ArticlePDF AvailableLiterature Review

Effects of myo-inositol in women with PCOS: A systematic review of randomized controlled trials

Taylor & Francis
Gynecological Endocrinology
Authors:

Abstract and Figures

Polycystic ovary syndrome (PCOS) affects 5%-10% of women in reproductive age, and it is the most common cause of infertility due to ovarian dysfunction and menstrual irregularity. Several studies have reported that insulin resistance is common in PCOS women, regardless of the body mass index. The importance of insulin resistance in PCOS is also suggested by the fact that insulin-sensitizing compounds have been proposed as putative treatments to solve the hyperinsulinemia-induced dysfunction of ovarian response to endogenous gonadotropins. Rescuing the ovarian response to endogenous gonadotropins reduces hyperandrogenemia and re-establishes menstrual cyclicity and ovulation, increasing the chance of a spontaneous pregnancy. Among the insulin-sensitizing compounds, there is myo-inosiol (MYO). Previous studies have demonstrated that MYO is capable of restoring spontaneous ovarian activity, and consequently fertility, in most patients with PCOS. With the present review, we aim to provide an overview on the clinical outcomes of the MYO use as a treatment to improve ovarian function and metabolic and hormonal parameters in women with PCOS.
Content may be subject to copyright.
Gynecological Endocrinology
2012
00
00
000
000
© 2012 Informa UK, Ltd.
10.3109/09513590.2011.650660
0951-3590
1473-0766
Gynecological Endocrinology, 2012; 1–7, Early Online
Copyright © 2012 Informa UK, Ltd.
ISSN 0951-3590 print/ISSN 1473-0766 online
DOI: 10.3109/09513590.2011.650660
Polycystic ovary syndrome (PCOS) affects 5%–10% of women in
reproductive age, and it is the most common cause of infertility
due to ovarian dysfunction and menstrual irregularity. Several
studies have reported that insulin resistance is common in PCOS
women, regardless of the body mass index. The importance
of insulin resistance in PCOS is also suggested by the fact that
insulin-sensitizing compounds have been proposed as putative
treatments to solve the hyperinsulinemia-induced dysfunction
of ovarian response to endogenous gonadotropins. Rescuing
the ovarian response to endogenous gonadotropins reduces
hyperandrogenemia and re-establishes menstrual cyclicity and
ovulation, increasing the chance of a spontaneous pregnancy.
Among the insulin-sensitizing compounds, there is myo-inosiol
(MYO). Previous studies have demonstrated that MYO is capable
of restoring spontaneous ovarian activity, and consequently
fertility, in most patients with PCOS. With the present review, we
aim to provide an overview on the clinical outcomes of the MYO
use as a treatment to improve ovarian function and metabolic
and hormonal parameters in women with PCOS.
Keywords: Infertility, insulin resistance, IVF, myo-inositol, oocyte
quality, PCOS
Introduction
Polycystic ovary syndrome (PCOS) is the most common cause
of infertility, ovarian dysfunction and menstrual irregularity,
aecting 5%–10% of women in reproductive age [1]. Both the
aetiology and diagnosis of the syndrome are controversial. Indeed,
the European Society of Human Reproduction and Embryology
and the American Society for Reproductive Medicine sponsored
a Consensus Meeting in Rotterdam (2003) [2,3], in order to reach
a general agreement of the scientic community on diagnostic
criteria for this syndrome.
Although nowadays the criteria established in Rotterdam are
widely accepted, they leave out a crucial condition related to
PCOS: insulin resistance.
Several studies have reported that insulin resistance is common
in PCOS women, regardless of the body mass index (BMI).
Indeed, hyperinsulinaemia due to insulin resistance occurs in
approximately 80% of women with PCOS and central obesity, as
well as in 30%–40% of lean women diagnosed with PCOS [4,5].
e exact cause of the insulin resistance observed in PCOS
women is unknown, although a post-receptor defect, that
could aect glucose transport, has been proposed [6,7]. Insulin
resistance is signicantly exacerbated by obesity, and it is a
key factor in the pathogenesis of anovulation and hyperandro-
genism [5,8]. e importance of insulin resistance in PCOS is
also suggested by the fact that insulin-sensitizing compounds,
such as metformin, pioglitazone and troglitazone, have been
proposed as treatment to solve the hyperinsulinemia-induced
dysfunction of ovarian response to endogenous gonadotropins.
Rescuing the ovarian response to endogenous gonadotropins
reduces hyperandrogenemia, re-establishes menstrual cyclicity
and ovulation, increasing the chance of a spontaneous pregnancy
[9–11].. In particular, metformin induces reduction of total and
free testosterone concentrations [12]. However, commonly used
insulin-sensitizing drugs, like metformin, can induce gastroin-
testinal side eects [13], possibly resulting in reduced patients’
compliance [13].
Further studies have suggested that impairment in the insulin
pathway could be due to a defect in the inositolphosphoglycans
(IPGs) second messenger [14,15]. IPGs are known to have a role
in activating enzymes that control glucose metabolism [16,17].
In PCOS women, a defect in tissue availability or altered metabo-
lism of inositol or IPGs mediators may contribute to insulin
resistance [18].
Inositol belongs to the vitamin B complex. Epimerization of
the six hydroxyl groups of inositol leads to the formation of up to
nine stereoisomers, including myo-inositol (MYO) and D-chiro-
inositol (DCI); both stereoisomers were used, as insulin sensitizer
drugs, in the treatment of PCOS treatments [19–23]. Human
adults consume approximately 1 g of inositol (mainly MYO) per
day in dierent biochemical forms [18]. Circulating free MYO
is taken up by most tissues by a membrane-associated sodium-
dependent inositol co-transporter; inositol uptake is inhibited by
glucose [24]. In particular, it was shown that MYO had 10 times
more anity for the transporter compared to DCI [25].
Data from other groups have shown that DCI is synthesized by
an epimerase that converts MYO into DCI, with each tissue having
its own particular conversion rate, likely due to the specic needs
for the two dierent molecules [26,27]. In particular, it was shown
that the DCI to mass index (MI) ratio was itself insulin depen-
dent. In fact, in subjects suering from type 2 diabetes, the DCI/
MI ratio was reduced [14,15,27,28], and less DCI was synthesized
due to a reduction in epimerase activity [14,15,27,28].
All of these studies were performed on insulin sensitive tissues,
such as muscle and liver. However, unlike tissues such as muscle
and liver, ovaries do not become insulin resistant [29–31].
REVIEW ARTICLE
Effects of myo-inositol in women with PCOS: a systematic review of
randomized controlled trials
V. Unfer1, G. Carlomagno1, G. Dante2 & F. Facchinetti2
1Gynecology Association Unfer Costabile (A.G.UN.CO.), Obstetrics and Gynecology Center, Rome, Italy and
2Mother-Infant Department, University of Modena and Reggio Emilia, Modena, Italy
Correspondence: Vittorio Unfer, Rome 00155, Italy. Tel: 3393690338. E-mail: vunfer@gmail.com
Gynecol Endocrinol Downloaded from informahealthcare.com by University Library Utrecht on 03/28/12
For personal use only.
2 V. Unfer et al
Gynecological Endocrinology
Furthermore, elevated concentrations of MYO in human folli-
cular uid play a role in follicular maturity and provide a marker
of good-quality oocytes [32,33].
Previous studies have demonstrated that MYO is capable of
restoring spontaneous ovarian activity, and consequently fertility,
in most patients with PCOS [21].
Here, we aim to provide an overview on the clinical outcomes
of MYO as a treatment to improve ovarian function and meta-
bolic and hormonal parameters in women with PCOS.
Methods
Systematic literature search was performed in December 2010
in the following electronic databases: Medline, Amed, and e
Cochrane Library. We performed a search over the period January
1999 to November 2010 and only randomized controlled clinical
trials (RCT) were included.
Search terms were as follows: “myo-inositol,” “inositol,” “poly-
cystic ovary syndrome,” “oocyte quality,” “ovarian stimulation,
“in vitro fertilization,” “ovarian function,” “insulin resistance.
No language restrictions were imposed. Further relevant
papers were located by hand-searching the reference lists of recent
systematic reviews. Only human studies were included. Data from
treatments with myo-inositol in combination with other drugs as
well as animal and in vitro investigations were excluded.
We obtained hard copies of all the papers listed through our
own university library or interlibrary loans. All sources of infor-
mation obtained were read and evaluated by one of us (G.D.), and
successively checked independently by the other authors (V.U.).
Results of the literature search
Decision tree is reported in Figure 1. A total of 70 studies were
identied; 49 out of 70 were excluded because of not involving
MYO treatment in women with PCOS. e remaining 21 trials
were considered eligible for this review. Six of them [20,22,34–37]
were RCTs (level of evidence Ib) and met the selection criteria for
this review and are presented in Table I. Four trials [22,34–36]
evaluated the eect of MYO administration on hormonal levels.
In one trial, it evaluated the eects of MYO on oocyte quality [20],
and in another one [37] data on ovarian function improvement
aer MYO administration were reported.
All the subjects analysed in these studies were PCOS patients.
Among the six studies that were RCTs, one trial was a random-
ized controlled MYO vs. folic acid [34]; two were double-blind
randomized controlled trial MYO vs. folic acid [22,35]; one was a
prospective randomized controlled MYO vs. folic acid [20]; one
was a randomized controlled MYO vs. metformin [36] and in
four of these trials [20,22,35,36] the dosage of 4 g of MYO was
used; in one trial [34] 2 g of MYO were used.
e last paper [37] described in Table I is not considered in our
discussion because it was performed on patients who were treated
with a multivitamin complex.
In the study of Genazzani et al. [34], 20 PCOS patients were
recruited, ve patients were amenorrheic and 15 oligomenorrheic.
Ten of the 20 patients (Group A) were randomly assigned to be
treated with MYO 2 g plus folic acid 200 g every day (Inofolic®,
LO.LI. Pharma, Rome, Italy). e other patients (Group B, control
group) were administered only folic acid at the daily dosage of 200
g. No changes of life style or diet were required from the patients.
Endocrine prole was evaluated aer treatment, on day 7 of the
rst menstrual cycle occurring aer the 10–12th week of treat-
ment. Consistent and signicant changes were observed in Group
A. Indeed, plasma luteinizing hormone (LH), prolactin (PRL), LH/
follicle-stimulating hormone (FSH) ratio and insulin levels signi-
cantly decreased; furthermore, the homeostatic model assessment
(HOMA) index that measures insulin resistance was also reduced.
On the other hand, the index of insulin sensitivity glucose/insulin
ratio signicantly increased. Furthermore, aer oral glucose load,
both insulin response and the area under the curve (AUC) were
signicantly lower (p < 0.01 and p < 0.05, respectively).
Ferriman-Gallway score decreased aer 12 weeks of MYO
administration although the reduction was not statistically signif-
icant (22.7 + 1.4 to 18 + 0.8). Ovarian volumes were signicantly
reduced (12.2 + 0.6 to 8.7 + 0.8 ml, p < 0.05). No changes were
observed in Group B.
Furthermore, as long as the patients were treated with
MYO, the normal menstrual cycles was restored, while patients
assigned to the Group B remained oligomenorrheic throughout
the study.
Figure 1. Flow chart of studies
Gynecol Endocrinol Downloaded from informahealthcare.com by University Library Utrecht on 03/28/12
For personal use only.
Myo-inositol in women with PCOS 3
Copyright ©  Informa UK, Ltd.
Table I. Eligible RCTs where MYO have been evaluated for the treatment of PCOS patients.
Reference Study design Duration Intervention N° of subjects Inclusion criteria Exclusion criteria Assessment of the response Results
34 Randomized,
controlled vs.
folic acid
12 weeks MYO 2 g FA
200 mg/day
N°= 20
Treatment: 10
Placebo: 10
Presence of micropolycystic
ovaries at ultrasound; mild-
to-severe hirsutism and/
or acne; oligomenorrhea or
amenorrhoea; absence of
enzymatic adrenal deciency
and/or other endocrine disease;
normal PRL levels (range
5–25 ng/ml); no hormonal
treatment for at least 6 months
before the study.
Not described LH, FSH, PRL, E2, A, 17OHP, T, insulin,
cortisol, OGTT* for insulin, glucose,
C-peptide determinations, vaginal
ultrasound examination Feriman-Gallway
score, BMI, HOMA
LH, PRL, T, insulin levels, LH/FSH results
weresignicantly reduced. Insulin sensitivity
results were signicantly improved. Menstrual
cyclicity was restored in all amenorrheic and
oligomenorrheic subjects.
35 Double-blind,
randomized,
controlled vs.
folic acid
12–16
weeks
MYO 4 g FA
400 mg/day
= 42
Treatment: 23
Placebo: 19
Presence of oligomenorrhea,
high serum-free testosterone
level and/or hirsutism presence
of micropolycystic ovaries at
ultrasound
Not described Systolic/diastolic blood pressure,
triglycerides, cholesterol, BMI, waist-
to-hip ratio, plasma glucose and insulin
sensitivity, total/free T, DHEAS, SHBG, A,
progesterone peak value
MI increased insulin sensitivity, improved
glucose tolerance and decreased glucose-
stimulated insulin release. ere was a
decrement in serum total T and serum-free
T concentrations. In addition, there was a
decrement in systolic and diastolic blood
pressure. Plasma triglycerides and total
cholesterol concentration decreased.
21 Prospective,
randomized,
controlled vs.
folic acid
During
ovulation
induction
for ICSI
MYO 4 g FA
200 mg/day
= 60
Treatment: 30
Placebo: 30
Age: <40 years PCOS
women diagnosed byoiligo
amenorrhea,hyperandroge
nism orhyperandrogenemia
andtypical features of ovarieson
ultrasound scan
Other medical
conditions causing
ovulatory disorders:
hyperinsulinemia,
hyperprolactinemia,
hypothyroidism, or
androgen excess,
such as adrenal
hyperplasia or
Cushing syndrome
Number of morphologically mature
oocytes retrieved, embryo quality,
pregnancy and implantation rates. Total
number of days of FSH stimulation, total
dose of gonadotropin administered, E2
level on the day of hCG administration,
fertilization rate per number of retrieved
oocytes, embryo cleavage rate, live birth
and miscarriage rate, cancellation rate,
and incidence of moderate or severe
ovarian hyperstimulation syndrome
Total r-FSH units and number of days of
stimulation were signicantly reduced in the
myo-inositol group. Peak E2 levels at hCG
administration were signicantly lower in
patients receiving myo-inositol. e mean
number of oocytes retrieved did not dier in
the two groups, whereas in the group cotreated
with myo-inositol the mean number of
germinal vesicles and degenerated oocytes was
signicantly reduced, with a trend for increased
percentage of oocytes in metaphase II
22 Double-blind,
randomized,
controlled vs.
folic acid
16 weeks MYO 4 g FA
200 mg/day
= 92
Treatment: 45
Placebo: 47
Age: <35 years women with
oiligoamenorrhea, amenorrhea
and PCOS ovaries. Ovaries were
described as polycystic (PCOs)
about the criteria of Adams
et al.**
Patients with
signicant
hyperprolactinemia,
abnormal thyroid
function tests and
congenital adrenal
hyperplasia.
Ovarian activity was monitored using
serum E2, P and LH. Ovulation frequency
was calculated using the ratio of luteal
phase weeks to observation weeks.
Inibin-b, fasting glucose, fasting insulin,
or insulin AUC, VLDL, LDL, HDL, total
cholesterol, triglycerides, BMI.
Benecial eect of MYO treatment upon
ovarian function, anthropometric measures
and lipid proles
36 Randomized,
controlled vs.
metformin
Until the
end of the
study or
positive
pregnancy
test
MYO 4 g FA
400 mg/day
= 120
Treatment:
60Placebo: 60
Age: <35 years Women with
PCOS dened by Rotterdam
Criteria
Other medical
condition
causing ovulatory
dysfunction, tubal
defects, semen
parameters defects.
Restoration of spontaneous ovarian
activity by weekly serum P dosage
and a transvaginal ultrasound scan
documenting the presence of follicular
growth or luteal cyst
Both metformin and MYO can be considered
as rst-line treatment for restoring normal
menstrual cycles in most patients with PCOS,
even if MI treatment seems to be more
37 Double-blind,
randomized,
controlled vs.
placebo
16 weeks Inositol
200 mg/day
= 283
Treatment: 136
Placebo: 147
Age: <35 years Women
with oligomenorrhea,
amenorrhea and PCOS ovaries.
Ovaries weredescribed as
polycystic(PCOs) about the
criteria ofAdams et al.**
Patients with
signicant
hyperprolactinemia,
abnormal thyroid
function tests and
congenital adrenal
hyperplasia.
Ovarian activity was monitored using
serum E2, P and LH. Ovulation frequency
was calculated using the ratio of luteal
phase weeks to observation weeks.
Inibin-b, fasting glucose, fasting insulin,
or insulin AUC, VLDL, LDL, HDL, total
cholesterol, triglycerides, BMI.
eective than metformin
FA, folic acid; PRL, prolactin; E2, oestradiol; A, androstenedione; 17OHP, 17-hydroxy-progesterone; T, testosterone; P, progesterone; OGTT, oral glucose tolerance; BMI, body mass index; LH, luteinizing hormone; FSH, follicle stimulating
hormone; DHEAS, dehydroepiandrosterone; SHBG, sex hormone binding globulin; AUC, area under the curve of OGTT; VLDL, very-low-density lipoprotein; LDL, low-density lipoprotein; HDL, high-density lipoprotein.
aOGTT performed sampling 15 minutes before and 30, 60, 90, 120 and 240 minutes aer the oral assumption of 75 g of glucose.
bAdams J, Polson JW, Franks S. Prevalence of polycystic ovaries in women with anovulation and idiopathic hirsutism. Br Med J 1986;293:355–359.
Gynecol Endocrinol Downloaded from informahealthcare.com by University Library Utrecht on 03/28/12
For personal use only.
4 V. Unfer et al
Gynecological Endocrinology
Costantino et al. [35] recruited 42 patients; aer randomiza-
tion, 23 received 2 g of MYO plus 200 g of folic acid (Inofolic®,
LO.LI. Pharma) twice a day and 19 received 400 g folic acid alone
as placebo. Among the 42 women, seven had impaired glucose
tolerance and were assigned as follows: four of them received
MYO and three received placebo.
e study started when patients were in the follicular phase of
the menstrual cycle; no changes in usual habits for food, sport and
lifestyle were required.
During the present study, patient treated with MYO showed a
reduction in both systolic and diastolic pressure values (131 ± 2
to 127 ± 2 mmHg and 88 ± 1 to 82 ± 3 mmHg, respectively),
while these values increased in the placebo group (128 ± 1 to
130 ± 1 mmHg; p = 0.002 and 86 ± 7 to 90 ± 1 mmHg; p = 0.001,
respectively).
Furthermore, in the MYO treatment group, plasma triglycer-
ides decreased from 195 ± 20 to 95 ± 17 mg/dl and total cholesterol
signicantly decreased from 210 ± 10 to 171 ± 11 mg/dl. Although
there was no change in the fasting plasma insulin and glucose
concentration in either group, AUC, for both insulin and glucose,
decreased during the oral glucose tolerance test (8.54 ± 1.149
to 5.535 ± 1.792 µU/ml/min; p = 0.03 and 12.409 ± 686 to
10.452 ± 414 mg/dl/min; p = 0.04, respectively) for MYO-treated
patients. No changes were observed in the placebo group.
Consequently, the composite whole body insulin sensi-
tivity index (ISI) signicantly increased from 2.80 ± 0.35 to
5.05 ± 0.59 mg/dl in the MYO group, while it did not change in
the placebo group. Ovulation was restored in 16 (69.5%) women
belonging to the MYO group and four(21%) belonging to the
placebo group (p = 0.001). Aer treatment, the progesterone (P)
peak was higher in the MYO group (15.1 ± 2.2 ng/ml) compared
to placebo. Furthermore, a signicant reduction in total serum T
(99.5 ± 7 to 34.8 ± 4.3 ng/dl, p = 0.003) and in free T (0.85 ± 0.11
to 0.24 ± 0.03 ng/dl, p = 0.01) was observed.
Testosterone reduction was accompanied by an increase in
serum sex hormone binding globulin. Furthermore, there was
reduction of more than 50% in the serum dehydroepiandros-
terone sulphate in the MYO group (366 ± 47 to 188 ± 24 µg/dl;
p = 0.003), while it was not signicant in the placebo group.
Papaleo et al. [20] broaden the clinical use of MYO by evaluating
its eect on oocyte quality and the ovarian stimulation protocol
for PCOS women. Sixty women were enrolled in the study; aer
randomization, 30 were assigned to receive 2 g MYO and 200 g
folic acid (Inofolic®, LO.LI. Pharma) twice a day (Group A); 30
received 400 g folic acid only (Group B).
In the Group A (Inofolic®), the stimulation protocol was
milder and shorter. Indeed, both the total recombinant FSH
(r-FSH units) (1958 ± 695 vs. 2383 ± 578; p = 0.01) and
number of stimulation days (11.4 ± 0.9 vs. 12.4 ± 1.4; p < 0.01)
were signicantly reduced. Furthermore, oestradiol (E2) levels
(2232 ± 510 vs. 2713 ± 595 pg/ml; p < 0.02) aer human chori-
onic gonadotropin administration were signicantly lower in
the Group A. ese resulted in signicant lower number of
cancelled cycles because of hyperstimulation risk (E2 > 4000
pg/ml). e number of oocytes retrieved did not dier between
the two groups, whereas in the group treated with MYO the
number of immature oocytes and degenerated oocytes was
signicantly reduced (1.0 ± 0.9 vs. 1.6 ± 1.0; p = 0.01), with a
trend for increased percentage of metaphase II stage oocytes
(0.82 ± 0.11% vs. 0.75 ± 0.15%).
No dierences were reported in fertilization and cleavage
rates, the number of transferred embryos, and the number of top-
quality transferred embryos and pregnancy rate.
Gerli et al. [22] enrolled 92 patients to evaluate MYO eects
on ovarian and metabolic factors; 45 received 2 g MYO combined
with 200 g folic acid twice a day (Inofolic®, LO.LI. Pharma), 47
received 400 g folic acid as placebo.
ere were eight conceptions and one miscarried in the rst
trimester. However, only 42 of the patients declared before the
study that they wished to conceive. Among these, the distribution
of pregnancy was: placebo one out of 19 patients, while in the
MYO group it was four out of 23 patients.
An intention to treat analysis revealed that eight of 45 MYO
patients failed to ovulate during treatment, compared with 17 out
of 47 placebo-treated patients (p = 0.04); the MYO-treated group
had a signicantly increased frequency of ovulation compared
with the placebo group. According to these data, the concen-
trations of P recorded during monitoring of ovarian function
indicated that most of the ovulations showed normal endocrine
proles during both MYO and placebo treatment. All patients
started treatment outside the luteal phase, and the delay to the
rst ovulation aer starting the program was signicantly shorter
in the MYO-treated group (24.5 vs. 40.5, p = 0.02).
e analysis of E2, inhibin-B, and T concentrations on the
rst and eighth day of treatment showed that the MYO-treated
group had a signicant (p = 0.03) increase in E2 levels, whereas
the control group showed no change. ere was no change in
circulating levels of inhibin-B or T concentrations.
e BMI decreased signicantly in the MYO group (p = 0.04).
No change was observed in the waist-to-hip ratio in either group.
Circulating leptin concentration declined in the MYO group, in
contrast to the control group, but there was no change recorded
in the fasting glucose, fasting insulin or insulin AUC in response
to the glucose challenge in either group.
e very-low-density lipoprotein showed little change during
the treatment period, but the low-density lipoprotein (LDL)
showed a trend toward reduction, and the high-density lipopro-
tein (HDL) increased signicantly in the MYO group.
Raone et al. [36] performed a study aiming to compare the
eects of metformin and MYO on PCOS patients: 120 women
were recruited; 60 patients were treated with metformin 1500 mg/
day (Glucophage®, Merck Pharma, Rome, Italy), while 60
received 4 g MYO plus 400 g folic acid (Inofolic®).
Among the patients treated with metformin, 50% restored
spontaneous ovulation activity; in these patients, ovulation
occurred aer 16.7 (+2.5) days from the day 1 of the menstrual
cycle. Pregnancy occurred spontaneously in 11 of these patients;
seven women dropped out. e remaining 42 patients were treated
with 1500 mg of metformin plus r-FSH for a maximum of three
cycles. Pregnancy occurred in 11 women, nine of these pregnan-
cies occurred in the metformin-resistant patients (n = 23), two in
the group which had ovulation restored with metformin alone.
e total pregnancy rate was 36.6%, ve of the 22 pregnancies
(22.7%) evolved in spontaneous abortion at 9 weeks of gestation.
Seven subjects in the metformin group dropped out because of
the development of side eects and loss of follow-up.
In the MYO group, 65% of patients restored spontaneous
ovulation activity, ovulation occurred aer a mean of 14.8 (+1.8)
days from the day 1 of the menstrual cycle. Pregnancy occurred
spontaneously in 18 of these patients and four women dropped
out. e remaining 38 patients were treated with 4 g/day MYO,
400 g/day folic acid and r-FSH in small doses (37.5 U/day for
three cycles). Pregnancy occurred in a total of 11 women, eight
of these pregnancies occurred in the MYO-resistant patients
(n = 17), three in the group which had ovulation restored with
MYO alone.
Gynecol Endocrinol Downloaded from informahealthcare.com by University Library Utrecht on 03/28/12
For personal use only.
Myo-inositol in women with PCOS 5
Copyright ©  Informa UK, Ltd.
e total pregnancy rate was 48.4%, six of the 29 pregnan-
cies (20.6%) evolved in spontaneous abortion. e ecacy in
restoring regular ovulation was evaluated by comparing both
the percentage of patients who responded to the treatment and
the median length of follicular phase in metformin and in MYO
group: ovulation occurred aer a mean 16.7 (+2.5) days from the
day 1 of the menstrual cycle in the metformin group and aer
a mean 14.8 (+1.8) in the MYO group. e median between
metformin and MYO diered signicantly (p < 0.003). Inclusion,
exclusion criteria and the main outcome measures for all studies
are described in Table I.
Conclusions
PCOS is one of the most common endocrine disorders aecting
women, it is the most common cause of female infertility and it
is characterized by a combination of hyperandrogenism, chronic
anovulation and irregular menstrual cycle [38,39]. Several
patients aected by PCOS are also aected by insulin resistance
although they do not show signs of diabetes [4,10]; furthermore,
insulin resistance is not linked to the BMI [7,40]. PCOS-induced
insulin resistance determines a higher risk for the development
of type 2 diabetes, hypertension and dyslipidemia, all elements of
the metabolic syndrome [41].
MYO is an important constituent of follicular microenviron-
ment, playing a determinant role in both nuclear and cytoplasmic
oocyte development. Indeed, inositol 1,4,5-triphosphate modu-
lates intracellular Ca2+ release.
Calcium signaling in oocytes has been extensively studied in
various species because of its putative role in oocyte maturation
and the early stages of fertilization [42,43]. It has been demon-
strated that fully grown mammalian germinal vesicles (GV), that
exhibit spontaneous intracellular calcium oscillations, are asso-
ciated with a higher incidence of GV breakdown. MYO supple-
mentation was suggested to promote meiotic progression of these
GV. Indeed, high concentration of MI in the uids of the human
follicles strongly associates with good-quality oocytes [44].
Despite the relatively high number of reports evaluating clin-
ical studies that used MYO as a treatment in women with PCOS,
only few of them were designed as RCTs (level of evidence Ib).
Four articles [22,34–36] evaluated the eects of MYO admin-
istration on hormonal and metabolic parameters in patients with
PCOS. e results of these studies support the hypothesis of a
primary role of IPG as second messenger of insulin signal and
demonstrate that MYO administration signicantly aects the
hormonal milieu in PCOS patients.
ere are specic positive eects of MYO treatment on insulin
plasma levels and on insulin response to oral glucose load.
Indeed, MYO decreased insulin plasma levels, glucose/insulin,
HOMA index as well as other hormonal parameters such as LH,
LH/FSH, testosterone and PRL. Furthermore, MYO inositol is
able to induce normal menstrual cycles. ese trials supported
the hypothesis that MYO supplementation induces the reduction
of insulin levels probably by inducing an increase of IPG levels;
therefore, higher IPG levels could be able to amplify insulin
signal.
In particular, in two studies [20,34] the authors suggest that
a deciency in the precursors of IPG such as MYO and/or DCI
might be an additional cofactor contributing to the pathophysi-
ology of the insulin resistance of PCOS patients [18].
All the PCOS patients showed a signicant improvement of
typical hormonal parameters: indeed, aer MYO treatment
LH levels, LH/FSH ratio, T and HOMA index were decreased.
Furthermore, the insulin AUC aer glucose load was reduced,
being a clear signal of the improved peripheral sensitivity.
Interestingly, PRL plasma levels also resulted signicantly lower
under MYO administration.
In addition to this, Gerli et al. demonstrated a signicant
reduction in weight in the patients treated with MYO, in contrast
to the placebo group where the BMI increased. Associated with
the weight loss, it was possible to observe a signicant reduction
in circulating leptin and an increase in HDL concentrations, while
LDL showed a trend toward reduction.
ese data on HDL cholesterol were the rst evidence showing
that MYO treatment could be useful in reducing the risk of
cardiovascular diseases in PCOS women.
Several trials showed that insulin sensitizer agents, such as
metformin and MYO, are the rst-line treatment to restore
normal menstrual cycles in women suering from PCOS [9–13],
suggesting that an endocellular defect of the precursor of IPG
such as MYO and/or DCI might trigger the compensatory hyper-
insulinemia in most PCOS subjects.
Moreover, Raone et al. showed that MYO slightly improves
pregnancy rate compared to metformin.
ese ndings further support the hypothesis that the reduc-
tion of insulin levels induced by MYO oral supplementation
depends on the increased availability of the main precursor of
IPG insulin second messenger.
Furthermore, in one trial [20] it has been shown that MYO is
eective in ovarian stimulation protocols in women with PCOS.
PCOS women have an increased risk of hyperstimulation
syndrome [45]. Indeed, high levels of serum ovarian androgens
are implicated in production of elevated serum E2 levels aer
gonadotropin ovarian stimulation. PCOS patients treated with
MYO + gonadotropin showed a signicant reduction in E2 levels
aer hGC administration. is was reected on the lower number
of in vitro fertilization (IVF) cycles cancelled because of high E2
levels (sign of hyperstimulation syndrome [20]).
Literature studies already suggested that MYO has positive
eect on developmental competence of maturing oocytes [46].
In line with this evidence, a recent clinical trial aiming to
compare the eect of MYO or DCI supplementation on oocyte
quality of PCOS patients showed that only MYO rather than DCI
is able to improve oocyte quality [47]. Based on these data, we
developed a theory that identied a “DCI paradox [48],” where we
suggest that ovaries in PCOS patients likely present an enhanced
MI to DCI epimerization that leads to a MI tissue depletion;
this, in turn, could eventually be responsible for the poor oocyte
quality characteristic of these patients [49].
Remarkably, in all the studies analysed, no side eects were
reported at the doses of both 2 and 4 g/day, thus resulting in a
high patient compliance. e 4 g/day treatment regimen is useful
to treat all the symptom spectrum, resulting in a more complete
and eective treatment.
In conclusion, by analyzing dierent studies focused on MYO
supplementation to improve several of the hormonal distur-
bances of PCOS, we provide a level Ia evidence of MYO eective-
ness. MYO mechanism of action appears to be mainly based on
improving insulin sensitivity of target tissues, resulting in a posi-
tive eect on the reproductive axis (MYO restores ovulation and
improves oocyte quality) and hormonal functions (MYO reduces
clinical and biochemical hyperandrogenism and dyslipidemia)
through the reduction of insulin plasma levels.
Declaration of Interest: e authors report no conict of
interest.
Gynecol Endocrinol Downloaded from informahealthcare.com by University Library Utrecht on 03/28/12
For personal use only.
6 V. Unfer et al
Gynecological Endocrinology
References
1. Homburg R. Polycystic ovary syndrome - from gynaecological curiosity
to multisystem endocrinopathy. Hum Reprod 1996;11:29–39.
2. Revised 2003 consensus on diagnostic criteria and long-term health
risks related to polycystic ovary syndrome (PCOS). Hum Reprod
2004;19:41–47.
3. Revised 2003 consensus on diagnostic criteria and long-term health
risks related to polycystic ovary syndrome. Fertil Steril 2004;81:19–25.
4. Genazzani AD, Battaglia C, Malavasi B, Strucchi C, Tortolani F, Gamba
O. Metformin administration modulates and restores luteinizing
hormone spontaneous episodic secretion and ovarian function in
nonobese patients with polycystic ovary syndrome. Fertil Steril
2004;81:114–119.
5. Ciampelli M, Fulghesu AM, Cucinelli F, Pavone V, Ronsisvalle E, Guido
M, Caruso A, Lanzone A. Impact of insulin and body mass index on
metabolic and endocrine variables in polycystic ovary syndrome.
Metab Clin Exp 1999;48:167–172.
6. Baillargeon JP, Nestler JE. Commentary: polycystic ovary syndrome:
a syndrome of ovarian hypersensitivity to insulin? J Clin Endocrinol
Metab 2006;91:22–24.
7. Dunaif A. Insulin resistance and the polycystic ovary syndrome:
mechanism and implications for pathogenesis. Endocr Rev
1997;18:774–800.
8. Cascella T, Palomba S, De Sio I, Manguso F, Giallauria F, De Simone
B, Tafuri D, Lombardi G, et al. Visceral fat is associated with
cardiovascular risk in women with polycystic ovary syndrome. Hum
Reprod 2008;23:153–159.
9. Baillargeon JP, Iuorno MJ, Nestler JE. Insulin sensitizers for polycystic
ovary syndrome. Clin Obstet Gynecol 2003;46:325–340.
10. De Leo V, la Marca A, Petraglia F. Insulin-lowering agents in
the management of polycystic ovary syndrome. Endocr Rev
2003;24:633–667.
11. Genazzani AD, Lanzoni C, Ricchieri F, Baraldi E, Casarosa E,
Jasonni VM. Metformin administration is more eective when
non-obese patients with polycystic ovary syndrome show both
hyperandrogenism and hyperinsulinemia. Gynecol Endocrinol
2007;23:146–152.
12. Nestler JE, Jakubowicz DJ. Lean women with polycystic ovary
syndrome respond to insulin reduction with decreases in ovarian
P450c17 alpha activity and serum androgens. J Clin Endocrinol Metab
1997;82:4075–4079.
13. Lord JM, Flight IH, Norman RJ. Metformin in polycystic ovary syndrome:
systematic review and meta-analysis. BMJ 2003;327:951–953.
14. Kennington AS, Hill CR, Craig J, Bogardus C, Raz I, Ortmeyer HK,
Hansen BC, et al. Low urinary chiro-inositol excretion in non-insulin-
dependent diabetes mellitus. N Engl J Med 1990;323:373–378.
15. Asplin I, Galasko G, Larner J. chiro-inositol deciency and insulin
resistance: a comparison of the chiro-inositol- and the myo-inositol-
containing insulin mediators isolated from urine, hemodialysate, and
muscle of control and type II diabetic subjects. Proc Natl Acad Sci U S
A 1993;90:5924–5928.
16. Cohen P. e twentieth century struggle to decipher insulin signalling.
Nat Rev Mol Cell Biol 2006;7:867–873.
17. Baillargeon JP, Nestler JE, Ostlund RE, Apridonidze T, Diamanti-
Kandarakis E. Greek hyperinsulinemic women, with or without
polycystic ovary syndrome, display altered inositols metabolism. Hum
Reprod 2008;23:1439–1446.
18. Baillargeon JP, Diamanti-Kandarakis E, Ostlund RE Jr, Apridonidze
T, Iuorno MJ, Nestler JE. Altered D-chiro-inositol urinary clearance
in women with polycystic ovary syndrome. Diabetes Care
2006;29:300–305.
19. Nestler JE, Jakubowicz DJ, Reamer P, Gunn RD, Allan G. Ovulatory and
metabolic eects of D-chiro-inositol in the polycystic ovary syndrome.
N Engl J Med 1999;340:1314–1320.
20. Papaleo E, Unfer V, Baillargeon JP, Fusi F, Occhi F, De Santis L.
Myo-inositol may improve oocyte quality in intracytoplasmic sperm
injection cycles. A prospective, controlled, randomized trial. Fertil
Steril 2009;91:1750–1754.
21. Papaleo E, Unfer V, Baillargeon JP, De Santis L, Fusi F, Brigante
C, Marelli G, et al. Myo-inositol in patients with polycystic ovary
syndrome: a novel method for ovulation induction. Gynecol Endocrinol
2007;23:700–703.
22. Gerli S, Papaleo E, Ferrari A, Di Renzo GC. Randomized, double blind
placebo-controlled trial: eects of myo-inositol on ovarian function
and metabolic factors in women with PCOS. Eur Rev Med Pharmacol
Sci 2007;11:347–354.
23. Minozzi M, D’Andrea G, Unfer V. Treatment of hirsutism with
myo-inositol: a prospective clinical study. Reprod Biomed Online
2008;17:579–582.
24. Coady MJ, Wallendor B, Gagnon DG, Lapointe JY. Identication
of a novel Na+/myo-inositol cotransporter. J Biol Chem
2002;277:35219–35224.
25. Ostlund RE Jr, Seemayer R, Gupta S, Kimmel R, Ostlund EL, Sherman
WR. A stereospecic myo-inositol/D-chiro-inositol transporter in
HepG2 liver cells. Identication with D-chiro-[3-3H]inositol. J Biol
Chem 1996;271:10073–10078.
26. Larner J. D-chiro-inositol--its functional role in insulin action and its
decit in insulin resistance. Int J Exp Diabetes Res 2002;3:47–60.
27. Sun TH, Heimark DB, Nguygen T, Nadler JL, Larner J. Both myo-inositol
to chiro-inositol epimerase activities and chiro-inositol to myo-inositol
ratios are decreased in tissues of GK type 2 diabetic rats compared to
Wistar controls. Biochem Biophys Res Commun 2002;293:1092–1098.
28. Larner J, Craig JW. Urinary myo-inositol-to-chiro-inositol ratios and
insulin resistance. Diabetes Care 1996;19:76–78.
29. Harwood K, Vuguin P, DiMartino-Nardi J. Current approaches to the
diagnosis and treatment of polycystic ovarian syndrome in youth.
Horm Res 2007;68:209–217.
30. Matalliotakis I, Kourtis A, Koukoura O, Panidis D. Polycystic
ovary syndrome: etiology and pathogenesis. Arch Gynecol Obstet
2006;274:187–197.
31. Rice S, Christoforidis N, Gadd C, Nikolaou D, Seyani L, Donaldson
A, Margara R, et al. Impaired insulin-dependent glucose metabolism
in granulosa-lutein cells from anovulatory women with polycystic
ovaries. Hum Reprod 2005;20:373–381.
32. Kane MT, Norris M, Harrison RA. Uptake and incorporation of inositol
by preimplantation mouse embryos. J Reprod Fertil 1992;96:617–625.
33. Chiu TT, Rogers MS, Law EL, Briton-Jones CM, Cheung LP, Haines CJ.
Follicular uid and serum concentrations of myo-inositol in patients
undergoing IVF: relationship with oocyte quality. Hum Reprod
2002;17:1591–1596.
34. Genazzani AD, Lanzoni C, Ricchieri F, Jasonni VM. Myo-inositol
administration positively aects hyperinsulinemia and hormonal
parameters in overweight patients with polycystic ovary syndrome.
Gynecol Endocrinol 2008;24:139–144.
35. Costantino D, Minozzi G, Minozzi E, Guaraldi C. Metabolic and
hormonal eects of myo-inositol in women with polycystic ovary
syndrome: a double-blind trial. Eur Rev Med Pharmacol Sci
2009;13:105–110.
36. Raone E, Rizzo P, Benedetto V. Insulin sensitiser agents alone and
in co-treatment with r-FSH for ovulation induction in PCOS women.
Gynecol Endocrinol 2010;26:275–280.
37. Gerli S, Mignosa M, Di Renzo GC. Eects of inositol on ovarian
function and metabolic factors in women with PCOS: a randomized
double blind placebo-controlled trial. Eur Rev Med Pharmacol Sci
2003;7:151–159.
38. Asunción M, Calvo RM, San Millán JL, Sancho J, Avila S, Escobar-
Morreale HF. A prospective study of the prevalence of the polycystic
ovary syndrome in unselected Caucasian women from Spain. J Clin
Endocrinol Metab 2000;85:2434–2438.
39. Knochenhauer ES, Key TJ, Kahsar-Miller M, Waggoner W, Boots LR,
Azziz R. Prevalence of the polycystic ovary syndrome in unselected
black and white women of the southeastern United States: a prospective
study. J Clin Endocrinol Metab 1998;83:3078–3082.
40. Nestler JE. Insulin resistance and the polycystic ovary syndrome: recent
advances. Curr Opin Endocrinol Diabetes Obes 2000;7:345–349.
41. Lobo RA, Carmina E. e importance of diagnosing the polycystic
ovary syndrome. Ann Intern Med 2000;132:989–993.
42. Pesty A, Lefèvre B, Kubiak J, Géraud G, Tesarik J, Maro B. Mouse
oocyte maturation is aected by lithium via the polyphosphoinositide
metabolism and the microtubule network. Mol Reprod Dev
1994;38:187–199.
43. DeLisle S, Blondel O, Longo FJ, Schnabel WE, Bell GI, Welsh
MJ. Expression of inositol 1,4,5-trisphosphate receptors changes
the Ca2+ signal of Xenopus oocytes. Am J Physiol 1996;270(4 Pt
1):C1255–C1261.
44. Chiu TT, Rogers MS, Briton-Jones C, Haines C. Eects of myo-inositol
on the in-vitro maturation and subsequent development of mouse
oocytes. Hum Reprod 2003;18:408–416.
45. Battaglia C, Mancini F, Persico N, Zaccaria V, de Aloysio D. Ultrasound
evaluation of PCO, PCOS and OHSS. Reprod Biomed Online
2004;9:614–619.
46. Goud PT, Goud AP, Van Oostveldt P, Dhont M. Presence and dynamic
redistribution of type I inositol 1,4,5-trisphosphate receptors in human
Gynecol Endocrinol Downloaded from informahealthcare.com by University Library Utrecht on 03/28/12
For personal use only.
Myo-inositol in women with PCOS 7
Copyright ©  Informa UK, Ltd.
oocytes and embryos during in-vitro maturation, fertilization and early
cleavage divisions. Mol Hum Reprod 1999;5:441–451.
47. Unfer V, Carlomagno G, Rizzo P, Raone E, Rose S. Myo-inositol
rather than D-chiro-inositol is able to improve oocyte quality in
intracytoplasmic sperm injection cycles. A prospective, controlled,
randomized trial. Eur Rev Med Pharmacol Sci 2011;15:452–457.
48. Carlomagno G, Unfer V, Rose S. e D-chiro-inositol paradox in the
ovary. Fertil Steril 2011;95:2515–2516.
49. Chattopadhayay R, Ganesh A, Samanta J, Jana SK, Chakravarty BN,
Chaudhury K. Eect of follicular uid oxidative stress on meiotic
spindle formation in infertile women with polycystic ovarian syndrome.
Gynecol Obstet Invest2010;69:197–202.
Gynecol Endocrinol Downloaded from informahealthcare.com by University Library Utrecht on 03/28/12
For personal use only.
... It has been shown that supplements have a high impact on the restoration of fertility. Myo-inositol has been demonstrated to improve ovarian function and increase the quality of oocytes, particularly in women with polycystic ovary syndrome (PCOS), leading to higher pregnancy rates [7,8]. In particular, several clinical trials demonstrate that its administration can have therapeutic effects in infertile women, and that it can also be useful as a preventive treatment during pregnancy [9]. ...
... (1) The participant refuses further participation or withdraws consent; (2) termination of the entire study; (3) occurrence of a spontaneous pregnancy; (4) failure to adhere to the study rules; (5) attendance of fewer than 75% of the exercise sessions by the participant in the intervention group; (6) occurrence of any other intervention or surgery during the study period that may alter the study outcome; (7) undesirable events related to the treatment, such as acute pain, musculoskeletal complaints, or acute mental or physical trauma preventing continuation of the study; (8) presence of a newly diagnosed malignancy; (9) any situation where oral therapy or physiotherapy is deemed unfit to continue according to the decision of the physician and physiotherapist. ...
Article
Full-text available
According to World Health Organization (WHO) data, 16% of people are affected by infertility across the globe. One underlying factor is the age-related decline of ovarian reserve (DOR), which can lead to a higher chance of infertility and has no widely accepted treatment currently. Specific supplements and moderate exercise have been shown to improve fertility; however, there is no consensus to date on the type of exercise providing the best results. Our goal is to develop a novel exercise program combined with natural supplements for the improvement of fertility. We also propose a single-centered, randomized, open-label clinical trial using our newly developed exercise in the intervention group, compared to walking and no exercise in the other groups, to investigate the benefits of this exercise program in the future. In this study, we developed a structured, novel combination of exercises focusing on the pelvic and ovarian regions, core strengthening and improvement of blood circulation in this region. The 70 min full body “reproductive gymnastics”, includes strengthening, stretching, and relaxation exercises combined with yoga-inspired moves and diaphragmatic breathing with meditation elements to activate the parasympathetic pathway and stress relief. We believe we can improve fertility through the combination of natural supplements and our targeted, moderate physiotherapy program in women with DOR.
... Позднее аналогичные результаты отмечены у худых пациенток с СПКЯ [22]. Начало истории инозитола свидетельствовало о его потенциале в лечении СПКЯ и требовало оценки механизмов его влияния на пострецепторный инсулиновый сигнал, так как разные ткани имеют различную выраженность ИР: мышцы, почки, печень становятся резистентными к инсулину, в отличие от ткани яичников [23]. Функции МИ и D-ХИ в яичниках различаются. ...
... Продолжение истории -оценка клинических результатов использования инозитола. Один из обзоров, включавших анализ 21 разнородного исследования, в том числе анализ использования МИ (500-1500 мг/сут), выявил значительное улучшение гормональных показателей: ЛГ, соотношение ЛГ/ФСГ, тестостерона, андростендиона, снижение концентрации общего холестерина и увеличение липопротеинов высокой плотности, инсулинемии и индекса HOMA-IR [23]. Наряду с улучшением метаболических показателей снизился ИМТ, улучшились менструальная функция и фертильность. ...
Article
Введение. Синдром поликистозных яичников является широко встречающейся патологией (ее распространенность варьирует в диапазоне 5–25%), которая сопровождается нарушениями фертильности и метаболизма. Избыточный вес или ожирение, как правило, висцеральное, – частые проявления данного синдрома, которые ассоциированы с инсулинорезистентностью, нарушениями гомеостаза глюкозы (пред- диабетом или сахарным диабетом 2-го типа) и гиперандрогенией. Потенциал коррекции метаболических нарушений включает наряду с изменением образа жизни использование инозитола. Материалы и методы. Обзор литературы проведен с использованием данных PubMed, Google Scholar, Elsevier. Для поиска применялись специфические слова и словосочетания: «синдром поликистозных яичников», «синдром поликистозных яичников и инсулинорезистентность», «инозитол», «мио-инозитол», «D-хиро-инозитол». Все найденные статьи были тщательно оценены и проанализированы. Результаты. Определена роль инозитолов в опосредовании действия инсулина и инсулинорезистентности, что базируется на их дефиците при синдроме поликистоза яичников. Приведены обоснования использования инозитолов в метаболической терапии данного синдрома и доказательства различий функций мио-инозитола и D-хиро-инозитола в яичниках. Продемонстрировано, что эффекты изомеров инозитола выходят за рамки метаболического воздействия на сигналы инсулина и оказывают влияние на гиперандрогению, менструальный цикл, овуляцию и другие показатели. Заключение. Проанализировав приведенные исследования, сделано заключение, что комбинация мио-инозитола и D-хиро-инозитола демонстрирует многоплановые преимущества в улучшении гормонального, гликемического и липидного профиля женщин с синдромом поликистоза яичников, высокую эффективность и хорошую переносимость. Introduction. Polycystic ovary syndrome is a common pathology (its prevalence varies in the range of 5–25%), accompanied by fertility disorders and metabolic disorders. Overweight or obesity, usually visceral, is a frequent manifestation of polycystic ovary syndrome, which are associated with insulin resistance, impaired glucose homeostasis (prediabetes or type 2 diabetes mellitus) and hyperandrogenism. The potential for correcting metabolic disorders includes, along with lifestyle modifications, the use of inositol. Materials and methods. The literature review was conducted using PubMed and Google Scholar, Elsevier data. Specific words were used for the search: "polycystic ovary syndrome", "polycystic ovary syndrome and insulin resistance", "inositol", "myo-inositol", "D-chiro-inositol". All the articles found have been carefully evaluated and analyzed. Results. The role of inositols in mediating the action of insulin and insulin resistance is substantiated, which is based on their deficiency in polycystic ovary syndrome. The substantiation of the use of inositols in the metabolic therapy of polycystic ovary syndrome and evidence of differences in the function of myo-inositol and D-chiro-inositol in the ovaries are presented. It has been demonstrated that the effects of inositol isomers go beyond the metabolic effect on insulin signals and have an effect on hyperandrogenism, menstrual cycle, ovulation, and other indicators. Conclusion. Summarizing the above studies, it was concluded that the combination of myo-inositol and D-chiro-inositol demonstrates multifaceted benefits in improving the hormonal, glycemic and lipid profiles of women with polycystic ovary syndrome with high efficacy and good tolerability.
... Weight management also showed gradual progress, with a slight reduction in BMI from 28 kg/m² to 26.5 kg/m², demonstrating the impact of dietary changes, Udvartana therapy, and regular exercise. 9 ...
Article
Full-text available
Background: Polycystic ovarian syndrome (PCOS) is a prevalent endocrine disorder in women of reproductive age characterized by hormonal imbalance, metabolic disruptions, and reproductive challenges. According to Ayurveda, PCOS is primarily associated with an imbalance of Kapha and Vata Doshas, requiring comprehensive holistic management. Objective: To assess the effectiveness of an integrative Ayurvedic and modern medical approach in managing symptoms and improving health outcomes in a case of PCOS. Methods: A single-case observational study was conducted on a 28-year-old woman diagnosed with PCOS exhibiting symptoms such as irregular menstruation, acne, and insulin resistance. Interventions included Ayurvedic herbal formulations (Kanchanar Guggulu, Shatavari, Ashokarishta), tailored dietary adjustments, lifestyle modifications, and supportive modern medical treatments over a period of three months. Results: Significant improvements were observed following integrative management, including regularization of menstrual cycles, substantial reduction in acne and associated discomfort, weight management, and notable improvements in insulin sensitivity and hormonal balance. Conclusion: The integrative Ayurvedic and modern medical approach demonstrates significant effectiveness in managing PCOS symptoms and improving overall reproductive and metabolic health in women, emphasizing the potential of holistic strategies in clinical practice.
... Yapılan klinik çalışmalarda miyoinositolün lipit profilinde iyileştirici etkide olduğu ve yüksek konsantrasyonlarda oositlerin kalitesini işlevini artırdığı görülmüştür. Bu sonuçlar bize miyo-inositolün adet döngüsünü düzenlediği, yumurtalamayı artırdığını, gebe kalma ihtimalini artırdığını göstermektedir (27). İnfertilite sorunu yaşayan ve diğer tedavilerden sonuç alınamayan hastalarda laparoskopik cerrahi yöntemi uygulanabilmektedir. ...
... When compared to metformin alone, the combination of ovarian morphology data in our study significantly reduced mean ovarian size and mean AFC. Likewise, Agarwal et al. revealed that MI enhanced the morphology of the ovaries in relations of size and stromal width (62). Metformin has a unique effect on the ovaries. ...
Article
Full-text available
Background: Polycystic ovary syndrome (PCOS) is the greatest mutual hormonal imbalance illness in females of childbearing age. In modern years, trainings have shown that inflammatory progressions stay complicated in ovulation and production a key character in ovarian follicular dynamics. Insulin sensitizers for instance myo-inositol and metformin are probable management selections for PCOS. Objective: evaluate the properties of combined metformin and ‎myoinositol against metformin alone on inflammatory markers (hs-CRP and ‎IL-18) in PCOS women. Methods: The patients' PCOS diagnosis was made using the Rotterdam criteria. Two groups of patients were identified: group MI+M received myo-inositol and metformin, and group M received metformin. Biochemical assessment, patient compliance, and reported adverse effects were among the examinations conducted at baseline and three months later. ‏. Results: The mean hs-CRP and IL-18 of the combination ‎and metformin groups before and after treatment are shown at baseline. After ‎treatment, the combination caused in a more important decrease in mean ‎hs-CRP and IL-18 than metformin.‎ Conclusion: This training unequivocally demonstrates the role myoinositol and metformin have in reducing inflammatory markers.Key: Polycystic Ovarian Syndrome, Myoinositol, c-reactive protein, interleukin 18.
Chapter
This chapter has attempted to streamline the concept of comprehensive, personalized dietetics treatment strategy for PCOS into one single-case example. A lot of emphasis has been put on the role of micronutrients in metabolic processing, hormone control, and reproductive functioning. The chapter stresses to fallacy the conventional dietetics approach that results in nonunique, uniform dietary therapeutic plans. By exploring the body’s most critical vitamins and minerals that are the direct causes of PCOS and are known for differential sparseness in the human body, the chapter demonstrated the potential to advance the next level of human nutrition. Namely, this point is also supported by the fact that artificial intelligence acts as a groundbreaking solution from another side, as it can work with complex health data and develop a schedule of customized microelement dosages for every human physiological profile that may cure such deficiency. Microelement optimization fused with AI-managed research then becomes a new frontier of personal nutrition, because it transfers a new, highly accurate, flexible, data-based tool from the hands of practicians to patients. This way, this study becomes a starting point that will guide a better future treatment for PCOS and analysis of personal dietetics concept.
Article
Systemic inflammation, often induced by elevated circulating lipopolysaccharide (LPS) levels, is a common consequence of intestinal epithelial barrier damage and microbial translocation. This condition is particularly prevalent in menopausal women, who are at increased risk for chronic inflammation and metabolic syndrome due to physiological changes during menopause. Myo-inositol has been shown to improve metabolic profiles in menopausal women with metabolic syndrome. In this study, we investigated whether small extracellular vesicles (sEVs) from human palatine tonsil-derived mesenchymal stem cells (T-MSCs) can restore circulating myo-inositol levels and promote myo-inositol synthesis in skeletal muscle under repeated LPS exposure, mimicking the intestinal leakage seen in menopausal women. Over 2 weeks period, LPS was repeatedly administered to mice, along with a group that also received T-MSC-derived sEVs. After 15 days, the expression of proteins involved in inositol synthesis in skeletal muscle, and serum inositol levels were measured. Additionally, intracellular inositol expression was compared in LPS-treated skeletal muscle cells with and without T-MSC sEVs treatment in vitro. Lastly, the protein and microRNA composition of T-MSC sEVs was analyzed. Our results demonstrated that T-MSC-derived sEVs significantly increased serum myo-inositol levels and enhanced the expression of myo-inositol synthesis proteins in mice exposed to LPS. Similarly, LPS-treated myotubes supplemented with T-MSC sEVs exhibited restored myo-inositol expression. Moreover, T-MSC sEVs were found to contain high levels of muscle-related proteins. These findings suggest that T-MSC sEVs may serve as a promising therapeutic strategy for mitigating the effects of intestinal leakage and chronic inflammation in menopausal women. By improving skeletal muscle mass and maintaining myo-inositol levels, T-MSC sEVs offer potential for addressing metabolic disturbances associated with menopause.
Article
Decreased Glucose transporter 4 (GLUT4) expression leads to abnormal glucose regulation. miR-93 regulates GLUT4 expression and studies have shown that, ginkgolide mediates miR-93 and GLUT4 in polycystic ovary syndrome (PCOS). 50 patients with PCOS and 50 healthy women were recruited. RT-qPCR detected miR-93 and GLUT4 expression. Luteinizing hormone (LH) (11.84 ± 4.08), T (76.87±30.24), FINs (20.06±11.37) and HOMA-IR (3.75±1.04) in the PCOS group was higher than control group (7.42±3.63, 43.58±13.9, 8.74±4.62, 1.55±0.39) ( P < 0.05). miR-93 expression in peripheral blood of PCOS group was significantly elevated, while GLUT4 mRNA expression was reduced ( P < 0.05). miR-93 negatively correlated with GLUT4. miR-93 in insulin resistance (IR) group was higher than non-IR group, while GLUT4 was lower ( P <0.05). miR-93 positively correlated with T (r=0.374, P =0.019), FINs (r=0.322, P =0.026) and HOMA-IR (r = 0.507, P = 0.005), while GLUT4 had a negative correlation. miR-93 and GLUT4 are abnormally expressed in PCOS, which is related to complications such as IR and endocrine metabolism.
Article
The sugar alcohol known as inositol and its derivatives are becoming more and more interesting in metabolism-related research studies due to its physiological roles. Inositol derivatives mainly myoinositol and d-chiroinositol have gained so much attention these days in the context of managing and treating metabolic issues such as diabetes and polycystic ovarian syndrome. For a long time, it was considered as the vitamin B, but later we got to know it is produced from glucose so considered non-essential. Nowadays rising rates of obesity which are further linked with diabetes and polycystic ovarian syndrome have fuelled demand for artificial sweeteners. Many people are searching for options to maintain their weight, which has led to the widespread usage of sucrose and fructose substitutes in the food industry. Sugar alcohols such as myoinositol and d chiroinositol (which are not been commonly used) in particular can be metabolized without the need for insulin control and have little effect on blood sugar levels. These sugar substitutes have the potential to provide sweetness without raising blood sugar levels and adding calories. These have half of the sweetness of sucrose. This paper aims to thoroughly investigate and understand the various roles played by inositol in the complex field of managing metabolic diseases. To find out the eligible papers for review paper screening has been performed through PubMed, Science Direct, Web of Science and Google Scholar. Different sets of keywords such as inositol, d chiro inositol, myo-inositol, insulin-resistance, diabetes, insulin sensitivity, DCI and type 2 diabetes mellitus were used. Studies which full text article were present used for the review. Research articles have been included the studies specifically to find out the relation between myoinositol and diabetes or any metabolic disease. The development of many diseases, such as PCOS and diabetes, is linked with inositol deficiency. Myoinositol is derived from the food we eat as well as from within. The low levels of MI can be the result of several factors, including decreased dietary intake, increased catabolism, alterations in the gut and reduced biosynthesis. These two forms, myoinositol and d-chiroinositol play significant roles in regulating lipid metabolism and glycaemic control. glycaemic metabolism and this are also supported by many research studies. Therefore, it can be a possible treatment or management option for people suffering from metabolic issues such as diabetes and PCOS. Future studies should focus on how we can increase the intake of myoinositol through diet and dietary food products.
Article
Full-text available
Polymers, both synthetic and natural, play a critical role in modern drug delivery systems by enhancing the efficacy, targeting, and release profiles of therapeutic agents. This comprehensive review delves into the various types of synthetic polymers such as poly (lactic-co-glycolic acid) (PLGA), polyethylene glycol (PEG), and polyvinyl alcohol (PVA), as well as natural polymers like chitosan, alginate, and gelatin. These polymers are explored for their potential to improve solubility, bioavailability, and controlled release of drugs. Moreover, their application in targeted drug delivery, particularly for cancer, cardiovascular, and inflammatory diseases, is highlighted. The review also compares the advantages and limitations of synthetic versus natural polymers, discussing their biodegradability, biocompatibility, and regulatory considerations. Advances in polymer-based drug delivery platforms such as nanoparticles, hydrogels, and micelles are also examined, offering insights into future directions in personalized medicine. The highlights of provide in the review article, initially basics of drug delivery, polymer, polymerization with role of polymer in polymerization. At intermediate, classification, sources of polymer with that some advanced approached in drug delivery and lastly, marketed, recent available products with future challenges and current status in the drug delivery. Keywords: Biodegradable polymer; natural; compatible; drug delivery; treatment; enhancing; polymers.
Article
Full-text available
Since the 1990 NIH‐sponsored conference on polycystic ovary syndrome (PCOS), it has become appreciated that the syndrome encompasses a broader spectrum of signs and symptoms of ovarian dysfunction than those defined by the original diagnostic criteria. The 2003 Rotterdam consensus workshop concluded that PCOS is a syndrome of ovarian dysfunction along with the cardinal features hyperandrogenism and polycystic ovary (PCO) morphology. PCOS remains a syndrome and, as such, no single diagnostic criterion (such as hyperandrogenism or PCO) is sufficient for clinical diagnosis. Its clinical manifestations may include: menstrual irregularities, signs of androgen excess, and obesity. Insulin resistance and elevated serum LH levels are also common features in PCOS. PCOS is associated with an increased risk of type 2 diabetes and cardiovascular events.
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
It is unknown whether hyperinsulinemia plays a role in the pathogenesis of polycystic ovary syndrome (PCOS) in normal weight or thin women. Evidence indicates that these women are insulin resistant and hyperinsulinemic, and this study was conducted to test the hypothesis that hyperinsulinemia stimulates ovarian cytochrome P450c17 alpha activity in nonobese women with PCOS, thereby increasing serum androgen concentrations. We assessed ovarian P450c17 alpha activity (by measuring the response of 17 alpha-hydroxyprogesterone to a GnRH agonist), fasting serum steroids, and oral glucose tolerance before and after oral administration of either metformin (500 mg) or placebo three times daily for 4-6 weeks in 31 nonobese women with PCOS. In the 19 women given metformin, the mean (+/-SE) area under the serum insulin curve after oral glucose administration decreased from 44 +/- 5 to 24 +/- 3 nmol/L.min (P = 0.003). Basal serum 17 alpha-hydroxyprogesterone decreased from 3.4 +/- 0.3 to 2.5 +/- 0.4 nmol/L (P = 0.05), and GnRH-stimulated peak serum 17 alpha-hydroxyprogesterone decreased from 12.2 +/- 1.6 to 7.5 +/- 0.7 nmol/L (P = 0.005). Serum 17 alpha-hydroxyprogesterone values did not change in the placebo group. In the metformin group, serum free testosterone decreased by 70% from 18.2 +/- 3.1 to 5.5 +/- 0.7 pmol/L (P < 0.001), and serum sex hormone-binding globulin increased from 84 +/- 6 to 134 +/- 15 nmol/L (P = 0.002). None of these values changed in the placebo group. These findings suggest that hyperinsulinemia stimulates ovarian P450c17 alpha activity in nonobese women with PCOS. They also indicate that decreasing serum insulin with metformin reduces ovarian cytochrome P450c17 alpha activity and ameliorates the hyperandrogenism of these women.
Article
Following the discovery of insulin, it took the rest of the twentieth century to understand how this hormone regulates intracellular metabolism. What are the main discoveries that led to our current understanding of this process? And how is this new knowledge being exploited in an attempt to develop improved drugs to treat the epidemic of type-2 diabetes?
Article
We studied the presence and distribution of the intracellular calcium channel regulating type I inositol 1,4,5-trisphosphate receptors (IP3R) in human immature and mature oocytes, pronuclear zygotes and cleaved embryos using a specific antibody. Two approaches were used: (i) fluorescence immunocytochemistry using a confocal laser scanning microscope (CLSM) and (ii) Western blotting. With confocal microscopy, the receptors were found in the oocytes, fertilized zygotes as well as cleaved embryos at all stages studied. The pattern and distribution of the receptor staining in the oocytes changed gradually from a diffuse granular patchy one at the germinal vesicle (GV) stage to a reticular and predominantly peripheral one through the metaphase and metaphase II (MII) stages. After fertilization, the distribution changed gradually to both, peripheral and central in the zygotes and early 2-4-cell embryos and predominantly perinuclear in the 6-8-cell embryos. Furthermore, an overall increase in the staining intensity was observed from GV to MII stage oocytes and from zygotes to 6-8-cell embryos. We also studied the spatial distribution of the receptor in detail by constructing three-dimensional images from the serial optical sections obtained on the CLSM. Peculiar peripheral aggregates of receptor clusters were noted in the MII stage oocytes, zygotes and some blastomeres from early cleaved embryos. Finally, Western blots performed on the extracts of 72 in-vitro matured oocytes and 50 spare cleavage stage embryos showed positive bands at similar to 260 kDa. These findings coincide with and thus possibly represent the dynamic changes occurring in the cellular Ca2+ release systems through oocyte maturation, fertilization and early embryogenesis. Thus, type I IP3R are likely to play a role during these stages of early development in the human.
Article
The polycystic ovary syndrome is a prevalent disorder characterized by chronic anovulation and hyperandrogenism. It has recently been appreciated that insulin resistance with a compensatory hyperinsulinemia is also a prominent feature of the disorder and plays a key role in its development in many affected women. The purpose of this review is to highlight clinical studies in this area that were published during the past year. Areas discussed include the use of insulin-sensitizing drugs for treatment, D-chiro-inositol and insulin resistance, syndrome X (ie, the dysmetabolic syndrome), genetics of polycystic ovary syndrome, precocious puberty and adrenarche as early manifestations of polycystic ovary syndrome, and early pregnancy loss in polycystic ovary syndrome.