ArticlePDF AvailableLiterature Review

Polycystic ovarian syndrome

Authors:

Abstract and Figures

Polycystic ovarian syndrome (PCOS) is a "multispeciality" disorder suspected in patients with irregular menses and clinical signs of hyperandrogenism such as acne, seborrhoea, hirsutism, irregular menses, infertility, and alopecia. Recently, PCOS has been associated with the metabolic syndrome. Patients may develop obesity, insulin resistance, acanthosis nigricans, Type 2 diabetes, dyslipidemias, hypertension, non-alcoholic liver disease, and obstructive sleep apnoea. Good clinical examination with hematological and radiological investigations is required for clinical evaluation. Management is a combined effort involving a dermatologist, endocrinologist, gynecologist, and nutritionist. Morbidity in addition includes a low "self image" and poor quality of life. Long term medications and lifestyle changes are essential for a successful outcome. This article focuses on understanding the normal and abnormal endocrine functions involved in the pathogenesis of PCOS. Proper diagnosis and management of the patient is discussed.
Content may be subject to copyright.
Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3310
Polycystic ovarian syndrome
Nina Madnani, Kaleem Khan, Phulrenu Chauhan
1
, Girish Parmar
1
ABSTRACT
Polycystic ovarian syndrome (PCOS) is a “multispeciality” disorder suspected in patients
with irregular menses and clinical signs of hyperandrogenism such as acne, seborrhoea,
hirsutism, irregular menses, infertility, and alopecia. Recently, PCOS has been associated
with the metabolic syndrome. Patients may develop obesity, insulin resistance, acanthosis
nigricans, Type 2 diabetes, dyslipidemias, hypertension, non‑alcoholic liver disease, and
obstructive sleep apnoea. Good clinical examination with hematological and radiological
investigations is required for clinical evaluation. Management is a combined effort involving
a dermatologist, endocrinologist, gynecologist, and nutritionist. Morbidity in addition includes
a low “self image” and poor quality of life. Long term medications and lifestyle changes are
essential for a successful outcome. This article focuses on understanding the normal and
abnormal endocrine functions involved in the pathogenesis of PCOS. Proper diagnosis and
management of the patient is discussed.
Key words: Hyperandrogenemia, hyperandrogenism, insulin resistance, metabolic syndrome,
polycystic ovary
Departments of Dermatology,
and 1Endocrinology,
P. D. Hinduja National
Hospital, Mahim, Mumbai,
India
Address for correspondence:
Dr. Nina Madnani,
Department of Dermatology,
P. D. Hinduja National
Hospital, Veer Savarkar
Marg, Mahim,
Mumbai ‑ 400 016, India.
E‑mail:
ninamadnani@gmail.com
How to cite this article: Madnani N, Khan K, Chauhan P, Parmar G. Polycystic ovarian syndrome. Indian J Dermatol Venereol Leprol
2013;79:310‑21.
Received: October, 2012. Accepted: December, 2012. Source of Support: Nil. Conict of Interest: None declared.
INTRODUCTION
In 1935, Stein and Leventhal reported a series of 7
women who presented with oligo/amenorrhoea,
hirsutism, obesity, infertility, and bilateral polycystic
ovaries (Stein‑Leventhal syndrome).[1] Bilateral
wedge resection of the enlarged ovaries was
therapeutic in normalizing the menses and fertility
of the women studied. They concluded that there
was a primary ovarian defect, and the disorder was
known as polycystic ovarian disease (PCOD). Today,
PCOD has been associated with various metabolic
disorders, and is now known as polycystic ovarian
syndrome (PCOS). Following terminologies are
important for better understanding of the clinical
syndrome:
• Hyperandrogenemia is the presence of
abnormally high amounts of androgens
detectable in the blood circulation.
• Hyperandrogenism is the clinical manifestation
of excessive androgen production/secretion,
characterized by oligo/amenorrhoea, infertility,
acne, seborrhoea, hirsutism, and alopecia.
• Cutaneous hyperandrogenism is considered
when there is no documented evidence of
hyperandrogenemia but the clinical signs of
hyperandrogenism are present.
• Hypertrichosis is the presence of excessive hair
in non‑androgen dependent areas.
• Hirsutism is the presence of excess and coarse
terminal hairs in a male‑pattern distribution,
such as upper lip, chin, cheeks, shoulders,
abdomen, lower back and inner thighs.
• Polycysticovariandisease(PCOD)wasdescribed
by Stein and Leventhal[1] in women with oligo/
amenorrhoea, hirsutism, obesity, infertility
and bilateral polycystic ovaries, managed with
wedge ovarian resection.
• Polycystic ovarian syndrome (PCOS) is
PCOD+ metabolic disease.
PCOS is a common disorder with an
incidence varying from 5% to 10%. It is
Review
Article
Access this article online
Quick Response Code: Website:
www.ijdvl.com
DOI:
10.4103/0378-6323.110759
PMID:
*****
Madnani, et al. Polycystic ovarian syndrome
311Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3
observed in women of child‑bearing age
across all cultures, and ethnicities.[2] A
prospective study of Indian adolescents
reported an incidence of 9.13%.[3]
The common features of PCOS are
irregular or anovulatory cycles with signs
of hyperandrogenism like acne, seborrhoea,
hirsutism, alopecia, frank virilization, and
with polycystic ovaries on pelvic sonography.
Recently, PCOS has been associated
with obesity, insulin‑resistance (IR) and
a risk of developing Type 2 diabetes
mellitus (T2DM).[4] The metabolic and
reproductive abnormalities have predisposed
women to develop infertility and endometrial
cancer, necessitating early diagnosis
and appropriate treatment.[5] A better
understanding of the complexities involved
in PCOS necessitates the understanding of
what is “normal ovulation”.
• Anormalovulatorycycleismaintainedduetoa
complex interplay of hormones secreted from the
pituitary, hypothalamus, adrenals, and ovaries.
Up‑regulation or down‑regulation of these
hormones is carried out by an efficient feed‑back
mechanism to maintain equilibrium [Figure 1].
The hypothalamus secretes gonadotropin
releasing hormone (GnRH) in a fixed, pulsatile
manner. This acts upon the cells of the anterior
pituitary to secrete two important hormones,
the follicle stimulating hormone (FSH) and the
leutinising hormone (LH), both targeting the
follicles in the outer cortex of the ovary which
are in various stages of development. FSH
secretion is regulated by a negative feedback
from estradiol, inhibin and progesterone. The
inner lining of the ovarian follicles is composed
of granulosa cells and theca cells. Receptors for
FSH are exclusively present on the granulosa
cells. Under the influence of FSH, the follicles
are recruited, but only one developing follicle
undergoes maturation to become the dominant
or graffian follicle. Interestingly, early stages of
follicular development up to 2 to 5 mm seem
to be independent of the need for FSH. FSH
is also essential for induction of aromatase
activitywithinthegranulosacells.LHreceptors
are expressed by the theca cells of all follicles
and the granulosa cells of large pre‑ovulatory
follicles. It is the stimulation of the latter
receptors by the LH surge seen in mid‑cycle,
which is responsible for ovulation. Estradiol has
a positive feedback response for LH release at
thelevelof pituitary.Also, LHplaysa vitalrole
in androgen production by the theca cells.
OVARIAN STEROIDOGENESIS
Cholesterol, the precursor for steroid synthesis in the
ovaries, undergoes a series of conversions to form
androstenedione (enzymes 3β‑HSD, 17α‑hydroxylase
and 17, 20–lyase mediate this conversion).
Androstenedione gets converted to testosterone (by
17β‑HSD)andtoestrone(byaromatase)inthegranulosa
cells [Figure 2]. The ovaries produce 25% of circulating
Figure 1: Feedback mechanism for a normal menstrual cycle
+\SRWKDODPXV
3LWXLWDU\
2YDULDQ
IROOLFOH
*Q5+
)6+
/+
(VWUDGLRO
3URJHVWHURQH
Figure 2: Ovarian steroidogenesis: Granulosa cells form the inner
lining of the ovarian follicle. The thecal cells abut them and form
the outer layer. Granulosa cells are dependent on the diffusing
androstenedione from the theca cells for substrate as they are not
in direct connection with the circulation (Two cell hypothesis). All
steroid hormones are derived from cholesterol. Its entry into the
mitochondria is mediated by StAR (Steroidogenic acute regulatory
protein) and is the rate limiting step in ovarian steroidogenesis
3 β HSD: Hydroxysteroid dehydrogenenase
&KROHVWHURO 3UHJHQRORQH
ȕ+6'
3URJHVWHURQH
ĮK\GUR[\ODVH
2+SURJHVWHURQH
O\DVH$URPDWDVH
$QGURVWHQHGLRQH (VWURQH
ȕ+6' 
ȕ+6'
7HVWRVWHURQH (VWUDGLRO
7KHFDFHOO *UDQXORVDFHOO
.H\ )6+)6+5HFHSWRU
/+ /+5HFHSWRU
Madnani, et al. Polycystic ovarian syndrome
Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3312
testosterone,whichisdependentonLHsecretedbythe
anterior pituitary. The ovaries also secrete 50% of the
androstenedione and 20% of dehydroepiandrosterone
(DHEA)[Table1].Testosteroneisusedasamarkerof
ovarian androgen secretion.
EXTRA OVARIAN STEROIDOGENESIS
The adrenals synthesize androgens from cholesterol,
under the influence of adreno corticotropic hormone
(ACTH) (secreted by the anterior pituitary), through
multiple steps. The adrenal androgens consist of all
the dehydroepiandrosterone sulphate (DHEAS) and
80% of the DHEA. The adrenals also secrete 50% of
androstenedione and 25% of circulating testosterone.
DHEAS and 11‑androstenedione are not secreted
by the ovaries, and therefore are used as markers
of adrenal androgen secretion. Both prolactin and
estrogen can affect adrenal androgen production.
The peripheral tissues like the skin, liver, and
adipose tissue also take part in androgen synthesis by
converting the weaker androgens to the more potent
ones. Androgen receptor‑activation takes place only
withtestosteroneanddihydrotestosterone(DHT).25%
of the circulating testosterone is produced individually
by the ovaries and adrenals. The remaining 50% by
the peripheral conversion of androstenedione.[6] Only
1% of the androgens produced circulates freely. Of the
remaining 99%, 80% is bound to sex hormone binding
globulin(SHBG),and18%toalbumin.IfSHBGlevels
are reduced, the amount of free circulating testosterone
increases. DHEAS, DHEA and androstenedione are
bound to albumin and freely available to tissues.
There is no regulatory feedback mechanism in both
the adrenals and ovaries in response to androgen
production.
ROLE OF INSULIN
Insulin plays an important role in the normal ovarian
cycle in the following ways: It enhances the amplitude of
LHpulsesatthelevelofpituitary,butnottheirfrequency.
It modulates the LH‑mediated androstenedione
production in theca cells and FSH‑mediated estradiol
production in granulosa cells, probably by increasing
ovarian cytochrome P457 enzyme activity. In the liver,
increased insulin levels inhibit production of sex
hormonebindingglobulin(SHBG).It inhibits hepatic
production of IGFBP‑1 which results in increased levels
of circulating IGF‑1 (Insulin like growth factor 1) levels.
It binds to the IGF‑1 receptors on the ovary and directly
stimulates androgen production.[7]
EVOLUTION OF POLYCYSTIC OVARIAN SYNDROME
The key steps in the evolution of PCOS are enumerated
below, but not necessarily in chronological order.
Pituitary dysfunction
There is an increase in the frequency and amplitude
ofGnRHpulsewhichresultsinelevatedlevelsofLH.
However, whether this alteration in LH secretion is
due to an intrinsic defect of the hypothalamus or due
to external factors such as loss of negative feedback
mechanism from the ovary, or increase in insulin
levels, remains an unresolved debate. Excessive
secretion of LH has traditionally been considered as
ahallmarkofclassicPCOS.AnLH/FSHratioofmore
than 2 especially in obese women is significant.
Enhanced ovarian androgen production
AbnormalLHinfluenceonthethecalcellsoftheovaries
results in overproduction of androgens. However,
all patients of PCOS do not have elevated levels of
LH but still have elevated levels of androgens. This
can be explained on the findings that ovarian thecal
cells of women with PCOS have elevated CYP17 gene
expression. The encoded enzyme (P450c17) is capable of
both, 17‑hydroxylation and 17, 20‑layse functions which
is a key step in ovarian steroidogenesis.[8] Neelson et al.,
in 2001 showed that the p450c17 and 3β‑HSDenzyme
activity is disproportionately higher in the thecal cells
of PCOS women as compared to normal women. So, the
enhanced production of testosterone precursors is the
probably the cause of elevated androgen levels.
Development of insulin resistance
Women with PCOS have decreased sensitivity to
circulating insulin irrespective of weight (obesity),
thus suggesting that this insulin insensitivity is
intrinsic to PCOS.[9] This has been attributed to
a defect in phosphorylation of tyrosine kinase
in the insulin receptor. Normally, tyrosine
autophosphorylation increases the insulin ‑ mediated
Table 1: Source of circulating hormones (approximate
concentration)
Site Testosterone
(%)
DHEA
(%)
Androstenedione
(%)
DHEAS
(%)
Ovaries 20 20 50 -
Adrenals 25 80 50 100
DHEAS: Dehydroepiandrosterone sulfate, DHEA-Dehydroepiandrosterone
sulfate
Madnani, et al. Polycystic ovarian syndrome
313Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3
metabolic changes within muscle and adipose cells.
Serine phosphorylation inhibits this action. Also,
serine phosphorylation has a stimulatory effect on
17,20‑lyase enzyme activity in the ovaries which is
responsible for production of androstenedione. Up
to 50% of women with PCOS have a genetic defect
coding for serine phosphorylation of insulin receptors.
The resulting increased insulin levels, directly and
indirectly, increase androgen production.[10] So
conversely, a reduction in serum insulin levels should
result in reduction in androgen levels.
ETIOPATHOGENESIS
Although the exact etiology is unknown, PCOS is a
multi‑factorial disease with a strong genetic influence.
Various animal models have been used to understand
the pathophysiology of PCOS. Each model has its own
limitation but rodents are the most widely used.[11]
Others include mice, rhesus monkeys, ewes and more
recently, sheep. These models have provided evidence
that intra‑uterine fetal exposure to androgen excess is
associated with development of PCOS in later life.[12]
Genetic basis of this disease can be seen from studies
evaluating the genetic pool of patients with PCOS.
Legro and colleagues studied the siblings of 80 patients
diagnosed with PCOS. Out of a total of 115 sisters,
46% had evidence of hyperandrogenemia and amongst
them, almost half fulfilled the criteria for PCOS. It was
also noted that brothers of the patients with PCOS had
increasedlevelsof DHEASwhencompared withage
matched controls.[13] Fathers of adolescent girls have
a very high incidence of obesity (94%) and metabolic
syndrome (79%). Also, mothers of such patients tend
to be obese (54.4%) or overweight (11.4%) with the
incidence of metabolic syndrome being 34%.[14]
CLINICAL FEATURES
The patient seeks a dermatology consultation for one
or more complaints like acne, hirsutism, alopecia,
acanthosis nigricans, skin tags and occasionally,
darkening of complexion with weight gain. If
irregular menstrual cycles or primary infertility
are the main complaints, the patient may consult a
gynaecologist. An endocrinologist may be consulted
for hirsutism and the metabolic syndrome. Very rarely
do patients present with all the clinical signs and
symptoms of PCOS and some may not be forthcoming
with information of concurrent treatment from a
gynaecologist or an endocrinologist. An alert clinician
should be able to link the symptoms together, pointing
to a possible underlying defect of hyperandrogenism.
Acne vulgaris
Patients with PCOS complain of inflammatory acne
minimally responsive to conventional line of treatment.
Even if responsive, lesions promptly recur on
stopping treatment, necessitating treatment with oral
isotretinoin and/or hormonal therapy. An important
feature seen in these patients is the development of
multiple closed comedones which rapidly transform
into tender, lumpy nodules, distributed in the lower
half of face and jaw‑line (V distribution) [Figure 3].
These tend to persist beyond the usual course of
5‑7 days. A pre‑menstrual flare is also common. Acne
lesions may not only be localized to the face, but may
also be present on the chest, shoulders and back.
Prompt relapse after stopping the treatment, strongly
suggests a hormonal basis. Patients may in addition,
have a history of irregular periods, and evidence of
hirsutism, alopecia, or a positive family history of
PCOS. The severity of hirsutism may not be matching
the severity of acne, and would be dependent on the
balance of activity between the alfa‑hydroxy type 2
vis‑a‑vis type 1.[15]
Hirsutism
Excessive facial hair is a racial trait for the Indian
sub‑continent, running within families and especially
strong in certain ethnic groups. This should be kept
in mind while evaluating patients complaining of
excessive facial/body hair. Androgens affect various
aspects of follicular activity. Acting via androgen
receptors and secretory factors, they increase the
growth rate, diameter and melanization of hair in
androgen‑sensitive areas. It is these thick, coarse,
terminal hair, in androgen‑dependent areas which are
unsightly on a female[16] and point to an underlying
hyper‑androgenic state [Figure 4]. Evaluation of the
degree of hirsutism is done by adopting a modified
Ferriman‑Galway score which evaluates 9 body areas
on a scale of 1 to 4. The total scores are significant if
more than 6 to 8.[15,17]
Alopecia
Notallcases of femalepatternhair loss (FPHL)may
be of androgenic origin.[18] Patterned hair loss in PCOS
may be difficult to distinguish from those secondary
to other hyperandrogenic states [Figure 5]. Various
clinical presentations include those described by
Ludwig (diffuse), Hamilton (male pattern), and
Olsen (frontal accentuation).[18] Women with early
Madnani, et al. Polycystic ovarian syndrome
Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3314
onsetFPHLaremuchmorelikelytohaveanassociated
hyperandrogenism. Hormonal influences convert
terminal to vellus hair, making the scalp appear bald.
Acanthosis nigricans
Typically thick dark velvety skin situated on the
nape of the neck, axillae, groins and other frictional
areas may often be the first clue of insulin resistance
[Figure 6]. The thickening occurs due to the
stimulation of tyrosine kinase growth factor ‑
signaling pathways in the epidermis. Insulin ‑ like
growth factor receptor 1 (IGF1R) is present in many
tissues including the epidermis and ovary. High
levels of insulin directly or indirectly stimulate the
IGF1R resulting in the skin changes. Skin tags in the
frictional areas like the neck, axillae, groins, infra
mammary or even under a pendulous abdominal
fold are common, especially in obese individuals.
Irregular menses and infertility
PCOS is characterised by chronic menstrual
irregularities or changes in menstrual pattern with
reduced fertility. The anovulatory or oligoovulatory
cycles result in continuous endometrial stimulation
with estrogens, resulting in endometrial hyperplasia,
thus increasing the risk of endometrial cancers.[19]
Metabolic complications
The incidence of type 2 Diabetes and gestational
diabetes is markedly increased in both lean and obese
women with PCOS.[4] Other morbidities include sleep
apnoea, non‑alcoholic liver disease, hypertension
and dyslipidemia. The increased incidence of
cardiovascular disease although probable, is still not
supported in controlled studies.
QUALITY OF LIFE ISSUES
Dokras et al. did a meta‑analysis and systematic review
of studies comparing anxiety symptoms in women
with PCOS compared to normal, and concluded that
all women with PCOS should be screened for anxiety
symptoms.[20]
Figure 4: Hirsutism in a patient with PCOS
Figure 6: Acanthosis nigricans and hirsutism in an obese girl
with PCOS
Figure 5: Patterned hair loss with bi‑temporal recession in a
patient with PCOS
Figure 3: Acne with hirsutism in a patient with PCOS
Madnani, et al. Polycystic ovarian syndrome
315Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3
DIAGNOSTIC CRITERIA
Since 1990, various bodies have laid down criteria for the
diagnosis of PCOS, based on oligo or anovulation, signs
of hyperandrogenism, and ovarian sonography [Table 2].
As mentioned in the Rotterdam diagnostic criteria,
important causes of androgen excess must be ruled
out before diagnosing PCOS and investigations
should be directed towards detecting these conditions
like late‑onset congenital adrenal hyperplasia,
hyperprolactinemia, pituitary tumors, ovarian tumors,
etc., [Table 3]. When a diagnosis of PCOS is made, it is
almost one of exclusion.
Hormonalevaluationshouldbedoneinthepre‑ovulatory
phase. Since the levels of various hormones fluctuate
considerably with a single menstrual cycle, it is best to
carry out investigations in a fixed time within the cycle.
The fourteenth day seems ideal, but since most patients
have anovulatory or irregular cycles, the only fixed
window for investigation would be immediately after
the menstrual bleeding begins i.e. between the second
tofourthdayofthecycle.Normally,inthisphase,FSH
levelsare3to4timeshigherthanLH, butin patients
withPCOS,thisisreversedandaLH/FSHratiogreater
than 2 is very suggestive of pituitary dysfunction.
A highly raised serum testosterone level (more than
3 times the upper normal range) is very suggestive of a
neoplasm. If level of androstenedione is concomitantly
elevated, it is almost diagnostic of an ovarian or
adrenal malignancy, the former being much more
frequent than the latter. Hence all patients with
severely raised testosterone levels must undergo a
trans‑vaginal ultrasound to rule out any ovarian tumor.
Adrenal tumors are more virilising and are associated
with increased levels of DHEAS (>8 mmol/ml). The
raised testosterone levels in such cases is secondary
to increased peripheral conversion. A CT scan of the
abdomen is the investigation of choice to rule out
adrenal tumors.
Non‑classical adrenal hyperplasia (NCAH) is a very
close mimic to the PCOS and an 8:00 am serum
17‑hydroxyprogesterone level should be done in
suspected cases (normal is<2 ug/ml). While a level of
more than 8 ug/ml is diagnostic, it is the range in between
(2‑8 ug/ml) which requires further investigation.[21]
Prolactinoma may be an important cause for androgen
excess. Although elevated plasma prolactin levels
are highly suggestive of a prolactinoma, other causes
of hyperprolactemia like hypothyroisim, Cushing’s
disease, medications, etc., should be ruled out. Isolated
increase in prolactin level is not uncommon in today’s
stressful life, and its significance should be in context
with the clinical presentation.
Insulin levels may be increased in a majority of patients
with PCOS. The hyperinsulinemic‑euglycemic clamp
technique is gold standard for assessing insulin sensitivity,
but is impractical in everyday practice. Rather, a ratio of
fasting glucose to fasting insulin is a simple and effective
alternative. A ratio of less than 7 is highly suggestive of
a hyperinsulenemic state. In the authors’ experience,
a fasting serum insulin level more than 15, and serum
insulin level post 75gm glucose of more than 25 can also
serve as a rough guide to a hyperinsulinemic status.
In 2003, Rotterdam criteria was the first to introduced
the role of ultrasound examination of the ovaries as a
diagnostic criteria. Although ovarian morphology is
better depicted on trans‑vaginal ultrasound examination,
trans‑abdominal ultrasound examination is preferred in
adolescents and mandatory in unmarried women. Unlike
blood investigations, the ultrasonography (USG) may be
performed during any phase of the menstrual cycle but
to avoid any confusion with a dominant follicle, it is
best performed within few days of menstrual bleeding.
Table 2: Various diagnostic criteria dened for PCOS
Criteria NIH 1990 Rotterdam 2003 Androgen Excess Society 2006
Diagnostic criteria Must include Two of following three Must include
Chronic anovulation 1. Oligo/anovulation Ovarian dysfunction
Oligo/anovulation
Polycystic ovaries on USG
Clinical and/or biochemical
signs of hyperandrogenism
2. Clinical and/or biochemical
signs of hyperandrogenism
Androgen Excess
Hirsutism
hyperandrogenemia
3. Polycystic ovaries on USG
All criteria assume that other causes of androgen excess have been ruled out, PCOS: Polycystic ovarian syndrome
Madnani, et al. Polycystic ovarian syndrome
Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3316
The USG classically shows multiple subcentimeter
cysts (>12, 2‑9 mm) within the ovaries [Figure 7]. The
“string of pearls” appearance (peripheral arrangement
of the follicles surrounding a central echogenic stroma),
though characteristic, is not a requirement for diagnosis.
The other ultrasonographic feature is that the volume
of the ovaries should be each more than 10cc. These
changes need not be seen in both ovaries to make a
diagnosis of polycystic ovaries.
Investigations must be done to rule out underlying
metabolic abnormality and include blood sugars
and lipid profile. Physical evaluation must include
BMI (wt/ht2) calculation and whether patient has
central obesity (waist: hip ratio), as these patients are
at a higher risk of developing metabolic syndrome.[22]
PCOS MANAGEMENT
PCOS is a highly prevalent heterogeneous syndrome
of clinical and/or biochemical androgen excess,
ovulatory dysfunction and polycystic ovaries (PCO).
Despite it being one of the most common reproductive
health problems of women, its effective treatment
remains a significant challenge to medical profession.
Treatment of women with PCOS tends to be symptom
based. It is often difficult to treat all complaints at
the same time. The most difficult one is the desire
to treat both anovulatory infertility and hirsutism
concurrently. Oral contraceptives are contra‑indicated
in infertility treatments because they block ovulation
and anti‑androgens in view of their potential
teratogenic effects in a male fetus. Because of these
conundrums in clinical care, treatment tends to fall
into two categories, either the treatment of anovulatory
infertility or the long term maintenance treatment for
PCOS related symptoms (i.e., hirsutism, menstrual
disorders, obesity, etc.).
LONG TERM MAINTENANCE OF PCOS
The term maintenance acknowledges the fact that
there is no known cure for PCOS. Often the triad of
hirsutism, oligomenorrhea, and obesity forms the key
presenting symptoms. It may make sense to choose
a primary metabolic parameter upon which to base
initial treatment. Glucose intolerance is the strongest
risk factor for diabetes and is also an independent
risk factor for cardiovascular events in these women.
Additional targeted therapies for hirsutism and/
or oligomenorrhea could be added depending on
response to the initial therapy. Contraception should be
considered if the patient is trying to avoid pregnancy.
THERAPIES AIMED AT IMPROVING INSULIN SENSITIVITY
TO TREAT ANOVULATION AND ANDROGEN EXCESS
A logical approach to the management of PCOS
includes using life style measures and medications that
improve insulin sensitivity in target tissues, achieving
reductions in insulin secretion, and stabilizing glucose
tolerance.
Figure 7: Ultrasound image of both ovaries showing multiple
follicles arranged peripherally, giving a “string of pearls”
appearance
Table 3: Important investigations to be done in suspect
cases of PCOS
Name of Investigation Signicance
Lipid prole May be deranged in metabolic syndrome
Blood sugar (fasting) May be deranged in metabolic syndrome
Blood sugar (post
75 gm glucose)
May be deranged in metabolic syndrome
Insulin (fasting) >12 mIU/ml suggestive of insulin
resistance
Insulin (Post 75 gm
glucose)
>25 mIU/ml suggestive of insulin
resistance
TSH Hypothyroidism may cause oligo/
amenorrhoea
FSH/LH ratio Normally FSH> 3-4 LH
Reversal of ratio signicant
Total testosterone >3.6 ng/ml signicant
Androstenedione Elevated in ovarian pathology
Prolactin May be raised in PCOS but very high
levels indicate a pituitary tumor
17 hydroxy progesterone Elevated in late-onset CAH
DHEAS Elevated in adrenal pathology
DHT Indicative of peripheral conversion of
testosterone
Ultrasonography of
pelvis
Volume of atleast 1 ovary more than
10 cc with “string of pearl“ appearance
PCOS: Polycystic ovarian syndrome
Madnani, et al. Polycystic ovarian syndrome
317Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3
Lifestyle modication
The gold standard for improving insulin sensitivity
in obese PCOS women should be weight loss, diet,
and exercise. It is recommended as the first line of
treatment in obese women who present with infertility.
Hypo‑caloricdietsresultinappropriateweightlossin
women with PCOS. Unfortunately, there have been few
studies on the effect of exercise alone on symptoms in
PCOS women,[23] although it is reasonable to assume
that exercise would have the same beneficial effects
in women with PCOS as it does in women with type 2
DM.Howevertheexerciseprogrammustbetailoredto
the degree of obesity, and the patient’s baseline fitness.
Biguanide
Metformin may be most useful in the long term
maintenance of PCOS. Metformin does lower serum
androgens, and improves ovulatory and menstrual
frequency.[24] One study estimated that menstrual
frequency improved by roughly a third from baseline.
Metformin is the drug of choice to treat glucose
intolerance and elevated diabetes risk in women with
PCOS. Its use throughout pregnancy has shown to have
beneficial effects in reducing early pregnancy loss and
have a favorable effect on plasma glucose levels and
other metabolic aspects.[25] Metformin is also associated
with weight loss in women with PCOS, although the
results in other populations are inconsistent. Metformin
is often used in conjunction with lifestyle therapy to treat
PCOS. Gastrointestinal symptoms (diarrhea, nausea,
vomiting, abdominal bloating, flatulence, and anorexia)
are the most common adverse reactions and may be
ameliorated by starting at a small dose and gradually
increasing the dose or by using the sustained‑release
versions. The dose is usually 1500‑2000 mg/day given
in divided doses. Metformin carries a small risk of lactic
acidosis, most commonly among women with poorly
controlled diabetes and impaired renal function. Other
reported side effects include vitamin B12 deficiency,[26]
peripheral neuropathy,[27] and even hepatotoxicity.[28]
Thiazolidinediones
Pioglitazone and rosiglitazone are pharmacological
ligands for the nuclear receptor peroxisome proliferator
activated receptor γ (PPARγ). They improve the action
of insulin in the liver, skeletal muscles, adipose
tissue and have only modest effect on hepatic glucose
output. Improving insulin sensitivity with these drugs
is associated with a decrease in circulating androgen
levels, improved ovulation rate, and improved
glucose tolerance.[29‑32] However the concern about
hepatotoxicity, cardiovascular risk, weight gain, and
the pregnancy category C have limited the use of these
drugs in women with PCOS.
THERAPIES AIMED AT TREATING OLIGOMENORRHEA
If the patient does not wish to conceive, medical
therapy is directed towards interruption of the
effect of unopposed estrogen on the endometrium.
Nonfluctuating levels of unopposed estradiol in
the absence of progesterone cause irregular uterine
bleeding, amenorrhea, infertility and increased risk of
endometrial cancer.
Combination of oral contraceptives
Oral contraceptives have been the mainstay of long‑term
management of PCO. They offer benefit through a variety
ofmechanisms,includingsuppressionofpituitaryLH
secretion, suppression of ovarian androgen secretion,
and increased circulating SHBG levels. Individual
OC preparations may have different doses and drug
combinations and thus have varying risk–benefit ratios.
Most oral contraceptives (OC) preperations contain
estrogen (ethiny lestradiol 0.030 mg) in combination
with anti‑androgens. Anti‑androgens include
cyproterone acetate, drosperinone, levonorgestril,
norgestimate and desogestril. The “best” oral
contraceptive for women with PCOS is unknown.
Because women with PCOS may have multiple
risk factors for serious adverse events on oral
contraceptives, they must be screened carefully
for risk factors for these events including smoking
history, presence of obesity and hypertension, and
history of clotting diathesis to mention some of the
important factors. There is no convincing evidence
that use of oral contraceptives contributes to the risk
of diabetes in women with PCOS, although there are
often adverse effects on insulin sensitivity that may
be dose dependent.[33] A low dose oral contraceptive
pill is therefore recommended. Oral contraceptives
may also be associated with a significant elevation
in circulating triglycerides as well as in high density
lipoprotein(HDL)levels,thoughthesedonotappearto
progress over time. There is no evidence to suggest that
women with PCOS experience more cardiovascular
events than the general population when they use
oral contraceptives. If a woman is taking an oral
contraceptive that contains drospirenone, a progestin
with anti‑mineralocorticoid properties, it may be
necessary to reduce her dose of spironolactone if used
concomitantly. Regular evaluation of potassium levels
are necessary.
Madnani, et al. Polycystic ovarian syndrome
Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3318
Progestin
Both depot and intermittent oral medroxyprogesterone
acetate (10 mg for 10 days) have been shown to suppress
pituitary gonadotropins and circulating androgens in
women with PCOS.[34] No studies have addressed the
long‑term use of these compounds to treat hirsutism.
There is also a paucity of data to address the varying
risk benefit ratios of varying classes of progestins.
Progestin‑only oral contraceptives are an alternative
for endometrial protection, but they are associated
with a high incidence of breakthrough bleeding.
THERAPIES AIMED AT TREATING HIRSUITISM
No oral contraceptive has been approved by the
FDA for the treatment of hirsutism. A number of
observational or non‑randomized studies have noted
improvement in hirsutism in women with PCOS who
use oral contraceptives, but no studies of adequate
power confirm their benefit in improving hirsutism in
PCOS.[35]
Spironolactone
It is primarily used to treat hirsutism and appears
effective, though the evidence is weak. It is a diuretic
and aldosterone antagonist, also binds to the androgen
receptor as an antagonist. It has other mechanisms of
action, including inhibition of ovarian and adrenal
steroidogenesis, competition for androgen receptors in
hair follicles, and direct inhibition of 5 alpha reductase
activity. The usual dose is 25–100 mg twice a day, and
is titrated to balance efficacy while avoiding side effects
such as orthostatic hypotension. A full clinical effect
may take 6 months or more. About 20% of women using
spironolactone will experience increased menstrual
frequency.[36] Because it can cause and exacerbate
hyperkalemia, spironolactone should be used cautiously
in women with renal impairment. Rarely, exposure has
resulted in ambiguous genitalia in male infants.
Cyproterone acetate
It is a progestogen with anti‑androgen properties. It is
frequently combined in an oral contraceptive tablets and
is popular in the treatment of PCOS. A newer progestin
from the same class, drospirenone has been marketed
in the U.S. as especially effective for the treatment of
female hyperandrogenism, although the data suggesting
this is superior to other formulations is not based on
head to head randomized trials.[37] When given as
100mg/day, it inhibits testosterone production resulting
in upto 75% decrease in circulating testosterone levels.
Flutamide
It is an androgen‑receptor antagonist, and is another
non steroidal anti‑androgen that has been shown to be
effective against hirsutism in smaller trials The most
common side effect is dry skin, but its use has been
associated with hepatitis in rare cases. The common
dosage is 250 mg/day. The risk of teratogenicity with
this compound is significant, and contraception
should be used. Flutamide has also been combined
with lifestyle and metformin therapy for treatment of
PCOS and may have additive effects.[38]
Finasteride
It is a specific inhibitor of type II 5α reductase enzyme
found in the hair follicles on the top of the scalp and
in the sebaceous gland ducts. Its use is restricted to
women in the post‑menopausal group or women with
documented hyperandrogenic state in the dosage of
5 mg/day. Finasteride is better tolerated than other
anti‑androgens, with minimal hepatic and renal
toxicity; however, it has well‑documented risk for
teratogenicity and feminising in a male fetuses, and
adequate contraception should be used. Overall,
randomized trials have found that spironolactone,
flutamide and finasteride to have similar efficacy in
improving hirsutism.
Ornithine decarboxylase inhibitors
These have been developed for the treatment of female
hirsutism. Ornithine decarboxylase is necessary
for the production of polyamines, and inhibition of
this enzyme limits cell division and function in the
pilosebaceous unit. Recently a potent inhibitor of this
enzyme, eflornithine, has been found to be effective
as a facial cream for the treatment of unwanted facial
hair.[39] It is available as a 13.9% cream of eflornithine
hydrochloride, and is applied to affected areas twice
daily. In clinical trials, 32% of patients had marked
improvement after 24 weeks compared to 8% of
placebo treated women, and the benefit was first
noted at eight weeks. It is pregnancy category C drug.
It appears to be well tolerated, with only about 2% of
patients developing skin irritation or other adverse
reactions. Relapse is common after stopping.
Mechanical and cosmetic means of hair reduction and
destruction
Mechanical hair removal techniques like shaving,
plucking, waxing, depilatory creams, electrolysis, and
laser hair reduction (LHR) offer good cosmetic relief
and often are the front line of treatment used by women.
A word of caution regarding facial waxing as it can
Madnani, et al. Polycystic ovarian syndrome
319Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3
precipitate folliculitis. Various lasers are used for hair
reduction and include the Diode (800nm), Alexandrite
(755 nm), Long pulsed Nd‑YAG (neodymium‑doped
yttrium aluminium garnet; Nd: Y3Al5O12) (1064nm)
andIPLdevices.Thereis no“goldstandard”forLHR
but the long pulsed Nd‑YAG is the safest in pigmented
skins. It must be emphasized that LHR would be
effective only after anti‑androgen therapy has been
initiated at least 2 to 3 months prior. It results in
reduction in density and thickness of hair but if at any
point in time, there is uncontrolled androgen excess,
there will be a relapse in hirsutism.
THERAPIES AIMED AT TREATING ANOVULATORY
INFERTILITY
The recommended first‑line treatment for ovulation
induction remains the anti‑estrogen clomiphene
citrate (CC). Recommended second‑line intervention
is either exogenous gonadotropins or laparoscopic
ovarian surgery.[40] There appears to be some benefit
of addition of metformin to clomiphene, especially in
obese subjects (modified first line treatment).
Clomiphene citrate
It is a triphenylethylene derived nonsteroidal
agent that is theorized to function at the level of
the hypothalamus as an anti‑estrogen to improve
gonadotropin secretion. An important concern is the
relatively high rate of multiple pregnancies (7.8%)
after conception, majority being twins.[41]
Gonadotropins
These are frequently used to induce ovulation in
women with PCOS for whom clomiphene treatment
has failed. Low‑dose therapy with gonadotropins
offers a higher rate of ovulation, monofollicular
development, with a significantly lower risk of ovarian
hyperstimulation syndrome.[42] Pregnancy rates are
comparable with other regimens, but there have been
no adequately powered trials to answer this question.
Ovarian surgery
This is primarily recommended as second line infertility
therapy. Multiple pregnancy rates are reduced in those
women who conceive following laparoscopic drilling.
In some cases, the fertility benefits of ovarian drilling
may be temporary and adjuvant therapy after drilling
with clomiphene may be necessary.[43] Ovarian drilling
does not appear to improve metabolic abnormalities in
women with PCOS.[44]
Aromatase inhibitors
Aromatase inhibitors such as letrozole and anastrazole
have been proposed as both first and secondary
treatment for ovulation induction (in women with
PCOS and also for unexplained infertility).[45] Results in
women with PCOS appear comparable to clomiphene
from small trials. Proposed benefits include oral
administration, a shorter half life than clomiphene,
more favorable effects on the endometrium, potentially
higher implantation rates, and lower multiple
pregnancy rates due to monofollicular ovulation.
Their use is still experimental at this point.
Thiazolidinediones
Smaller trials have shown some benefit to this class
of drugs for the treatment of infertility usually in
conjunction with clomiphene.[46,47] However the
concern about hepatotoxicity, cardiovascular risk,
weight gain, and the pregnancy Category C have
limited the use of these drugs in women with PCOS.
RESEARCH ONGOING
Statins
Another area where there is emerging support in the
literature for a cardiovascular and endocrine benefit
in women with PCOS is the use of statins. They have
been shown to improve hyperandrogenemia, lipid
levels, and reduce inflammation.[48] However, their
long term effects in preventing cardiovascular disease
in young women with PCOS is unknown. There are
concerns about teratogenecity with the use of this drug
in reproductive age women, as it is FDA pregnancy
category X. The use of these drugs is still experimental
in women with PCOS.
APPROACH TO A PATIENT WITH PCOS
Suspect PCOS in women (obese/lean) with/without
a history of irregular periods with one or more of
the following complaints, namely, acne, hirsutism,
alopecia, acanthosis nigricans and metabolic
syndrome. Investigations both, hematological and
imaging are geared to rule out other hyperandrogenic
states as there is no single investigation to diagnose
PCOS. Patients must be counseled about the long
duration of treatment which includes life‑style
modifications along with the systemic treatment.
Success in effective management of a woman with
PCOS is through a synchronized effort between
the dermatologist, endocrinologist, gynecologist,
nutritionist and physical trainer.
Madnani, et al. Polycystic ovarian syndrome
Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3320
REFERENCES
1. Stein IF, Leventhal ML. Amenorrhea associated with bilateral
polycystic ovaries. Am J Obstet Gynecol 1935;29:181‑91.
2. Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES,
Yildiz BO. The prevalence and features of the polycystic ovary
syndrome in an unselected population. J Clin Endocrinol Metab
2004;89:2745‑9.
3. Nidhi R, Padmalatha V, Nagarathna R, Amritanshu R. Prevalence
of polycystic ovarian syndrome in Indian adolescents. J Pediatr
Adolesc Gynecol 2011;24:223‑7.
4. Legro RS, Kunselman AR, Dodson WC, Dunaif A. Prevalence
and predictors of risk for type 2 diabetes mellitus and impaired
glucose tolerance in polycystic ovary syndrome: A prospective,
controlled study in 254 affected women. J Clin Endocrinol
Metab 1999;84:165‑9.
5. Fearnley EJ, Marquart L, Spurdle AB, Weinstein P, Webb PM.
Australian ovarian cancer study group and australian national
endometrial cancer study group. Polycystic ovary syndrome
increases the risk of endometrial cancer in women aged less
than 50 years: An Australian case‑control study. Cancer Causes
Control 2010;21:2303‑8.
6. Adashi EY. The climacteric ovary as a functional
gonadotropin‑driven androgen‑producing gland. Fertil Steril
1994;62:20‑7.
7. Dunaif A. Insulin resistance and the polycystic ovary syndrome
mechanism and implications for pathogenesis. Endocr Rev
1997;18:774‑800.
8. Tsilchorozidou T, Overton C, Conway GS. The pathophysiology
of polycystic ovary syndrome. Clin Endocrinol (Oxf)
2004;60:1‑17.
9. Dunaif A, Segal KR, Futterweit W, Dobrjansky A. Profound
peripheral insulin resistance, independent of obesity in
polycystic ovary syndrome. Diabetes 1989;38:1165‑74.
10. Cara JF, Rosenfield RL. Insulin‑like growth factor I and
insulin potentiate luteinizing hormone‑induced androgen
synthesis by rat ovarian thecal‑interstitial cells. Endocrinology
1988;123:733‑9.
11. Shi D, Vine DF. Animal models of polycystic ovary syndrome:
A focused review of rodent models in relationship to
clinical phenotypes and cardiometabolic risk. Fertil Steril
2012;98:185‑93.
12. Xita N, Tsatsoulis A. Review: Fetal programming of polycystic
ovary syndrome by androgen excess: Evidence from
experimental, clinical and genetic association studies. J Clin
Endocrinol Metab 2006;91:1660‑6.
13. Legro RS, Kunselman AR, Demers L, Wang SC, Bentley‑Lewis R,
Dunaif A. Elevated dehydroepiandrosterone sulfate levels
as the reproductive phenotype in the brothers of women
with polycystic ovary syndrome. J Clin Endocrinol Metab
2002;87:2134.
14. Leibel NI, Baumann EA, Kocherginsky M, Rosenfield RL.
Relationship of adolescent polycystic ovary syndrome to
parental metabolic syndrome. J Clin Endocrinol Metab
2006;91:1275‑83.
15. ArcherJS,Chang RJ.Hirsutismand acnein polycysticovary
syndrome. Best Pract Res Clin Obstet Gynaecol 2004;18:737‑54.
16. Azziz R. The evaluation and management of hirsutism. Obstet
Gynecol 2003;101:995‑1007.
17. Morin‑Papunen LC, Vauhkonen I, Koivunen RM, Ruokonen A,
Martikainen HK, Tapanainen JS. Endocrine and metabolic
effects of metformin versus ethinyl estradiol‑cyproterone
acetate in obese women with polycystic ovary syndrome:
A randomized study. J Clin Endocrinol Metab 2000;85:3161‑8.
18. Olsen EA. Female pattern hair loss. J Am Acad Dermatol
2001;45:70‑80.
19. ParkJC,LimSY,JangTK,BaeJG,KimJI,RheeJH.Endometrial
histology and predictable clinical factors for endometrial
disease in women with polycystic ovary syndrome. Clin Exp
Reprod Med 2011;38:42‑6.
20. Dokras A, Clifton S, Futterweit W, Wild R. Quality of Life Issues
and PCOS. Fertil Steril 2012;97:225‑30.
21. Escobar‑MorrealeHF,SanchonR,SanMillanJL.Aprospective
study of the prevalence of nonclassical congenital
adrenal hyperplasia among women presenting with
hyperandrogenic symptoms and signs. J Clin Endocrinol Metab
2008;93:527‑33.
22. Mandrelle K, Kamath MS, Bondu DJ, Chandy A, Aleyamma T,
George K. Prevalence of metabolic syndrome in women with
polycystic ovarysyndrome attending an infertility clinic
in a tertiary care hospital in south India. J Hum Reprod Sci
2012;5:26‑31.
23. Knowler WC, Barrett‑Connor E, Fowler SE, Hamman RF,
Lachin JM, Walker EA, et al. Reduction in the incidence of
type 2 diabetes with lifestyle intervention or metformin. N Engl
J Med 2002;346:393‑403.
24. Lord JM, Flight IH, Norman RJ. Metformin in polycystic
ovary syndrome: systematic review and meta‑analysis. BMJ
2003;327:951‑3.
25. Kumar P, Khan K. Effects of metformin use in pregnant
patients with polycystic ovary syndrome. J Hum Reprod Sci
2012;5:166‑9.
26. Kumthekar AA, Gidwani HV, Kumthekar AB. Metformin
associated B12 deficiency. J Assoc Physicians India
2012;60:58‑60.
27. Bell DS. Metformin‑induced vitamin B12 deficiency presenting
as a peripheral neuropathy. South Med J 2010;103:265‑7.
28. Miralles‑Linares F, Puerta‑Fernandez S, Bernal‑Lopez MR,
TinahonesFJ,AndradeRJ,Gomez‑HuelgasR.Metformin‑induced
hepatotoxicity. Diabetes Care 2012;35:e21.
29. Azziz R, Ehrmann D, Legro RS, Whitcomb RW, Hanley R,
Fereshetian AG, et al. Troglitazone improves ovulation and
hirsutism in the polycystic ovary syndrome: A multicenter,
double blind, placebo‑controlled trial. J Clin Endocrinol Metab
2001;86:1626‑32.
30. Baillargeon JP, Jakubowicz DJ, Iuorno MJ, Jakubowicz S,
Nestler JE. Effects of metformin and rosiglitazone, alone and
in combination, in nonobese women with polycystic ovary
syndrome and normal indices of insulin sensitivity. Fertil Steril
2004;82:893‑902.
31. Vessey MP, Painter R. Endometrial and ovarian cancer and oral
contraceptives‑findings in a large cohort study. Br J Cancer
1995;71:1340‑2.
32. Schlesselman JJ. Risk of endometrial cancer in relation to use
of combined oral contraceptives. A practitioner’s guide to
meta‑analysis.HumReprod1997;2:1851‑63.
33. Costello MF, Shrestha B, Eden J, Johnson NP, Sjoblom P.
Metformin versus oral contraceptive pill in polycystic ovary
syndrome:ACochranereview.HumReprod2007;22:1200‑9.
34. Anttila L, Koskinen P, Erkkola R, Irjala K, Ruutiainen K. Serum
testosterone, androstenedione and luteinizing hormone levels
after short‑term medroxyprogesterone acetate treatment in
women with polycystic ovarian disease. Acta Obstet Gynecol
Scand 1994;73:634‑6.
35. Swiglo BA, Cosma M, Flynn DN, Kurtz DM, Labella ML,
Mullan RJ, et al. Antiandrogens for the treatment of hirsutism:
A systematic review and metaanalyses of randomized
controlled trials. J Clin Endocrinol Metab 2008;93:1153‑60.
36. Helfer EL, Miller JL, Rose LI. Side‑effects of spironolactone
therapy in the hirsute woman. J Clin Endocrinol Metab
1988;66:208‑11.
37. Palep‑Singh M, Mook K, Barth J, Balen A. An observational
study of Yasmin in the management of women with
polycysticovarysyndrome. JFamPlann ReprodHealth Care
2004;30:163‑5.
38. Gambineri A, Patton L, Vaccina A, Cacciari M,
Morselli‑Labate AM, Cavazza C, et al. Treatment with flutamide,
metformin, and their combination added to a hypocaloric diet
in overweight‑obese women with polycystic ovary syndrome:
Madnani, et al. Polycystic ovarian syndrome
321Indian Journal of Dermatology, Venereology, and Leprology | May-June 2013 | Vol 79 | Issue 3
A randomized, 12‑month, placebo‑controlled study. J Clin
Endocrinol Metab 2006;91:3970‑80.
39. WolfJEJr,ShanderD,HuberF,JacksonJ,LinCS,MathesBM,
et al. Randomized, double‑blind clinical evaluation of the
efficacy and safety of topical eflornithine HCl 13.9% cream
in the treatment of women with facial hair. Int J Dermatol
2007;46:94‑8.
40. Thessaloniki ESHRE/ASRM‑Sponsored PCOS Consensus
Workshop Group. Consensus on infertility treatment related to
polycystic ovary syndrome. Fertil Steril 2008;89:505‑22.
41. Asch RH, Greenblatt RB. Update on the safety and efficacy
of clomiphene citrate as a therapeutic agent. J Reprod Med
1976;17:175‑80.
42. Christin‑Maitre S, Hugues JN. A comparative randomized
multicentric study comparing the step‑up versus step‑down
protocol in polycystic ovary syndrome. Hum Reprod
2003;18:1626‑31.
43. Farquhar C, Brown J, Marjoribanks J. Laparoscopic ‹drilling›
by diathermy or laser for ovulation induction in anovulatory
polycystic ovary syndrome. Cochrane Database Syst Rev
2012;6:CD001122.
44. Lemieux S, Lewis GF, Ben‑Chetrit A, Steiner G, Greenblatt EM.
Correction of hyperandrogenemia by laparoscopic ovarian
cautery in women with polycystic ovarian syndrome is not
accompanied by improved insulin sensitivity or lipid‑lipoprotein
levels. J Clin Endocrinol Metab 1999;84:4278‑82.
45. Gysler M, March CM, Mishell DR Jr, Bailey EJ. A decade›s
experience with an individualized clomiphene treatment
regimen including its effect on the postcoital test. Fertil Steril
1982;37:161‑7.
46. GhazeeriG,KuttehWH,Bryer‑AshM,HaasD,KeRW.Effectof
rosiglitazone on spontaneous and clomiphene citrate‑induced
ovulation in women with polycystic ovary syndrome. Fertil
Steril 2003;79:562‑6.
47. Rouzi AA, Ardawi MS. A randomized controlled trial of
the efficacy of rosiglitazone and clomiphene citrate versus
metformin and clomiphene citrate in women with clomiphene
citrate‑resistant polycystic ovary syndrome. Fertil Steril
2006;85:428‑35.
48. Sathyapalan T, Kilpatrick ES, Coady AM, Atkin SL. The effect
of atorvastatin in patients with polycystic ovary syndrome:
A randomized double‑blind placebo‑controlled study. J Clin
Endocrinol Metab 2009;94:103‑8.
Multiple Choice Questions
1. Ovaries are responsible for ___% of circulating testosterone
a. 20% b. 30%
c. 40% d. 50%
2. Development of insulin resistance in patients with PCOS is
a. Independent of weight b. Directly proportional to weight
c. Very rare d. Genetically determined
3. PCOS is associated with increased incidence of
a. Type 2 Diabetes Meliitus b. Gestational diabetes
c. Deranged lipid profile d. All of the above
4. All are true about metformin except
a. Lowers serum androgens b. Improves ovulatory cycles
c. Unsafeinpregnancy d. Helpstoreduceweight
5. Treatment for hirsutism includes all except
a. Spiranolactone b. Medroxyprogesterone acetate
c. Flutamide d. Finasteride
6. Ultrasound diagnostic criteria for PCOS includes
a. Volume of each ovary more than 5 cc b. Total volume of both ovaries equal to 8 cc
c. Volume of at least one ovary larger than 10 cc d. Volume of at least one ovary more than 7.5 cc
7. Hyperinsulinemialeadsto
a. IncreasedfrequencybutnottheamplitudeofLHsecretion b. IncreasedfrequencyandamplitudeofLHsecretion
c. IncreasedamplitudebutnotthefrequencyofLHproduction d. Noneoftheabove
8. The progesterones which are not androgenetic are
a. Desogestrel b. Levonorgestrel
c. Norgestrel d. All of the above
9. Adverse effects of thiazolidinediones include
a. Hepatotoxicity b. Cardiovascularrisk
c. Weight gain d. All of the above
10. Percentage of women experiencing menstrual irregularities with spirinolactone
a. 25% b. 30%
c. 20% d. 40%
Answers:
1. a, 2. a, 3. d, 4. c, 5. b, 6. c, 7. c, 8. a, 9. d, 10. c
... Additionally, patients may have a history of irregular menstrual cycles, as well as hirsutism, alopecia, or a positive family history of PCOS. The balance of activity between the alpha hydroxy types 2 and 1 will determine whether the severity of hirsutism matches the severity of acne [19] . ...
... By using a modified Ferriman Galway score, which rates nine body regions on a scale of 1 to 4, the degree of hirsutism is assessed. If the overall scores range from 6 to 8, then they are important [19] . ...
... Patterned hair loss associated with PCOS may be challenging to distinguish from secondary hyperandrogenic conditions. The clinical manifestations mentioned by Ludwig (diffuse), Hamilton (male pattern), and Olsen (frontal accentuation) are just a few examples [19] .Hyperandrogenism is substantially more prevalent in women with early-onset FPHL. Hormonal effects change ISSN: 2320-4850 [22] CODEN (USA): AJPRHS terminal hair into vellus hair, giving the appearance of a bald scalp. ...
... There are different screening suggestions for each of these pathologies, but the clinician must have a low threshold for workup if any manifestation is shown in PCOS patients. Good clinical examination with haematological and radiological investigations is required for clinical assessment [9] [10]. The prevalence ranges between 5% and 15% depending on the diagnostic scale applied. ...
... Delay in diagnosis can lead to the advancement of comorbidities making it more difficult to implement lifestyle intervention, which is critical to improve the features of PCOS and quality of life [10]. Management is a combined effort involving an endocrinologist, gynaecologist, dermatologist, and nutritionist [9]. An unhealthy diet is a key component of the "deadly quartet" of metabolic risk factors such as obesity, diabetes mellitus, hypertension, and hypertriglyceridemia associated with PCOS pathophysiology, along with low-grade inflammation, Hyperinsulinemia, and Hyperandrogenism [13]. ...
Article
Polycystic ovarian syndrome (PCOS) is a very common endocrine disorder encountered in women and is associated with several complications, such as menstrual irregularities; hirsutism; obesity; insulin resistance; acne; and, in later years, infertility with diabetes mellitus, cardiovascular risk, depression, obstructive sleep apnea (OSA), and uterine cancer. Early diagnosis is essential for early interventions to minimize the immediate and chronic consequences. The exact facts about PCOS has not yet been clearly determined. This study is an attempt to assess its influence of food usage and their relationship with PCOS. We undertook a survey among the PCOS affected women between the age group of 18 – 40 from the state of Tamil Nadu. The study population represents a random sample of PCOS female (139 women). The study period was six months from July 2021 to December 2021. This study definitely created awareness among the women in respect to PCOS. This will help them to modify their life style especially food habit and to have better reproductive life later. During this study we identified the adolescents with risk for developing PCOS, we ask them to take proper diagnosis and treatment with consultation of a gynaecologist and also dieticians. In married women, especially in the presence of other risk factors for infertility, early conception is advised and to find the effectiveness of awareness programme.
... As a result, LH levels increased, follicle-stimulating hormone (FSH) levels decreased and thereby the LH/FSH ratio increased (48). The unfavourable feedback of oestradiol influences the generation of FSH as well (49). The activation of androgen synthesis by LH release in endometrial cells results in an increase in androgen levels in the body. ...
Article
Full-text available
Polycystic ovary syndrome (PCOS), a hormonal and metabolic disorder manifested in women of reproductive age, is still being treated using drugs with side effects. As an alternative to these drugs, isoflavone, also identified as phytoestrogen, has anti-PCOS activity. Isoflavone can help relieve PCOS symptoms by lowering the level of testosterone, which causes hyperandrogenism, thereby normalizing the menstrual cycle and restoring normal ovarian morphology. Furthermore, isoflavone influences the improvement of the metabolic profile, which changes because of PCOS, as well as the reduction of inflammatory markers and oxidative stress. However, both significant and non-significant results have been generated on the activity of isoflavones in PCOS. The present review aims to discuss the existing literature on the effect of isoflavone on PCOS symptoms based on in vivo and clinical trial studies.
... The World Health Organization (WHO) data suggests that over 166 million women are affected by PCOS on a global basis [7]. Despite PCOS being the most common endocrine health problem of women of reproductive age, its management remains a significant challenge to the medical profession [8]. ...
Article
Full-text available
Polycystic ovarian syndrome (PCOS) is a known endocrine disorder that has affected many women of childbearing age, and is accompanied by various neurodegenerative conditions. Hence, this study investigates the impact of butyrate in reversing hypothalamic-related disorder, possibly through γ aminobutyric acid (GABA) in a rat model of PCOS. Eight-week-old female Wistar rats were allotted into four groups (n = 5), which include control, butyrate, letrozole, and letrozole + butyrate groups. PCOS was induced by administering 1 mg/kg of letrozole (oral gavage) for 21 days. After confirmation of PCOS, 200 mg/kg of butyrate (oral gavage) was administered for 6 weeks. Rats with PCOS were characterized by elevated levels of plasma insulin and testosterone. Increases in plasma and hypothalamic triglyceride levels, inflammatory biomarker (SDF-1), apoptotic marker (caspase-6), and decreased plasma GnRH were observed. Additionally, a decrease in hypothalamic GABA was revealed. Nevertheless, the administration of butyrate attenuated these alterations. The present study suggests that butyrate ameliorates hypothalamic inflammation in an experimental model of PCOS, a beneficial effect that is accompanied by enhanced GABA production.
... Met-SR therapy also showed reduction testosterone, increase SHBG levels, ovulatory frequency and menstrual irregularities, which is similar to other studies. 19,20 In the present study we have confirmed that, women with PCOS showed correlation between IR and BMI before and after therapy, and also observed a positive correlation between SHBG and TT with HOMA-IR after six months of Met-SR therapy. ...
... Polycystic ovarian syndrome (PCOS) is the most common endocrinopathy in women during their reproductive ages, and depending on different criteria, the prevalence can be up to 6-20% [1][2][3]. It is clinically characterized by hyperandrogenisms, like acne, seborrhoea, hirsutism, irregular menses, infertility, and alopecia [4]. The diagnosis and treatment of PCOS are not complex, but the pathogenic mechanisms remain obscure, studies have reported that PCOS is associated with insulin resistance and obesity [5,6]. ...
Article
Introduction: Polycystic ovarian syndrome (PCOS) is a common endocrinopathy in women. MicroRNAs (miRNAs) have been proven to play a crucial role in balancing the proliferation and apoptosis of granulosa cells (GCs) in PCOS. Material and methods: The miRNA of PCOS was screened by bioinformatics analysis, and microRNA 646 (miR-646) was found to be involved in insulin-related pathways by enrichment analysis. The cell counting kit-8 (CCK-8), cell colony formation, and the 5-ethynyl-2'-deoxyuridine (EdU) assays were used to explore the effect of miR-646 on proliferation of GCs, flow cytometry was used to measure the cell cycle and apoptosis, and Western blot and quantitative real-time polymerase chain reaction (qRT-PCR) were used to explore the biological mechanism of miR-646. The human ovarian granulosa cells KGN were selected by measuring the miR-646 and via insulin-like growth factor 1 (IGF-1) levels and used for cell transfection. Results: Overexpressed miR-646 inhibited KGN cell proliferation, and silenced miR-646 advanced it. Most cells were arrested in the S phase of cell cycle with overexpressed-miR-646, while after silencing miR-646, cells were arrested in the G2/M phase. And the miR-646 mimic raised apoptosis in KGN cells. Also, a dual-luciferase reporter proved the regulation effect of miR-646 on IGF-1, miR-646 mimic inhibited IGF-1, and miR-646 inhibitor advanced IGF-1. The cyclin D1, cyclin-dependent kinase 2 (CDK2), and B-cell CLL/lymphoma 2 (Bcl-2) levels were inhibited with overexpressed-miR-646, while silenced-miR-646 promoted their expression, and the bcl-2-like protein 4 (Bax) level was the opposite. This study found that silenced-IGF1 antagonized the promotive effect of the miR-646 inhibitor on cell proliferation. Conclusions: MiR-646 inhibitor treatment can promote the proliferation of GCs by regulating the cell cycle and inhibiting apoptosis, while silenced-IGF-1 antagonizes it.
... It either directly affects the hepatic metabolism of free fatty acids or indirectly acts by lowering hyperinsulinemia to improve dyslipidemia [86]. Additionally, TZDs (pioglitazone and rosiglitazone) lower insulin levels by increasing insulin sensitivity, which reduces levels of androgens in the blood [87]. Women with PCOS have reported that pioglitazone had an impact on reducing insulin resistance, hyperandrogenism, and ovulatory dysfunction. ...
Article
Full-text available
Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorder, typically characterized by anovulation, infertility, obesity, insulin resistance, and polycystic ovaries. Lifestyle or diet, environmental pollutants, genetics, gut dysbiosis, neuroendocrine alterations, and obesity are among the risk factors that predispose females to PCOS. These factors might contribute to upsurging metabolic syndrome by causing hyperinsulinemia, oxidative stress, hyperandrogenism, impaired folliculogenesis, and irregular menstrual cycles. Dysbiosis of gut microbiota may play a pathogenic role in the development of PCOS. The restoration of gut microbiota by probiotics, prebiotics, or a fecal microbiota transplant (FMT) might serve as an innovative, efficient, and noninvasive way to prevent and mitigate PCOS. This review deliberates on the variety of risk factors potentially involved in the etiology, prevalence, and modulation of PCOS, in addition to plausible therapeutic interventions, including miRNA therapy and the eubiosis of gut microbiota, that may help treat and manage PCOS.
Article
Polystyrene microplastics, extensively considered endocrine disrupting chemicals, disturb the reproductive system of living organisms. Polycystic ovary syndrome (PCOS), the reproductive endocrinopathy, is longstanding concern due to its eternal impacts as reproductive disorder and infertility. Despite several reports in reproductive and endocrine toxicity, there is inadequate literature regarding the daily intake of polystyrene-microplastics via drinking water in causing PCOS and leading to ovarian fibrosis in long-term. The present study investigated whether daily consumption of polystyrene-microplastics at doses equivalent to human exposure can cause PCOS and progress to ovarian fibrosis, using female zebrafish as model. Resembling letrozole-PCOS zebrafish model, daily intake of polystyrene-microplastics displayed hallmark PCOS pathophysiology; like excess body weight and %Gonadosomatic index, decreased Follicle Stimulating Hormone and β-estradiol, increased Luteinising Hormone, brain and ovarian Testosterone (39.3% and 75% respectively). Correspondingly, ovarian histology revealed more developing (stage I and II) oocytes and less mature oocytes alongwith cystic lesions; like follicular membrane disorganization, zona pellucida invagination, theca hypertrophy, basophilic granular accumulation and oocyte buddings. Lipid deposition in intestinal and ovarian tissues was evidenced and increased fasting blood glucose manifesting insulin resistance. The expression of PCOS biomarkers (tox3, dennd1a, fem1a) was significantly disturbed. Polystyrene microplastics played vital role in inducing PCOS further enhancing oxidative stress, which positively influences inflammation and aggravate ovarian mitophagy, shedding light on its ability to harshen PCOS into ovarian fibrosis, which is characterized by collagen deposition and upregulation of pro-fibrogenic biomarker genes. These findings illustrate the potential of daily microplastics intake via drinking water in triggering PCOS and its progression to ovarian fibrosis.
Article
Full-text available
Polycystic ovarian syndrome (PCOS) is a complex disorder marked by elevated androgen levels, menstrual abnormalities and presence of small cysts on one or both ovaries. The disorder can be morphological (polycystic ovaries) or predominantly biochemical (hyperandrogenemia). This article aims to review the various aspects of pharmacological amd non pharmacological management of PCOS.
Article
Full-text available
Use of metformin throughout pregnancy in women with polycystic ovary syndrome (PCOS) has shown to reduce the rates of early pregnancy loss, preterm labor, and prevention of fetal growth restriction. Metformin has been shown to have encouraging effects on several metabolic aspects of polycystic ovarian syndrome, such as insulin sensitivity, plasma glucose concentration and lipid profile and since women with PCOS are more likely than healthy women to suffer from pregnancy-related problems like early pregnancy loss, gestational diabetes mellitus and hypertensive states in pregnancy, the use of metformin therapy in these patients throughout pregnancy may have beneficial effects on early pregnancy loss and development of gestational diabetes.
Article
Full-text available
The aim of the present study was to evaluate the prevalence of metabolic syndrome in women with polycystic ovary syndrome (PCOS). Infertility clinic in a tertiary care hospital. A prospective cross-sectional study. All the women attending the infertility clinic categorized as polycystic ovary syndrome according to Rotterdam criteria (2003) during the study period were included in the study. The women with PCOS underwent screening for metabolic syndrome as defined by the modified American Heart Association/National Heart Lung Blood Institute (AHA/NHLBI) modified ATP 111 (2005) definition. A multivariate logistic regression analysis was applied and significant predictors identified for the prediction of metabolic syndrome. The overall prevalence of metabolic syndrome according to the modified AHA/NHLBI ATP III (2005) criteria was 37.5%. A total of 5.8 % cases were detected to have diabetes mellitus, 8.3% had impaired fasting glucose, and 11.7 % had an impaired glucose test. Dyslipidemia was present in 93.3% cases of PCOS. Among all the risk factors, age and waist hip ratio ≥0.85 were strongly associated with the presence of metabolic syndrome. Infertile women with PCOS, particularly those with age ≥25 years or with central obesity (a waist hip ratio of ≥0.85), are at a higher risk of developing metabolic syndrome and should be offered screening tests.
Article
Objective Polycystic ovary syndrome (PCOS) is a common endocrine disorder of premenopausal women, characterized by hyperandrogenism, polycystic ovaries, and chronic anovulation along with insulin resistance and abdominal obesity as frequent metabolic traits. Although, PCOS manifests clinically during adolescence, emerging data suggest that the natural history of PCOS may originate in intrauterine life. Evidence Acquisition Evidence from experimental, clinical, and genetic research supporting the hypothesis for the fetal origins of PCOS has been analyzed. Evidence Synthesis Female primates, exposed in utero to androgen excess, exhibit the phenotypic features of PCOS during adult life. Clinical observations also support a potential fetal origin of PCOS. Women with fetal androgen excess disorders, including congenital 21-hydroxylase deficiency and congenital adrenal virilizing tumors, develop features characteristic of PCOS during adulthood despite the normalization of androgen excess after birth. The potential mechanisms of fetal androgen excess leading to a PCOS phenotype in humans are not clearly understood. However, maternal and/or fetal hyperandrogenism can provide a plausible mechanism for fetal programming of PCOS, and this, in part, may be genetically determined. Thus, genetic association studies have indicated that common polymorphic variants of genes determining androgen activity or genes that influence the availability of androgens to target tissues are associated with PCOS and increased androgen levels. These genomic variants may provide the genetic link to prenatal androgenization in human PCOS. Conclusion Prenatal androgenization of the female fetus induced by genetic and environmental factors, or the interaction of both, may program differentiating target tissues toward the development of PCOS phenotype in adult life.
Article
Objective: To assess the effectiveness of metformin in improving clinical and biochemical features of polycystic ovary syndrome. Design: Systematic review and meta-analysis. Data sources: Randomised controlled trials that investigated the effect of metformin compared with either placebo or no treatment, or compared with an ovulation induction agent. Selection of studies: 13 trials were included for analysis, including 543 women with polycystic ovary syndrome that was defined by using biochemical or ultrasound evidence. Main outcome measure: Pregnancy and ovulation rates. Secondary outcomes of clinical and biochemical features of polycystic ovary syndrome. Results: Meta-analysis showed that metformin is effective in achieving ovulation in women with polycystic ovary syndrome, with odds ratios of 3.88 (95% confidence interval 2.25 to 6.69) for metformin compared with placebo and 4.41 (2.37 to 8.22) for metformin and clomifene compared with clomifene alone. An analysis of pregnancy rates shows a significant treatment effect for metformin and clomifene (odds ratio 4.40, 1.96 to 9.85). Metformin has an effect in reducing fasting insulin concentrations, blood pressure, and low density lipoprotein cholesterol. We found no evidence of any effect on body mass index or waist:hip ratio. Metformin was associated with a higher incidence of nausea, vomiting, and other gastrointestinal disturbance. Conclusions: Metformin is an effective treatment for anovulation in women with polycystic ovary syndrome. Its choice as a first line agent seems justified, and there is some evidence of benefit on variables of the metabolic syndrome. No data are available regarding the safety of metformin in long term use in young women and only limited data on its safety in early pregnancy. It should be used as an adjuvant to general lifestyle improvements and not as a replacement for increased exercise and improved diet.
Article
• Decrease in hair density in the central (vertex, mid and frontal) scalp, bitemporal and parietal regions in women. • Miniaturization of affected hairs. • Two ages of onset: early (post-puberty to third decade) and late (age 40+ years). • Signs of hyperandrogenism (hirsutism, irregular periods) or hyperandrogenemia occur in a subset of women with female pattern hair loss (FPHL) but most women with FPHL have neither. • Many, but not all, affected women respond to antiandrogens or 5α-reductase inhibitors with increased hair growth indicating an androgen etiology in at least some cases of FPHL.