgens triggering the condition; and the action of androgens on the brain is important to the development of PCOS.

That means if we can find a way to stop those excess androgens in the brain, it could signal a new way to treat PCOS.

“These data highlight the previously overlooked importance of extraovarian [outside the ovary] neuroendocrine androgen action in the origins of PCOS,” the researchers explain.

To be clear, this study has only looked at mice so far, and the results need to be replicated in humans before we can get an idea of whether the same thing is happening in our own reproductive systems.

But this is a big deal because, until now, the focus when looking for effective treatments and preventions has been on the ovaries – and we haven’t had much luck.

The new study, though it’s still early days, gives researchers a new target to look into, and it could hopefully lead to new, more effective treatments for people with the condition.

Source:http://www.sciencealert.com

Omega-3 fatty acid supplementation does not affect insulin resistance in PCOS


Women with polycystic ovary syndrome likely receive no benefit from daily omega-3 fatty acid supplementation, according to a meta-analysis of three randomized controlled trials.

“Reducing the levels of serum insulin and increasing insulin sensitivity are considered to be of paramount importance for therapeutic targets in PCOS,”Alirez Sadeghi, of the department of cellular and molecular nutrition at the School of Nutritional Sciences and Dietetics at Tehran University of Medical Sciences, Iran, and colleagues wrote. “Omega-3 fatty acids may lead to insulin sensitivity by producing and secreting anti-inflammatory adipokines, such as adiponectin, and also through reducing inflammation and proinflammatory cytokines. Although it is said that omega-3 fatty acids have positive effects on insulin resistance, various studies have indicated contradictory results.”

Sadeghi and colleagues analyzed data from three studies that measured the association between oral omega-3 supplementation and insulin resistance inwomen with PCOS. Studies were conducted in Australia, Iran and the United States, and included 72 women with PCOS and 73 controls. All studies were double blind and published between 2009 and 2012 with follow-up between 6 and 8 weeks. In all three studies, PCOS groups received 1.2 g to 3.6 g daily omega-3 supplementation containing eicosapentaenoic acid and docosahexaenoic acid (median, 2.7 g); control groups received an oral placebo (olive oil, soybean oil or other placebo). Researchers assessed plasma fatty acid composition in one study; participants maintained their usual diet during intervention in two of the studies; daily energy intake was assessed at baseline and end of intervention in two studies.

In women with PCOS, researchers found that omega-3 fatty acid supplementation did not affect insulin plasma level (mean difference: 6.018; 95% CI, –3.347 to 15.382) or homeostasis model assessment of insulin resistance (HOMA-IR; mean difference: 0.276; 95% CI, –1.428 to 1.981), with high heterogeneity observed for both. The researchers noted that samples sizes in the included studies were low, and further, high-quality randomized controlled trials are needed to validate the findings. – by Regina Schaffer

Metabolic syndrome rate, severity in PCOS reduced following bariatric surgery


Bariatric surgery can improve cardiometabolic health in women with polycystic ovary syndrome and obesity, according to findings of a retrospective cohort study presented here.

McAnto Antony, MBBS, a second-year resident at Medstar Washington Hospital Center in Washington, D.C., and colleagues evaluated data from Medstar facilities on 19 women with PCOS (mean age, 18.4 years; 53% black; 41% white; 6% Asian) who had undergone a bariatric surgical procedure. The most common procedure was gastric sleeve, followed by lap band with fewer Roux-en-Y gastric bypass, according to Antony. Researchers compared BMI, blood pressure, HbA1c, and triglyceride and HDL levels before and at least 6 months after surgery (mean time between surgery and follow-up, 7.9 months).

McAnto Antony

McAnto Antony

Compared with presurgical values, postsurgical reductions were observed in body weight (mean, 271 kg vs. 205.4 kg; P< .0001), BMI (mean, 45.9 kg/m2 vs. 35 kg/m2; P < .0001), systolic BP (mean, 133.4 mm Hg vs. 119.5 mm Hg; P = .0002), diastolic BP (mean, 81.9 mm Hg vs. 73.1 mm Hg; P= .007), triglycerides (mean, 143.2 mg/dL vs. 111.5 mg/dL; P = .04) and HbA1c (mean 6.6% vs. 5.8%; P = .03); mean HDL level increased (44.8 mg/dL vs. 52.5 mg/dL; P = 0.04). Before surgery, participants had a mean 2.7 components of metabolic syndrome on average, which decreased to 1.9 after their procedure (P < .01). Forty-seven percent of participants had at least three of the five components of metabolic syndrome, meeting criteria for the condition, before surgery. Following surgery, prevalence dropped to 21%.

“Bariatric surgery is definitely an option in the obese woman with PCOS to reduce her risk of developing cardiovascular disease in the future,” Antony told Endocrine Today. “ – by Jill Rollet

Fertility in women with PCOS improves with weight loss, exercise


Ovulation in women with polycystic ovary syndrome improves with weight loss and exercise, according to recent study findings published in The Journal of Clinical Endocrinology & Metabolism.

“The findings confirm what we have long suspected — that exercise and a healthy diet can improve fertility in women who have PCOS,” Richard S. Legro, MD, vice chair of research and professor of obstetrics and gynecology and public health sciences at Penn State College of Medicine, said in a press release. “Making preconception lifestyle changes is beneficial, either alone or in combination with other pretreatment options.”

Richard Legro

Richard S. Legro

Legro and colleagues evaluated 149 women aged 18 to 40 years with overweight or obesity and infertility due to PCOS to determine the efficacy of preconception intervention on reproductive and metabolic abnormalities. Main outcome measures were weight, ovulation and live birth.

During the preconception intervention, women were randomly assigned to 16 weeks of one of the three following treatments: continuous oral contraceptive pills (pills; n = 49); lifestyle modification consisting of caloric restriction with meal replacements, weight-loss medication and increased physical activity to promote a 7% weight loss (lifestyle; n = 50); or combined treatment with both oral contraceptive pills and lifestyle modification (combined; n = 50).

Participants underwent standardized ovulation induction with clomiphene citrate, as well as timed intercourse for four cycles after the intervention, and pregnancies were followed at trimester visits until delivery.

Compared with the pill group, the lifestyle and combined groups achieved weight loss (P < .0001) and a decrease in waist circumference (P = .03) after intervention.

Compared with baseline, there was a significant increase in metabolic syndrome within the pill group (OR = 2.47; 95% CI, 1.42-4.27); no changes were found with the lifestyle or combined groups.

The combined group achieved ovulation more commonly than the pill group (P < .05). The rate of live births nearly reached statistical significance after combining the lifestyle and combined groups compared with the pill group (P = .05). Among patients who ovulated, fecundity was higher among the lifestyle group compared with the pill group (P = .04).

“The research indicates preconception weight loss and exercise improve women’s reproductive and metabolic health,” Legro said. “In contrast, using oral contraceptives alone may worsen the metabolic profile without improving ovulation. Lifestyle change is an important part of any fertility treatment approach for women with PCOS who are overweight or obese.” – by Amber Cox

CVD risk higher for women aged at least 30 years with PCOS


Among women with polycystic ovary syndrome, those aged 30 years or older are potentially at higher risk for developing early atherosclerosis, based on elevated lipid levels, lipid ratios and hypertension rates, compared with younger women with or without polycystic ovary syndrome, according to research in the International Journal of Endocrinology.

Subclinical cardiovascular disease was more prevalent in women aged at least 30 years with PCOS regardless of BMI, according to researchers.

“If we consider that women with PCOS are exposed to risk factors for CVD early in life, the diagnosis of subclinical atherosclerosis in this population would be of importance,” the researchers wrote.

Djuro Macut, MD, of the University of Belgrade, Serbia, and colleagues compared data from 100 women with PCOS (26.32 ± 5.26 years; BMI, 24.98 ± 6.38 kg/m²) with 50 healthy women (27.96 ± 5.6 years; BMI, 24.66 ± 6.74 kg/m²). Baseline blood samples collected after 12 hours of fasting during the follicular phase of the menstrual cycle, or randomly in the case of amenorrhea, were analyzed for levels of total cholesterol, HDL cholesterol, LDL cholesterol, triglycerides, apolipoprotein A, ApoB, glucose, insulin, total testosterone, sex hormone-binding globulin, androstenedione and dehydroepiandrosterone sulfate.

Patients aged at least 30 years with PCOS (n = 24) had higher BMI (P < .001) waist-to-hip ratio (P = .008), systolic blood pressure (P < .001), diastolic BP (P < .001), all lipids and their ratios, and ApoB (P = .014) than younger women with PCOS (n = 76), according to researchers. After adjustment for BMI, significant differences remained for systolic BP (P = .003), diastolic BP (P = .003), triglycerides (P = .05), insulin (P = .028) and free androgen index (P = .043).

In the older subgroups, women with PCOS had a significantly higher prevalence of hypertension than women without PCOS (n = 18; 61% vs. 17%, P = .003).

“A more proper assessment of the clinical phenotypes and use of specific metabolic indicators could be a valuable tool for the evaluation of [CV] potential and outcomes in future randomized studies on women with PCOS,” the researchers wrote. – by Regina Schaffer