Gynecological Endocrinology

ISSN: 0951-3590 (Print) 1473-0766 (Online) Journal homepage: http://www.tandfonline.com/loi/igye20

Hormonal contraceptive choice for women with PCOS: a systematic review of randomized trials and observational studies Nicolas Mendoza, Tommaso Simoncini & Alessandro D. Genazzani To cite this article: Nicolas Mendoza, Tommaso Simoncini & Alessandro D. Genazzani (2014) Hormonal contraceptive choice for women with PCOS: a systematic review of randomized trials and observational studies, Gynecological Endocrinology, 30:12, 850-860, DOI: 10.3109/09513590.2014.943725 To link to this article: http://dx.doi.org/10.3109/09513590.2014.943725

Published online: 25 Sep 2014.

Submit your article to this journal

Article views: 656

View related articles

View Crossmark data

Citing articles: 6 View citing articles

Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=igye20 Download by: [Australian Catholic University]

Date: 28 September 2017, At: 08:08

http://informahealthcare.com/gye ISSN: 0951-3590 (print), 1473-0766 (electronic) Gynecol Endocrinol, 2014; 30(12): 850–860 ! 2014 Informa UK Ltd. DOI: 10.3109/09513590.2014.943725

POLYCISTIC OVARY

Hormonal contraceptive choice for women with PCOS: a systematic review of randomized trials and observational studies Nicolas Mendoza1, Tommaso Simoncini2, and Alessandro D. Genazzani3 Departamento de Obstetricia y Ginecologı´a, Universidad de Granada, Granada, Spain, 2Dipartimento di Medicina Clinica e Sperimentale, Div. Ginecologia e Ostetricia, Universita` di Pisa, Pisa, Italy, and 3Clinica Ostetrica Ginecologica, University of Modena, Modena, Italy

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

1

Abstract

Keywords

Introduction: This paper provides a critical review of the data concerning the effects of combined hormonal contraceptives (CHC) for polycystic ovary syndrome (PCOS). The aim is to determine the best treatment option for each PCOS phenotype. Study design: A literature search of the PubMed database was conducted for randomized clinical trials (RCTs) and observational studies published in any language prior to October 2013. Hyperandrogenism (HA) is the essential diagnostic criterion for PCOS and is frequently associated with insulin resistance (IR) or obesity. The combinations of these criteria define the different PCOS phenotypes and establish the scale of metabolic and cardiovascular risks. Results and conclusions: 19 RCTs and eight observational studies evaluated issues related to the current objectives. CHC represent an effective and safe treatment in women with any PCOS phenotype. In HA/PCOS patients, any CHC analyzed in this review can be used for symptom relief. For patients with metabolic risk, overweight or moderate IR that does not require metformin, a vaginal contraceptive ring appears to be preferred to oral EE/DRP. In these patients, the combination of CHC and myo-inositol may be more effective in controlling endocrine and metabolic profiles. However, further research is needed to define the optimal duration and to clarify the effects of treatment on long-term metabolic outcomes. Future research should also focus on new CHC.

Hirsutism, hormonal contraception, ovary, polycystic ovary syndrome

Introduction Polycystic ovary syndrome (PCOS) is a complex and heterogeneous disease that involves reproductive and metabolic elements. An expert conference by the European Society for Human Reproduction and Embryology and the American Society for Reproduction Medicine determined that PCOS should be diagnosed when at least two of the following three features (Rotterdam criteria) are present [1]: oligo- and/or anovulation, hyperandrogenism (HA) and polycystic ovaries. This definition is more generic than the previous definition and increases the already high PCOS prevalence to the point that it is the most common endocrinopathy in women. Currently, PCOS has a worldwide prevalence of 7–14% of women of child-bearing age. Moreover, the additional phenotypes defined by the Rotterdam criteria identify women with primarily reproductive rather than metabolic dysfunction. The treatment choice for PCOS is partially determined by the scale of metabolic risk, defined by the presence of HA and the combination with obesity or insulin resistance (IR) [2]. Combined hormonal contraceptives (CHC) are the most common treatment for PCOS. However, there are doubts concerning their impact on the cardiovascular system and carbohydrate metabolism such that an ideal CHC does not seem to exist [3,4].

Address for correspondence: Prof. Nicolas Mendoza, University of Granada, Granada, Spain. E-mail: [email protected]

History Received 4 April 2014 Revised 6 May 2014 Accepted 8 July 2014 Published online 25 September 2014

The aim of this paper is to provide a critical review of the data concerning the effects of CHC in patients with PCOS to determine which option is best for each phenotype. There is a special focus on monotherapy strategies (CHC use without combination with other drugs for PCOS, e.g. metformin or flutamide) without and on the long-term health balance between the risks and benefits.

Methods Selection of studies Using the strategy described in Appendix A, we searched the PubMed database for all articles, in all languages, published in peer-reviewed journals through October 2013. First, we included articles concerning the choice of CHC in PCOS women. Articles describing the treatment or prevention of PCOS symptoms or the consequences of PCOS were also included. The inclusion criteria were the following: any randomized controlled trial (RCT), non-RCT or non-controlled studies focusing on CHC and PCOS. Papers that did not discuss CHC use in PCOS patients were excluded. Assessment of study quality and data synthesis We followed PRISMA guidelines for systematic reviews [5]. PICOS (population, interventions, comparators, outcomes, studies design) criteria were formulated a priori to guide the review’s scope and the search procedure, selection and synthesis of the literature. The selection criteria were as follows: (population)

Contraceptives and PCOS

DOI: 10.3109/09513590.2014.943725

PCOS patients of any age; (intervention) treatment with CHC; (outcome) efficacy and safety; (study design) RCTs non-RCT or non-controlled studies; no language restrictions. Two reviewers independently screened titles and abstracts and subsequently extracted data from the selected studies including quality items, and on outcomes of interest. A meta-analysis was performed for safety issues. The authors independently conducted the search and screened studies for inclusion, extracted and checked the data, and synthesized the findings. The authors independently determined the adequacy of the design of the studies and their main methodological characteristics in order to ascertain the validity of eligible research. Disagreements were resolved by discussion and consensus. When duplicates of a study were found, we selected the more detailed or complete data from the multiple reports indicating the corresponding references.

Discussion Some of the most relevant weaknesses in the literature used for this study are due to limitations in the research designs. There are only a limited number of RCT and longitudinal studies with long-term follow-ups that occur after the completion of the intervention (see Table 1). In addition, data on effect sizes is often lacking. In many studies, the sample sizes are small and no information regarding power calculations to estimate the appropriate sample size is available. Furthermore, outcome measures are diverse, which makes it difficult to compare findings. Although components of combined interventions are usually described in detail, analyses of component-specific, separated effects are often not reported. Because the study designs, participants, interventions and reported outcome measures varied markedly, we focused on describing the studies, their main characteristics and results, and on qualitative synthesis rather than a meta-analysis. Nevertheless, several issues remain controversial or unknown, and future research should properly address these concerns. In addition to our reservations regarding the quality of the research included here, our review also suffers from several limitations. The most important limitation refers to the exclusion of reports due to the document type (e.g. non-peer-reviewed reports) or to their conditions (e.g. language, unavailability). PCOS phenotypes In the most recent implementation of the Rotterdam diagnostic criteria, different PCOS phenotypes have been defined. Severe PCOS, or the type I phenotype (HA, chronic anovulation and polycystic ovaries), is the most common. The type II phenotype (HA and chronic anovulation but normal ovaries) is much less

Records identified through database searching (n=167)

Records screened (n= 167)

Eligibility

Screening

Idenficaon

The literature search returned 167 studies; 129 were removed after reading titles or abstracts, leaving 38 studies of which 11 studies were excluded for various reasons (basically by the use of other treatments different than CHC), leaving 27 studies for final analysis. publications involving 19 RCT [6–32] (see flow diagram, Figure 1). Full articles that met the inclusion criteria were reviewed in detail. Data items to be considered were discussed by the review authors and appear in Table 1. Other relevant papers were used as a reference. When some of these informations were not available or clear, we treated it as missing data. Eleven RCTs were used to evaluate the best CHC method for women with hyperandrogenism (HA). One of the studies also evaluated women with idiopathic hirsutism [13], and we separated two RCTs that evaluated CHC in combination with anti-androgen drugs and therefore did not examine CHC monotherapy [13,14]. With the exception of the Battaglia study, which included a vaginal ring method, all of these RCTs compared oral CHCs [6]. Most of the RCTs used ethinyl-estradiol (EE) at a dose of 30 mcg as the estrogen component, while the progestin component varied between drospirenone (DRP), cyproterone acetate (CPA), chlormadinone acetate (CMA) and desogestrel (DSG). In general terms, when used as monotherapy, all CHC regimens were shown to relieve HA symptoms after 6 months. The study by Colonna et al. [12] in 2012, reported greater reductions in acne and seborrhea compared to DRP methods, while the only study that provided an assessment at 12 months indicated that CPA is the most potent drug for reducing HA symptoms and signs [11]. Most of the observational studies included in this review were limited to six months of intervention/follow-up and used CHC with 30 mcg EE. In a recent Italian study, an oral

Full text articles assessed for eligibility (n= 38)

Included

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

CHC containing estradiol valerate plus dienogest (E2V/DNG) was tested [8]. When assessing patients with IR or any carbohydrate metabolism disorder, most RCTs have compared the above-mentioned CHC and metformin [17–23]. Among the treatment groups, CHC-metformin seems to be a more effective therapeutic option due to the additional beneficial effect on IR. Moreover, the CHC-metformin combination may be particularly beneficial in overweight women with PCOS because it improves vascular endothelial function. When CHC is compared in monotherapy, oral CHC containing DRP has a better lipid profile and a more favorable glycemic and hormonal profile than oral CHC with DSG. In one report, the vaginal ring appeared to be preferable to oral CHC with DRP in PCOS/hyperinsulinemic patients [6].

Limitations

Results

Figure 1. Flow diagram.

851

Studies included in qualitative synthesis (n= 27)

Full text articles excluded (n= 11)

Records excluded on a title/abstract basic (N= 129)

Duplicated (n= 0)

Randomization

Allocation in a 1:1 ratio. Random number table. Double blind

Computer-generated randomization

Not described

Random number table

Computer-generated randomization table

Computer-generated randomization tables in a 1:1:1 ratio. Double blind

Reference

Battaglia et al. [6]

Kriplani et al. [7]

De Leo V et al. [8]

Harris-Glocker et al. [9]

Oner [10]

Bhattacharya et al. [11]

The treatment of hirsutism with both COC containing DRP offered comparable effects and was well tolerated. No difference in effects after 6 months. At 12 months, CPA showed the strongest antiandrogen activities (Ferriman Galwey score). Effects on metabolic parameters were identical. CPA significantly increased SHBG (change ¼ 142.91) compared with DSG (change ¼ 99.53); DRP significantly increased SHBG (change ¼ 131.52) compared with DSG; and CPA significantly decreased the Free Androgen Index (change ¼ 10.57) compared with DSG (change ¼  5.58).

Hirsutism, Hormonal levels

 COC (DRP 3 mg + EE 30 mg + metformin  COC + placebo. 6 months  DRP 3 mg + EE 30 mg  DRP 3 mg + EE 20 mg 6 months  DRP 3 mg + EE 30 mg  CPA 2 mg + EE 30 mg  DSG 150 mg + EE 30 mg 12 months

Endocrine and metabolic parameters

N. Mendoza et al.

50 women (no age data) with moderate to severe hirsutism 171 Women (18–35 years) with HA/PCOS Androgen Excess Society criteria

36 post-menarchal (12–18 years) with HA/PCOS NIH criteria

Quality-of-life

HA

    3

40 women (35 years) with HA/PCOS Rotterdam criteria

DRP 3 mg + EE 30 mg CMA 2 mg + EE 30 mg GES 75 mg + EE 30 mg DSG 150 mg + EE 30 mg months

Endocrine and metabolic parameters

 DRP 3 mg + EE 30 mg or  DSG 150 mg + EE 30 mg 6 months

60 women (16–40 years) not desirous of conception + PCOS Rotterdam criteria

Both treatments improved hirsutism, hyperandrogenemia and ultrasound and color Doppler ovarian parameters. Both induced a slight but significant increase of diurnal and 24-hour blood pressure. Although both therapies worsened the lipid profile, the oral pill administration was associated with a more evident increase of circulating triglycerides. The 6-month treatment with the vaginal ring significantly improved the area under the curve for glucose, insulin, and Cpeptide, whereas the DRP-EE pill induced an increase in the insulinogenic index and homeostatic model assessment estimate for insulin resistance values. COC with DRP has better outcome in regular cycles, antiandrogenic effect, fall in BMI and BP, better lipid profile, favorable glycemic and hormonal profile than COC with DSG:  BMI fell by 0.52 kg/m2;  LDL was decreased (p50.01) and HDL was elevated (p50.05),  Systolic BP reduced (1.6% fall) and diastolic BP (0.5% fall) (p50.05)  A significant fall in fasting/postprandial blood sugar and insulin and total testosterone with both COC without differences between groups. In all groups, mean concentrations of free T, total T and A dropped by 40–60%, and concentrations of DHEAS dropped by 20–50%. Formulations with DRSP and CMA caused a greater reduction of androgens and a progressive increase in serum concentrations of SHBG than those with DSG and GSD (without significant differences). The addition of metformin does not add improvement to quality-of-life.

CVD risk, endocrine and metabolic parameters

 DRP 3 mg + EE 30 mg  Vaginal ring (EE 15 mg + ENG 120 mg) 6 months

Key results

40 women 418 years for treatment of HA/PCOS and contraceptive necessities Rotterdam criteria

Outcomes

Intervention

Participants*/PCOS criteria

Randomized controlled trial involved in hyperandrogenism (HA) PCOS patients.

Table 1. Summary of studies.

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

852 Gynecol Endocrinol, 2014; 30(12): 850–860

Not described

Not described

Not described

Non-blind, based on patients’ chronological presence (indifferent of BMI and IR)

Computer-generated randomization tables

Colonna et al. [12]

Karakurt et al. [13]

Hadzˇiomerovic´Pekic´ et al. [14]

Panidis et al. [15]

Romualdi et al. [16]

Endocrine and metabolic parameters

AMH levels

 Metformin (M)  Naltrexone (N)  COC (EE 35 mg + CPA 2 mg) + prednisolone (Pr) 3 month.

   6

29 women (18–36 years) HA/PCOS + IR Rotterdam criteria

45 HA/PCOS NHI criteria

30 PCOS (19–30 years) Rotterdam criteria

 DRP 3 mg + EE 30 mg  DRP 3 mg + EE 20 mg 12 months

Endocrine and metabolic parameters

Hirsutism

 COC (EE 35 mg + CPA 2 mg) + SPL 100 mg  Flutamide 250 mg 6 months

29 with hirsutism (HA/ PCOS and idiopatic)

EE 35 mg + CPA 2 mg EE 30 mg + DRP 3 mg Metformine months

HA

59 women (19–29 years) HA/PCOS

 DRP 3 mg + EE 30 mg  CMA 2 mg + EE 30 mg 6 months

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

Contraceptives and PCOS (continued )

Both treatments were well tolerated and showed a significant improvement of HA, although EE/ DRP showed a more potent effect on acne and seborrhea at the end of the study (p50.05 between treatments). Seborrhoea (mcg/cm2 of skin) 2412.26 ± 1692.03 versus 4076.69 ± 2497.66 p Ferryman–Gallwey score 9.43 ± 2.02 versus 11.24 ± 2.35 delta-4-androstenedione (ng/mL) 1.98 ± 0.72 versus 2.72 ± 0.64 total testosterone (nmol/L) 0.99 ± 0.37 versus 1.28 ± 0.37 Both treatments are effective in the treatment of hirsutism: A significant decrease in Ferryman– Gallwey score was observed with flutamide (from 11.2 ± 3.3 to 7.6 ± 4.0) and SPL + CPA/EE (from 9.9 ± 1.9 to 7.1 ± 2.0). However, there was no statistically significant difference between the two groups. A statistically significant decrease in free androgen index was seen in all groups. Testosterone concentrations declined significantly in the M and N groups after 3 months, and in the COC/Pr group, only after the first month. DHEAS decreased significantly only in the N group. SHBG levels more than doubled during COC use and was not affected by M or N. The only significant within-group changes in the metabolic state were an average decrease in total cholesterol by 10 mg/dL and BP by 9 mm Hg in the M group (p ¼ 0. 01, p ¼ 0.03, respectively). HDL cholesterol increased in the OC/Pr group by 7 mg/dL, whereas it remained unchanged in the other two groups (p ¼ 0.046). The diastolic BP increased in the OC/Pr group by 10.6 mm Hg, whereas it decreased in the M group and remained unchanged in the N group (p ¼ 0.02). AMH serum levels were significantly decreased under treatment with EE 35 mg + CPA 2 mg (p ¼ 0.002 at 3 months and p ¼ 0.001 at 6 months). EE/DRP and metformin did not significantly affect serum AMH levels. AMH was significantly decreased under COCs treatment compared to metformin (p ¼ 0.005). Both COC induced a significant improve in hirsutism score, testosterone, DHEAS and SHBG levels. Total Cholesterol, LDL, HDL and triglycerides

DOI: 10.3109/09513590.2014.943725

853

Randomization

Participants*/PCOS criteria

Intervention

Case-control study followed by a RCT

Not described

Not described

Luque-Ramı´rez et al. [18]

Moran et al. [19]

Kebapcilar et al. [20]

48 women with PCOS Rotterdam criteria + IR

Overweight, age and BMImatched women with PCOS (no ¼ 80) or without PCOS (no ¼ 27)

40 PCOS patients and 20 non-HA women.

Blood clotting tests and endothelial function

Inflammatory markers, endocrine and metabolic parameters Endocrine and metabolic parameters

 metformin  EE 35 mg + CPA 2 mg 6 months A. COC (EE 35 mg + CPA 2 mg) B. Metformin C. COC + metformin D. COC + SPL 3 months

Outcomes CVD risk factor

Outcomes

 COC (EE 35 mg + CPA 2 mg)  metformin 24 weeks

Randomized controlled trial involved in PCOS patients with insulin resistance (IR) or obesity. Reference Randomization Participants*/PCOS criteria Intervention  COC (35 mg EE + 2 mg Luque-Ramı´rez Randomized, open-label, 34 HA/PCOS patients and CPA) et al. [17] parallel clinical trial 40 non-HA control  Metformin women NIH criteria

Reference

Randomized controlled trial involved in hyperandrogenism (HA) PCOS patients.

Table 1. Continued

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

Key results Obesity is the main determinant of serum uric acid concentrations in PCOS patients, yet amelioration of androgen excess with an antiandrogenic contraceptive pill results in a significant decrease in these levels, an effect that is not observed with metformin. There were no statistically significant differences in uric acid levels between PCOS and non-hyperandrogenic control women. Obese women showed higher uric acid concentrations than lean and overweight women, In PCOS women, COC decreased in uric acid levels (p ¼ 0.018), whereas no changes were observed with metformin. COC and metformin may exert deleterious effects on blood clotting tests of PCOS women, yet the effects of metformin appear to be milder. Because smoking potentiates some of these effects and deteriorates endothelial function, smoking cessation should be promoted in PCOS patients. Alterations in leptin between women with and without PCOS and following pharmacological interventions are primarily related to adiposity and not IR. Aldosterone was reduced equivalently with metformin and the COC. In all treatment groups were observed reduced levels of coagulation parameters, improvement of hormonal, hematological and metabolical variables. by most probably reducing insulin levels. Among the treatment groups, EE/CA-metformin may be a more effective therapeutic option than the other protocols and this may be due to the beneficial effect of EE/CA-metformin on IR: All women lost weight significantly after the treatment (Group A: 3.5 kg, 4.8%; Group B: 2.8 kg, 3.6%; Group C: 2.7 kg, 3.7%; Group D: 2.6 kg, 3.4%). Lipid profiles: Group A: no change; Group B: significant decrease in total and LDL cholesterol and increase in HDL; there were no changes in triglycerides; Group C: triglycerides, total and LDL cholesterol levels were significantly reduced after the treatment whereas HDL concentrations

increased in both groups. Triglycerides more markedly increased with EE 30 mg.

Key results

854 N. Mendoza et al. Gynecol Endocrinol, 2014; 30(12): 850–860

Computerized random number generator using a 1:1 ratio

Not described

Not described

Essah et al. [21]

Orbetzova et al. [22]

Kilic et al. [23]

96 women (18–35 years) non-HA and oligoamenorrheic/

66 PCOS + overweight + IR Rotterdam criteria

18 women (no data of age) PCOS/HA Rotterdam criteria

CVD risk and carbohydrate metabolism

Adipose tissue hormones and hypothalamic neuropeptide Y

 COC (CMA 2 mg + EE 30 mg) + metformin  COC + rosiglitazone 3-months

1. Obese + metformin. 2. Obese + COC (DSG 150 mg + EE 30 mg) 3. Non-obese + metformin

Endocrine and metabolic parameters

 COC (EE 35 mg + norgestimate 0.18– 0.25 mg) + metformin  COC + placebo 3 months

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

Contraceptives and PCOS (continued )

were increased significantly; Group D: LDL and total cholesterol were significantly reduced but there were no significant changes in triglycerides and HDL levels. Activated partial thromboplastin time, prothrombin time levels D-dimer, WBC and MPV were reduced significantly in all the groups (p50.05). HOMA-IR, insulin and androgen levels decreased in all treatment groups. EE/CA–metformin and metformin alone groups resulted in a higher proportional reduction of Ddimer levels. EE/CA–metformin group showed higher proportional reduction fasting insulin concentrations, HOMA-IR and free testosterone levels than metformin alone and EE/CA–SPL groups. DHEAs levels significantly decreased in group EE/ CA–metformin than EE/CA alone and EE/CA– SPL groups. In multiple stepwise regression analyses, reduction in proportional insulin levels was independently and positively associated with decrease of MPV, D-dimer, free testosterone levels. Both treatments had similar effects on androgen levels, lipid profile, insulin sensitivity, and serum inflammatory markers, but flow-mediated dilatation increased by 69.0% in the metformin plus COC group while it remained unchanged in the COC group. beneficial effects of the treatment on potential cardiovascular risk in IR PCOS women Significant decrease of leptin (p50.01; p ¼ 0.001, respectively), resistin (p50.01; p50.01, respectively), TNF a) (p ¼ 0.001; p50.001, respectively.), and neuropeptide Y (p50.05; p50.001, respectively) was observed in both groups after treatment. Decrease in the anthropometric parameters of body weight in the metformin group only. No significant changes in hormonal characteristics of the groups were found except for a decrease in androstenedione and DHEA-S (p50.05) in the metformin group and in 17-OH-progesterone (p50.05) in the rosiglitazone group. HDL-cholesterol rose and diastolic BP fell (p50.05) in the metformin group. A significant decrease was observed in ADMA, homocysteine and HOMA-IR levels in Groups 1 and 3. An increase in ADMA and hs-CRP levels was observed in Groups 2 and 4.

DOI: 10.3109/09513590.2014.943725

855

1:1 proportion using a randomizer program

Randomization

49 HA/PCOS women  20 PCOS who wanted HC  20 control required HC  20 control wanted nonHC 28 lean PCOS and 28 control. Rotterdam criteria

20 women (18–33 years) PCOS Rotterdam criteria

Banaszewska et al. [27]

Gomes et al. [28]

De Leo et al. [30]

Aydin et al. [29]

155 patients with PCOS prospective, open-label clinical study.

Participants*/PCOS criteria 50 non-obese women: 30 HA/PCOS and 20 control

50 women (18–35 years) Rotterdam criteria (no describe if HA or not)

PCOS + impaired glucose tolerance

Participants*/PCOS criteria

Minozzi et al. [26]

Observational studies involved in PCOS patients. Reference Uras et al. [25]

Vieira et al. [24]

Reference

Randomized controlled trial involved in hyperandrogenism (HA) PCOS patients.

Table 1. Continued

Endocrine and metabolic parameters Endocrine and metabolic parameters

 EE 30 mg + GTD 75 mg  EE/GTD + myo-inositol 12 months. COC (EE 20 mg + 150 DSG) 6 months. COC: CMA 2 mg + EE 30 mg 6 months

Anthropometric, endocrine and metabolic parameters

Anthropometric, and metabolic parameters

EE 30 mg/DRP 3 mg 6 months.

E2V/DNG 3 months

Plasma concentrations of Matrix metalloproteinases (MMPs)

Outcomes Endocrine and metabolic parameters

Arterial function

Outcomes

Intervention PCOS: CMA 2 mg + EE 30 mg 6 months

 COC (CMA 2 mg + EE 30 mg)  COC + SPL 100 mg 12 months

4. Non-obese + COC 6 months

Intervention

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

After 6 months of treatment in the PCOS patients, total fat percentage increased from 24.5% ± 7.1% to 26.0% ± 6.1% (p ¼ 0.035) and trunk fat percentage increased from 20.2% ± 8.9% to 22.2% ± 7.1% (p ¼ 0.014), although weight, BMI and waist to hip ratio remained unchanged. Median values of insulin, Homeostasis model assessment of insulin resistance and oral glucose tolerance test after treatment were lower than values before treatment. No significant difference was observed in BMI

Reduced plasma MMP-2 concentrations in both healthy and PCOS women.

Key results In the EE/CMA group, androgen levels and bioavailability, hirsutism and acne score were significantly lower, whereas they did not change in the control group. In both groups, glucose-insulin metabolism and body composition parameters were not affected. Combination of COC and myo-inositol may be more effective in controlling endocrine, metabolic, and clinical profile in patients with PCOS than COC alone, and reduce insulin levels and IR. Decrease in the testosterone level but negative effect on total cholesterol and triglycerides level

In this study, metformin treatment leads to improvement in hormonal and metabolic parameters and decreases endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA) and homocysteine levels. COC impaired glucose tolerance, increases ADMA and high sensitive C-reactive protein levels and creates an increase in the metabolic risk. The increase in mean total cholesterol levels was greater in the COC + SPL group than in the COC group (27 versus 13%, respectively; p ¼ 0.02). The increase in SHBG levels was greater in the COC group than in the COC + SPL group (424 versus 364%, respectively; p ¼ 0.01).

Key results

856 N. Mendoza et al. Gynecol Endocrinol, 2014; 30(12): 850–860

Contraceptives and PCOS *All without contraindications for hormonal contraceptives. BMI: Body mass index; BP: blood pressure; CMA: chlormadinone acetate; COC: combined oral contraceptive; CPA: ciproterone acetate; CVD: cardio vascular disease; DNG: dienogest; DSG: desogestrel; DRP: drospirenone; E2V: estradiol valerate; EE: etinilestradiol; ENG etonogestrel; GSD: gestodene; HA: hyperandrogenism; IR: insulin resistance; MMPs: Matrix metalloproteinases; SHBG: sex hormone-binding globulin; NIH: National Institutes of Health; PCOS: polycystic ovary syndrome; SPL: spironolactone.

Endocrine and metabolic parameters 46 lean women: 23 PCOS and 23 control Harmanci et al. [32]

PCOS: EE-DRP + SPL 6 months

Anthropometric, endocrine and metabolic parameters  EE/DRP (30 mg/3 mg)  EE/DRP + metformin 6 months 45 lean HA/PCOS women Cinar et al. [31]

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

EE/DRSP alone or in combination with metformin improves clinical and biochemical HA in lean PCOS. Both treatments similarly alter lipid profile. EE/DRSP alone does not affect insulin sensitivity, whereas combining EE/DRSP with metformin might improve it. Improves HA in lean PCOS without any adverse effects on adiposity, glucose tolerance status or lipid profile, and increases high-sensitivity Creactive protein and homocysteine levels.

DOI: 10.3109/09513590.2014.943725

857

frequent but has similar clinical and endocrine characteristics as phenotype I. The Ovulatory PCOS, or type III, phenotype (HA and polycystic ovaries) is relatively common and presents milder clinical and endocrine alterations than phenotypes I and II. The normoandrogenic PCOS, or type IV, phenotype is relatively uncommon and characterizes patients with normal body mass index (BMI), insulin sensitivity and free androgen index with increased levels of LH and LH/FSH ratio [33,34]. All PCOS phenotypes display changes due to aging, suggesting an amelioration of ovarian dysfunction as indicated by the increase in number of regular menstrual cycles, decrease in serum androgen levels, and decrease in IR [35]. Apart from these signs and symptoms, a considerable proportion of women with PCOS display IR and overweight/obesity. The mechanisms responsible for IR are currently unknown, although it is likely a result of altered adipose and muscular cell glucose uptake due to a genetic defect that is independent of obesity. Although IR does not occur exclusively in overweight/ obese women with PCOS, IR is more frequent among these patients [36]. Similarly, a direct association between IR and HA has been observed although the cause-and-effect relationship is not well understood. Androgens have not been shown to alter the physiology or glucose homeostasis of adipocytes. In addition, tests have indicated that hyperinsulinism tends to be a cause rather than a result of excess androgens [37]. Other markers of cardiovascular disease (CVD) are increased in women with PCOS; however, whether this apparent risk translates into an increased incidence of CVD later in life remains to be elucidated [38,39]. Obesity likely plays a major role, but adjustments for BMI do not affect these findings, suggesting that the increased CVD risk is not exclusively related to a higher BM. Instead, the increased CVS risk may be due to excess androgen concentrations [28]. A common genetic origin of PCOS and diabetes mellitus has also been proposed [40]. A number of studies have linked PCOS with an increased risk of gestational diabetes, but a 2009 meta-analysis did not find sufficient evidence to confirm this hypothesis [41]. This is likely because the new definition of PCOS includes other phenotypes that are exempted from this risk. In this sense, the main link between PCOS and cardiovascular disease remains IR. Moreover, the risk for adverse obstetric or neonatal outcomes increases in patients with PCOS phenotypes with ovarian dysfunction plus biochemical HA, whereas no significant effect was detected for clinical HA without anovulation [42]. Thus, some clinicians prefer to delay fertility treatment until the patients have reached an appropriate weight or have leveled out their insulin levels [43]. In summary, HA serves as the essential diagnostic criterion of the syndrome and is frequently associated with IR or obesity. The combinations of these conditions define the different PCOS phenotypes and establish the scale of metabolic and CVD risk [44,45]. The long-term health risks of PCOS patients demand that their general health be monitored even after their reproductive needs have been fulfilled. Metabolic and cardiovascular risk prevention in women with PCOS should start as early as possible [46]. To address this issue, the National Institutes of Health (NIH) Office for Disease Prevention-sponsored Evidence-based Methodology Workshop on Polycystic Ovary Syndrome recently proposed dividing the PCOS phenotypes into two distinct diseases. Those with primarily reproductive concerns should continue to be referred to as PCOS patients, and those with important metabolic consequences should have a new disease designation [47].

858

N. Mendoza et al.

PCOS phenotypes: hormonal contraception choice

PCOS with metabolic risk or obesity

To date, CHC have been the most common treatment for PCOS and its symptoms. However, there are no clinical guidelines that recommend a specific CHC for the different PCOS phenotypes. There are still doubts concerning the metabolic and cardiovascular effect of CHC in this population. In general terms, it has been recommended that the ideal contraceptive for women with PCOS should limit follicle development to reduce androgens synthesis, block the peripheral action of androgens and achieve good menstrual cycle control with minimal metabolic or thromboembolic risk.

There are doubts concerning the impact of CHC on cardiovascular function and carbohydrate metabolism, which hinders the choice of CHC method. Oral CHCs may worsen glucose tolerance in healthy users, but there is no finding indicating an increased risk of CVD in women with PCOS. These discrepancies could be due to inadequate adjustments for confounding factors that might influence body weight or IR (lifestyle, dietary composition, ethnicity, non-compliance, etc.) [56,57]. Although the metabolic effects of CHC in PCOS could be dependent on body weight, few studies have focused on the possible differences in the effect of CHC on lean versus obese patients. In lean-PCOS women, CHC do not change clinical anthropometric measures and do not exhibit adverse effects on adiposity, insulin sensitivity/glucose tolerance status and lipid profile [25,29,31,32]. In three meta-analyses that examined the effect of CHC on metabolic outcomes, the use of CHC was not associated with clinically significant adverse metabolic consequences. When changes in the lipid spectrum are compared, the vast majority of healthy users remain within reference limits, and it is unlikely that this could have a clinically significant effect on the risk of CVD [58–60]. In contrast, according to the World Health Organization’s recommendations, when arterial hypertension or an elevated risk of thromboembolism is present, levonorgestrelintrauterine system or progestogen-only hormonal contraceptives should be used instead of CHC [61]. Thus, the selected studies of this review suggest that progestogens with minimal metabolic side-effects, such as DRSP, could be more advantageous in overweight women or women whose glucose tolerance is a concern. In addition, the combination of CHC with myo-inositol may be more effective in controlling the endocrine, metabolic and clinical profiles in patients with PCOS than CHC alone, and may reduce insulin levels and IR [26]. With respect to estrogen, a recent European review indicates that the use of low-dose-oral CHC is preferable in patients with glucose intolerance or IR [62]. However, even though we have little evidence, it is interesting that the vaginal ring and oral E2V/DNG formulations seem to be preferred over CHC with EE/DRP [6,8].

HA/PCOS (type I, II and III phenotypes)

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

Gynecol Endocrinol, 2014; 30(12): 850–860

As stated in the previous section, the key to defining the phenotype and severity of PCOS is HA. In HA/PCOS phenotypes, the CHC of choice would alleviate the HA without a negative impact on metabolism or cardiovascular risk. A recent practical guideline recommends CHC (oral, patch or vaginal ring) as firstline management for HA/PCOS patients, but there are insufficient data to define the optimal duration of treatment [48]. For the signs and symptoms of androgens excess, the most important parameter in CHC selection is the type of progestin. CPA, CMA, DNG and DRSP are progestogens with powerful anti-androgen activity. These progestogens mainly act by blocking androgen receptors in target organs; however, they also reduce 5 alpha-reductase skin activity. Studies on the efficacy of CHC containing DRSP or CPA in the treatment of hirsutism show a reduction in clinical manifestations of HA after 6–12 months of therapy, a significant reduction of circulating androgens and a significant increase in SHBG. Moreover, it is speculated that an interference with androgen synthesis may also reduce the risk of later metabolic disturbances and health complications [49]. In addition to this, estrogen increases SHBG, thereby reducing bioavailable androgens. However, there are no data providing direct comparisons regarding the type or dose of estrogen. Furthermore, research has shown a 2-fold increase in the risk of venous thromboembolism among women with PCOS who were taking oral CHC and a 1.5-fold increased risk among women with PCOS not taking oral CHC [50]. Therefore, it is recommended that women with PCOS use a CHC with the lowest effective estrogen dose. In addition, the dose of EE correlates with the concentration of SHBG, a critical factor in decreasing circulating free androgen. The residual follicular activity and adrenal steroidogenesis are more frequent with low estrogen dose. However, there are few studies comparing EE doses [10]. E2V/DNG has an impact on various metabolic and hemostatic parameters that is comparable to or less than that of EE/LNGcontaining oral CHC. The effects on prothrombin, D-dimer, HDL, LDL, insulin and carbohydrate metabolism are, in general, more favorable with E2V/DNG than with EE/LNG [51]. E2V/DNG could thus be recommended as an oral CHC in PCOS women with IR or overweight [8]. The specific CHC regimen does not appear to influence the metabolism of androgens; however, extended regimens improve follicular control and hormonal suppression [52]. One extended regimen consisting of 2 mg CMA plus 30 mcg EE showed an improvement in acne after 6 months [53,54], and another study evaluating 30 mcg EE plus 2 mg DNG reported increased SHBG and decreased testosterone levels, which is similar to previous observations with the conventional regimen [55]. Non-HA/PCOS phenotype Only one RCT analyzed PCOS women without HA; however, all of the participants had obesity or impaired glucose tolerance [23].

Conclusions CHC represent an effective and safe treatment in women with any phenotype of PCOS. In HA/PCOS patients, any CHC analyzed in this review can be used for symptom relief. In patients with metabolic risk, overweight or moderate IR that does not need metformin, a vaginal contraceptive ring appears to be preferred to oral EE/DRP. Moreover, a combination of CHC and myo-inositol may be more effective in controlling the endocrine and metabolic profile. However, further research is needed to define the optimal duration and to clarify the effects of treatment on long-term metabolic outcomes. Future research should also focus on new CHC.

Declaration of interest All authors participated in the review and approved the final version of the manuscript. None of them have conflict of interest with. No one is on speaker’s bureaus, received research funding or consulting.

References 1. Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group. Revised 2003, consensus on diagnostic criteria and longterm health risks related to polycystic ovary syndrome (PCOS). Human Reprod 2004;19:41–7.

Contraceptives and PCOS

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

DOI: 10.3109/09513590.2014.943725

2. de Groot PC, Dekkers OM, Romijn JA, et al. PCOS, coronary heart disease, stroke and the influence of obesity: a systematic review and meta-analysis. Hum Reprod Update 2011;17:495–500. 3. Vrbı´kova´ J, Cibula D. Combined oral contraceptives in the treatment of polycystic ovary syndrome. Hum Reprod Update 2005;11: 277–91. 4. Bird ST, Hartzema AG, Brophy JM, et al. Risk of venous thromboembolism in women with polycystic ovary syndrome: a population-based matched cohort analysis. CMAJ 2013;185: E115–20. 5. Prisma. The Prisma Statement. Available at: http://www.prismastatement.org/statement.htm. 6. Battaglia C, Mancini F, Fabbri R, et al. Polycystic ovary syndrome and cardiovascular risk in young patients treated with drospirenoneethinylestradiol or contraceptive vaginal ring. A prospective, randomized, pilot study. Fertil Steril 2010;94:1417–25. 7. Kriplani A, Periyasamy AJ, Agarwal N, et al. Effect of oral contraceptive containing ethinyl estradiol combined with drospirenone vs. desogestrel on clinical and biochemical parameters in patients with polycystic ovary syndrome. Contraception 2010;82: 139–46. 8. De Leo V, Di Sabatino A, Musacchio MC, et al. Effect of oral contraceptives on markers of hyperandrogenism and SHBG in women with polycystic ovary syndrome. Contraception 2010;82: 276–80. 9. Harris-Glocker M, Davidson K, Kochman L, et al. Improvement in quality-of-life questionnaire measures in obese adolescent females with polycystic ovary syndrome treated with lifestyle changes and oral contraceptives, with or without metformin. Fertil Steril 2010; 93:1016–19. 10. Oner G, Muderris II. A prospective randomized trial comparing low-dose ethinyl estradiol and drospirenone 24/4 combined oral contraceptive vs. ethinyl estradiol and drospirenone 21/7 combined oral contraceptive in the treatment of hirsutism. Contraception 2011; 84: 508–11. 11. Bhattacharya SM, Jha A. Comparative study of the therapeutic effects of oral contraceptive pills containing desogestrel, cyproterone acetate, and drospirenona in patients with polycystic ovary syndrome. Fertil Steril 2012;98:1053–9. 12. Colonna L, Pacifico V, Lello S, et al. Skin improvement with two different oestroprogestins in patients affected by acne and polycystic ovary syndrome: clinical and instrumental evaluation. J Eur Acad Dermatol Venereol 2012;26:1364–71. 13. Karakurt F, Sahin I, Gu¨ler S, et al. Comparison of the clinical efficacy of flutamide and spironolactone plus ethinyloestradiol/ cyproterone acetate in the treatment of hirsutism: a randomized controlled study. Adv Ther 2008;25:321–8. 14. Hadzˇiomerovic´-Pekic´ D, Wildt L, Weiss JM, et al. Metformin, naltrexone, or the combination of prednisolone and antiandrogenic oral contraceptives as first-line therapy in hyperinsulinemic women with polycystic ovary syndrome. Fertil Steril 2010;94:2385–8. 15. Panidis D, Georgopoulos NA, Piouka A, et al. The impact of oral contraceptives and metformin on anti-Mu¨llerian hormone serum levels in women with polycystic ovary syndrome and biochemical hyperandrogenemia. Gynecol Endocrinol 2011;27:587–92. 16. Romualdi D, De Cicco S, Busacca M, et al. Clinical efficacy and metabolic impact of two different dosages of ethinyl-estradiol in association with drospirenone in normal-weight women with polycystic ovary syndrome: A randomized study. J Endocrinol Invest 2013;36:636–41. 17. Luque-Ramı´rez M, Alvarez-Blasco F, Uriol Rivera MG, EscobarMorreale HF. Serum uric acid concentration as non-classic cardiovascular risk factor in women with polycystic ovary syndrome: effect of treatment with ethinyl-estradiol plus cyproterone acetate versus metformin. Hum Reprod 2008;23:1594–601. 18. Luque-Ramı´rez M, Mendieta-Azcona C, del Rey Sa´nchez JM, et al. Effects of an antiandrogenic oral contraceptive pill compared with metformin on blood coagulation tests and endothelial function in women with the polycystic ovary syndrome: influence of obesity and smoking. Eur J Endocrinol 2009;160:469–80. 19. Moran LJ, Meyer C, Hutchison SK, et al. Novel inflammatory markers in overweight women with and without polycystic ovary syndrome and following pharmacological intervention. J Endocrinol Invest 2010;33:258–65. 20. Kebapcilar L, Taner CE, Kebapcilar AG, et al. Comparison of four different treatment regimens on coagulation parameters, hormonal

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

859

and metabolic changes in women with polycystic ovary syndrome. Arch Gynecol Obstet 2010;281:35–42. Essah PA, Arrowood JA, Cheang KI, et al. Effect of combined metformin and oral contraceptive therapy on metabolic factors and endothelial function in overweight and obese women with polycystic ovary syndrome. Fertil Steril 2011;96:501–4. Orbetzova MM, Pehlivanov BK, Mitkov MM, et al. Effect of shortterm standard therapeutic regimens on neuropeptide Y and adipose tissue hormones in overweight insulin-resistant women with polycystic ovary syndrome. Folia Med (Plovdiv) 2011;53:15–24. Kilic S, Yilmaz N, Zulfikaroglu E, et al. Inflammatory-metabolic parameters in obese and nonobese normoandrogenemic polycystic ovary syndrome during metformin and oral contraceptive treatment. Gynecol Endocrinol 2011;27:622–9. Vieira CS, Martins WP, Fernandes JB, et al. The effects of 2 mg chlormadinone acetate/30 mcg ethinylestradiol, alone or combined with spironolactone, on cardiovascular risk markers in women with polycystic ovary syndrome. Contraception 2012;86:268–75. Uras R, Orru` M, Pani F, et al. Endocrinological, metabolic and clinical features of treatment with oral contraceptive formulation containing ethinylestradiol plus chlormadinone acetate in nonobese women with polycystic ovary syndrome. Contraception 2010;82: 131–8. Minozzi M, Costantino D, Guaraldi C, Unfer V. The effect of a combination therapy with myo-inositol and a combined oral contraceptive pill versus a combined oral contraceptive pill alone on metabolic, endocrine, and clinical parameters in polycystic ovary syndrome. Gynecol Endocrinol 2011;27:920–4. Banaszewska B, Spaczyn´ski RZ, Ozegowska K, Pawelczyk L. [The influence of low-dose oral contraceptive pill on clinical and metabolic parameters in young women with polycystic ovary syndrome]. Ginekol Pol 2011;82:430–5. Gomes VA, Vieira CS, Jacob-Ferreira AL, et al. Oral contraceptive containing chlormadinone acetate and ethinylestradiol reduces plasma concentrations of matrix metalloproteinase-2 in women with polycystic ovary syndrome. Basic Clin Pharmacol Toxicol 2012;111:211–16. Aydin K, Cinar N, Aksoy DY, et al. Body composition in lean women with polycystic ovary syndrome: effect of ethinyl estradiol and drospirenone combination. Contraception 2013;87:358–62. De Leo V, Fruzzetti F, Musacchio MC, et al. Effect of a new oral contraceptive with estradiol valerate/dienogest on carbohydrate metabolism. Contraception 2013;88:364–8. Cinar N, Harmanci A, Bayraktar M, Yildiz BO. Ethinyl estradioldrospirenone vs ethinyl estradiol-drospirenone plus metformin in the treatment of lean women with polycystic ovary syndrome. Clin Endocrinol (Oxf) 2013;78:379–84. Harmanci A, Cinar N, Bayraktar M, Yildiz BO. Oral contraceptive plus antiandrogen therapy and cardiometabolic risk in polycystic ovary syndrome. Clin Endocrinol (Oxf) 2013;78:120–5. Guastella E, Longo RA, Carmina E. Clinical and endocrine characteristics of the main polycystic ovary syndrome phenotypes. Fertil Steril 2010;94:2197–201. Rehme MF, Pontes AG, Goldberg TB, et al. Manifestac¸o˜es clı´nicas, bioquı´micas, ultrassonogra´ficas e metabo´licas da sı´ndrome dos ova´rios policı´sticos em adolescentes [Clinical manifestations, biochemical, ultrasonographic and metabolic of polycystic ovary syndrome in adolescents]. Rev Bras Ginecol Obstet 2013;35: 249–54. Brown ZA, Louwers YV, Fong SL, et al. The phenotype of polycystic ovary syndrome ameliorates with aging. Fertil Steril 2011;96:1259–65. Baranova A, Tran TP, Birerdinc A, Younossi ZM. Systematic review: association of polycystic ovary syndrome with metabolic syndrome and non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2011;33:801–14. Baptiste CG, Battista MC, Trottier A, Baillargeon JP. Insulin and hyperandrogenism in women with polycystic ovary syndrome. J Steroid Biochem Mol Biol 2010;122:42–52. Toulis KA, Goulis DG, Mintziori G, et al. Meta-analysis of cardiovascular disease risk markers in women with polycystic ovary syndrome. Hum Reprod Update 2011;17:741–60. de Groot PC, Dekkers OM, Romijn JA, et al. PCOS, coronary heart disease, stroke and the influence of obesity: a systematic review and meta-analysis. Hum Reprod Update 2011;17:495–500.

Downloaded by [Australian Catholic University] at 08:08 28 September 2017

860

N. Mendoza et al.

Gynecol Endocrinol, 2014; 30(12): 850–860

40. Mendoza N. Common genetic aspects between polycystic ovary syndrome and diabetes mellitus. Curr Diabetes Rev 2011;7:377–91. 41. Toulis KA, Goulis DG, Kolibianakis EM, et al. Risk of gestational diabetes mellitus in women with polycystic ovary syndrome: a systematic review and a meta-analysis. Fertil Steril 2009;92: 667–77. 42. Palomba S, Falbo A, Russo T, et al. Pregnancy in women with polycystic ovary syndrome: the effect of different phenotypes and features on obstetric and neonatal outcomes. Fertil Steril 2010;94: 1805–11. 43. Moran LJ, Hutchison SK, Norman RJ, Teede HJ. Lifestyle changes in women with polycystic ovary syndrome. Cochrane Database Syst Rev 2011;(7):CD007506. 44. Moran L, Teede H. Metabolic features of the reproductive phenotypes of polycystic ovary syndrome. Hum Reprod Update 2009;15: 477–88. 45. Moghetti P, Tosi F, Bonin C, et al. Divergences in insulin resistance between the different phenotypes of the polycystic ovary syndrome. J Clin Endocrinol Metab 2013;98:E628–37. 46. ESHRE Capri Workshop Group. Health and fertility in World Health Organization group 2 anovulatory women. Hum Reprod Update 2012;18:586–99. 47. Dunaif A, Fauser BC. Renaming PCOS – a two-state solution. J Clin Endocrinol Metab 2013;98:4325–8. 48. Legro RS, Arslanian SA, Ehrmann DA, et al. Diagnosis and treatment of polycystic ovary syndrome: an endocrine society clinical practice guideline. J Clin Endocrinol Metab 2013;98:4565–92. 49. Vrbı´kova´ J, Cibula D. Combined oral contraceptives in the treatment of polycystic ovary syndrome. Hum Reprod Update 2005;11: 277–91. 50. Bird ST, Hartzema AG, Brophy JM, et al. Risk of venous thromboembolism in women with polycystic ovary syndrome: a population-based matched cohort analysis. CMAJ 2013;185: E115–20. 51. Fruzzetti F, Tre´mollieres F, Bitzer J. An overview of the development of combined oral contraceptives containing estradiol: focus on estradiol valerate/dienogest. Gynecol Endocrinol 2012;28: 400–8.

52. Kallio S, Puurunen J, Ruokonen A, et al. Antimu¨llerian hormone levels decrease in women using combined contraception independently of administration route. Fertil Steril 2013;99:1305–10. 53. Anthuber S, Schramm G, Heskamp ML. Six-month evaluation of the benefits of the low-dose combined oral contraceptive chlormadinone acetate 2mg/ethinylestradiol 0.003mg in young women. Clin Drug Investig 2010;30:211–20. 54. Go¨retzlehner G, Waldmann-Rex S, Schramm G. Extended cycles with the combined oral contraceptive chlormadinone acetate 2 mg/ ethinylestradiol 0.003 mg. Clin Drug Investig 2011;31:269–77. 55. Sa¨nger N, Stahlberg S, Manthey T, et al. Effects of an oral contraceptive containing 30 mcg ethinyl estradiol and 2 mg dienogest on thyroid hormones and androgen parameters: conventional vs. extended-cycle use. Contraception 2008;77:420–5. 56. Bozdag G, Yildiz BO. Combined oral contraceptives in polycystic ovary syndrome- indications and cautions. Front Horm Res 2013;40: 115–27. 57. Westhoff CL, Torgal AT, Mayeda ER, et al. Predictors of noncompliance in an oral contraceptive clinical trial. Contraception 2012;85:465–9. 58. Halperin IJ, Kumar SS, Stroup DF, Laredo SE. The association between the combined oral contraceptive pill and insulin resistance, dysglycemia and dyslipidemia in women with polycystic ovary syndrome: a systematic review and meta-analysis of observational studies. Hum Reprod 2011;26:191–201. 59. Costello MF, Shrestha B, Eden J, et al. Metformin versus oral contraceptive pill in polycystic ovary syndrome: a Cochrane review. Hum Reprod 2007;22:1200–9. 60. Jing Z, Liang-Zhi X, Tai-Xiang W, et al. The effects of Diane-35 and metformin in treatment of polycystic ovary syndrome: an updated systematic review. Gynecol Endocrinol 2008;24:590–600. 61. World Health Organization. Medical eligibility criteria for contraceptive use. 4th ed. Available from: http://whqlibdoc.who.int/ publications/2010/9789241563888_eng.pdf. 2009. 62. Verhaeghe J. Hormonal contraception in women with the metabolic syndrome: a narrative review. Eur J Contracept Reprod Health Care 2010;15:305–13.

Appendix A: Search strategy

‘‘contraceptives’’[All Fields])) OR ((‘‘polycystic ovary syndrome’’[MeSH Terms] OR (‘‘polycystic’’[All Fields] AND ‘‘ovary’’[All Fields] AND ‘‘syndrome’’[All Fields]) OR ‘‘polycystic ovary syndrome’’[All Fields] OR (‘‘polycystic’’[All Fields] AND ‘‘ovary’’[All Fields]) OR ‘‘polycystic ovary’’[All Fields]) AND (‘‘contraception’’[MeSH Terms] OR ‘‘contraception’’[All Fields]))

((‘‘polycystic ovary syndrome’’[MeSH Terms] OR (‘‘polycystic’’[All Fields] AND ‘‘ovary’’[All Fields] AND ‘‘syndrome’’[All Fields]) OR ‘‘polycystic ovary syndrome’’[All Fields]) AND hormonal[All Fields] AND (‘‘contraceptive agents’’[Pharmacological Action] OR ‘‘contraceptive agents’’[MeSH Terms] OR (‘‘contraceptive’’[All Fields] AND ‘‘agents’’[All Fields]) OR ‘‘contraceptive agents’’[All Fields] OR

Hormonal contraceptive choice for women with PCOS: a systematic review of randomized trials and observational studies.

This paper provides a critical review of the data concerning the effects of combined hormonal contraceptives (CHC) for polycystic ovary syndrome (PCOS...
503KB Sizes 0 Downloads 6 Views