Scolaris Content Display Scolaris Content Display

胰岛素增敏药物(二甲双胍、罗格列酮、吡格列酮、D‐手性肌醇)治疗患有多囊卵巢综合征、少发性闭经和不孕症的女性

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Ayaz 2013 {published data only}

Ayaz A, Alwan Y, Farooq MU. Efficacy of combined metformin‐clomiphene citrate in comparison with clomiphene citrate alone in infertile women with polycystic ovarian syndrome (PCOS). Journal of Medicine and Life 2013;6:199‐201. CENTRAL
Ayaz A, Alwan Y, Farooq MU. Metformin‐clomiphene citrate vs. clomiphene citrate alone: polycystic ovarian syndrome. Journal of Human Reproductive Sciences 2013;6:15‐8. CENTRAL

Baillargeon 2004 {published data only}

Baillargeon JP, Jakubowicz DJ, Iuorno MJ, Jakubowicz S, Nestler JE. Effects of metformin and rosiglitazone, alone and in combination, in nonobese women with polycystic ovary syndrome and normal indices of insulin sensitivity. Fertility and Sterility 2004;82:893‐902. CENTRAL

Begum 2014 {published data only}

Begum H, Chowdhury TA, Begum F, Begum SN, Kabir N, Haque MDA. Effect of metformin and clomiphene citrate in improving fertility in subfertility women with polycystic ovary syndrome: a randomised controlled trial. Bangladesh Journal of Obstetrics and Gynaecology 2014;29:15‐20. CENTRAL

Ben Ayed 2009 {published data only}

Ben Ayed B, Dammak Dit Mlik S, Ben Arab H, Trabelssi H, Chahtour H, Mathlouthi N. Metformin effects on clomifene‐induced ovulation in the polycystic ovary syndrome. Tunisie Medicale 2009;87(1):43‐8. CENTRAL

Boudhraa 2010 {published data only}

Boudhrâa K, Jellouli MA, Amri M, Farhat M, Torkhani F, Gara MF. Indication of metformin in the management of hormonal dysfunction secondary to polycystic ovarian syndrome: prospective comparative study of 63 cases. [La metformine dans la prise en charge de l'infertilite du SOPK hors FIV etude prospective comparative a propos de 63 cas]. Tunisie Medicale 2010;88(5):335‐40. CENTRAL

Brettenthaler 2004 {published data only}

Aigner E, Bachofner N, Klein K, De Geyter C, Hohla F, Patsch W, et al. Retinol‐binding protein 4 in polycystic ovary syndrome‐‐association with steroid hormones and response to pioglitazone treatment. Journal of Clinical Endocrinology and Metabolism 2009;94(4):1229‐35. CENTRAL
Brettenthaler N, De Geyter C, Huber PR, Keller U. Effect of the insulin sensitizer pioglitazone on insulin resistance, hyperandrogenism, and ovulatory dysfunction in women with polycystic ovary syndrome. Journal of Clinical Epidemiology 2004;89:3835‐40. CENTRAL

Carmina 2004 {published data only}

Carmina E, Lobo RA. Does metformin induce ovulation in normoandrogenic anovulatory women?. American Journal of Obstetrics and Gynecology 2004;191:1580‐4. CENTRAL

Chou 2003 {published data only}

Chou KH, von Eye Corleta H, Capp E, Spritzer PM. Clinical, metabolic and endocrine parameters in response to metformin in obese women with polycystic ovary syndrome: a randomized, double‐blind and placebo‐controlled trial. Hormone and Metabolic Research 2003;35:86‐91. CENTRAL

Eisenhardt 2006 {published data only}

Eisenhardt S, Schwarzmann N, Henschel V, Germeyer A, von Wolff M, Hamann A, et al. Early effects of metformin in women with polycystic ovary syndrome: a prospective randomized, double‐blind, placebo‐controlled trial. Journal of Clinical Endocrinology and Metabolism 2006;91:946‐52. CENTRAL

El‐Biely 2001 {published data only}

El‐Biely MM, Habba M. The use of metformin to augment the induction of ovulation in obese infertile patients with polycystic ovary syndrome. Middle East Fertility Society Journal 2001;6(1):43‐9. CENTRAL

Fleming 2002 {published data only}

Fleming R, Hopkinson ZE, Wallace AM, Greer IA, Sattar N. Ovarian function and metabolic factors in women with oligomenorrhea treated with metformin in a randomized double blind placebo‐controlled trial. Journal of Clinical Endocrinology and Metabolism 2002;87(2):569‐74. CENTRAL
Fleming R, Hopkinson ZE, Wallace E, Greer IA, Sattar N. Metformin treatment improves ovulation frequency in women with oligomenorrhoea: a randomized, double blind placebo trial. Human Fertility2000; Vol. 3, issue 4:389‐90. CENTRAL

Gerli 2003 {published data only}

Gerli S, Mignosa M, Di Renzo GC. Effects of inositol on ovarian function and metabolic factors in women with PCOS: a randomized double blind placebo‐controlled trial. European Review for Medical and Pharmacological Sciences 2003;7:151‐9. [Original article]CENTRAL
Gerli S, Papaleo E, Ferrari A, Di Renzo GC. Randomized, double blind placebo‐controlled trial: effects of myo‐inositol on ovarian function and metabolic factors in women with PCOS. European Review for Medical and Pharmacological Sciences 2007;11(5):347‐54. CENTRAL

Glintborg 2005 {published data only}

Glintborg D, Frystyk J, Hojlund K, Andersen KK, Henriksen JE, Hermann A, et al. Total and high molecular weight (HMW) adiponectin levels and measures of glucose and lipid metabolism following pioglitazone treatment in a randomized placebo‐controlled study in polycystic ovary syndrome. Clinical Endocrinology (Oxford) 2008;68(2):165‐74. CENTRAL
Glintborg D, Hermann AP, Andersen M, Hagen C, Beck‐Nielsen H, Veldhuis JD, et al. Effect of pioglitazone on glucose metabolism and luteinizing hormone secretion in women with polycystic ovary syndrome. Fertility and Sterility 2006;86:385‐97. CENTRAL
Glintborg D, Hojlund K, Andersen M, Henriksen JE, Beck‐Nielsen H, Handberg A. Soluble CD36 and risk markers of insulin resistance and atherosclerosis are elevated in polycystic ovary syndrome and significantly reduced during pioglitazone treatment. Diabetes Care 2008;31:328‐34. CENTRAL
Glintborg D, Stoving RK, Hagen C, Hermann AP, Frystyk J, Veldhuis JD, et al. Pioglitazone treatment increases spontaneous growth hormone (GH) secretion and stimulated GH levels in polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 2005;90:5605‐12. [original article]CENTRAL

Hoeger 2004 {published data only}

Hoeger KM, Kochman L, Wixom N, Craig K, Miller RK, Guzick DS. A randomized, 48‐week, placebo‐controlled trial of intensive lifestyle modification and/or metformin therapy in overweight women with polycystic ovary syndrome: a pilot study. Fertility and Sterility 2004;82:421‐9. CENTRAL

Hwu 2005 {published data only}

Hwu YM, Lin SY, Huang WY, Lin MH, Lee RK. Ultra‐short metformin pretreatment for clomiphene citrate‐resistant polycystic ovary syndrome. International Journal of Gynaecology and Obstetrics 2005;90:39‐43. CENTRAL

Jakubowicz 2001 {published data only}

Jakubowicz DJ, Seppala M, Jakubowicz S, Rodriguez‐Armas O, Rivas‐Santiago A, Koistinen H, et al. Insulin reduction with metformin increases luteal phase serum glycodelin and insulin‐like growth factor‐binding protein 1 concentrations and enhances uterine vascularity and blood flow in the polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 2001;86(3):1126‐33. CENTRAL

Kar 2015 {published data only}

Kar S. Metabolic risks of the lean PCOS woman. Fertility and Sterility ASRM abstracts 2013;100(S359):P‐730. CENTRAL
Kar S, Sanchita S. Clomiphene citrate, metformin or a combination of both as the first line ovulation induction drug for Asian Indian women with polycystic ovarian syndrome: a randomized controlled trial. Journal of Human Reproductive Science 2015;8:197‐201. CENTRAL

Karimzadeh 2007 {published data only}

Karimzadeh M, Tayebi N, Eftekhar M, Sakhavat L, Taheripanah R, Zare F. The effect of administration of metformin on lipid profile changes and insulin resistance in patients with polycystic ovary syndrome. Middle East Fertility Society Journal 2007;12(3):174‐8. CENTRAL

Karimzadeh 2010 {published data only}

Karimzadeh MA, Javedani M. An assessment of lifestyle modification versus medical treatment with clomiphene citrate, metformin, and clomiphene citrate‐metformin in patients with polycystic ovary syndrome. Fertility and Sterility 2010;94(1):216‐20. CENTRAL

Khorram 2006 {published data only}

Khorram O, Helliwell JP, Katz S, Bonpane CM, Jaramillo L Khorram O, et al. Two weeks of metformin improves clomiphene citrate‐induced ovulation and metabolic profiles in women with polycystic ovary syndrome. Fertility and Sterility 2006;85:1448‐51. CENTRAL

Lam 2011 {published data only}

Lam PM, Tam WH, Ma RC, Cheung LP, Tsui MH, Tong PC, et al. The reproductive and metabolic effect of rosiglitazone on Chinese women with polycystic ovarian syndrome ‐ a double‐blind randomized placebo‐controlled study. Fertility and Sterility 2011;96(2):445‐51. CENTRAL

Legro 2007 {published data only}

Cataldo N, Barnhart H, Legro R, Myers E, Schlaff W, Carr B, et al. Extended‐release metformin does not reduce the clomiphene citrate dose required to induce ovulation in polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 2008;93(8):3147‐7. CENTRAL
Legro RS, Barnhart HX, Schlaff WD, Carr BR, Diamond MP, Carson SA, et al. Clomiphene, metformin, or both for infertility in the polycystic ovary syndrome. New England Journal of Medicine 2007;356:551‐66. [original article]CENTRAL

Lord 2006 {published data only}

Lord J, Thomas R, Fox B, Acharya U, Wilkin T. The effect of metformin on fat distribution and the metabolic syndrome in women with polycystic ovary syndrome‐‐a randomised, double‐blind, placebo‐controlled trial. British Journal of Obstetrics and Gynaecology 2006;113:817‐24. CENTRAL

Machado 2012 {published data only}

Machado RC, Machado NA, Geber S. Evaluation of the use of metformin for ovulation in patients with PCOS resistant to isolated use of clomiphene citrate. Brazilian Journal Assisted Reproduction 2012;16(1):27‐31. CENTRAL

Maciel 2004 {published data only}

Maciel GA, Soares Jr JM, Alves da Motta EL, Abi Haidar M, de Lima GR, Baracat EC, et al. Nonobese women with polycystic ovary syndrome respond better than obese women to treatment with metformin. Fertility and Sterility 2004;81:355‐60. CENTRAL

Malkawi 2002 {published data only}

Malkawi HY, Qublan HS. The effect of metformin plus clomiphene citrate on ovulation and pregnancy rates in clomiphene‐resistant women with polycystic ovary syndrome. Saudi Medical Journal 2002;23(6):663‐6. [MEDLINE: 22065897]CENTRAL

Moghetti 2000 {published data only}

Moghetti P, Castello R, Negri C, Tosi F, Perrone F, Caputo M, et al. Metformin effects on clinical features, endocrine and metabolic profiles, and insulin sensitivity in polycystic ovary syndrome: a randomized, double‐blind, placebo‐controlled 6‐month trial, followed by open, long‐term clinical evaluation. Journal of Clinical Endocrinology and Metabolism 2000;85(1):139‐46. CENTRAL

Moll 2006 {published data only}

Moll E, Bossuyt PM, Korevaar JC, Lambalk CB, Van der Veen F. Effect of clomifene citrate plus metformin and clomifene citrate plus placebo on induction of ovulation in women with newly diagnosed polycystic ovary syndrome: randomised double blind clinical trial. BMJ 2006;332:1485. [original article]CENTRAL
Moll E, Korevaar JC, Bossuyt PM, van der Veen F. Does adding metformin to clomifene citrate lead to higher pregnancy rates in a subset of women with polycystic ovary syndrome?. Human Reproduction 2008;23(8):1830‐4. CENTRAL
Moll E, Van Wely M, Lambalk CB, Bossuyt PM, Van der Veen F. Health‐related quality of life in women with newly diagnosed polycystic ovary syndrome randomized between clomifene citrate plus metformin or clomifene citrate plus placebo. Human Reproduction 2012;27:3273‐8. CENTRAL

Morin‐Papunen 2012 {published data only}

Morin‐Papunen L, Rantala AS, Unkila‐Kallio L, Tiitinen A, Hippeläinen M, Perheentupa A, et al. Metformin improves pregnancy and live‐birth rates in women with polycystic ovary syndrome (PCOS): a multicenter, double‐blind, placebo‐controlled randomized trial. Journal of Clinical Endocrinology and Metabolism 2012;97:1492‐1500. CENTRAL

Nestler 1998 {published data only}

Nestler JE, Jakubowicz DJ, Evans WS, Pasquali R. Effects of metformin on spontaneous and clomiphene‐induced ovulation in the polycystic ovary syndrome. New England Journal of Medicine 1998;338(26):1876‐80. CENTRAL

Nestler 1999 {published data only}

Nestler JE, Jakubowicz DJ, Reamer P, Gunn RD, Allan G. Ovulatory and metabolic effects of D‐chiro‐inositol in the polycystic ovary syndrome. New England Journal of Medicine 1999;340(17):1314‐20. CENTRAL

Ng 2001 {published data only}

Ng EHY, Wat NMS, Ho PC. Effects of metformin on ovulation rate, hormonal and metabolic profiles in women with clomiphene‐resistant polycystic ovaries: a randomized, double‐blinded placebo‐controlled trial. Human Reproduction 2001;16(8):1625‐31. CENTRAL

Onalan 2005 {published data only}

Onalan G, Goktolga U, Ceyhan T, Bagis T, Onalan R, Pabuccu R. Predictive value of glucose‐insulin ratio in PCOS and profile of women who will benefit from metformin therapy: obese, lean, hyper or normoinsulinemic?. European Journal of Obstetrics, Gynecology, and Reproductive Biology 2005;123:204‐11. CENTRAL

Otta 2010 {published data only}

Otta CF, Wior M, Iraci GS, Kaplan R, Torres D, Gaido MI, et al. Clinical, metabolic, and endocrine parameters in response to metformin and lifestyle intervention in women with polycystic ovary syndrome: a randomized, double‐blind, and placebo control trial. Gynecological Endocrinology 2010;26(3):173‐8. CENTRAL

Palomba 2005 {published data only}

Palomba S, Orio F, Falbo A, Manguso F, Russo T, Cascella T, et al. Prospective parallel randomized, double‐blind, double‐dummy controlled clinical trial comparing clomiphene citrate and metformin as the first‐line treatment for ovulation induction in nonobese anovulatory women with polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 2005;90(7):4068‐74. [original article]CENTRAL
Palomba S, Orio F, Falbo A, Russo T, Tolino A, Zullo F. Effects of metformin and clomiphene citrate on ovarian vascularity in patients with polycystic ovary syndrome. Fertility and Sterility 2006;86:1694‐701. CENTRAL

Pasquali 2000 {published data only}

Casimirri F, Biscotti M, Gambineri A, Calzoni F, Eliana B, Pasquali R. Metformin improves insulin, body fat distribution, and androgens in obese women with and without the polycystic ovary syndrome. International Journal of Obesity 1997;21 Suppl 2:61. CENTRAL
Pasquali R, Gambineri A, Biscotti D, Vicennati V, Gagliardi L, Colitta D, et al. Effect of long‐term treatment with metformin added to hypocaloric diet on body composition, fat distribution, and androgen and insulin levels in abdominally obese women with and without the polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 2000;85(8):2767‐74. CENTRAL

PCOSMIC 2010 {published data only}

Johnson NP, Bontekoe S, Stewart AW. Analysis of factors predicting success of metformin and clomiphene treatment for women with infertility owing to PCOS‐related ovulation dysfunction in a randomised controlled trial. Australian & New Zealand Journal of Obstetrics & Gynaecology 2011;51:252‐6. CENTRAL
Johnson NP, Stewart AW, Falkiner J, Farquhar CM, Milsom S, Singh V‐P, et al. on behalf of REACT‐NZ (REproduction And Collaborative Trials in New Zealand), a multi‐centre fertility trials group. PCOSMIC: a multi‐centre randomized trial in women with Polycystic Ovary Syndrome evaluating Metformin for Infertility with Clomiphene. Human Reproduction 2010;25(7):1675‐83. [MEDLINE: 10.1093/humrep/deq100]CENTRAL

Rautio 2006 {published data only}

Rautio K, Tapanainen JS, Ruokonen A, Morin‐Papunen LC. Endocrine and metabolic effects of rosiglitazone in overweight women with PCOS: a randomized placebo‐controlled study. Human Reproduction 2006;21:1400‐7. CENTRAL
Rautio K, Tapanainen JS, Ruokonen A, Morin‐Papunen LC. Rosiglitazone treatment alleviates inflammation and improves liver function in overweight women with polycystic ovary syndrome: a randomized placebo‐controlled study. Fertility and Sterility 2007;87(1):202‐6. CENTRAL

Romualdi 2010 {published data only}

Romualdi D, Giuliani M, Cristello F, Fulghesu AM, Selvaggi L, Lanzone A, et al. Metformin effects on ovarian ultrasound appearance and steroidogenic function in normal‐weight normoinsulinemic women with polycystic ovary syndrome: a randomized double‐blind placebo‐controlled clinical trial. Fertility and Sterility 2010;93(7):2303‐10. CENTRAL

Sahin 2004 {published data only}

Sahin Y, Yirmibes U, Kelestimur F, Aygen E. The effects of metformin on insulin resistance, clomiphene‐induced ovulation and pregnancy rates in women with polycystic ovary syndrome. European Journal of Obstetrics, Gynecology, and Reproductive Biology 2004;113:214‐20. CENTRAL

Siebert 2009 {published data only}

Siebert T, Kruger T, Lombard C. Evaluating the equivalence of clomiphene citrate with and without metformin in ovulation induction in PCOS patients. Journal of Assisted Reproduction and Genetics 2009;26(4):165‐71. CENTRAL

Sturrock 2002 {published data only}

Sturrock NDC, Lannon B, Fay TN. Metformin does not enhance ovulation induction in clomiphene resistant polycystic ovary syndrome in clinical practice. British Journal of Clinical Pharmacology 2002;53(5):469‐73. [MEDLINE: 2002180629]CENTRAL

Tang 2006 {published data only}

Tang T, Glanville J, Hayden CJ, White D, Barth JH, Balen AH. Combined lifestyle modification and metformin in obese patients with polycystic ovary syndrome. A randomized, placebo‐controlled, double‐blind multicentre study. Human Reproduction 2006;21:80‐9. CENTRAL

Trolle 2007 {published data only}

Trolle B, Flyvbjerg A, Kesmodel U, Lauszus FF. Efficacy of metformin in obese and non‐obese women with polycystic ovary syndrome: a randomized, double‐blinded, placebo‐controlled cross‐over trial. Human Reproduction 2007;22(11):2967‐73. [original article]CENTRAL
Trolle B, Flyvbjerg A, Kesmodel U, Lauszus FF. Efficacy of metformin in obese and non‐obese women with polycystic ovary syndrome: a randomized, double‐blinded, placebo‐controlled cross‐over trial. Obstetrics and Gynecological Survey 2008;63(2):96‐8. CENTRAL

Vandermolen 2001 {published data only}

Vandermolen DT, Ratts VS, Evans WS, Stovall DW, Kauma SW, Nestler JE. Metformin increases the ovulatory rate and pregnancy rate from clomiphene citrate in patients with polycystic ovary syndrome who are resistant to clomiphene citrate alone. Fertility and Sterility 2001;75:310‐5. CENTRAL

Williams 2009 {published data only}

Williams SC, Pastore LM, Shelly WB, Bailey AP, Baras DC, Bateman BG. A randomized, placebo‐controlled study of the influence of instant‐release metformin on response to clomiphene citrate and time to conception in polycystic ovary syndrome. Fertility and Sterility 2009;92 Suppl(3):105. CENTRAL

Yarali 2002 {published data only}

Yarali H, Yildiz BO, Demirol A, Zeyneloglu HB, Yigit N, Bukulmez O, et al. Co‐administration of metformin during rFSH treatment in patients with clomiphene citrate‐resistant polycystic ovarian syndrome: a prospective randomized trial. Human Reproduction 2002;17(2):289‐94. CENTRAL
Yarali H, Yyldyz B, Demirol A, Zeyneloglu H, Yigit N, Bukulmez O. Co‐administration of metformin during recombinant follicle stimulating hormone (recombinant FSH) treatment using the low‐dose step protocol in patients with clomiphene citrate resistant polycystic ovary syndrome (PCOS): a prospective randomized trial. Fertility and Sterility 2001;76 Suppl(3):36. CENTRAL

Zain 2009 {published data only}

Zain MM, Jamaluddin R, Ibrahim A, Norman RJ. Comparison of clomiphene citrate, metformin, or the combination of both for first‐line ovulation induction, achievement of pregnancy, and live birth in Asian women with polycystic ovary syndrome: a randomized controlled trial. Fertility and Sterility 2009;91(2):514‐21. CENTRAL

References to studies excluded from this review

Abuelghar 2013 {published data only}

Abuelghar WM, Elkady OS, Khamees AA. Clomiphene citrate alone, in combination with metformin or in combination with pioglitazone as first line therapy in induction of ovulation in infertile women with polycystic ovary syndrome, a randomised controlled trial. Middle East Fertility Society Journal 2013;18:135‐41. CENTRAL

Aroda 2009 {published data only}

Aroda V, Ciaraldi T, Burke P, Mudaliar S, Clopton P, Phillips S, et al. Metabolic and hormonal changes induced by pioglitazone in polycystic ovary syndrome: a randomized, placebo‐controlled clinical trial. Journal of Clinical Endocrinology and Metabolism 2009;94(2):469‐76. CENTRAL

Azziz 2001 {published data only}

Azziz R, Ehrmann D, Legro RS, Whitcomb RW, Hanley R, Fereshetian AG, et al. Troglitazone improves ovulation and hirsutism in the polycystic ovary syndrome: a multicenter, double blind, placebo‐controlled trial. Journal of Clinical Endocrinology and Metabolism 2001;86(4):1626‐32. [original article]CENTRAL
Guyatt G, Weaver B, Cronin L, Dooley JA, Azziz R. Health‐related quality of life in women with polycystic ovary syndrome, a self‐administered questionnaire, was validated. Journal of Clinical Epidemiology 2004;57:1279‐87. CENTRAL
Legro RS, Azziz R, Ehrmann D, Fereshetian AG, O'Keefe M, Ghazzi MN. Minimal response of circulating lipids in women with polycystic ovary syndrome to improvement in insulin sensitivity with troglitazone. Journal of Clinical Endocrinology and Metabolism 2003;88:5137‐44. CENTRAL

Azziz 2003 {published data only}

Azziz R, Ehrmann DA, Legro RS, Fereshetian AG, O'Keefe M, Ghazzi MN. Troglitazone decreases adrenal androgen levels in women with polycystic ovary syndrome. Fertility and Sterility 2003;79:932‐7. CENTRAL

Chaudhry 2016 {published data only}

Chaudhry I, Nisa SU, Nia SU. Comparison between pioglitazone and metformin in terms of efficacy in patients with polycystic ovarian syndrome. Pakistan Journal of Medical and Health Sciences 2016;10:574‐7. CENTRAL

Chaudhury 2008 {published data only}

Chaudhury K, Chaudhury S, Chowdhury S. Does metformin augment the ovulation inducing effects of clomiphene in non‐obese women with polycystic ovary syndrome?. Journal of the Indian Medical Association 2008;106(10):643‐8. CENTRAL

Constantino 2009 {published data only}

Costantino D, Minozzi G, Minozzi F, Guaraldi C. Metabolic and hormonal effects of myo‐inositol in women with polycystic ovary syndrome: a double‐blind trial. European Review for Medical and Pharmacological Sciences 2009;13(2):105‐10. CENTRAL

Crave 1995 {published data only}

Crave JC, Fimbel S, Lejeune H, Cugnardey N, Dechaud H, Pugeat M. Effects of diet and metformin administration on sex hormone‐binding globulin, androgens, and insulin in hirsute and obese women. Journal of Clinical Endocrinology and Metabolism 1995;80(7):2057‐62. CENTRAL

Curi 2012 {published data only}

Curi DD, Fonseca AM, Marcondes JA, Almeida JA, Bagnoli VR, Soares JM, et al. Metformin versus lifestyle changes in treating women with polycystic ovary syndrome. Gynecological Endocrinology 2010;28:182‐5. CENTRAL

De Leo 1999 {published data only}

De Leo V, La Marca A, Ditto A, Morgante G, Cianci A. Effects of metformin on gonadotropin‐induced ovulation in women with polycystic ovary syndrome. Fertility and Sterility 1999;72(2):282‐5. CENTRAL

Dunaif 1996 {published data only}

Dunaif A, Scott D, Finegood D, Quintana B, Whitcomb R. The insulin‐sensitizing agent troglitazone improves metabolic and reproductive abnormalities in the polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 1996;81(9):3299‐306. CENTRAL

Elter 2002 {published data only}

Elter K, Imir G, Durmusoglu F. Clinical, endocrine and metabolic effects of metformin added to ethinyl estradiol‐cyproterone acetate in non‐obese women with polycystic ovarian syndrome: a randomized controlled study. Human Reproduction 2002;17(7):1729‐37. [MEDLINE: 785]CENTRAL

Farzadi 2006 {published data only}

Farzadi L, Salman S. Metformin‐therapy effects in 50 clomiphene citrate resistant PCOS patients. Journal of Medical Science 2006;6:765‐71. CENTRAL

Heathcote 2013 {published data only}

Heathcote G, Forbes K, Lee A, Gregor M, Luscombe G, Boothroyd C. Live birth rate after metformin and clomiphene vs clomiphene alone in polycystic ovary syndrome (PCOS): a randomized, double‐blind, placebo‐controlled trial. Draft paper supplied by authors. CENTRAL

Hou 2000 {published data only}

Hou J, Yu J, Wei M. Study on treatment of hyperandrogenism and hyperinsulinism in polycystic ovary syndrome with Chinese herbal formula "tiangui fang". Zhongguo Zhong Xi Yi Jie He Za Zhi 2000;20(8):589‐92. CENTRAL

Ibanez 2002 {published data only}

Ibanez L, Valls C, Ferrer A, Ong K, Dunger D, De‐Zegher F. Additive effects of insulin‐sensitizing and anti‐androgen treatment in young, nonobese women with hyperinsulinism, hyperandrogenism, dyslipidemia, and anovulation. Journal of Clinical Endocrinology and Metabolism 2002;87(6):2870‐4. [MEDLINE: 22045399]CENTRAL

Kazerooni 2009 {published data only}

Kazerooni T, Ghaffarpasand F, Kazerooni Y, Kazerooni M, Setoodeh S. Short‐term metformin treatment for clomiphene citrate‐resistant women with polycystic ovary syndrome. International Journal of Gynaecology and Obstetrics 2009;107(1):50‐3. CENTRAL

Kelly 2002 {published data only}

Kelly CJ, Gordon D. The effect of metformin on hirsutism in polycystic ovary syndrome. European Journal of Endocrinology 2002;147(2):217‐21. [MEDLINE: 782]CENTRAL

Kocak 2002 {published data only}

Kocak M, Caliskan E, Simsir C, Haberal A. Metformin therapy improves ovulatory rates, cervical scores, and pregnancy rates in clomiphene citrate‐resistant women with polycystic ovary syndrome. Fertility and Sterility 2002;77(1):101‐6. CENTRAL

Ladson 2011 {published data only}

Ladson G, Dodson WC, Sweet SD, Archibong AE, Kunselman AR, Demers LM, et al. The effects of metformin with lifestyle therapy in polycystic ovary syndrome: a randomized double‐blind study. Fertility and Sterility 2011;95(3):1059‐66. CENTRAL

Leanza 2014 {published data only}

Leanza V, Coco L, Grasso F, Leanza G, Zarbo G, Palumbo M. Ovulation induction with clomiphene citrate and metformin in women with polycystic ovary syndrome. Minerva Ginecologica 2014;66(3):299‐301. CENTRAL

Mantzoros 1997 {published data only}

Mantzoros CS, Dunaif A, Flier JS. Leptin concentrations in the polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 1997;82(6):1687‐91. CENTRAL

Morin‐Papunen 2000 {published data only}

Morin‐Papunen LC, Vauhkonen I, Koivunen RM, Ruokonen A, Martikainen HK, Tapanainen JS. Endocrine and metabolic effects of metformin versus ethinyl estradiol‐cyproterone acetate in obese women with polycystic ovary syndrome: a randomized study. Journal of Clinical Endocrinology and Metabolism 2000;85(9):3161‐8. [DOI: 10.1210/jcem.85.9.6792]CENTRAL

Morin‐Papunen 2010 {published data only}

Morin‐Papunen L, Rantala A, Unkila‐Kallio L, Tiitinen A, Hippelainen M, Tinkanen H, et al. Metformin improves pregnancy and live birth rates in women with polycystic ovary syndrome ‐ a multicentre placebo‐controlled randomised trial. 26th Annual Meeting of the European Society of Human Reproduction and Embryology, ESHRE Rome Italy 2010;25:i67‐8. CENTRAL

Nestler 1996 {published data only}

Nestler JE, Jakubowicz DJ. Decreases in ovarian cytochrome P450c17 alpha activity and serum free testosterone after reduction of insulin secretion in polycystic ovary syndrome. New England Journal of Medicine 1996;335(9):617‐23. CENTRAL

Nestler 1997 {published data only}

Nestler JE, Jakubowicz DJ. Lean women with polycystic ovary syndrome respond to insulin reduction with decreases in ovarian P450c17 alpha activity and serum androgens. Journal of Clinical Endocrinology and Metabolism 1997;82(12):4075‐9. CENTRAL

Palomba 2011 {published data only}

Palomba S, Falbo A, Di Cello A, Cappiello F, Tolino A, Zullo F. Does metformin affect the ovarian response to gonadotropins for in vitro fertilization treatment in patients with polycystic ovary syndrome and reduced ovarian reserve? A randomized controlled trial. Fertility and Sterility 2011;96:1128‐33. CENTRAL

Papaleo 2009 {published data only}

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. Fertility and Sterility 2009;91:1750‐4. CENTRAL

Ramzy 2003 {published data only}

Ramzy A, El‐Kateb S, Al‐Inany H, Aboulmaaty Z, Badie M. The use of metformin in overweight and lean infertile patients with polycystic ovarian syndrome: a randomised controlled trial. Middle East Fertility Society Journal 2003;8:143‐9. CENTRAL

Refaie 2005 {published data only}

Refaie A, Ibrahim G, Oash S. Characteristics of polycystic ovary syndrome with and without insulin resistance and the role of insulin sensitizing drug (metformin) in its management. Middle East Fertility Society Jounal 2005;10:142‐9. CENTRAL

Rouzi 2006 {published data only}

Rouzi AA, Ardawi MS. A randomized controlled trial of the efficacy of rosiglitazone and clomiphene citrate versus metformin and clomiphene citrate in women with clomiphene citrate‐resistant polycystic ovary syndrome. Fertility and Sterility 2006;85:428‐35. CENTRAL

Salman 2014 {published data only}

Salman SA, Farghaly TA, Attallah DA, Abdel‐Hafeez HA, Shaaban OM. Insulin sensitizing agent (metformin) improves clinical pregnancy rate in clomiphene citrate resistant polycystic ovarian syndrome patients with acanthosis nigricans. Fertility and Sterility 2014;102(Supplement 3):e139. CENTRAL

Santonocito 2009 {published data only}

Santonocito V, Rapisarda V, Abruzzo SRM, Pollicino R, Coco L, Zarbo G. Comparison between clomiphene citrate and metformin for induction of ovulatory cycles in infertile nonobese women with polycystic ovary syndrome. Giornale Italiano di Ostetricia e Ginecologia 2009;31(11‐12):455‐60. CENTRAL

Shobokshi 2003 {published data only}

Shobokshi A, Shaarawy M. Correction of insulin resistance and hyperandrogenism in polycystic ovary syndrome by combined rosiglitazone and clomiphene citrate therapy. Journal of the Society for Gynecologic Investigation 2003;10:99‐104. CENTRAL

Unfer 2011 {published data only}

Unfer V, Carlomagno G, Rizzo P, Raffone E, Roseff S. Myo‐inositol rather than D‐chiro‐inositol is able to improve oocyte quality in intracytoplasmic sperm injection cycles. A prospective, controlled, randomized trial. European Review for Medical and Pharmacological Sciences 2011;15:452‐7. CENTRAL

Abu Hashim 2016

Abu Hashim H. Twenty years of ovulation induction with metformin for PCOS; what is the best available evidence?. Reproductive BioMedicine Online 2016;32:44‐53.

Alebic 2014

Alebić MS, Bulum T, Stojanović N, Duvnjak L. Definition of insulin resistance using the homeostasis model assessment (HOMA‐IR) in IVF patients diagnosed with polycystic ovary syndrome (PCOS) according to the Rotterdam criteria. Endocrine 2014;47:625‐30.

Alvarez‐Blasco 2006

Alvarez‐Blasco F, Botella‐Carretero JI, San Millán JL, Escobar‐Morreale HF. Prevalence and characteristics of the polycystic ovary syndrome in overweight and obese women. Archives of Internal Medicine 2006;166:2081‐6.

Apridonidze 2005

Apridonidze T, Essah PA, Iuorno MJ, Nestler JE. Prevalence and characteristics of the metabolic syndrome in women with polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 2005;90:1929‐35.

Azziz 2005

Azziz R, Marin C, Hoq L, Badamgarav E, Song P. Health care‐related economic burden of the polycystic ovary syndrome during the reproductive life span. Journal of Clinical Endocrinology and Metabolism 2005;90:4650‐8.

Balen 2014

Balen AH. Infertility in Practice. 4th Edition. Boca Raton: CRC Press, 2014. [13:978‐1‐84184‐5]

Balen 2016

Balen AH, Morley LC, Misso M, Franks S, Legro RS, Wijeyaratne CN, et al. The management of anovulatory infertility in women with polycystic ovary syndrome: an analysis of the evidence to support the development of global WHO guidance. Human Reproduction Update 2016;22:687‐708. [DOI: 10.1093/humupd/dmw025]

Bordewijk 2017

Bordewijk EM, Nahuis M, Costello MF, Van der Veen F, Tso LO, Mol BWJ, et al. Metformin during ovulation induction with gonadotrophins followed by timed intercourse or intrauterine insemination for subfertility associated with polycystic ovary syndrome. Cochrane Database of Systematic Reviews 2017, Issue 1. [DOI: 10.1002/14651858.CD009090.pub2]

Cassina 2014

Cassina M, Donà M, Di Gianantonio E, Litta P, Clementi M. First‐trimester exposure to metformin and risk of birth defects: a systematic review and meta‐analysis. Human Reproduction Update 2014;20:656‐69.

Cedergren 2004

Cedergren MI. Maternal morbid obesity and the risk of adverse pregnancy outcome. Obstetrics and Gynecology 2004;103:219‐24.

Celik 2014

Celik C, Tasdemir N, Abali R, Bastu E, Yilmaz M. Progression to impaired glucose tolerance or type 2 diabetes mellitus in polycystic ovary syndrome: a controlled follow‐up study. Fertility and Sterility 2014;101:1123‐8.

Colca 2013

Colca JR, McDonald WG, Cavey GS, Cole SL, Holewa DD, Brightwell‐Conrad AS, et al. Identification of a mitochondrial target of thiazolidinedione insulin sensitizers (mTOT)‐‐relationship to newly identified mitochondrial pyruvate carrier proteins. PLoS One 2013;8:Epub ahead of print.

Costello 2003

Costello MF, Eden JA. A systematic review of the reproductive system effects of metformin in patients with polycystic ovary syndrome. Fertility and Sterility 2003;79:1‐13.

Costello 2007

Costello M, Shrestha B, Eden J, Sjoblom P, Johnson N. Insulin‐sensitising drugs versus the combined oral contraceptive pill for hirsutism, acne and risk of diabetes, cardiovascular disease, and endometrial cancer in polycystic ovary syndrome. Cochrane Database of Systematic Reviews 2007, Issue 1. [DOI: 10.1002/14651858.CD005552.pub2; PUBMED: 17253562]

De Frene 2014

De Frène V, Vansteelandt S, T'Sjoen G, Gerris J, Somers S, Vercruysse L, et al. A retrospective study of the pregnancy, delivery and neonatal outcome in overweight versus normal weight women with polycystic ovary syndrome. Human Reproduction 2014;29:2333‐8.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG (editors). Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Diamanti‐Kandarakis 2010

Diamanti‐Kandarakis E, Christakou CD, Kandarakis E, Economou F. Metformin: an old medication of new fashion: evolving new molecular mechanisms and clinical implications in polycystic ovary syndrome. European Journal of Endocrinology 2010;162:193‐212.

Ehrmann 2006

Ehrmann DA, Liljenquist DR, Kasza K, Azziz R, Legro RS, Ghazzi MN. Prevalence and predictors of the metabolic syndrome in women with polycystic ovary syndrome. Journal of Clinical Endocrinology and Metabolism 2006;91:48‐53.

EMA 2011

European Medicines Agency. European Medicines Agency clarifies opinion on pioglitazone and the risk of bladder cancer. www.ema.europa.eu/ema/index.jsp?curl=pages/news_and_events/news/2011/10/news_detail_001368.jsp&mid=WC0b01ac058004d5c1&jsenabled=trueAccessed 2011.

ESHRE/ASRM 2004

The Rotterdam ESHRE/ASRM‐sponsored PCOS consensus workshop group. Revised 2003 consensus on diagnostic criteria and long‐term health risks related to polycystic ovary syndrome (PCOS). Human Reproduction 2004;19(1):41.

ESHRE/ASRM 2008

The Thessaloniki ESHRE/ASRM‐sponsored PCOS consensus workshop group. Consensus on infertility treatment related to polycystic ovary syndrome. Human Reproduction 2008;23:462‐77.

Fauser 2012

Fauser BC, Tarlatzis BC, Rebar RW, Legro RS, Balen AH, Lobo R, et al. Consensus on women's health aspects of polycystic ovary syndrome (PCOS): the Amsterdam ESHRE/ASRM‐sponsored 3rd PCOS Consensus Workshop Group. Fertility and Sterility 2012;97:28‐38. [PUBMED: 22153789]

FDA 2002

FDA (Federal Drug Administration). www.fda.gov Accessed 14 January 2003.

Ford 2004

Ford ES. The metabolic syndrome and mortality from cardiovascular disease and all‐causes: findings from the National Health and Nutrition Examination Survey II Mortality Study. Atherosclerosis 2004;173:309‐14.

Franik 2014

Franik S, Kremer JAN, Nelen W, Farquhar C. Aromatase inhibitors for subfertile women with polycystic ovary syndrome. Cochrane Database of Systematic Reviews 2014, Issue 2. [DOI: 10.1002/14651858.CD010287.pub2]

Gillies 2007

Gillies CL, Abrams KR, Lambert PC, Cooper NJ, Sutton AJ, Hsu RT, et al. Pharmacological and lifestyle interventions to prevent or delay type 2 diabetes in people with impaired glucose tolerance: systematic review and meta‐analysis. BMJ 2007;334:299.

GRADEpro GDT 2015 [Computer program]

McMaster University (developed by Evidence Prime). GRADEpro GDT. Hamilton (ON): McMaster University (developed by Evidence Prime), Accessed 2015‐2017.

Graham 2003

Graham DJ, Green L, Senior JR, Nourjah P. Troglitazone‐induced liver failure: a case study. The American Journal of Medicine 2003;114:299‐306.

Hahn 2007

Hahn S, Tan S, Sack S, Kimmig R, Quadbeck B, Mann K, et al. Prevalence of the metabolic syndrome in German women with polycystic ovary syndrome. Experimental and Clinical Endocrinology & Diabetes 2007;115:130‐5.

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327:557‐60.

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Huber‐Buchholz 1999

Huber‐Buchholz MM, Carey DG, Norman RJ. Restoration of reproductive potential by lifestyle modification in obese polycystic ovary syndrome: role of insulin sensitivity and luteinizing hormone. Journal of Clinical Endocrinology and Metabolism 1999;84:1470‐4.

Jensterle 2014

Jensterle M, Kocjan T, Pfeifer M, Janez A. Short‐term intervention with liraglutide improved eating behaviour in obese women with polycystic ovary syndrome. Endocrine Research 2014;40:133‐8.

Kiddy 1992

Kiddy DS, Hamilton‐Fairley D, Bush A, Short F, Anyaoku V, Reed MJ, et al. Improvement in endocrine and ovarian function during dietary treatment of obese women with polycystic ovary syndrome. Clinical Endocrinology (Oxford) 1992;36:105‐11.

Knowler 2002

Knowler WC, Barrett‐Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. New England Journal of Medicine 2002;346:393‐403.

Lago 2007

Lago RM, Singh PP, Nesto RW. Congestive heart failure and cardiovascular death in patients with prediabetes and type 2 diabetes given thiazolidinediones: a meta‐analysis of randomised clinical trials. Lancet 2007;370(9593):1129‐36.

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Legro 2006a

Legro RS. Type 2 diabetes and polycystic ovary syndrome. Fertility and Sterility 2006;86 Suppl 1:16‐7.

Legro 2006b

Legro RS, Myers ER, Barnhart HX, Carson SA, Diamond MP, Carr BR, et al. and Reproductive Medicine Network. The Pregnancy in Polycystic Ovary Syndrome Study: baseline characteristics of the randomized cohort including racial effects. Fertility and Sterility 2006;86(4):914‐33.

Lord 2002

Lord J, Wilkin T. Polycystic ovary syndrome and fat distribution: the central issue?. Hum Fertil (Camb) 2002;5:67‐71.

March 2010

March WA, Moore VM, Willson KJ, Phillips DI, Norman RJ, Davies MJ. The prevalence of polycystic ovary syndrome in a community sample assessed under contrasting diagnostic criteria. Human Reproduction 2010;25:544‐51.

Misso 2013

Misso ML, Costello MF, Garrubba M, Wong J, Hart R, Rombauts L, et al. Metformin versus clomiphene citrate for infertility in non‐obese women with polycystic ovary syndrome: a systematic review and meta‐analysis. Human Reproduction Update 2013;19:2‐11.

Moran 2011

Moran LJ, Hutchison SK, Norman RJ, Teede HJ. Lifestyle changes in women with polycystic ovary syndrome. Cochrane Database of Systematic Reviews 2011, Issue 7. [DOI: 10.1002/14651858.CD007506.pub3]

Moran 2012

Moran LJ, Deeks AA, Gibson‐Helm ME, Teede HJ. Psychological parameters in the reproductive phenotypes of polycystic ovary syndrome. Human Reproduction 2012;27:2082‐8.

Palomba 2009

Palomba S, Pasquali R, Orio Jr F, Nestler JE. Clomiphene citrate, metformin or both as first‐step approach in treating anovulatory infertility in patients with polycystic ovary syndrome (PCOS): a systematic review of head‐to‐head randomized controlled studies and meta‐analysis. Clinical Endocrinology 2009;70:311‐21.

Pasquali 2003

Pasquali R, Pelusi C, Genghini S, Cacciari M, Gambineri A. Obesity and reproductive disorders in women. Human Reproduction Update 2003;9:359‐72.

Pernicova 2014

Pernicova I, Korbonits M. Metformin‐‐mode of action and clinical implications for diabetes and cancer. Nature Reviews. Endocrinology 2014;10:143‐56. [DOI: 10.1038/nrendo.2013.256]

Rautio 2006a

Rautio K, Tapanainen JS, Ruokonen A, Morin‐Papunen LC. Endocrine and metabolic effects of rosiglitazone in overweight women with PCOS: a randomized placebo‐controlled study. Human Reproduction 2006;21:1400‐7.

Rautio 2006b

Rautio K, Tapanainen JS, Ruokonen A, Morin‐Papunen LC. Rosiglitazone treatment alleviates inflammation and improves liver function in overweight women with polycystic ovary syndrome: a randomized placebo‐controlled study. Fertility and Sterility 2007;87(1):202‐6.

RevMan 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Robinson 1993

Robinson S, Kiddy D, Gelding SV, Willis D, Niththyananthan R, Bush A, et al. The relationship of insulin insensitivity to menstrual pattern in women with hyperandrogenism and polycystic ovaries. Clinical Endocrinology (Oxford) 1993;39:351‐5.

Schünemann 2011

Schünemann HJ, Oxman AD, Higgins JPT, Vist GE, Glasziou P, Guyatt GH. Chapter 11: Presenting results and ‘Summary of findings' tables. In: Higgins JPT, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Schünemann 2013

Schünemann H, Brożek J, Guyatt G, Oxman A, editors. GRADE handbook for grading quality of evidence and strength of recommendations. Updated October 2013. The GRADE Working Group, 2013. Available from guidelinedevelopment.org/handbook.

Siebert 2012

Siebert, TI, Viola MI, Steyn DW, Kruger TF. Is metformin indicated as primary ovulation induction agent in women with PCOS? A systematic review and meta‐analysis. Gynecologic and Obstetric Investigation 2012;73:304‐13.

Soares 2007

Soares EM, Azevedo GD, Gadelha RG, Lemos TM, Maranhao TM. Prevalence of the metabolic syndrome and its components in Brazilian women with polycystic ovary syndrome. Fertility and Sterility 2007;88(5):1389‐95.

Stein 1935

Stein. Amenorrhea associated with bilateral polycystic ovaries. American Journal of Obstetrics and Gynecology 1935;29:181‐91.

Stepto 2013

Stepto NK, Cassar S, Joham AE, Hutchison SK, Harrison CL, Goldstein RF, et al. Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycaemic‐hyperinsulaemic clamp. Human Reproduction 2013;28:544‐51.

Sterne 2011

Sterne JAC, Egger M, Moher D (editors). Chapter 10: Addressing reporting biases. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Intervention. Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Tso 2014

Tso L, Costello MF, Albuquerque LE, Andriolo RB, Macedo CR. Metformin treatment before and during IVF or ICSI in women with polycystic ovary syndrome. Cochrane Database of Systematic Reviews 2014, Issue 11. [DOI: 10.1002/14651858.CD006105.pub3]

Uusitupa 2000

Uusitupa M, Louheranta A, Lindstrom J, Valle T, Sundvall J, Eriksson J, et al. The Finnish Diabetes Prevention Study. British Journal of Nutrition 2000;83 Suppl 1:137‐42.

Velazquez 1997

Velazquez E, Acosta A, Mendoza SG. Menstrual cyclicity after metformin therapy in polycystic ovary syndrome. Obstetrics and Gynecology 1997;90:392‐5.

Weerakiet 2007

Weerakiet S, Bunnag P, Phakdeekitcharoen B, Wansumrith S, Chanprasertyothin S, Jultanmas R, et al. Prevalence of the metabolic syndrome in Asian women with polycystic ovary syndrome: using the International Diabetes Federation criteria. Gynecological Endocrinology 2007;23:153‐60.

Wijeyaratne 2002

Wijeyaratne CN, Balen AH, Barth JH, Belchetz PE. Clinical manifestations and insulin resistance (IR) in polycystic ovary syndrome (PCOS) among South Asians and Caucasians: is there a difference?. Clinical Endocrinology (Oxford) 2002;57:343‐50.

Wijeyaratne 2004

Wijeyaratne CN, Nirantharakumar K, Balen AH, Barth JH, Sheriff R, Belchetz PE. Plasma homocysteine in polycystic ovary syndrome: does it correlate with insulin resistance and ethnicity?. Clinical Endocrinology (Oxford) 2004;60:560‐7.

Yki‐Jarvinen 2004

Yki‐Jarvinen H. Thiazolidinediones. New England Journal of Medicine 2004;351:1106‐18.

Zhang 2012

Zhang Y, Ye J. Mitochondrial inhibitor as a new class of insulin sensitizer. Acta Pharmacologica Sinica 2012;2:341‐9.

References to other published versions of this review

Lord 2003

Lord JM, Flight IH, Norman RJ. Insulin‐sensitising drugs (metformin, troglitazone, rosiglitazone, pioglitazone, D‐chiro‐inositol) for polycystic ovary syndrome. Cochrane Database of Systematic Reviews 2003, Issue 3. [DOI: 10.1002/14651858.CD003053]

Tang 2009

Tang T, Lord JM, Norman RJ, Yasmin E, Balen AH. Insulin‐sensitising drugs (metformin, rosiglitazone, pioglitazone, D‐chiro‐inositol) for women with polycystic ovary syndrome, oligo amenorrhoea and subfertility. Cochrane Database of Systematic Reviews 2009, Issue 3. [DOI: 10.1002/14651858.CD003053.pub4; PUBMED: 20091537]

Tang 2012

Tang T, Lord JM, Norman RJ, Yasmin E, Balen AH. Insulin‐sensitising drugs (metformin, rosiglitazone, pioglitazone, D‐chiro‐inositol) for women with polycystic ovary syndrome, oligo amenorrhoea and subfertility. Cochrane Database of Systematic Reviews 2012, Issue 5. [DOI: 10.1002/14651858.CD003053.pub5]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Ayaz 2013

Methods

RCT

Setting: Saudi Arabia

Method of randomisation: unclear

Blinding: double

Number randomised: 42

Participants

Summary: metformin and CC versus CC alone

Inclusion criteria: PCOS (Rotterdam criteria)

Exclusion criteria: other endocrine disorders, male factor infertility, recent PID, tubal infertility

Baseline characteristics of each group: metformin and CC versus CC alone
Mean age (SD) 32 (3.5), 31.3 (2.9)
BMI > 25 14 (56.7)), 15 (71.4)
Mean thyroid stimulating hormone mIU/L (SD) 4.6 (1.3), 3.9 (1.7)

Free thyroxin nmol/L (SD) 4.81 (1.6), 5.2 (1.8)
Mean total testosterone: mmol/L (SD) 2.60 (0.78), 2.74 (0.65)

Sex hormone‐binding globulin: nmol/L (SD) 21.7 (3.7), 18.9 (4.3)

Dropouts: none

Interventions

Main intervention: metformin 500 mg 3/d

Duration: 6 months until 8 weeks of a confirmed pregnancy

Co‐interventions: CC 50 mg from day 2 until day 6 of cycle

Outcomes

Ovulation: follicle tracking on transvaginal US

Others: menstrual pattern, pregnancy rate, multiple pregnancy rate

Notes

Endocrine and metabolic outcomes not recorded

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Low risk

Sealed envelopes used

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No women were lost to follow‐up

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Unclear

Baillargeon 2004

Methods

RCT

Setting: Venezuela

Method of randomisation: fixed block of 8 randomisation which was performed by the investigational pharmacist.

Blinding: double

Number randomised: 128

Participants

Summary: non‐obese PCOS

Inclusion criteria: PCOS (oligomenorrhoea < 8 periods/year, hyperandrogenism total testosterone > 2.43 nmol/L
Normal prolactin and TFT, fasting insulin < 15 μIU/mL and fasting glucose to insulin ration > 4.5
Normal OGTT
Hormonal contraceptives were not used before the trial.

Exclusion criteria: late onset adrenal hyperplasia, hypertension. Previous insulin‐sensitiser users

Baseline characteristics of each group:

  • mean age (SD) metformin 27.7 (4.7), rosiglitazone 27.9 (5.16), placebo 27.2 (4.9)

  • mean BMI (SD) metformin 24.6 (1.1), rosiglitazone 24.3(1.4), placebo 24.6 (1.9)

  • mean fasting insulin mIU/L (SD) metformin 6.3 (5.8), rosiglitazone 11.2(5.6), placebo 7.9 (2.0)

  • mean total testosterone mol/L (SD) metformin 3.8 (2.0), rosiglitazone 3.5 (1.9), placebo 4.67 (2.0

Dropouts: 4 (12.5%) in the metformin arm, 10 (31.3%) in the rosiglitazone group and 2 (6.3%) in the placebo group

Interventions

Main intervention: metformin 850 mg, rosiglitazone 4 mg or placebo tablets twice daily

Duration: 6 months

Co‐interventions: none

Outcomes

Ovulation: weekly progesterone measurement with a level > 4 ng/mL was considered to be ovulation

Anthropometric: weight, BMI, WHR, BP

Hormones: testosterone, SHBG, free testosterone, DHEAS

Metabolic markers: fasting glucose, AUC glucose and fasting glucose:insulin ratio

Others: menstrual pattern

Notes

This study randomised 128 women into 4 groups (metformin alone, rosiglitazone alone, combined metformin and rosiglitazone, placebo alone). We included the combined group in our analysis. We analysed the metformin and rosiglitazone groups separately and compared the results from these 2 groups with the same group of women who took placebo.
Women were predominantly white European emigrants to Venezulea
Delays in the delivery of the drug rosiglitazone to the research centre resulted in higher dropout rates in this group after randomisation.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Fixed block of 8 randomisation which was performed by the investigational pharmacist

Allocation concealment (selection bias)

Low risk

Trial drugs packed in coded boxes allocated by the research nurse. Trial drugs were similar in appearance

Blinding (performance bias and detection bias)
All outcomes

Low risk

"Double blinded"

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 4 (12.5%) in the metformin arm, 10 (31.3%) in the rosiglitazone group and 2 (6.3%) in the placebo group. Details not provided

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Unclear

Begum 2014

Methods

RCT

Setting: Bangladesh (Infertility Department of women and children's hospital)

Method of randomisation: envelopes used, but no other information

Blinding: unclear

Number randomised: 71

Participants

Summary: PCOS meeting the Rotterdam criteria for diagnosis

Inclusion criteria: subfertile women between 20‐35 years with a diagnosis of PCOS according to Rotterdam criteria

Exclusion criteria: Age > 35 years, hypo‐ or hyperthyroidism, hyperprolactinaemia, diabetes mellitus and male factor infertility

Baseline characteristics of each group:

  • no significant difference in age (years), BMI, WHR, duration of infertility (months)

  • no significant difference is FSH, LH, TSH, prolactin, glucose tolerance

Dropouts: none stated

Interventions

Main intervention: Group 1: metformin 1500 mg/d. Group 2: CC 100 mg/d for 5 d

Duration: 6 months

Co‐interventions: none

Outcomes

Ovulation rate

Pregnancy rate

Notes

We have contacted study authors for further information regarding methodology

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of generating random sequence for distribution in envelopes is not stated.

Allocation concealment (selection bias)

High risk

Allocation to each group revealed in envelopes but not stated if opaque and sealed. Due to high risk of allocation concealment bias, Begum 2014 is excluded from subgroup analysis by study quality.

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

No information given

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

None stated

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

None stated

Ben Ayed 2009

Methods

RCT

Setting: Tunisia

Method of randomisation: not stated

Blinding: not stated

Number randomised: 32

Participants

Summary: non‐obese PCOS

Inclusion criteria: Rotterdam criteria

Exclusion criteria: late onset adrenal hyperplasia, Cushing's Syndrome, abnormal TFT, hyperprolactinaemia, androgen‐secreting tumour

Baseline characteristics of each group:

  • Mean age 32.81, 29.38

  • Mean BMI; 28, 28

Interventions

Main intervention: metformin 1700 mg/d or placebo

Duration: unclear

Co‐interventions: CC 100 mg from day 3 to day 7 of the cycle. Lifestyle advice on the obese subjects

Outcomes

Ovulation: USS follicular tracking with follicular size > 16 mm

Notes

Inadequate information in the protocol to assess the quality of the trial

No reply from study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Inadequate information

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Inadequate information

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Boudhraa 2010

Methods

RCT 

Setting: Tunisia 

Method of randomisation: not stated* 

Blinding: unblinded

Number randomised: 63

Participants

Summary: PCOS non‐obese 

Inclusion criteria: unclear. ? diagnostic criteria of PCOS used

Exclusion criteria: male factor infertility, tubal disease

Baseline characteristics of each group:

  • mean age 30.55, 30.72

  • mean BMI 29.9, 29.77

Dropouts: none

Interventions

Main intervention: metformin 850 mg

Duration: not stated

Co‐interventions: recommendations on healthy diet. 5 d 100 mg CC treatment

Outcomes

Ovulation: method to confirm ovulation not stated

Live birth

Notes

Study protocol is too brief. Inadequate information to assess the quality of the study. No reply from study author*

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Inadequate information

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Inadequate information

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Brettenthaler 2004

Methods

RCT 

Setting: Switzertland 

Method of randomisation: unclear* 

Blinding: double 

Number randomised: 40

Participants

Summary: PCOS non‐obese 

Inclusion criteria: menstrual dysfunction (oligo‐ or amenorrhoea), hirsutism with Ferriman‐Gallwey score > 7 or serum total testosterone > 2.5 nmol/L and SHBG < 50 nmol/L

Exclusion criteria: adrenal disease, thyroid dysfunction, diabetes, hyperprolactinaemia

Pregnancy or desire for pregnancy, basal FSH > 20 IU/L

Medication known to affect reproductive or metabolic functions

Previous hysterectomy

History of liver disease or alcohol abuse

Abnormal liver function tests.

Baseline characteristics of each group:

  • mean age (SD) 30.2 (5.7), 30.6 (5.1)

  • mean BMI (SD) 29.4 (7), 27.5 (5.1)

  • mean fasting  insulin mIU/L (SD)

  • mean total testosterone nmol/L (SD) 

Dropouts: 3 in the treatment group and 2 in the placebo group. The details were not given (lost in follow‐up and protocol violation)

 

Interventions

Main intervention: pioglitazone 30 mg or placebo tablet once daily

Duration: 3 months 

Co‐interventions: recommendations on healthy diet and physical activity for weight maintenance 4 weeks prior to the study

Outcomes

Ovulation: progesterone > 9 nmol/L 

Anthropometric: BMI, WHR 

Hormones: testosterone, SHBG, DHEAS 

Metabolic markers: insulin, glucose, AUC insulin, AUC glucose, cholesterol, triglyceride

Others: hirsutism

Notes

Participants in this study were very heterogeneous (65% European, 30% Turkish and 5% Asian)

No serious side effects or abnormal liver function tests were reported. Nevertheless, women who took pioglitazone experienced more side effects compared with those who took placebo; mild peripheral oedema (18% vs 0%), mastopathy (11.7% vs 5%), sleeping disorders (23% vs 5%), headache (23% vs 5%) and stomach arch (23% vs %%)

 *No reply from the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Inadequate information

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Identical trial and placebo tablets. Inadequate information to assess the methodology

Incomplete outcome data (attrition bias)
All outcomes

High risk

Missing data not reported

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Participants in this study were very heterogeneous (65% European, 30% Turkish and 5% Asian). Inadequate information to assess. No reply from study author

Carmina 2004

Methods

RCT 

Setting: USA

Method of randomisation: random table 

Blinding: not stated

Number randomised: 24

Participants

Summary: non‐obese PCOS

Inclusion criteria: chronic anovulation with serum progesterone < 2 ng/mL on day 22 of cycle, in 2 consecutive cycles

Normal TFT

No clinical and biochemical features of hyperandrogenism

Exclusion criteria:

Baseline characteristics of each group:

  • mean age 24.6, 24.2

  • mean BMI 25.2, 25.8

  • mean fasting  insulin mIU/L 11.8, 12.0

  • mean total testosterone nmol/L 1.3, 1.4 

Dropouts: none

Interventions

Main intervention: metformin 500 mg 3/d, placebo

Duration: 3 months 

Co‐interventions:

Outcomes

Ovulation: method to confirm ovulation not stated

Notes

This study evaluated the efficacy of metformin in women with anovulation who do not have evidence of hyperandrogenism; although > 79% of included women had USS evidence of PCO; hence, met the Rotterdam diagnostic criteria of PCOS.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Inadequate information

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Limited information to assess

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Only 79% of the subjects were PCOS

Chou 2003

Methods

RCT

Setting: Brazil

Method of randomisation: participants were randomised by a 3rd party by using a table with random numbers (odd number assigned for the metformin group, even number assigned for the placebo group)*

Blinding: double

Number randomised: 32

Participants

Summary: obese PCOS

Inclusion criteria: oligomenorrhoea (< 6 menstrual cycles), clinical or biochemical hyperandrogenism
BMI > 30
Non‐smoker. Participants had not used any medication 3 months before the start of the trial
Exclusion criteria: renal or liver disease. CAH (serum 17‐hydroxyprogesterone > 12 ng/dL 1 h after 0.25 mg ACTH intramuscular injection)

Baseline characteristics of each group:

  • mean age (SD) 24 (5), 24.5 (6.1)

  • mean BMI (SD) 35.6 (4.9), 37.4 (6)

  • mean fasting insulin mIU/L (SD) 44.3 (21.6), 46.8 (41.4)

  • mean total testosterone mmol/L (SD) 2.02 (0.70), 2.41 (1.1)

Dropouts: 1 in each arm (protocol violation)

Interventions

Main intervention: metformin 500 mg or placebo tablet 3/d

Duration: 3 months

Co‐interventions:

Outcomes

Anthropometric: BMI, WHR, BP*

Hormones: testosterone, SHBG*

Metabolic markers: insulin, glucose, cholesterol, LDL, HDL and triglyceride*

Others: menstrual pattern

Notes

This study was designed to evaluate the benefit of using metformin in obese women (BMI > 30) with PCOS. 3 participants in each arm were found to have glucose intolerance according to WHO criteria

The results of the women who dropped out from the study were excluded from the analysis.

*Information kindly provided by the study author that was not in the original paper

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised by a 3rd party by using a table with random numbers. Odd number assigned for the metformin group, even number assigned for the placebo group

Allocation concealment (selection bias)

Low risk

Trial drugs were similar in appearance. Randomisation carried out by a 3rd party who kept the code until the end of the study.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

The results of the women who dropped out from the study were excluded from the analysis. Details of the excluded women were not given

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Eisenhardt 2006

Methods

RCT

Setting: Germany

Method of randomisation: computer‐generated random numbers with randomisation in block of 6. The code was sealed by a 3rd party until the end of the study period.

Blinding: double

Number randomised: 45

Participants

Summary: obese PCOS

Inclusion criteria: oligomenorrhoea (cycle length > 35days or < 9 periods/year) or amenorrhoea (cycle length > 12 weeks), PCO on USS (Rotterdam consensus 2003), clinical or biochemical hyperandrogenism (testosterone > 2.1 nmol/L or androstenedione > 10.1 nmol/L)
Age between 21‐36 years

Exclusion criteria: hyperprolactinaemia, diabetes, thyroid disease, CAH, Cushings's syndrome
Medications that influence hormonal profiles or anti‐obesity drugs ≤ 6 months before the start of the study

Baseline characteristics of each group:*

  • median age 27, 29.7

  • median BMI 28.9, 32.4

  • median fasting insulin (mIU/L) 20.0, 22.0

  • median testosterone (mmol/L) 1.59, 1.66

Dropouts: 1 in the metformin arm, 3 in the placebo arm. Details were not given. Furthermore, 1 in the metformin group and 2 in the placebo became pregnant and were also excluded from the analysis.

Interventions

Main intervention: metformin 500 mg or placebo tablet 3/d

Duration: 12 weeks

Co‐interventions: none

Outcomes

Anthropometric: BMI, weight*

Hormones: testosterone, androstenedione, SHBG, oestradiol, DHEAS, LH, FSH*

Metabolic markers: glucose, insulin, AUC glucose, AUC insulin*

Others: hirsutism, menstrual pattern*

Notes

The objective of this study was to evaluate the effects of metformin in women with PCOS according to the status of insulin resistance. Insulin resistance was defined as fasting glucose to insulin ratio < 4.5. 32 out of 45 women (71.1%) were classified as insulin‐resistant PCOS.

Insulin‐resistant PCOS women responded better than non insulin‐resistant PCOS women in terms of improvement in menstrual cyclicity.

The results were presented in median and range. Hence, we could not include these data in the meta‐analysis. We are currently still waiting for a reply from the study author for the converted results in a format of mean and standard deviation.

*still awaiting for a reply from the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers with randomisation in block of 6

Allocation concealment (selection bias)

Low risk

The code was sealed by a 3rd party until the end of the study period. Trial drugs were provided by a pharmaceutical company not involved in study design and data analysis

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropouts: 1 in the metformin arm, 2 in the placebo arm. Details not given

Selective reporting (reporting bias)

Low risk

All primary outcome measures (menstrual frequency and metabolic parameters) reported

Other bias

Unclear risk

Inadequate information

El‐Biely 2001

Methods

RCT

Setting: Egypt

Method of randomisation: computer‐based, blocked (block size not stated)

Blinding: not stated; presumed to be unblinded

Number randomised: 90

Participants

Summary: PCOS, obese

Inclusion criteria: PCOS (oligomenorrhoea, US findings of ≥ 10 ovarian cysts measuring 2‐8 mm around a dense stroma), hyperinsulinaemia (fasting insulin > 30 mIU/L)
BMI > 28 kg/m2
WHR > 0.85
Normal semen analysis
No tubal disease

Exclusion criteria: diabetes mellitus, thyroid dysfunction, raised prolactin

Baseline characteristics of each group:

  • mean age (± SD) 26.4 (4.5), 25.7 (4.3)

  • mean BMI (± SD) 28.7 (5.9), 27.4 (3.6)

  • mean fasting insulin mIU/L (± SD) 39.3 (8.1), 39.2 (8.5)

  • mean total testosterone mmol/L (± SD) 3.1 (2.1), 3.0 (1.5)

Dropouts: only as a result of pregnancy (13 from metformin group, 4 from no metformin)

Interventions

Main intervention: 1 of: metformin 500 mg 3/d, no treatment

Duration: 6 months

Co‐interventions: CC 50 mg on days 5‐9, increased each cycle if not ovulated by 50 mg up to a maximum of 150 mg
hCG 10,000 IU given to trigger ovulation

Outcomes

Ovulation: by serum progesterone (> 15.9 nmol/L) 9 d after hCG

Metabolic markers: fasting insulin

Others: pregnancy
Number of mature follicles
Diameter of largest follicle
Premature ovulation rate
Ovarian hyperstimulation syndrome (not defined)

Notes

The inclusion criteria did not include previous response to CC. Overall, 65% of those receiving CC and placebo ovulated (compared to 85% of those receiving CC and metformin).

This trial reported a significantly higher mean number of mature follicles in the metformin group (3.1 versus 1.9, P < 0.0001), but a significantly lower rate of ovarian hyperstimulation syndrome (4 versus 31, P < 0.001).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer randomised

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Only as a result of pregnancy (13 from metformin group, 4 from no metformin)

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

The inclusion criteria did not include previous response to CC. Overall, 65% of those receiving CC and placebo ovulated (compared to 85% of those receiving CC and metformin).

Fleming 2002

Methods

RCT

Setting: UK

Method of randomisation: computer‐generated randomisation by pharmacy in blocks of 4

Blinding: double‐blind

Number randomised: 94

Participants

Summary: obese PCOS

Inclusion criteria: PCOS (oligomenorrhoea < 8 cycles/year, exclusion of other endocrinopathy, US finding of PCO)
Age < 35 years

Exclusion criteria: diabetes mellitus, adrenal hyperplasia, thyroid dysfunction, hyperprolactinaemia, medication likely to influence hormonal profiles

Baseline characteristics of each group:

  • mean age (+/‐ SD) 28.6 (5.8), 29.2 (5.6)

  • mean BMI (± SD) 34.2 (8.6), 35.0 (8.2)

  • mean fasting insulin mIU/L (± SD) 16.7 (12.7), 18.4 (13.6)

  • mean total testosterone mol/L (± SD) 3.0 (1.5), 3.8 (1.6)

Dropouts: 30 (32%), with 22 in the treatment arm and 8 in the placebo, mainly due to gastrointestinal side effects in metformin group. Overall, 58% of the metformin arm completing the trial and 83% of the placebo arm. Included in ITT analysis

Interventions

Main intervention: 1 of metformin 850 mg 2/d, placebo

Duration: 12‐16 weeks

Co‐interventions: 1st week of treatment at 850 mg 1/d

Outcomes

Ovulation: by twice‐weekly serum oestradiol. Where oestradiol > 300 pmol/L, LH and progesterone (> 8 nmol/L in ≥ 2 successive samples defined ovulation*) were determined

Anthropometric: BMI, WHR

Reproductive hormones: total testosterone, free testosterone, androstenedione, estradiol, SHBG, FSH, LH

Metabolic markers: fasting glucose, fasting insulin, AUC insulin during GTT, leptin, inhibin‐B, cholesterol (HDL, LDL, VLDL), triglycerides

Others: ovarian US, pregnancy, adverse effects

Notes

Diagnostic criteria different to other trials ‐ using US not hyperandrogaenemia (although 90% did have raised androgens, and mean entry‐FAI 10 with 5% CI 8.6). Subgroup analysis showed that those who ovulated in response to metformin had significantly lower androgens.

High rate of background ovulation (64% on placebo ovulated at some stage)

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated randomisation by pharmacy in blocks of 4

Allocation concealment (selection bias)

Low risk

Remote allocation. Identical metformin and placebo tablets. Randomisation code kept in the pharmacy department until the end of the trial

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 30 (32%), with 22 in the treatment arm and 8 in the placebo, mainly due to gastrointestinal side effects in metformin group. Overall, 58% of the metformin arm completed the trial and 83% of the placebo arm.

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Gerli 2003

Methods

RCT

Setting: Italy

Method of randomisation: computer‐generated random number table*

Blinding: double

Number randomised: 283

Participants

Summary: obese PCOS

Inclusion criteria: PCO on USS, oligomenorrhoea (cycle length > 40 d or < 8 cycles/year) or amenorrhoea, clinical and biochemical hyperandrogenism
Age < 35 years
Exclusion criteria: CAH, thyroid dysfunction, hyperprolactinaemia

Baseline characteristics of each group:

  • mean age (SD) 28.6 (10.1), 29.2 (9.3)

  • mean BMI (SD) 34.2 (14.8), 35 (15.4)

  • mean fasting insulin mIU/L (SD) 16.7 (22), 18.4 (24.2)

  • mean total testosterone mol/L (SD) 3.0 (2.3), 3.8 (2.4)

Dropouts: significantly more women withdrew in the treatment group (n = 15) compared with the placebo group (n = 5). Reasons not given

Interventions

Main intervention: inositol 100 mg or placebo tablet twice daily

Duration: 16 weeks

Co‐interventions: none

Outcomes

Ovulation: progesterone > 6 nmol/L

Anthropometric: BMI, WHR*

Hormones:

Metabolic markers: insulin, glucose, AUC insulin, leptin, VLDL, LDL, HDL, triglyceride*

Others: menstrual pattern, pregnancy*

Notes

This is the largest study published so far on the effects of inositol on ovarian function and metabolic factors in women with PCOS. Women were recruited from gynaecology, endocrine and infertility outpatient clinics in the study centre. Nearly half of the participants presented with history of infertility. However, only 42 women declared a wish to conceive before the start of the trial. Therefore, it would be difficult to interpret the pregnancy rate accurately. 

 

*No further information from the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random number table

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: significantly more women withdrew in the treatment group (n = 15) compared with the placebo group (n = 5). Reasons not given

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Glintborg 2005

Methods

RCT

Setting: Denmark

Method of randomisation: computer‐generated numbers. Randomisation was conducted in the local pharmacist. The code was kept in the pharmacist department until the end of the trial*

Blinding: double*

Number randomised: 30

Participants

Summary: obese PCOS

Inclusion criteria: oligomenorrhoea (cycle length > 35 days), free testosterone > 0.035 nmol/L or clinical evidence of hirsutism

Fasting insulin level > 50 pmol/L and/or BMI > 30
Participants stopped oral contraceptives for at least 3 months before the trial

Normal TFT and prolactin levels

Exclusion criteria: diabetes, hypertension, renal dysfunction, heart disease or abnormal liver function tests

Baseline characteristics of each group:

  • mean age (SD) 32 (2.4), 34 (2.25)

  • mean BMI (SD) 33.4 (3.3), 33.6 (5.9)

  • mean fasting insulin mIU/L (SD) 15.8 (10.8), 11.5 (4.25)

  • mean total testosterone mmol/L (SD) 1.93 (0.99), 1.74 (0.75)

Dropouts: 2 in total. 1 in the placebo group due to pregnancy. Another subject in the treatment group experienced side effects from pioglitazone (ankle oedema, anxiety, dizziness). No serious side effects were reported in this study and all women had normal liver function tests at the end of the trial.

Interventions

Main intervention: pioglitazone 30 mg or placebo once daily

Duration: 16 weeks

Co‐interventions:

Outcomes

Anthropometric: BMI, WHR, waist circumference

Hormones: testosterone, SHBG, free testosterone

Metabolic markers: fasting insulin

Others: menstrual pattern, hirsutism

Notes

The main objective of this study was to investigate the effect of pioglitazone on growth hormone levels in women with PCOS. The secondary endpoint measures included changes in anthropometric and hormonal parameters.

The participants were recruited from the local endocrine and infertility clinics. All the women were instructed to use barrier contraception combined with spermatocidal cream provided by the department throughout the trial period due to the potential risks in pregnancy.

No serious side effects were reported. All participants had normal liver functions at the end of the trial period.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated numbers. Randomisation was conducted by the local pharmacist

Allocation concealment (selection bias)

Low risk

The code was kept in the pharmacist department until the end of the trial

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 2 in total, 1 in each group

Selective reporting (reporting bias)

Low risk

All primary measures reported

Other bias

Low risk

Primary objective investigated the effect of pioglitazone on growth hormone levels in women with PCOS. All women were instructed to use contraception

Hoeger 2004

Methods

RCT

Setting: USA

Method of randomisation: computer‐generated random number, randomisation conducted by the pharmacy department*

Blinding: double

Number randomised: 38

Participants

Metformin vs placebo
Summary: PCOS, obese 

Inclusion criteria: PCOS (oligomenorrhoea with < 6 menses/year and evidence of hyperandrogenism), BMI > 25, normal TSH, prolactin and FSH concentrations

No hormonal treatment within 2 months before the trial commenced. 

Exclusion criteria: adrenal disease

Baseline characteristics of each group:

  • mean age (SD) 29.5 (6.4), 27.1 (4.5)

  • mean BMI (SD) 37.1 (4.9), 37.1 (4.6)

  • mean fasting  insulin mIU/L (SD) 21.6 (11.1), 21.08( 7.4)

  • mean total testosterone nmol/L (SD) 2.1 (0.8), 2.0 (0.60)

Dropouts: 3 (33.3%) in the metformin arm and 2 (22.2%)in the placebo arm at 24 months of the trial

Lifestyle advice + metformin vs lifestyle advice alone

Summary: PCOS, obese 

Inclusion criteria: PCOS (oligomenorrhoea with < 6 menses/year and evidence of hyperandrogenism), BMI > 25, normal TSH, prolactin and FSH concentrations

No hormonal treatment within 2 months before the trial commenced

Exclusion criteria: adrenal disease

Baseline characteristics of each group:

  • mean age (SD) 30.4 (5.4), 27.1 (4.3)

  • mean BMI (SD) 41.7 (6.2), 40 (7.4)

  • mean fasting  insulin mIU/L (SD) 24.6 (7.2), 20.5 (9.6)

  • mean total testosterone nmol/L (SD) 2.43 (0.59), 2.00 (0.66)

Dropouts: 4 (44.4%) in the metformin/lifestyle arm and 2 (18.2%)in the placebo/lifestyle arm at 24 months of the trial

Interventions

Main intervention: metformin 850 mg 2/d or placebo 

Duration: 24 months 

Co‐interventions: lifestyle modification programme to reduce calorie intake by 500‐1000 kcal/d. All women were provided with an individual, healthy, balanced meal plan. The lifestyle team consisted of a dietitian and exercise physiology. No lifestyle modification for the non‐obese group

Outcomes

Anthropometric: weight, BMI, hirsutism

Hormones: total testosterone, SHBG, FAI, AUC glucose, AUC insulin, fasting glucose, fasting insulin*

Metabolic markers:

Others: menstrual pattern*

Notes

This trial was designed to investigate the combined effects of metformin and intensive lifestyle modification in overweight women with PCOS. The women were recruited through a direct advertisement, referral from physician and reproductive endocrinology outpatient clinic in the same study centre. The women were randomised into 4 groups (metformin alone, placebo alone, combined lifestyle changes and metformin, and lifestyle changes alone). We decided to separate the analysis into 2 groups; metformin versus placebo and combined lifestyle and metformin versus lifestyle 

We also decided to analyse the results for those who completed the trial at 24 weeks as there were too many dropouts at the end of the trial period at 48 weeks.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random number

Allocation concealment (selection bias)

Low risk

Randomisation conducted by the pharmacy department

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double. Drug and placebo packaged and labelled according to participant number by the pharmacy

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 3 (33.3%) in the metformin arm and 2 (22.2%)in the placebo arm at 24 months of the trial. Further 4 (44.4%) in the metformin/lifestyle arm and 2 (18.2%) in the placebo/lifestyle arm at 24 months of the trial. Baseline characteristics between the subjects completed and the drop outs were similar

Selective reporting (reporting bias)

Low risk

Study protocol available. Pre‐specified outcome measures (ovulation and testosterone levels) were reported

Other bias

Low risk

Hwu 2005

Methods

RCT

Setting: Taiwan

Method of randomisation: computer‐generated random numbers, block of 2 randomisation process*

Blinding: no*

Number randomised: 80

Participants

Summary: CC‐resistant PCOS
Inclusion criteria: oligomenorrhoea (< 6 menses/year), clinical or biochemical hyperandrogenism (total testosterone > 2.42 nmol/L), PCO on USS (≥ 12 follicles 2‐9 mm in diameter per ovary)

CC resistance was defined as no follicular development after 2 cycles up to 150 mg CC treatment for 5 d

Exclusion criteria: not mentioned

Baseline characteristics of each group:

  • mean age (SD) 29.07 (4.45), 27.8 (3.75)

  • mean BMI (SD) 25.27 (3.3), 24.11 (3.58)

  • mean fasting insulin mIU/L (SD)

  • mean total testosterone mmol/L (SD) 2.5 (0.1), 2.4 (0.3)

Dropouts: none

Interventions

Main intervention: metformin 500 mg 3/d versus no treatment. Metformin was commenced on day 1 after induced menstruation followed by a 5‐d course of CC 150 mg treatment from day 13 of the cycle. When there was evidence of follicles > 12 mm in diameter 3 days after the last dose of CC, metformin was continued until the dominant follicles reached 20 mm. Intramuscular hCG 5000 IU was then administrated and the participants were instructed to have intercourse in the following 2 days

Duration: 1 cycle

Co‐interventions: CC 150 mg, hCG 5000 IU (Pregnyl; Organon, Holland)

Outcomes

Ovulation: confirmed by USS and serum progesterone > 5 ng/mL on day 7 after hCG injection

Anthropometric:

Hormones:

Metabolic markers:

Others: pregnancy and miscarriage rates

Notes

This study was to evaluate the effect of a short course of metformin as a co‐therapy in ovulation induction with CC 150 mg in women with PCOS who developed CC resistance in the previous treatment cycles. Compared with the other included studies, CC treatment was commenced at day 13 of the menstrual cycle rather than at early follicular phase.

Intramuscular hCG (5000 IU) was used to trigger ovulation when a dominant follicle reached a diameter of 20 mm.

The sequence of allocation was not concealed and this study was unblinded. Therefore, bias cannot be excluded.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers, block of 2 randomisation process*

Allocation concealment (selection bias)

High risk

The sequence of allocation was not concealed and this study was unblinded.

Blinding (performance bias and detection bias)
All outcomes

High risk

Not blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

High risk

Using hCG injection triggering ovulation rather than natural ovulation

Jakubowicz 2001

Methods

RCT

Setting: Venezuela (63% white, 31% Hispanic, 4% Arabic, 2% South American Indian)

Method of randomisation: sequentially numbered, identical containers of identical drugs*

Blinding: double‐blind

Number randomised: 48

Participants

Summary: obese PCOS, CC‐sensitive

Inclusion criteria: PCOS (oligomenorrhoea ≤ 8 cycles/year, elevated free testosterone, exclusion of other endocrinopathy, ultrasonographic finding of PCO), ovulation with CC 150 mg (demonstrated by serum progesterone > 12.7 pmol/L and US)

Exclusion criteria: adrenal hyperplasia, thyroid dysfunction, hyperprolactinaemia, diabetes mellitus, failure to ovulate with CC as described above, medication that could affect insulin sensitivity*

Baseline characteristics of each group:

  • mean age (± SD) 27 (5.1), 27 (4.7)

  • mean BMI (± SD) 31.8 (1.5), 31.7 (1.4)

  • mean fasting insulin mIU/L (±‐ SD) 34.33 (23.0), 54.67 (40.7)

  • mean total testosterone mmol/L (± SD) 3.4 (1.8), 3.8 (2.7)

Dropouts: after randomisation, 8 (14%), 2 in metformin arm and 6 in placebo. Not included in analysis

Interventions

Main intervention: 1 of metformin 500 mg 3/d, placebo

Duration: 4‐5 weeks prior to CC, then for a further 19 d after commencing CC

Co‐interventions: CC 150 mg for 5 d

Outcomes

Ovulation: by serum progesterone > 12.7 pmol/L and US. Ovulation checked on 2 occasions on day 23: once after metformin/placebo cycle and once after subsequent metformin/placebo with CC.

Anthropometric: BMI, WHR

Reproductive hormones: total testosterone, free testosterone, androstenedione, DHEAS, 17‐beta estradiol, SHBG

Metabolic markers: fasting glucose, fasting insulin, AUC insulin and glucose during GTT

Others: glycodelin, IGFBP‐1, endometrial thickness, endometrial vascular penetration, resistance index of uterine spiral arteries

Notes

Women that were given metformin and ovulated received an extra week's course of treatment when compared with the placebo group.

High dropout rate between recruitment and randomisation (24%) as only those who ovulated with CC prior to randomisation were included.

The primary outcome measures are not relevant to this review, but the other parameters reported are.

It is assumed that the units quoted for testosterone are mmol/dL and not mmol/L.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Low risk

Sequentially numbered, identical containers of identical drugs

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: after randomisation, 8 (14%), 2 in metformin arm and 6 in placebo. Not included in analysis. Missing data not reported. High dropout rate between recruitment and randomisation (24%) as only those who ovulated with CC prior to randomisation were included.

Selective reporting (reporting bias)

Unclear risk

The primary outcome measures are not relevant to this review, but the other parameters such as ovulation reported are.

Other bias

Low risk

Women that were given metformin and ovulated received an extra week's course of treatment when compared with the placebo group.

Kar 2015

Methods

RCT

Setting: India (private hospital)

Method of randomisation: envelopes prepared by a nurse "naive to this study"

Blinding: double‐blind

Number randomised: 105

Participants

Summary: Asian Indian women with "treatment naive" PCOS
Inclusion criteria: history of infertility and oligomenorrhoea, meeting the Rotterdam criteria for PCOS. Normal male factor, at least 1 patent tube by hysterosalpingography, treatment naive

Exclusion criteria: any major systemic illness

Baseline characteristics of each group: no significant difference in age (years), duration of infertility (years), BMI, Ferriman‐Galloway score, waist circumference, hip circumference. No significant difference in biochemical parameters, such as FSH, LH, TSH, prolactin, insulin, fasting blood glucose, insulin resistance and metabolic syndrome

Dropouts: 24 (81 women completed the study)

Interventions

Main intervention: 3 equal groups. Group 1: CC 50‐150 mg/d. Group 2: metformin 1700 mg/d. Group 3: CC plus metformin, doses as above)

Duration: 6 months, or until pregnant, or until resistant to CC

Co‐interventions: not applicable

Outcomes

Primary: live birth rate

Secondary: ovulation rate, pregnancy rate, early pregnancy loss rate

Notes

We have contacted the study authors for further information regarding methodology

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Method of generating random sequence for distribution in envelopes not stated

Allocation concealment (selection bias)

Unclear risk

Allocation revealed in envelopes but not clear if opaque or sealed

Blinding (performance bias and detection bias)
All outcomes

Low risk

A member of staff separate to the investigators supplied the envelopes containing the allocation.

Incomplete outcome data (attrition bias)
All outcomes

High risk

22.9% dropout rate, without reasons given

Data analysis not performed as ITT

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Low risk

None noted

Karimzadeh 2007

Methods

RCT 

Setting: Iran 

Method of randomisation: computer‐generated sequences that was sealed in envelopes

Blinding: double 

Number randomised: 200 

Participants

Summary: non‐obese PCOS 

Inclusion criteria: Rotterdam criteria 2003 

Exclusion criteria: hyperprolactinaemia, CSH, thyroid disease, Cushings syndrome, androgen‐secreting tumour

Baseline characteristics of each group:

  • mean age (SD) 27.2 (6.8), 28.6 (7.4)

  • mean BMI (SD) 28.3 (3.18), 29.5 (4.75)

Dropouts: not mentioned

Interventions

Main intervention: metformin 500 mg 3/d, placebo 

Duration: 3 months 

Co‐interventions: nil

Outcomes

Ovulation: progesterone > 10 ng/mL

Metabolic markers: cholesterol, triglycerides 

Others: pregnancy

Notes

Women were recruited from a single centre. The primary objective of this study was to investigate the effect of metformin on lipid profile. The duration of the trial was relatively short. Therefore, it was difficult to ascertain the reliability on both of the ovulation rates and the improvement in menstrual patterns.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated sequences that were sealed in envelopes

Allocation concealment (selection bias)

Low risk

Sequences sealed in envelopes and code kept in the pharmacy department

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Not stated

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information to assess other bias. Sample size calculation not mentioned. Unspecified recruitment period

Karimzadeh 2010

Methods

RCT 

Setting: Iran 

Method of randomisation: not stated

Blinding: not stated 

Number randomised: 343

Participants

Summary: non‐obese PCOS 

Inclusion criteria: Rotterdam criteria 2003. Age between 19 and 35, BMI 25‐29, primary infertility, normal prolactin levels, TFT, liver and renal functions

Exclusion criteria: male factor infertility

Baseline characteristics of each group:

  • mean age: CC only 27.47 metformin only 27.33, CC + met 27.34, lifestyle 27.48

  • mean BMI: CC only 27.2 metformin only 27.17, CC + met 27.96, lifestyle 27.92

Dropouts: none

Interventions

Main intervention: metformin 500 mg 3/d, no placebo

Duration: 3‐6 months 

Co‐interventions: CC 100 mg day 3‐7; lifestyle group were advised to increase daily exercise for 30 min along with high carbohydrate diet

Outcomes

Ovulation: USS follicular tracking

Notes

This study compared the effect of CC, metformin, combined CC and metformin, and lifestyle modification on subfertile women with PCOS.

Very little information can be extracted from the study protocol.

A large sample size without any dropouts

Some of the women may have been included in the previous trial Karimzadeh 2007.

No reply from study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Inadequate information

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Inadequate information

Selective reporting (reporting bias)

Low risk

Not all the primary outcome measures (endocrine parameters, lipid profile) data available

Other bias

Low risk

A large sample size without any dropouts

Some of the women may have been included in the previous trial Karimzadeh 2007.

No reply from study author

Khorram 2006

Methods

RCT 

Setting: USA 

Method of randomisation: picking a card out of a box

Blinding: none

Number randomised: 31

Participants

Summary: obese PCOS 

Inclusion criteria: oligomenorrhoea (< 8 cycles/year), PCO on USS, clinical (acne, hirsutism, alopecia) or biochemical hyperandrogenism (elevated testosterone level)

BMI > 29 

Exclusion criteria: pregnancy, hepatic or renal disease, heart disease, alcoholism, pulmonary disorder, abnormal TFT, hyperprolactinaemia, CAH or androgen‐secreting tumour

Baseline characteristics of each group:

  • mean age (SD) 28.2 (3.12), 28 (4.26)

  • mean BMI (SD) 35.3 (4.0), 38.8 (6.2)

  • mean fasting insulin mIU/L (SD)  17 (11.2), 15.8 (10.8)

  • mean total testosterone nmol/L (SD) 1.79 (0.79), 1.5 (0.97)

Dropouts: none

Interventions

Main intervention: metformin 500 mg 3/d. Placebo was not used

Duration: 2 weeks from the start of the menstrual cycle. 1 trial cycle only

Co‐interventions: CC 100 mg for 5 d from day 5 of the cycle

Outcomes

Ovulation: method to detect ovulation was not stated

Hormones: free testosterone, testosterone, SHBG

Metabolic markers: insulin, glucose

Notes

This study was designed to evaluate the effect of a shot course of metformin treatment on the outcomes of CC ovulation induction therapy. 

All participants were Hispanic except 1 African American in the CC‐only group and 1 white woman in the combined group. None of the participants had taken CC before. 

The trial was unblinded. The method of randomisation and concealment were inadequate. Therefore, potential bias may have been introduced.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Picking a card out of a box

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

High risk

Unblinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No missing data

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Insufficient information

Lam 2011

Methods

RCT 

Setting: Hong Kong

Method of randomisation: computer‐generated random number, block of 10

Blinding: double

Number randomised: 70

Participants

Summary: non‐obese PCOS 

Inclusion criteria: Rotterdam criteria

Exclusion criteria: CAH, Cushing's syndrome, endometrial hyperplasia, diabetes, cardiovascular, hepatic or renal disease

Baseline characteristics of each group:

  • mean BMI 25.9, 23.5

  • mean fasting insulin mIU/L 14.2, 14.9

  • mean total testosterone nmol/L 2.41, 2.38

Dropouts: 11 in metformin, 5 in placebo

Interventions

Main intervention: rosiglitazone 4 mg or placebo

Duration: 6 months

Co‐interventions:

Outcomes

Menstrual cycle frequency

Metabolic parameters: lipid profiles, testosterone, SHBG, glucose and insulin

Notes

This study investigated the effect of using rosiglitazone on Chinese women with PCOS. It is unclear whether the subjects were infertile.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random number

Allocation concealment (selection bias)

Low risk

Trial drug and placebo similar appearance, and packaged according to the trial number. The code kept in the local pharmaceutical company and concealed from the research team until the end of the trial.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

High risk

A much higher dropout rate in the rosiglitazone group than the placebo group. Missing data not reported

Selective reporting (reporting bias)

Low risk

A clear, detailed study protocol and all primary outcome measures reported

Other bias

Unclear risk

Inadequate information

Legro 2007

Methods

RCT

Setting: USA

Method of randomisation: a large multi‐centre, randomised, placebo‐controlled study. (see Legro 2006b for detail)

Blinding: double

Number randomised: 626

Participants

Summary: obese PCOS

Inclusion criteria: oligomenorrhoea (< 8 periods/year), biochemical hyperandrogenism (elevated testosterone level documented within the previous year on the basis of local laboratory results)
Women should have at least 1 proven patent fallopian tube. Normal uterine cavity. Normal semen analysis (sperm concentration > 20 million/mL)

Exclusion criteria: hyperprolactinaemia, CSH, thyroid disease, Cushings's syndrome, androgen‐secreting tumour

Baseline characteristics of each group:
Mean age (SD) 28.3 (4.0), 27.9 (4.0), 28.1 (4)
Mean BMI (SD) 34.2 (8.4), 36.0 (8.9), 35.6 (8.5)
Mean fasting insulin mIU/L (SD) 22.4 (30), 22.6 (20.7), 24 (28.4)
Mean total testosterone mmol/L (SD) 2.21 (0.98), 2.13 (1.1), 2.13 (0.87)

Dropouts: 49 (23.7%) in the metformin and CC group, 55 (26.3%) in the placebo and CC group, 72 (34.6%) in the metformin group. The differences were not significant.

Interventions

Main intervention: 2 extended‐release metformin 500 mg or 2 placebo tablets twice daily

Duration: up to 6 cycles or 30 weeks

Co‐interventions: CC 50 mg or second matching placebo tablet was commenced concurrently from day 3‐7 of the cycle. When women had no or poor response, the dose was increased by 50 mg or 1 additional placebo tablet with the maximum dose of 150 mg or 3 placebo tablets

Outcomes

Ovulation: progesterone > 5 ng/mL

Anthropometric: BMI, WHR

Hormones: testosterone, SHBG

Metabolic markers: insulin, proinsulin, glucose

Others: pregnancy, live birth, miscarriage, side effects, serious adverse events in pregnancy

Notes

This is the largest RCT published so far on the effects of metformin on women with PCOS. A total of 626 infertile women with PCOS were randomised into 3 groups (metformin and placebo, metformin and CC, CC and placebo).

The sample size calculation was based on the live birth rates. The secondary outcomes included the rate of pregnancy loss, singleton birth and ovulation.

Based on the initial sample size calculation, 678 was needed to detect a 15% absolute difference in live birth rates with a power of 80% and a type I error of 0.05. Due to limitations in the supplying metformin and the matching placebo tablets, the number of required women was reduced to 626. This was approved after the assessment by the data safety and monitoring board. Because the observed live birth rate was lower than projected, the number of recruited participants (626) was sufficient to detect a 15% difference with the same magnitude of power and type I error.

The backgrounds of the participants were relatively heterogeneous. Two‐thirds of the participants were white and about one‐third was Hispanic or Latino origin. Only 40% of the women had no previous exposure to metformin or CC.

Ovulation was confirmed when 2 consecutive measurements of progesterone levels > 5 ng/mL in 1‐2 weeks apart.

US monitoring of ovarian response was not included in the study protocol. Ovulation triggering with hCG and intrauterine insemination were not employed in this study.

Metformin combined with CC did not achieve a better live birth rate compared with CC therapy. The metformin group was found to have a significantly inferior pregnancy and live‐birth rate compared with the combined therapy (metformin and CC) and the CC groups. This study also demonstrated that BMI poses a significant negative impact on live births.

In this most recent update, ITT analysis was used to determine ovulation rate per woman. This was calculated from the first 3 treatment cycles, taking into account the number of women who became pregnant.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated; participants were randomised by means of an interactive voice system and stratified based on study site and previous exposure to study drugs

Allocation concealment (selection bias)

Low risk

Each participant received a medication package on a monthly basis that consisted of a bottle M (metformin or placebo) and a bottle C (CC or placebo). Data co‐ordinating centre at the clinical research institute Legro 2006b

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 49 (23.7%) in the metformin and CC group, 55 (26.3%) in the placebo and CC group, 72 (34.6%) in the metformin group. A much higher dropout rate at the metformin‐only group. The differences were not significant. Characteristics of the subjects who dropped out were not given.

Selective reporting (reporting bias)

Low risk

All primary and secondary outcome measures reported

Other bias

High risk

The original sample size was 678 to detect a 15% absolute difference in live birth rates. However, due to drug supply logistics, the sample size later reduced to 626 after the data safety and monitoring board review.

Lord 2006

Methods

RCT

Setting: UK

Method of randomisation: randomisation was conducted centrally by computer at the hospital pharmacy department using a block with sequential numbers. The code was kept sealed until the trial was completed.*

Blinding: double

Number randomised: 44

Participants

Summary: obese PCOS

Inclusion criteria: oligomenorrhoea (< 6 periods/year), biochemical hyperandrogenism (FAI > 5.0)
Age between 18‐40 years

Exclusion criteria: diabetes, thyroid disease, hyperprolactinaemia, CAH, the use of ovulation‐induction agents or drugs that could affect insulin metabolism within 2 months before the start of the trial

Baseline characteristics of each group:

  • mean age (SD) 27.76 (4.89), 30.63 (4.84)

  • mean BMI (SD) 33.74 (6.74),36.37 (7.46)

  • mean fasting insulin mIU/L (SD) 21.57 (15.54), 18.85 (6.04)

  • mean total testosterone mmol/L (SD) 2.60 (0.78), 2.74 (0.65)

Dropouts: 3 women in the metformin group and 1 in the placebo were excluded after they were assigned to the group (did not meet the inclusion criteria). Furthermore, 3 (2 due to pregnancy and 1 lost to follow‐up) in the metformin arm and 5 (3 due to pregnancy and 2 lost to follow‐up) in the placebo arm did not complete the study.
Overall, 6 (27.2%) in the metformin group and 6 (27.2%) in the placebo group withdrew from the study after they had been randomised.

Interventions

Main intervention: metformin 500 mg or placebo tablet 3/d

Duration: 12 weeks

Co‐interventions: general advice on diet and exercise

Outcomes

Ovulation: progesterone > 30 nmol/L

Anthropometric: the distributions of subcutaneous and visceral fat were measured by areal planimetry (CT scan), weight, BMI, waist circumference, WHR, BP

Hormones: testosterone, SHBG, DHEAS

Metabolic markers: insulin, glucose, LDL, HDL, triglyceride

Others: menstrual pattern, pregnancy

Notes

This study was to ascertain the effects of metformin on metabolic parameters, visceral and subcutaneous fat distributions in women with PCOS.

The fat distribution was measured with areal planimetry (CT scan). There were no significant changes in any of the measures of fat distribution between the metformin and the placebo groups. Although, metformin significantly reduced serum cholesterol concentrations, treatment effects on androgens, insulin, triglycerides, ovulation and pregnancy were not observed.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was conducted centrally by computer at the hospital pharmacy department using a block with sequential numbers.

Allocation concealment (selection bias)

Low risk

The code was kept sealed until the trial was completed.

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Overall, 6 (27.2%) in the metformin group and 6 (27.2%) in the placebo group withdrew from the study after they had been randomised. Details of dropouts were not provided

Selective reporting (reporting bias)

Low risk

All outcome measures reported

Other bias

Unclear risk

Inadequate information

Machado 2012

Methods

RCT

Setting: Brazil

Method of randomisation: numbered, sealed, opaque envelopes

Number randomised: 36

Participants

Summary: CC‐resistant PCOS

Inclusion criteria: oligomenorrhoea or amenorrhoea, Rotterdam criteria for PCOS, lack of response to previous ovulation induction with CC

Exclusion criteria: male factor and tubal infertility, endocrinology and chronic health conditions, the use of hormonal treatments within 60 days of the trial commencing

Baseline characteristics of each group: placebo, metformin

  • mean age (SD) 27.1 (4.2), 27.65 (3.6)

  • mean BMI (SD) 28 (3.55), 30 (2.9)

  • insulin resistance (%) 32.15, 18.0

Dropouts*: 67 women were initially included in the study. 21 women did not respond to CC alone and 13 became pregnant. 36 women were then randomised to receive metformin or placebo. All 36 women completed the study, with no women dropping out

Interventions

Main intervention: metformin 850 mg 2/d or placebo tablet 2/d

Duration: 60 days

Co‐interventions: CC 100 mg day 5‐9 with concurrent use of metformin or placebo

Outcomes

Ovulation: visible follicular growth on USS with subsequent formation of the corpus luteum. Free fluid in the POD and change of endometrial thickness also. Plasma progesterone > 3000 pg/mL on day 21

Anthropometric: BMI, WHR

Metabolic markers: insulin, glucose, glucose‐insulin ratio, LFTs, creatinine

Others: pregnancy rate

Notes

This study aimed to evaluate the efficacy of metformin with CC on ovulation in women previously resistant to CC alone. We did not perform a subgroup analysis by BMI in our analysis due to the small number of women in the study.

*Additional information was provided by the study author on request.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Numbered envelopes used

Allocation concealment (selection bias)

Low risk

Sealed, opaque envelopes used

Blinding (performance bias and detection bias)
All outcomes

Low risk

The author has confirmed in private correspondence that women and healthcare providers were blinded for the duration of the study.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data were available for all 36 women who participated in the study.

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Low risk

No other potential bias detected

Maciel 2004

Methods

RCT

Setting: Brazil

Method of randomisation: computer‐generated random numbers

Blinding: double

Number randomised: 30

Participants

Summary: non‐obese PCOS

Inclusion criteria: amenorrhoea or oligomenorrhoea (< 6 periods/year), clinical or biochemical hyperandrogenism. USS evident of PCO was not part of the diagnostic criteria.
Age between 17‐32 years

Exclusion criteria: other causes of amenorrhoea. Use of lipid‐lowering drugs, antidiabetic medications or hormonal contraception within 3 months of the recruitment; Cushing's syndrome, CAH, androgen‐secreting tumours, diabetes, renal or hepatic disease

Baseline characteristics of each group:

  • mean age (SD) 22.5 (5), 19.9 (1.1)

  • mean BMI (SD) 25.3 (5.5), 25.1 (4.5)

  • mean fasting insulin mIU/L (SD) 12.1 (6.3), 13.6 (8.8)

  • mean total testosterone mmol/L (SD) 3.67 (1.1), 3.38 (1.2)

Dropouts: details of the dropouts were not available

Summary: obese PCOS 

Inclusion criteria: amenorrhoea or oligomenorrhoea (< 6 periods/year), clinical or biochemical hyperandrogenism. USS evident of PCO was not part of the diagnostic criteria.

Age between 17‐32 years

Exclusion criteria: other causes of amenorrhoea. Use of lipid‐lowering drugs, antidiabetic medications or hormonal contraception within 3 months of the recruitment; Cushing’s syndrome, CAH, androgen‐secreting tumours, diabetes, renal or hepatic disease. 

Baseline characteristics of each group:

Mean age (SD) 20.5 (5.4), 21.1 (1.7)

Mean BMI (SD) 37.2 (4.8), 35.8 (3.7)

Mean fasting insulin mIU/L (SD) 22.6 (11.6) 20.9 (4.6)

Mean total testosterone nmol/L (SD) 4.1 (0.8), 3.5 (2.4)) 

Dropouts: details of the dropouts were not available

Interventions

Main intervention: metformin 500 mg or placebo tablet 3/d

Duration: 6 months

Co‐interventions: none

Outcomes

Anthropometric: BMI, BP

Hormones: testosterone, SHBG, free testosterone, androstenedione

Metabolic markers: insulin, glucose, AUC insulin, AUC glucose, LDL, HDL and triglyceride

Others: menstrual pattern, hirsutism

Notes

The primary objective of this study was to compare the clinical, hormonal and biochemical effects of metformin therapy in the obese PCOS group (BMI > 30) with the non‐obese group (BMI < 30). We entered the results of the obese group separately in the analysis.

The results indicated that non‐obese participants responded better than obese participants with PCOS to metformin 1.5 g/d. Non‐obese women experienced an improvement in menstrual cyclicity, decrease in serum androgen levels and fasting insulin concentrations; whilst, obese women showed a significant reduction of free testosterone levels. Caution is needed to interpret the results as 5 of the original 34 enrolled participants did not complete the trial and these findings were not included in the analysis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers

Allocation concealment (selection bias)

Low risk

Participants received a sealed envelope that contained the study number. An independent clinician recorded side effects and clinical measurements

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

5 participants were not evaluated because of pregnancy. Details were not given

Selective reporting (reporting bias)

Unclear risk

All pre‐specified outcome measures (androgens and metabolic parameters)

Other bias

Unclear risk

Although USS evidence of PCO was not employed as part  of the diagnostic criteria for PCOS,  the diagnostic criteria used in this study would have met the Rotterdam criteria.

Malkawi 2002

Methods

RCT

Setting: Jordan

Method of randomisation: centralised randomisation process with women receiving a sequential number*

Blinding: double‐blind*

Number randomised: 28

Participants

Summary: non‐obese PCOS, CC resistance

Inclusion criteria: US findings of polycystic ovaries together with 3 of: oligomenorrhoea < 6 cycles in preceding year, Ferriman‐Gallwey score > 7, hyperandrogaenemia (free testosterone, androstenedione, DHEAS), elevated LH or LH:FSH > 2 CC resistance defined as failure to ovulate with 150 mg day 5‐9 for 3 months. Normal uterine cavity and patent tubes on hysterosalpingography. Normal semen analysis

Exclusion criteria: raised prolactin, adrenal hyperplasia, thyroid dysfunction, Cushing's syndrome.

Baseline characteristics of each group:

  • mean age (± SD) 29 (3.1), 29 (7.3)

  • mean BMI (± SD) 27.5 (4.1), 27.8 (3.3)

  • mean fasting insulin mIU/L (± SD) 20.5 (4.2), 21.2 (5.3)

  • mean total testosterone mmol/L (± SD) 1.14 (0.17), 1.07 (0.18)

Dropouts: nil

Interventions

Main intervention: 1 of metformin 850 mg 2/d, placebo

Duration: 6 months

Co‐interventions: CC 50 mg day 5‐9 in the first cycle, increasing by 50 mg up to 200 mg in each subsequent cycle until ovulation achieved

Outcomes

Ovulation: serum progesterone on day 21 and 28 > 15.9 nmol/L

Others: pregnancy

Notes

Units of testosterone assumed to be ng/mL

*Information kindly provided by the study author that was not in the original paper

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Centralised randomisation process with women receiving a sequential number

Allocation concealment (selection bias)

Unclear risk

Centralised randomisation process with women receiving a sequential number

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Moghetti 2000

Methods

RCT

Setting: Italy

Method of randomisation: sequentially numbered, identical containers of identical drugs*

Blinding: double‐blind

Number randomised: 23

Participants

Summary: non‐obese PCOS

Inclusion criteria: PCOS (oligomenorrhoea ≤ 6 cycles/year or anovulation confirmed with luteal‐phase progesterone, hyperandrogaenemia (either raised serum androgens, or clinical hyperandrogaenemia*). Exclusion of other endocrinopathy

Exclusion criteria: adrenal hyperplasia, Cushing's syndrome, thyroid dysfunction, hyperprolactinaemia, androgen‐secreting tumour,
concomitant disease, taking any medication.

Baseline characteristics of each group:

  • mean age (± SD) 23.9 (4.0), 21.4 (4.9)

  • mean BMI (± SD) 27.1 (5.0), 32.6 (3.8)

  • mean fasting insulin mIU/L (± SD) 15.2 (15.3), 20.1 (13.9)

  • mean total testosterone nmol/L (± SD)* 2.9 (0.6), 2.4 (0.6)

Dropouts: nil*

Interventions

Main intervention: 1 of metformin 500 mg 3/d, placebo

Duration: 26 weeks

Co‐interventions: no modification in usual eating habits

Outcomes

Anthropometric: BMI, WHR,

Reproductive hormones: free testosterone, androstenedione, DHEAS, SHBG, FSH, LH

Metabolic markers: fasting glucose, fasting insulin, 120‐min insulin and glucose levels after GTT, insulin sensitivity, HDL, LDL, triglycerides, systolic BP, diastolic BP

Others: menstrual pattern, 17‐alpha‐hydroxyprogesterone response to buserelin

Notes

Placebo group had significantly higher BMI (P < 0.05) at baseline and higher fasting insulin (non‐significant), but similar insulin sensitivity. Metformin group had higher androgens (non‐significant)

Mild side effects in 5 in metformin group and 2 in placebo group

It is assumed that the figures quoted in the publication are for standard errors.

*Information kindly provided by the study author that was not in the original paper

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Low risk

Sequentially numbered, identical containers of identical drugs

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Low risk

All primary outcome measures reported (menstrual frequency and metabolic parameters)

Other bias

High risk

Placebo group had a significantly higher BMI than the metformin group. It was assumed that the figures quoted in the publication are for standard errors.

Moll 2006

Methods

Multicentre RCT

Setting: the Netherlands

Method of randomisation: computer‐generated blocks of 4

Blinding: double‐blind

Number randomised: 225

Participants

Summary: non‐obese women with PCOS

Inclusion criteria: PCOS (according to Rotterdam consensus), normal FSH concentrations

Exclusion criteria: age > 40 years, abnormal liver function tests or creatinine levels > 95 umol/L, history of heart disease, history of male factor infertility with total motile sperm count < 10 x 106

Baseline characteristics of each group:

  • mean age (SD) 27.9 (3.7), 28.4 (4.7)

  • mean BMI (SD) 28.5 (7.1), 27.8 (6.7)

  • mean fasting insulin mIU/L (SD)

  • mean total testosterone nmol/L (SD) 3.49 (3.68), 3.55 (3.54)

Dropouts: no significant difference in the dropout rates, 28 (25%) in the metformin arm, 21 (18%) in the placebo arm

Interventions

Main intervention: metformin 2000 mg/d (increased from 500 mg to 2000 mg over a period of 7 days in order to limit the side effects) or placebo

Duration: all women received metformin or placebo for 1 month before starting CC treatment (a maximum of 6 cycles for those who ovulated with CC)

Co‐interventions: CC 50 mg from day 3 (spontaneous menstruation) or day 5 (progestogen induced menstruation) for a period of 5 days. If ovulation did not occur with this dose, CC was increased with steps of 50 mg with a maximum of 150 mg/d in the next cycles

Outcomes

Ovulation: progesterone > 14 nmol/L in the second half of menstrual cycle, biphasic basal body temperature curve, follicular diameter > 16 mm on transvaginal USS or pregnancy

Anthropometric:

Hormones:

Metabolic markers:

Others: pregnancy, miscarriage and CC resistance

Notes

A large, multicentre RCT. The sample size calculation was based on the ovulation rate. In total, 228 women were initially screened and 3 were subsequently excluded. 111 women were randomised to receive metformin and CC; whilst 114 received placebo and CC.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated blocks of 4

Allocation concealment (selection bias)

Low risk

Randomisation was carried out in the co‐ordinating centre (Amsterdam) and the list was kept until inclusion was completed

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double blind. Each centre received blinded, numbered container

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropouts: no significant difference in the dropout rates, 28 (25%) in the metformin arm, 21 (18%) in the placebo arm. Details of the dropout participants not mentioned; although number of dropouts in each group were similar

Selective reporting (reporting bias)

Low risk

Primary outcome (ovulation) and secondary outcome (pregnancy, miscarriage rates) measures reported

Other bias

Unclear risk

Inadequate information

Morin‐Papunen 2012

Methods

Multicentre RCT (parallel‐group study)

Setting: Finland

Method of randomisation: randomisation codes remained concealed. Metformin and placebo identically packaged and consecutively numbered

Blinding: double

Number randomised: 320

Participants

Summary: metformin and pregnancy outcomes in PCOS

Inclusion criteria: anovulatory infertility for at least 6 months and 3 months since the last infertility treatment. Age range 18‐39 years

Exclusion criteria: type 1 diabetes mellitus, liver, cardiac or renal disease, hormone medication, alcohol use, regular smoking

Baseline characteristics of each group: Metformin, placebo

  • mean age (SD) 28.4 (3.9), 27.9 (4.1)

  • mean BMI (SD) 27.1 (6.3), 27.4 (6.2)

  • mean fasting insulin (microIU/ml) 11.0 (11.2), 11.4 (11.8)

  • testosterone (ng/dL) 43.2 (17.3), 45.8 (20.2)

Dropouts: 61 women were lost to follow‐up or discontinued but their data were included in the ITT analysis

Interventions

Main intervention: metformin 500 mg 1/d for 1 week, then increased weekly by 1 extra tablet/d to 1.5 g in non‐obese and 2 g/d in obese women versus placebo

Duration: 3‐9 months

Co‐interventions: if pregnancy has not occurred by 3 months, ovulation induction was started with CC. If unsuccessful after 4‐6 cycles, gonadotrophins or aromatase inhibitors were used

Outcomes

Anthropometric: WHR, waist (cm), hirsutism score, BMI, ovarian volume

Others: pregnancy rate, miscarriage rate, pregnancy complications, live birth rate

Notes

This study was to ascertain the effects of metformin on pregnancy and live birth rates. Endocrine/metabolic outcomes not measured. Additional information sought from the study authors

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Performed by hospital pharmacy with 1:1 allocation in random blocks of 10 using computer‐generated lists

Allocation concealment (selection bias)

Low risk

Randomisation codes remained blinded until database lock had taken place

Blinding (performance bias and detection bias)
All outcomes

Low risk

Metformin and placebo identically packaged and consecutively numbered

Incomplete outcome data (attrition bias)
All outcomes

Low risk

61 women were lost to follow‐up or discontinued but their data were included in the ITT analysis

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Unclear

Nestler 1998

Methods

Multicentre RCT

Setting: USA (3 participants), Venezuela (54 participants), Italy (4 participants)*

Method of randomisation: centralised randomisation process*.

Blinding: single‐blind, participants blinded

Number randomised: 61

Participants

Summary: PCOS, obese

Inclusion criteria: PCOS (oligomenorrhoea < 6 cycles/year, hyperandrogaenemia (elevated free testosterone), exclusion of other endocrinopathy, US finding of PCO), BMI >28

Exclusion criteria: diabetes mellitus, adrenal hyperplasia, thyroid dysfunction, hyperprolactinaemia, taking any medication for previous 2 months

Baseline characteristics of each group:

  • mean age (± SD) 29 (5.9), 28 (5.1)

  • mean BMI (± SD) 32.3 (4.7), 32.2 (5.1)

  • mean fasting insulin mIU/L (± SD) 19 (11.8), 22( 30.6)

  • mean total testosterone mmol/L (± SD) 2.44 (1.0), 2.20 (0.9)

Dropouts: none

Interventions

Main intervention: 1 of metformin 500 mg 3/d, placebo

Duration: 34 d, then those who did not ovulate continued for a further 19 d

Co‐interventions: those that did not ovulate after 34 days had CC 50 mg for 5 d and continued metformin/placebo for a total of 53 d

Outcomes

Ovulation: by serum progesterone (≥ 25.6 nmol/L) measured on days 14, 28, 35 (and 44 & 53 in those that went on to receive CC)

Anthropometric: BMI, WHR

Reproductive hormones: total testosterone, free testosterone, androstenedione, DHEAS, SHBG, 17‐beta estradiol

Metabolic markers: fasting glucose, fasting insulin, AUC of insulin and glucose during GTT

Notes

89% of participants were recruited in Venezuela

Most of the outcome measures were only reported for those that failed to ovulate during the metformin vs placebo phase of the trial. These have not been included in the analysis as a further analysis to include all participants was not possible.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Centralised randomisation process

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

High risk

Single‐blinded (participant only)

Incomplete outcome data (attrition bias)
All outcomes

High risk

No dropouts. Most of the outcome measures were only reported for those that failed to ovulate during the metformin vs placebo phase of the trial.

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Nestler 1999

Methods

Multicentre RCT

Setting: Venezuela (white 73%, Hispanic 16%, Afro‐Hispanic 4.5%, Arabic 4.5%, Asian 2%)

Method of randomisation: drug and placebo packaged at same time and labelled according to participant number. Randomisation in blocks of 4
Blinding: double‐blind

Number randomised: 44

Participants

Summary: PCOS, obese

Inclusion criteria: PCOS (oligomenorrhoea ≤ 8 cycles/year, hyperandrogaenemia (elevated free testosterone)* or hirsutism (physician reported ‐ subjective)*, exclusion of other endocrinopathy), BMI > 28

Exclusion criteria: diabetes mellitus, thyroid dysfunction, hyperprolactinaemia, taking any medication for previous 2 months.

Baseline characteristics of each group:

  • mean age (± SD) 29 (6), 26 (5)

  • mean BMI (± SD) 31.3 (2.4), 31.0 (2.2)

  • mean fasting insulin mIU/L (± SD) 35 (40), 38 (51)

  • mean total testosterone mmol/L (± SD) 3.14 (1.64), 2.79 (1.50)

Dropouts: none

Interventions

Main intervention: 1 of D‐chiro inositol 1200 mg 1/d, placebo

Duration: 6 weeks; those who ovulated continued for a further 2 weeks
Co‐interventions: no change in usual eating habits, physical activity or lifestyle

Outcomes

Ovulation: by serum progesterone (≥ 25 nmol/L) weekly

Anthropometric: BMI, WHR

Reproductive hormones: total testosterone, free testosterone, androstenedione, DHEAS, SHBG, 17‐beta estradiol

Metabolic markers: fasting glucose, fasting insulin, AUC of insulin and glucose during GTT, systolic BP, diastolic BP, HDL, LDL, triglycerides

Others: LH response to leuprolide, 17‐alpha‐hydroxyprogesterone response to leuprolide

Notes

All women had US features of PCO, but this was not an inclusion criteria

None of the participants had diabetes mellitus, but 10 (23%) had impaired glucose tolerance (6 in treatment arm, 4 in placebo arm)
*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Randomisation in blocks of 4

Allocation concealment (selection bias)

Low risk

Drug and placebo packaged at same time and labelled according to participant number

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No dropouts

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Insufficient information

Ng 2001

Methods

RCT

Setting: Hong Kong (Chinese women)

Method of randomisation: computer‐generated list in sealed envelopes

Blinding: double‐blind

Number randomised: 20

Participants

Summary: non‐obese PCOS, CC resistance

Inclusion criteria: PCOS (irregular cycles of ≤ 21 days or ≥ 35 days and cycle‐to‐cycle variation of > 4 days*, anovulation with mid‐luteal progesterone < 16 nmol/L whilst taking CC 100 mg for 5 d over 3 cycles, exclusion of other endocrinopathy (raised prolactin, thyroid disorder*), US findings of PCO, age < 40, day 2 FSH < 10, bilateral patent tubes demonstrated by laparoscopy, normal semen parameters

Exclusion criteria: taking any sex hormones in previous 3 months, smokers, renal impairment.

Baseline characteristics of each group*:

  • mean age (± SD) 30.4 (2.1), 31.2 (2.6)

  • mean BMI (± SD) 25.5 (4.6), 23.5 (4.4)

  • mean fasting insulin mIU/L (± SD) 10.4 (4.9), 12.4 (5.9)

  • mean total testosterone mol/L (± SD) 2.0 (0.9), 1.6 (1.2)

Dropouts: 5 (25%), 3 in placebo arm, 2 in metformin. Analysis on ITT

Interventions

Main intervention: 1 of metformin 500 mg 3/d, placebo

Duration: 3 months. Those who did not ovulate continued for a further cycle

Co‐interventions: CC 100 mg for 5 d was given after 3 months if there was no ovulation

Outcomes

Ovulation: by serum progesterone (> 16 nmol/L) weekly

Anthropometric: BMI

Reproductive hormones: total testosterone, androstenedione, DHEA, SHBG, FSH, LH

Metabolic markers: fasting glucose, fasting insulin, 120‐min glucose levels after GTT, fasting leptin, HDL, LDL, triglycerides

Other: live birth

Notes

The BMI was lower than in other trials

In spite of the fact that anovulation and CC resistance was an inclusion criteria, 7 out of 9 women taking placebo ovulated (3 with placebo alone, and 4 out of the 6 remaining in the trial who had CC and placebo)

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated list

Allocation concealment (selection bias)

Low risk

In sealed envelopes. Double, identical appearance and packed by the hospital pharmacy. Code kept in the pharmacy department until the end of the trial

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropouts: 5 (25%), 3 in placebo arm, 2 in metformin. Analysis on ITT. Details not provided

Selective reporting (reporting bias)

Low risk

All primary outcome measures reported

Other bias

Unclear risk

In spite of the fact that anovulation and CC resistance was an inclusion criteria, 7 out of 9 women taking placebo ovulated (3 with placebo alone, and 4 out of the 6 remaining in the trial who had CC and placebo)

Onalan 2005

Methods

RCT

Setting: Turkey

Method of randomisation: computer‐generated randomisation in blocks of 4

Blinding: double*

Number randomised: 139 were randomised into 6 main groups according to the fasting glucose/insulin ratio (with a level < 4.5 classified as hyperinsulinaemia) and BMI (< 25, 25‐29.9 and > 30)

Participants

Summary: non‐obese PCOS

Inclusion criteria: oligomenorrhoea (< 6 periods/year), clinical hyperandrogenism (Ferrriman‐Gallwey score > 7) and/or biochemical hyperandrogenism (free testosterone > 4 ng/dL)

Exclusion criteria: other causes of hyperandrogenism, Cushing's syndrome, CAH, hyperprolactinaemia, thyroid dysfunction

Baseline characteristics of each group:

  • mean age (SD) hyperinsulinaemic lean 25.7 (4.9), 24.2 (4.7); hyperinsulinaemic overweight 27.5 (5.7), 24.8 (6.6); normoinsulinaemic lean 26.4 (4.1), 27.1 (4.8); normoinsulinaemic overweight 24.6 (4.8), 27.3 (4.4)

  • mean BMI (SD) hyperinsulinaemic lean 21.55 (3.07), 21.8 (1.76); hyperinsulinaemic overweight 28.4 (0.7), 28.4 (0.9); normoinsulinaemic lean 21.6 (2.25), 21.96 (1.52); normoinsulinaemic overweight 28.1 (1.0), 28.2 (0.7)

  • mean fasting insulin mIU/L (SD) hyperinsulinaemic lean 20.5 (0.68), 22.0 (3.95); hyperinsulinaemic overweight 22.7 (3.0), 23.1 (6.0); normoinsulinaemic lean 14.9 (2.2), 15.6 (2.52); normoinsulinaemic overweight 14.6 (1.5), 13.8 (1.6)

Summary: obese PCOS 

Inclusion criteria: oligomenorrhoea (< 6 periods/year), clinical hyperandrogenism (Ferrriman‐Gallwey score > 7) and/or biochemical hyperandrogenism (free testosterone >4 ng/dL)

Exclusion criteria: other causes of hyperandrogenism, Cushing's syndrome, CAH, hyperprolactinaemia, thyroid dysfunction 

Baseline characteristics of each group:

  • Mean age (SD) hyperinsulinaemic obese 25.1 (3.6), 28.4 (6.9); normoinsulinaemic obese 31.8 (4.0)

  • Mean BMI (SD) hyperinsulinaemic obese 31.7 (1.9), 34.9 (3.5); normoinsulinaemic obese 31.6 (1.1), 32.2 (3.2)

  • Mean fasting insulin mIU/L (SD) hyperinsulinaemic obese 27.8 (10.3), 23.3 (2.8); normoinsulinaemic obese 18.8 (2.3), 21.2 (1.3)

Dropouts: 15 in total, mainly due to gastro‐intestinal side effects. Further 8 women were excluded in the analysis because of pregnancy*

Dropouts: 15 in total, mainly due to gastro‐intestinal side effects*

Interventions

Main intervention: metformin 850 mg or placebo tablet twice daily

Duration: 6 months

Co‐interventions: none

Outcomes

Ovulation: progesterone > 5 ng/mL

Anthropometric: BMI, weight, WHR*

Hormones: testosterone, free testosterone, androstenedione, DHEAS, cortisol*

Metabolic markers: glucose, insulin, LDL, HDL, triglyceride*

Others: hirsutism*

Notes

The objective of this study was to investigate the effects of hyperinsulinaemia (fasting glucose/insulin ratio < 4.5mg/10‐4 U and obesity (BMI > 30) on the responses to metformin treatment in women with PCOS. There were 6 subgroups, normoinsulinaemic lean (BMI < 25), overweight (BMI 25‐29.9) and obese (BMI >30); hyperinsulinaemic lean (BMI < 25), overweight (BMI 25‐29.9) and obese (BMI > 30)

The results of the non‐obese subgroups were entered separately from the obese subgroup in the meta‐analysis

We have written to the study author regarding the details of randomisation and concealment. Additionally, we also asked the study author to provide further information of the anthropometric, hormonal and metabolic results at the end of the trial period. 

*No reply from study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated blocks of 4 randomisation

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 15 in total (11%), mainly due to gastro‐intestinal side effects. Missing outcomes not addressed. Imbalance in missing data between the intervention and placebo groups.

Selective reporting (reporting bias)

Unclear risk

Primary outcome measures not stated. Inadequate study protocol reporting

Other bias

Unclear risk

Inadequate information to assess

Otta 2010

Methods

RCT

Setting: Argentina

Method of randomisation: computer‐generated

Blinding: double

Number randomised: 30

Participants

Summary: obese PCOS

Inclusion criteria: oligomenorrhoea (cycle length > 35 days), biochemical hyperandrogenism (level not defined)

Exclusion criteria: other causes of hyperandrogenism, Cushing's syndrome, CAH, hyperprolactinaemia, thyroid dysfunction, abnormal renal, liver functions, diabetes, infection

Baseline characteristics of each group:

  • mean age 25, 24

  • mean BMI 32.4, 31.5

  • mean fasting insulin mIU/L 14.2, 17.18

Dropouts: 1 in metformin, poor compliance

Interventions

Main intervention: metformin 750 mg or placebo tablet twice daily

Duration: 4 months

Co‐interventions: lifestyle modification (high carbohydrate diet and increase exercise with a minimum of 40 min walk/d)

Outcomes

Ovulation: method of detecting ovulation not stated

Anthropometric: BMI, weight, WHR

Metabolic markers

Notes

This study investigated the effects of combined metformin and lifestyle changes on endocrine and metabolic parameters in women with PCOS

Methodology and study protocol were too brief. Unable to determine the quality of the trial

Only 5 out of 30 subjects were trying to conceive.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Not stated

Incomplete outcome data (attrition bias)
All outcomes

High risk

Missing data not reported. 1 in metformin group and excluded from analysis

Selective reporting (reporting bias)

Unclear risk

Primary outcome measures were unclear

Other bias

Low risk

Methodology and study protocol were too brief. Unable to determine the quality of the trial

Only 5 out of 30 subjects were trying to conceive.

Palomba 2005

Methods

RCT 

Setting: Italy

Method of randomisation: computer‐generated random allocation sequence in double block

Blinding: double 

Number randomised: 100

Participants

Summary: non‐obese PCOS 

Inclusion criteria: National Institutes of Health criteria, age 20‐34 years, BMI < 30 kg/m2, tubal patency confirmed by hysterosalpingogram, normal semen analysis

Exclusion criteria: metabolic disorders, hepatic or renal dysfunction, thyroid disease, hyperprolactinaemia, Cushing's syndrome, CAH, hormonal drugs, pelvic diseases, previous pelvic surgery

Baseline characteristics of each group:

  • mean age (SD) 26.4 (2.9), 25.9 (2.7)

  • mean BMI (SD) 27.0 (2.9), 26.7 (2.8)

  • mean fasting  insulin mIU/L (SD) 19.5 (5.4), 20.4 (5.6)

  • mean total testosterone mol/L (SD) 3.12 (1.04), 3.47 (1.0)

Dropouts: 5 in the metformin group and 3 in the metformin + CC group

Interventions

Main intervention: metformin 850 mg or matched placebo tablets twice daily 

Duration: 6 months 

Co‐interventions: CC 150 mg or matched placebo tablets, day 3‐7 of the cycle and timed intercourse

Outcomes

Ovulation: USS follicular tracking

Pregnancy, ovulation

Notes

This study was designed to compare the effectiveness of metformin and CC treatment as a first‐line therapy in non‐obese anovulatory women with PCOS. 

The primary end point measure was the pregnancy rate.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random allocation sequence in double block

Allocation concealment (selection bias)

Unclear risk

Allocation sequence concealed until the interventions were assigned

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 5 in the metformin group and 3 in the metformin + CC group

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Insufficient information

Pasquali 2000

Methods

RCT

Setting: Italy

Method of randomisation: block of 4. Drug and placebo packaged and labelled according to participant number

Blinding: double‐blind

Number randomised: 20

Participants

Summary: obese PCOS

Inclusion criteria: oligomenorrhoea (< 4 cycles in past 6 months), hyperandroenaemia, USS of PCO, BMI > 25, WHR > 0.8

Exclusion criteria: diabetes mellitus, adrenal hyperplasia, thyroid dysfunction, hyperprolactinaemia, cardiovascular, renal or liver dysfunction

Baseline characteristics:

  • age: 30.8, 32.3

  • BMI: 39.8, 39.39

  • mean fasting insulin mIU/L 43, 33.5

  • mean total testosterone mol/L 2.37, 1.78

Dropouts: 2 from metformin arm due to pregnancy. Not included in analysis

Interventions

Main intervention: metformin 850 mg 2/d or placebo

Duration: 6 months

Co‐interventions: standardised hypercaloric diet 1 month prior to treatment and continued throughout the trial

Outcomes

Anthropometric parameters

Reproductive hormones and metabolic markers

Notes

The trial was designed to investigate the combined effects of diet and metformin on fat distribution in women with PCOS. The study also included a control group who were matched for age, weight and WHR but with regular menstrual cycles.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Block of 4, random table

Allocation concealment (selection bias)

Low risk

Drug and placebo packaged and labelled according to participant number

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Missing data not reported

Selective reporting (reporting bias)

Low risk

A clear protocol published with all primary outcome measured reported

Other bias

Unclear risk

Inadequate information

PCOSMIC 2010

Methods

Multicentre RCT

Setting: New Zealand

Randomisation: double‐blind

Number randomised: 171

Participants

Inclusion criteria: women with PCOS according to Rotterdam consensus criteria

Exclusion criteria: couples had undergone previous fertility treatment involving > 5 months treatment with CC or metformin; tubal factor (at least 1 tube blocked); severe male factor (< 15 mil/mL); important medical disorders

Interventions

Women with BMI > 32 kg/m2 were randomised to receive either metformin 500 mg 3/d (increasing dose over 2 weeks) or matching placebo

Women with BMI ≤ 32 kg/m2 were randomised to receive either metformin 500 mg 3/d, CC 50 mg from day 2‐6 (increasing up to 150 mg over 3 months if no evidence of ovulation) or metformin 500 mg 3/d combined with CC 50 mg day 2‐6 (increasing up to 150 mg over 3 months if no evidence of ovulation)

Participants received up to 2 packages of 3 months' treatments. All study drugs were stopped once the participant was pregnant

Outcomes

Primary outcomes were clinical pregnancy (intrauterine gestation sac) and live birth

Secondary outcomes were ovulation, miscarriage, ectopic pregnancy or multiple pregnancy

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computerised block randomisation (blocks of 10)

Allocation concealment (selection bias)

Low risk

"allocation concealment was strictly maintained by a telephone call from the recruiting nurse to pharmacy, ...dispensing pre‐prepared drugs in a true third party randomisation"

Blinding (performance bias and detection bias)
All outcomes

Low risk

"Blinding of all parties was maintained in all cases ...until the end of the course of treatment or in the event of pregnancy, until after the pregnancy"

Incomplete outcome data (attrition bias)
All outcomes

Low risk

ITT analysis planned and protocol breach and losses to follow‐up were reported in figure 3

Selective reporting (reporting bias)

Low risk

Protocol published and all outcomes reported

Other bias

Unclear risk

Inadequate information

Rautio 2006

Methods

RCT

Setting: Finland

Method of randomisation: computer‐generated random numbers (block of 5). Randomisation was conducted within the department.*

Blinding: double*

Number randomised: 30

Participants

Summary: obese PCOS

Inclusion criteria: PCOS was defined according to the Rotterdam consensus 2003. PCO on USS. Oligomenorrhoea or amenorrhoea, clinical (Ferriman‐Gallwey score > 7) or biochemical (testosterone > 2.7 nmol/L) hyperandrogenism, BMI>25

Exclusion criteria: diabetes, abnormal liver function tests, smokers, history of alcohol abuse, hormonal drugs or drugs known to affect lipid metabolism

Baseline characteristics of each group:

  • mean age (SD) 26.7 (1.1), 30.1 (2.1)

  • mean BMI (SD) 33.1 (5.8), 33.6 (3.7)

  • mean fasting insulin mIU/L (SD) 12.4 (6.58), 15.0 (9.73)

  • mean total testosterone mmol/L (SD) 2.7 (0.35), 3.5 (1.12)

Dropouts: 3 (20%) in the rosiglitazone group (2 due to pregnancy); 1 (6.6%) in the placebo group (personal reasons)

Interventions

Main intervention: rosiglitazone 4 mg for 2 weeks followed by 4 mg twice daily for 4 months or placebo

Duration: 4 months

Co‐interventions: none

Outcomes

Anthropometric: BMI, WHR*

Hormones: testosterone, SHBG, androstenedione, DHEAS

Metabolic markers: insulin, glucose, AUC insulin, AUC glucose*, fasting C‐peptide, insulin resistance measured by euglycaemic hyperinsulinaemic clamp test

Others: hirsutism, menstrual pattern

Notes

The objective of this study was to assess the effects of rosiglitazone in obese women with PCOS. All the participants were recruited from a single endocrine clinic.

All the participants were advised to use some form of non‐hormonal contraception during the study as rosiglitazone is a category C drug.

Rosiglitazone improves menstrual cyclicity, insulin resistance and hyperandrogenism.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers (block of 5)

Allocation concealment (selection bias)

Unclear risk

Randomisation was conducted within the department. Unclear concealment, carried out by the departmental staff

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 3 (20%) in the rosiglitazone group (2 due to pregnancy); 1 (6.6%) in the placebo group (personal reasons)

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Romualdi 2010

Methods

RCT 

Setting: Italy 

Method of randomisation: block of 10 sealed envelopes containing randomisation codes assigning 5 women to metformin and 5 to placebo group 

Blinding: double

Number randomised: 28

Participants

Summary: non‐obese PCOS 

Inclusion criteria: Rotterdam consensus 2003, normal weight 

Exclusion criteria: abnormal TFT, LFT 

Baseline characteristics of each group:

  • mean age (SD):24.7 (4.4), 27.2 (2.3)

  • mean BMI (SD): 22.2 (2.2), 22.3 (3.9)

  • mean fasting insulin mIU/L (SD)

  • mean total testosterone nmol/L (SD): 1.94, 1.9 

Dropouts: 2 in metformin due to poor compliance; 3 in placebo group (lost to follow‐up)

Interventions

Main intervention: metformin 500 mg or placebo tablets twice daily 

Duration: 6 months 

Co‐interventions: lifestyle modification

Outcomes

Ovulation: 

Anthropometric: BMI, WHR 

Hormones: testosterone, SHBG 

Metabolic markers: lipid profiles

Notes

A small RCT investigated the effect of metformin on ovarian US appearance and steroidogenic function in normal‐weight normoinsulinaemic women with PCOS. Only the metabolic data was included in our analysis.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Not stated

Allocation concealment (selection bias)

Low risk

Blocks of 10 sealed opaque envelopes containing randomisation codes

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind, placebo‐controlled

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 2 in metformin due to poor compliance; 3 in placebo group (lost to follow‐up)

Selective reporting (reporting bias)

Low risk

None identified

Other bias

Unclear risk

Inadequate information

Sahin 2004

Methods

RCT 

Setting: Turkey 

Method of randomisation: not stated* 

Blinding: not stated* 

Number randomised: 21

Participants

Summary: obese PCOS

Inclusion criteria: PCO on USS (≥ 10 cysts 2‐10 mm), oligomenorrhoea (cycle length > 35 d) or amenorrhoea (no menstrual period > 6 months), clinical or biochemical hyperandrogenism (testosterone > 2.7 nmol/L); participants received no medication known to affect pituitary‐ovarian function or carbohydrate metabolism for at least 12 weeks before the study

Exclusion criteria: androgen‐secreting tumour, Cushing’s syndrome, thyroid dysfunctions, CAH, hyperprolactinaemia and diabetes

Baseline characteristics of each group:

  • median age 27, 24.5

  • median BMI 30.4, 25.7

 Dropouts: none

Interventions

Main intervention: metformin 850 mg twice daily. Placebo was not used.

Duration: metformin alone or no treatment for 3 months followed by combining CC ovulation‐induction therapy for further 6 cycles or until pregnancy occurred. 

Co‐interventions: CC 100 mg daily for 5 d from day 5 of the cycle. Ovulation was triggered by administration of 10,000 IU hCG (Pregnyl, Organon, Holland)

Outcomes

Ovulation: progesterone > 5.0 ng/mL

Anthropometric: BMI 

Hormones: testosterone, free testosterone, androstenedione, SHBG, DHEAS, estradiol, prolactin

Metabolic markers: insulin, glucose, AUC insulin, AUC glucose

Others: pregnancy, live birth, miscarriage

Notes

Women were recruited from a single infertility unit. All participants presented with primary infertility. Tubal disease and male‐factor infertility were excluded. Of the women, 90% presented with oligomenorrhoea and 10% amenorrhoea. In addition, half of the participants had Ferriman‐Gallwey score > 8. 

Since placebo was not used in the study, bias may exist in the trial period. We are still waiting for a reply from the study author regarding the method of randomisation and concealment. Furthermore, all the anthropometric, hormonal and metabolic data were presented in a format of median and range, which we cannot enter in the meta‐analysis. Hence, we asked the study author to provide the results in mean and standard deviation. 

Ovulation rates after the initial 3 months metformin treatment alone were not given. 

The response to CC treatment was monitored by serial USS. When there were < 4 follicles with diameter > 15 mm with a leading follicle of > 18 mm in diameter, 10,000 IU hCG was administrated intramuscularly. 

Pregnancy was defined by US evidence of a gestational sac and the presence of fetal heart activity.

In this most recent update, we have calculated ovulation rate per woman. In the paper, values are given as number of participants and percentage ovulation/cycle. These data have been used to infer the ovulation rate per person.

 

*No reply from the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Inadequate information

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Insufficient information to assess

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Insufficient information to assess

Siebert 2009

Methods

RCT 

Setting: South Africa 

Method of randomisation: computer‐generated random numbers

Blinding: unblinded 

Number randomised: 107

Participants

Summary: obese PCOS 

Inclusion criteria: PCOS (according to Rotterdam consensus 2003), confirmed tubal patency

Exclusion criteria: male factor subfertility 

Baseline characteristics of each group: 

  • median  BMI: 30.48, 30.71

  • median fasting  insulin mIU/L: 17.20, 13.6

  • median total testosterone nmol/L: 2.35, 2.00

Dropouts: no significant different in the dropout rates, 10 in metformin + CC group and 7 in CC‐only group

Interventions

Main intervention: metformin 850 mg twice daily 

Duration: 6 weeks before and throughout ovulation induction with CC 

Co‐interventions: CC 50‐150 mg day 4‐8 for 4 cycles + lifestyle modification

Outcomes

Ovulation: day 21 progesterone level (level not stated)

Notes

A single‐centre RCT investigated the benefit of using metformin in CC ovulation induction treatment. ITT was used in our analysis. Participant lost to follow‐up classified as non‐responder; whilst pregnant participants did not attend follow‐up visit (one in each arm) were classified as responder

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random numbers

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

High risk

Unblinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: no significant difference in the dropout rates, 10 in metformin + CC group and 7 in CC‐only group

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Sturrock 2002

Methods

Cross‐over RCT

Setting: UK

Method of randomisation: performed by pharmacy*

Blinding: double‐blind

Number randomised: 19

Participants

Summary: obese PCOS, CC resistance

Inclusion criteria: oligomenorrhoea cycle > 40 d for 6 months, anovulation demonstrated by day 20‐22 progesterone ≤ 10 nmol/L, lack of response to CC 100 mg for 5 d with US showing endometrial thickness ≤ 5 mm and no ovarian follicle ≥ 14 mm. Age 18‐40 years

Exclusion criteria: raised prolactin, adrenal hyperplasia, thyroid dysfunction, medication known to affect insulin action*
Baseline characteristics of each group*:

  • mean age (± SD) 29.1 (4.3), 31.1 (3.7)

  • mean BMI (± SD) 34.2 (4.0), 35.0 (3.6)

  • mean fasting insulin mIU/L (± SD) 14.6 (9.9), 17.2 (8.0)

  • mean total testosterone mmol/L (± SD) 2.4 (0.8), 2.2 (0.4)

Dropouts: 4 (40%) from metformin arm and 4 (44%) from placebo arm*. Not included in analysis

Interventions

Main intervention: 1 of metformin 500 mg 3/d, placebo

Duration: 6 months

Co‐interventions: 1st week of treatment at 500 mg 1/d, 2nd at 500 mg 2/d and 3rd at 500 mg 3/d. Those that did not ovulate after 3 months had CC 50 mg days 2‐6, increased to 100 mg for a total of 3 cycles

Outcomes

Ovulation: by monthly serum progesterone (> 10 nmol/L) and presence of follicle ≥ 14 mm on ovarian US*

Anthropometric: weight, BMI, WHR

Reproductive hormones: total testosterone, FAI, SHBG

Metabolic markers: fasting glucose, fasting insulin, insulin resistance, beta‐cell function, systolic BP, diastolic BP

Others: pregnancy, menstrual cycle, Ferriman‐Gallwey score

Notes

This was designed as a cross‐over trial, with 6 months in the treatment/placebo arm followed by a 1‐month washout and then a 3‐month cross‐over. In this review, we only considered the first phase.

The inclusion criteria were simply for CC‐resistant anovulation and not specifically PCOS. However only 2 women did not have US criteria of PCOS, and 75% had a raised FAI*
In this review, only those participants who had a raised FAI were included in the analysis*

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Performed by pharmacy

Allocation concealment (selection bias)

Unclear risk

Performed by pharmacy

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 4 (40%) from metformin arm and 4 (44%) from placebo arm*. Not included in analysis

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Tang 2006

Methods

Multicentre RCT

Setting: UK

Method of randomisation: randomisation was performed by the research pharmacy department centrally. Using a random table, a block of 4 randomisation technique was employed in the study. Medications were supplied centrally from the research pharmacy department. The code was kept in the pharmacy department until the end of the trial period.

Blinding: double

Number randomised: 143

Participants

Summary: obese PCOS

Inclusion criteria: PCO on USS (> 10 cysts 2‐8 mm in diameter), oligomenorrhoea (cycle length > 35 d) or amenorrhoea (no period in 6 months)
Age between 18‐39 years
BMI > 30
Normal semen analysis and the participant should have at least 1 proven patent fallopian tube

Exclusion criteria: concurrent hormone therapy within previous 6 weeks, metabolic or chronic disease, renal or liver disease, diabetes, CAH, androgen‐secreting tumour

Baseline characteristics of each group:

  • mean age (SD) 29.7 (3.7), 29.8 (3.8)

  • mean BMI (SD) 37.6 (5.0), 38.9 (9.5)

  • mean fasting insulin mIU/L (SD) 16.3 (12.7), 17.4 (19.6)

  • mean total testosterone mmol/L (SD) 2.2 (0.6), 2.5 (0.64)

Dropouts: 11 (15.9%) in the metformin arm, 6 (8.1%). The difference was not significant.

Interventions

Main intervention: metformin 850 mg or placebo tablet once twice daily

Duration: 6 months

Co‐interventions: lifestyle modification (combination of diet and exercise) aiming to reduce 500 kcal/d

Outcomes

Anthropometric: BMI, weight, WHR, BP

Hormones: total testosterone, SHBG

Metabolic markers: insulin, glucose, total cholesterol, triglyceride

Others: menstrual pattern, pregnancy

Notes

A large multicentre randomised placebo controlled study was conducted to investigate the combined effects of the lifestyle modification and the use of metformin in obese women with PCOS (BMI > 30). A total of 8 centres in UK took part in the recruitment.

All the participants were recruited from the infertility clinics. The ethnic origin of the participants was not recorded.

Both the metformin and the placebo groups experienced improvement in weight loss and in menstrual pattern. However, the differences between the 2 groups were not significant. Participants in the metformin arm showed a greater reduction in total testosterone levels compared with women in the placebo arm.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The randomisation was performed by the research pharmacy department centrally. Using a random table, a block of 4 randomisation technique was employed in the study

Allocation concealment (selection bias)

Low risk

Medications were supplied centrally from the research pharmacy department. The code was kept in the pharmacy department until the end of the trial period

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 11 (15.9%) in the metformin arm, 6 (8.1%). The difference was not significant. Details of the dropout participants were not mentioned

Selective reporting (reporting bias)

Low risk

Primary outcome measure (menstrual frequency) and secondary outcome measures (metabolic parameters) were reported

Other bias

Unclear risk

Inadequate information

Trolle 2007

Methods

Cross‐over RCT

Setting: Denmark

Method of randomisation: random number table

Blinding: double

Number randomised: 60

Participants

Summary: obese PCOS

Inclusion criteria: Rotterdam criteria, age between 18‐45 years

Exclusion criteria: elevated serum gonadotrophins levels, hyperprolactinaemia, diabetes, abnormal thyroid, renal or liver functions, pregnancy, a wish for fertility treatment

Baseline characteristics of each group:

  • mean age 32 years

  • mean BMI 33.8 kg/m2  

  • mean fasting  insulin 9.50 mIU/L

  • mean total testosterone 2.89 mmol/L

Dropouts: 2 in each group

Interventions

Main intervention: metformin 850 mg or placebo twice daily

Duration: 6 months

Co‐interventions: no

Outcomes

Anthropometric: weight, systolic BP*

Hormones: testosterone*

Metabolic markers: insulin, glucose, HDL*

Notes

This was a single‐centre randomised, double blinded, placebo controlled cross‐over study to assess the effects of metformin on menstrual frequency and metabolic parameters. Women were randomised to receive either metformin or placebo tablets for 6 months. After a 3‐month wash‐out period, the women received the alternate treatment. 

Women who wished for fertility treatment were excluded.

*Information that was not in the original article kindly provided by the study author.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random number table

Allocation concealment (selection bias)

Unclear risk

The randomisation code stored in a closed envelope until the end of recruitment. Identical trial drug and placebo tablet

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blinded

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 dropouts in each group. Similar dropout rates. Baseline characteristics were comparable between the dropout and the completed groups

Selective reporting (reporting bias)

Low risk

A clear study protocol. Power calculation reported. Primary outcome (menstrual frequency) reported

Other bias

High risk

Initial power calculation indicated minimum of 50 participants in the trial. However, due to an increased dropout rate, the number of recruitment subsequently increased to 60.

Vandermolen 2001

Methods

Multicentre RCT

Setting: USA

Method of randomisation: computer generation in blocks of 6

Blinding: double‐blind

Number randomised: 27

Participants

Summary: obese PCOS, CC resistance

Inclusion criteria: PCOS (oligomenorrhoea < 6 cycles/year, anovulation with CC 150 mg for 5 d confirmed by progesterone < 4 ng/mL or amenorrhoea by day 35, hyperandrogaenemia (elevated androstenedione, free testosterone or total testosterone)* or hirsutism, exclusion of other endocrinopathy, US findings of PCO; age 18‐35; normal semen analysis; tubal patency if previous pelvic surgery or infection

Exclusion criteria: diabetes mellitus, adrenal hyperplasia, thyroid dysfunction, hyperprolactinaemia, abnormal renal or liver function, medication known to affect insulin action*

Baseline characteristics of each group:

  • mean age (± SD) 29 (4.0), 30 (3.7)

  • mean BMI (± SD) 37.6 (14.3), 38.4 (8.2)

  • mean fasting insulin mIU/L (± SD) 8.9 (6.0), 12.5 (7.1)

  • mean total testosterone nmol/L (± SD) 2.90 (0.8), 3.04 (1.42)

Dropouts: 1 from each arm (7%); 1 in the placebo arm ovulated in response to CC but was excluded owing to non‐compliance. Not included in analysis

Interventions

Main intervention: 1 of metformin 500 mg 3/d, placebo

Duration: 7 weeks initially, then those who did not ovulate continued for a further 6 cycles

Co‐interventions: those that did not ovulate after 7 weeks had CC 50 mg for 5 d. If ovulation did not occur the dose was increased to 100 mg then 150 mg for a total of 6 cycles

No change in usual eating habits, physical activity or lifestyle

Outcomes

Ovulation: serum progesterone ≥ 12.7 nmol/L on days 10, 20, 30 and 40 (and days 21 and 28 of subsequent cycles if received CC)

Anthropometric: weight, BMI

Reproductive Hormones: total testosterone, free testosterone, androstenedione, DHEAS, SHBG, estradiol, FSH, LH, 17‐alpha hydroxyprogesterone

Metabolic markers: fasting glucose, fasting insulin, AUC of insulin and glucose during GTT

Others: live birth, pregnancy

Notes

Although obesity was not an inclusion criteria, the mean BMI was high in this study although similar in both arms.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer generation in blocks of 6

Allocation concealment (selection bias)

Unclear risk

Not stated

Blinding (performance bias and detection bias)
All outcomes

Low risk

Double‐blind

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Dropouts: 1 from each arm (7%); 1 in the placebo arm ovulated in response to CC but was excluded owing to non‐compliance. Not included in analysis. Details not provided

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Williams 2009

Methods

RCT

Setting: USA

Method of randomisation: not stated

Blinding: double‐blind

Number randomised: 55

Participants

Summary: PCOS

Inclusion criteria: not stated. Unknown BMI and age

Exclusion criteria: unknown

Baseline characteristics of each group: not stated

Dropouts: not stated

26 women underwent 99 blinded treatment cycles whilst 29 women underwent 88 blinded treatment cycles

Interventions

Main intervention: metformin 500 mg or placebo 3/d

Duration: 6 cycles

Co‐interventions: CC (dose unclear)

Outcomes

Ovulation

Pregnancy

Notes

A conference abstract presented in 57th Annual Meeting of The Pacific Coast Reproductive Society 2009.

No reply from study author regarding the detail of the study

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Inadequate information

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Inadequate information

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Yarali 2002

Methods

RCT

Setting: Turkey

Method of randomisation: computer‐generated numbers. Centralised randomisation process*

Blinding: double‐blind

Number randomised: 32

Participants

Summary: non‐obese PCOS, CC resistance

Inclusion criteria: PCOS (oligomenorrhoea < 6 cycles/year, anovulation confirmed with progesterone < 5 ng/mL, testosterone > 2.4 nmol/L, exclusion of other endocrinopathy, US findings of PCO, CC resistance to 250 mg for 5 d for up to 6 months, normal semen analysis, normal HSG or laparoscopy within 6 months

Exclusion criteria: diabetes mellitus, adrenal hyperplasia, Cushing's syndrome, thyroid dysfunction, hyperprolactinaemia, medication known to alter insulin action, previous gonadotrophin treatment, infertility other than that caused by PCOS, previous pelvic surgery

Baseline characteristics of each group:

  • mean age (± SD) 29.7 (5.6), 28.4 (5.1)

  • mean BMI (± SD) 28.6 (4.0), 29.6 (4.8)

  • mean fasting insulin mIU/L (± SD) 15.5 (21.4), 11 (5.5)

  • mean total testosterone mmol/L (± SD) 6.19 (3.57), 6.01 (2.93)

Dropouts: 2 (6%) from the metformin/placebo part of the trial owing to pregnancy. They were excluded from analysis

Interventions

Main intervention: 1 of metformin 850 mg 2/d, placebo

Duration: 6 weeks initially, then those who did not ovulate continued for1 cycle

Co‐interventions: those that did not ovulate after 6 weeks had recombinant FSH in a low‐dose, step‐up protocol

No change in usual eating habits

Outcomes

Ovulation: serum progesterone > 15.9 nmol/L weekly

Anthropometric: BMI, WHR

Reproductive hormones: total testosterone, free testosterone, androstenedione, DHEAS, estradiol, FSH, LH, 17‐alpha hydroxyprogesterone

Metabolic markers: fasting insulin, AUC insulin and glucose during GTT, insulin sensitivity, leptin,

Others: live birth, adverse events, pregnancy, duration of rFSH stimulation, total dose of FSH, oestradiol on day of hCG, monofollicular development
cycle cancellation rate

Notes

Free testosterone was significantly higher in the metformin group. Fasting insulin was non‐significantly higher with a wide SD compared with placebo.

*Information not in the original paper kindly provided by the study author

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated numbers. Centralised randomisation process*

Allocation concealment (selection bias)

Unclear risk

Inadequate information

Blinding (performance bias and detection bias)
All outcomes

Unclear risk

Inadequate information

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Dropouts: 2 (6%) from the metformin/placebo part of the trial owing to pregnancy. They were excluded from analysis

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Insufficient information

Zain 2009

Methods

RCT

Setting: Malaysia 

Method of randomisation: picking a card out of a box 

Blinding: no 

Number randomised: 124

Participants

Summary: obese PCOS 

Inclusion criteria: newly diagnosed with PCOS (Rotterdam criteria), age < 40 years

Exclusion criteria: diabetes, hepatic or renal dysfunction, heart disease, abnormal semen analysis (WHO criteria) 

Baseline characteristics of each group:

  • mean age (SD) 27.8 (3.6), 29.6 (4.3), 29.3 (4.9)

  • mean BMI (SD) 33.9 (3.6), 32.9 (4.2), 33.0 (4.1)

  • mean fasting insulin mIU/L (SD)

  • mean total testosterone nmol/L (SD) 0.57 (0.1), 0.41 (0.45), 0.77 (0.14) 

Dropouts: 4 (9.5%) in the metformin group, 2 (4.9%) in the CC group and 3 (7.3%) in the combined metformin and CC group

Interventions

Main intervention: metformin 1500 mg/d 

Duration: 6 months 

Co‐interventions: CC 50 mg from day 2‐6 of the cycle. If women did not respond to the treatment, the dose increased by 50 mg to a maximum dose of 200 mg

All the women were offered dietary advice.

Outcomes

Ovulation: USS follicular tracking

Hormones: testosterone

Others: live birth, pregnancy, miscarriage

Notes

This study was designed to compare the live birth rates in women who received CC, metformin and combined CC and metformin treatments. Placebo tablets were not used in this unblinded RCT. Therefore, potential bias may be introduced. 

Most women were Malay (about 90%)

Analysis was based on analysis per protocol, not ITT

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Picking a card out of a box

Allocation concealment (selection bias)

Unclear risk

Picking a card out of a box

Blinding (performance bias and detection bias)
All outcomes

High risk

Unblinded

Incomplete outcome data (attrition bias)
All outcomes

High risk

Dropouts: 4 (9.5%) in the metformin group, 2 (4.9%) in the CC group and 3 (7.3%) in the combined metformin and CC group. Analysis was based on analysis per protocol, not ITT

Selective reporting (reporting bias)

Unclear risk

Insufficient information in the study

Other bias

Unclear risk

Inadequate information

Baseline characteristics given in order of main intervention (drug, placebo).

Where the trial protocol included a statement such as, "all patients had ultrasound features of PCOS" then this has been included as an inclusion criteria (unless the authors specifically state that it was not in which case it is recorded under notes).

Abbreviations Table 1:

ACTH:
AUC: area under the curve
BMI: body mass index
BP: blood pressure
CAH:
CC: clomiphene citrate
CI: confidence interval
CT: computerised tomography scan
DHEAS: dehydroepiandrosterone sulphate
FAI: free androgen index
FSH: follicle stimulating hormone
GTT: glucose tolerance test
HbA1C: glycosylated haemoglobin
HDL: high density lipoprotein cholesterol
IGFBP‐1: insulin growth factor binding protein 1
ITT: intention‐to‐treat
LDL: low density lipoprotein cholesterol
LFT:
LH: luteinising hormone
OGTT:
RCT: randomised controlled trial
rFSH: recombinant follicle stimulating hormone
PCOS: polycystic ovary syndrome
PID:
SD: standard deviation
SE: standard error of the mean
SHBG: sex hormone‐binding globulin
TFT: thyroid function test
TSH:
US(S): ultrasound (scan)
VLDL: very low density lipoprotein cholesterol
WHO: World Health Organization
WHR: waist:hip ratio

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Abuelghar 2013

Human chorionic gonadotrophin hormone was used an an ovulation trigger, which may have added additional heterogeneity to the results. Reasons for losses to follow up not given. Not intention to treat analysis

Aroda 2009

The aim of the study was to evaluate the effects of pioglitazone on insulin action and ovarian androgen production in women with PCOS. This was a randomised, placebo‐controlled trial but the details of randomisation were not provided. Furthermore, the recruited participants did not have history of subfertility

Azziz 2001

Randomised, double‐blind study comparing troglitazone (150 mg, 300 mg, 600 mg daily) with placebo.

Randomisation method was unclear and no reply from the study author.

Troglitazone has been withdrawn from the market due to risk of hepatic damage

Azziz 2003

Randomised, double‐blind study comparing troglitazone (150 mg, 300 mg, 600 mg daily) with placebo.

Randomisation method was unclear and no reply from the study author.

Troglitazone has been withdrawn from the market due to risk of hepatic damage.

Chaudhry 2016

This study compared 2 insulin‐sensitising agents, which does not meet the inclusion criteria for this review

Chaudhury 2008

This study compared the efficacy of metformin with folic acid

Constantino 2009

This study compared the efficacy of metformin with folic acid

Crave 1995

Randomised, double‐blind trial comparing metformin 850 mg 2/d with placebo

Participants were women with hirsutism and obesity but not necessarily anovulation. 67% had regular menses. 63% had polycystic ovaries on US.

The results indicated that weight loss induced by a hypocalorific diet led to improvements in insulin and androgen levels, but that metformin gave no additional benefit over diet

Curi 2012

The aim of this study was to ascertain the effects of metformin on ovarian function. The outcomes of this review, such as ovulation rate, pregnancy or live birth rate were not measured as contraception was advised during the study

De Leo 1999

Randomised trial in women with CC‐resistant PCOS having 75 IU FSH for ovulation induction, comparing pre‐treatment with metformin 500 mg 3/d with no metformin pre‐treatment.

The aims and outcome measures were different from the other included trials (main outcome measures were number of FSH ampoules, days of treatment and markers of ovarian hyperstimulation). The trial reported treatment cycles rather than participants, and combined the results of each group in a cross‐over type analysis. Therefore the data were not suitable for inclusion in this meta‐analysis

Dunaif 1996

Randomised double blind trial in women with PCOS comparing troglitazone 200mg and troglitazone 400mg daily.

This trial only randomised for dose of troglitazone, and did not have a placebo or no treatment arm

Elter 2002

Randomised trial in non‐obese women with PCOS comparing metformin and the combined oral contraceptive (ethinyl estradiol/cyproterone acetate) with the combined contraceptive alone.

This trial did not compare metformin with placebo, no treatment or an ovulation‐induction agent

Farzadi 2006

In this study, the efficacy of metformin was compared with vitamin B, and we were unable to contact the study author

Heathcote 2013

No record of this publication

Hou 2000

Non‐English‐language trial in women with CC resistant PCOS, comparing metformin with the Chinese herbal formula tiangui fang.

The paper makes no mention of randomisation and has therefore been classified as a controlled clinical trial. Ovulation was assessed by menstrual cyclicity and basal body temperature change but not by a biochemical method.

This trial did not have a placebo or no‐treatment arm, and the only significant result reported was a reduction in testosterone and BMI in the tiangui fang arm compared with baseline

Ibanez 2002

Randomised trial in lean, young women with anovulation, hyperinsulinaemia, and hyperandrogaenemia. It had 3 arms: metformin only, flutamide only and metformin and flutamide together.

This trial was not included because it had no placebo arm, and the anti‐androgen flutamide is not an ovulation‐induction agent

Kazerooni 2009

This study evaluated the effect of short‐course pretreatment with metformin on hyperandrogenism, insulin resistance, cervical scores and pregnancy rates in women with CC‐resistant PCOS

Apart from receiving CC treatment, all participants received 10,000 U of hCG injection to stimulate ovulation followed by timed intercourse. Hence, all women received 2 ovulation‐induction agents per cycle of treatment. Therefore, it would not be appropriate to combine these subjects in the current review as all the included trial participants only received 1 type of ovulation‐induction agent with or without metformin

Kelly 2002

This study was published in 2002. The objective was to ascertain the effect of metformin on hirsutism in women with PCOS. This outcome measure has been removed in the update review

Kocak 2002

Quasi‐randomised trial comparing combined CC and metformin with CC on ovulation in CC‐resistant women with PCOS.

Inadequate randomisation and sequence generation (sequential by order of admission). Admission determined by day of menses. Allocation performed by nurse blinded to the study. Odd numbers allocated metformin, even numbers allocated placebo

Ladson 2011

In this RCT women were advised to avoid pregnancy, so the outcomes of interest in the review were not investigated

Leanza 2014

Patients in this study underwent intrauterine insemination and assisted reproduction is an exclusion criteria for this review. Aspects of the methodology are missing from the article

Mantzoros 1997

Randomised, double‐blind study in women with PCOS comparing troglitazone 200 mg and troglitazone 400 mg daily.

This trial only randomised for dose of troglitazone, and did not have a placebo or no‐treatment arm

Morin‐Papunen 2000

Randomised trial in obese women with PCOS comparing metformin 500 mg 2/d and 1 g 2/d with combined oral contraceptive (ethinyl estradiol/cyproterone acetate).

This trial was not blinded and did not compare metformin with placebo, no treatment or an ovulation‐induction agent

Morin‐Papunen 2010

Conference abstract. Not enough information to separate the data for analysis. Data from a published paper by the same author is included in the 3rd update of this review

Nestler 1996

This study investigated whether hyperinsulinaemia stimulates ovarian cytochrome P450c17α activity in women with PCOS. Some of the participants were not infertile

Nestler 1997

Partially randomised trial in lean women with PCOS comparing metformin 500 mg 3/d with placebo.

The method of randomisation was initially by pulling pieces out of a hat, but then continued as an observational study. The trial was initially single‐blind, with the patient blinded

The published data included both randomised and non‐randomised participants, and an analysis to include only the randomised participants was not possible

Palomba 2011

Participants in this study underwent assisted conception using IVF

Papaleo 2009

This study investigated the effect of insulin sensitisers on oocyte quality in IVF cycles, and therefore does not meet the inclusion criteria for this review

Ramzy 2003

An open‐labelled, randomised trial comparing metformin 500 mg 3/d with placebo 6 weeks prior to CC treatment. In addition, randomisation was performed using alternate numbers. These factors introduced significant bias.

Refaie 2005

Attempts to contact study author for more information unsuccessful

Rouzi 2006

Randomised trial comparing CC and metformin with CC 1.5 g and rosiglitazone 4 mg in CC‐resistant women with PCOS.

This trial did not compare metformin/CC with CC/placebo

Salman 2014

This was a conference abstract only, with not enough detail to warrant inclusion. Literature search found no subsequent publication

Santonocito 2009

The objective of this study was to compare CC with metformin on ovulation rates. However, all participants also received 2000 U of hCG injection once follicular diameter > 15 mm on USS

Shobokshi 2003

The objective of this study was to compare the effects of combined rosiglitazone and CC with CC monotherapy. Since placebo was not employed in the trial, both the clinician and participants were not blinded. Therefore, bias may exist in this study.

It was unclear whether the study was randomised. We are currently still waiting for a response from the study author

Unfer 2011

This study investigated the effect of insulin sensitisers on oocyte quality in IVF cycles, and therefore does not meet the inclusion criteria for this review

CC: clomiphene citrate; FSH: follicle‐stimulating hormone; IVF: in vitro fertilisation; PCOS: polycystic ovary syndrome; RCT: randomised controlled trial; US(S): ultrasound (scan)

Data and analyses

Open in table viewer
Comparison 1. Metformin versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Live birth rate Show forest plot

4

435

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.00, 2.51]

Analysis 1.1

Comparison 1 Metformin versus placebo or no treatment, Outcome 1 Live birth rate.

Comparison 1 Metformin versus placebo or no treatment, Outcome 1 Live birth rate.

1.1 Participants with BMI < 30 kg/m2

3

370

Odds Ratio (M‐H, Fixed, 95% CI)

1.51 [0.94, 2.44]

1.2 Participants with BMI ≥ 30 kg/m2

1

65

Odds Ratio (M‐H, Fixed, 95% CI)

2.87 [0.51, 16.01]

2 Adverse events (gastrointestinal side effects) Show forest plot

7

670

Odds Ratio (M‐H, Fixed, 95% CI)

4.76 [3.06, 7.41]

Analysis 1.2

Comparison 1 Metformin versus placebo or no treatment, Outcome 2 Adverse events (gastrointestinal side effects).

Comparison 1 Metformin versus placebo or no treatment, Outcome 2 Adverse events (gastrointestinal side effects).

2.1 Participants with BMI < 30 kg/m2

4

393

Odds Ratio (M‐H, Fixed, 95% CI)

5.61 [2.89, 10.88]

2.2 Participants with BMI ≥ 30 kg/m2

3

277

Odds Ratio (M‐H, Fixed, 95% CI)

4.13 [2.28, 7.49]

3 Clinical pregnancy rate Show forest plot

9

1027

Odds Ratio (M‐H, Fixed, 95% CI)

1.93 [1.42, 2.64]

Analysis 1.3

Comparison 1 Metformin versus placebo or no treatment, Outcome 3 Clinical pregnancy rate.

Comparison 1 Metformin versus placebo or no treatment, Outcome 3 Clinical pregnancy rate.

3.1 Participants with BMI < 30 kg/m2

5

733

Odds Ratio (M‐H, Fixed, 95% CI)

1.89 [1.35, 2.65]

3.2 Participants with BMI ≥ 30 kg/m2

4

294

Odds Ratio (M‐H, Fixed, 95% CI)

2.21 [0.98, 4.98]

4 Ovulation rate Show forest plot

14

701

Odds Ratio (M‐H, Fixed, 95% CI)

2.55 [1.81, 3.59]

Analysis 1.4

Comparison 1 Metformin versus placebo or no treatment, Outcome 4 Ovulation rate.

Comparison 1 Metformin versus placebo or no treatment, Outcome 4 Ovulation rate.

4.1 Participants with BMI < 30 kg/m2

5

229

Odds Ratio (M‐H, Fixed, 95% CI)

4.15 [2.31, 7.45]

4.2 Participants with BMI ≥ 30 kg/m2

10

472

Odds Ratio (M‐H, Fixed, 95% CI)

1.96 [1.28, 3.01]

5 Menstrual frequency Show forest plot

7

427

Odds Ratio (M‐H, Fixed, 95% CI)

1.72 [1.14, 2.61]

Analysis 1.5

Comparison 1 Metformin versus placebo or no treatment, Outcome 5 Menstrual frequency.

Comparison 1 Metformin versus placebo or no treatment, Outcome 5 Menstrual frequency.

5.1 Participants with BMI < 30 kg/m2

1

23

Odds Ratio (M‐H, Fixed, 95% CI)

21.15 [1.01, 445.00]

5.2 Participants with BMI ≥ 30 kg/m2

6

404

Odds Ratio (M‐H, Fixed, 95% CI)

1.57 [1.03, 2.41]

6 Miscarriage rate per woman Show forest plot

4

748

Odds Ratio (M‐H, Fixed, 95% CI)

1.08 [0.50, 2.35]

Analysis 1.6

Comparison 1 Metformin versus placebo or no treatment, Outcome 6 Miscarriage rate per woman.

Comparison 1 Metformin versus placebo or no treatment, Outcome 6 Miscarriage rate per woman.

6.1 Participants with BMI < 30 kg/m2

3

683

Odds Ratio (M‐H, Fixed, 95% CI)

1.19 [0.52, 2.71]

6.2 Participants with BMI ≥ 30 kg/m2

1

65

Odds Ratio (M‐H, Fixed, 95% CI)

0.5 [0.04, 5.80]

7 Sensitivity analysis: miscarriage rate per pregnancy Show forest plot

4

200

Odds Ratio (M‐H, Fixed, 95% CI)

0.58 [0.25, 1.34]

Analysis 1.7

Comparison 1 Metformin versus placebo or no treatment, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.

Comparison 1 Metformin versus placebo or no treatment, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.

7.1 Participants with BMI < 30 kg/m2

3

188

Odds Ratio (M‐H, Fixed, 95% CI)

0.63 [0.26, 1.53]

7.2 Participants with BMI ≥ 30 kg/m2

1

12

Odds Ratio (M‐H, Fixed, 95% CI)

0.25 [0.02, 4.00]

8 Body mass index (kg/m2) Show forest plot

16

827

Mean Difference (IV, Fixed, 95% CI)

‐0.08 [‐0.33, 0.17]

Analysis 1.8

Comparison 1 Metformin versus placebo or no treatment, Outcome 8 Body mass index (kg/m2).

Comparison 1 Metformin versus placebo or no treatment, Outcome 8 Body mass index (kg/m2).

8.1 Participants with BMI < 30 kg/m2

7

419

Mean Difference (IV, Fixed, 95% CI)

‐0.06 [‐0.33, 0.21]

8.2 Participants with BMI ≥ 30 kg/m2

10

408

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐0.92, 0.52]

9 Waist‐hip ratio Show forest plot

11

702

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.01, ‐0.00]

Analysis 1.9

Comparison 1 Metformin versus placebo or no treatment, Outcome 9 Waist‐hip ratio.

Comparison 1 Metformin versus placebo or no treatment, Outcome 9 Waist‐hip ratio.

9.1 Participants with BMI < 30 kg/m2

5

389

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.01, ‐0.00]

9.2 Participants with BMI ≥ 30 kg/m2

6

313

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.02, 0.01]

10 Blood pressure ‐ systolic (mm Hg) Show forest plot

7

379

Mean Difference (IV, Fixed, 95% CI)

‐3.59 [‐5.13, ‐2.04]

Analysis 1.10

Comparison 1 Metformin versus placebo or no treatment, Outcome 10 Blood pressure ‐ systolic (mm Hg).

Comparison 1 Metformin versus placebo or no treatment, Outcome 10 Blood pressure ‐ systolic (mm Hg).

10.1 Participants with BMI < 30 kg/m2

3

96

Mean Difference (IV, Fixed, 95% CI)

‐3.52 [‐5.29, ‐1.76]

10.2 Participants with BMI ≥ 30 kg/m2

5

283

Mean Difference (IV, Fixed, 95% CI)

‐3.80 [‐5.00, ‐0.60]

11 Blood pressure ‐ diastolic (mm Hg) Show forest plot

6

292

Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐1.35, 1.07]

Analysis 1.11

Comparison 1 Metformin versus placebo or no treatment, Outcome 11 Blood pressure ‐ diastolic (mm Hg).

Comparison 1 Metformin versus placebo or no treatment, Outcome 11 Blood pressure ‐ diastolic (mm Hg).

11.1 Participants with BMI < 30 kg/m2

3

96

Mean Difference (IV, Fixed, 95% CI)

‐0.21 [‐1.55, 1.13]

11.2 Participants with BMI ≥ 30 kg/m2

4

196

Mean Difference (IV, Fixed, 95% CI)

0.18 [‐2.65, 3.02]

12 Serum testosterone (nmol/L) Show forest plot

15

863

Mean Difference (IV, Fixed, 95% CI)

‐0.49 [‐0.59, ‐0.39]

Analysis 1.12

Comparison 1 Metformin versus placebo or no treatment, Outcome 12 Serum testosterone (nmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 12 Serum testosterone (nmol/L).

12.1 Participants with BMI < 30 kg/m2

7

419

Mean Difference (IV, Fixed, 95% CI)

‐0.71 [‐0.86, ‐0.56]

12.2 Participants with BMI ≥ 30 kg/m2

9

444

Mean Difference (IV, Fixed, 95% CI)

‐0.29 [‐0.44, ‐0.15]

13 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

15

823

Mean Difference (IV, Fixed, 95% CI)

0.49 [‐1.82, 2.81]

Analysis 1.13

Comparison 1 Metformin versus placebo or no treatment, Outcome 13 Serum sex hormone‐binding globulin (nmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 13 Serum sex hormone‐binding globulin (nmol/L).

13.1 Participants with BMI < 30 kg/m2

6

387

Mean Difference (IV, Fixed, 95% CI)

‐0.22 [‐6.73, 6.28]

13.2 Participants with BMI ≥ 30 kg/m2

10

436

Mean Difference (IV, Fixed, 95% CI)

0.60 [‐1.88, 3.07]

14 Fasting glucose (mmol/L) Show forest plot

15

849

Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐0.21, ‐0.07]

Analysis 1.14

Comparison 1 Metformin versus placebo or no treatment, Outcome 14 Fasting glucose (mmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 14 Fasting glucose (mmol/L).

14.1 Participants with BMI < 30 kg/m2

5

364

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.22, ‐0.04]

14.2 Participants with BMI ≥ 30 kg/m2

11

485

Mean Difference (IV, Fixed, 95% CI)

‐0.16 [‐0.27, ‐0.05]

15 Fasting insulin (mIU/L) Show forest plot

14

573

Mean Difference (IV, Fixed, 95% CI)

‐4.13 [‐5.67, ‐2.58]

Analysis 1.15

Comparison 1 Metformin versus placebo or no treatment, Outcome 15 Fasting insulin (mIU/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 15 Fasting insulin (mIU/L).

15.1 Participants with BMI < 30 kg/m2

4

85

Mean Difference (IV, Fixed, 95% CI)

‐6.20 [‐8.56, ‐3.84]

15.2 Participants with BMI ≥ 30 kg/m2

11

488

Mean Difference (IV, Fixed, 95% CI)

‐2.57 [‐4.62, ‐0.53]

16 Total cholesterol (mmol/L) Show forest plot

10

562

Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐0.31, 0.02]

Analysis 1.16

Comparison 1 Metformin versus placebo or no treatment, Outcome 16 Total cholesterol (mmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 16 Total cholesterol (mmol/L).

16.1 Participants with BMI < 30 kg/m2

5

276

Mean Difference (IV, Fixed, 95% CI)

‐0.02 [‐0.26, 0.22]

16.2 Participants with BMI ≥ 30 kg/m2

6

286

Mean Difference (IV, Fixed, 95% CI)

‐0.26 [‐0.48, ‐0.03]

17 Triglyceride levels (mmol/L) Show forest plot

7

309

Mean Difference (IV, Fixed, 95% CI)

0.14 [‐0.05, 0.32]

Analysis 1.17

Comparison 1 Metformin versus placebo or no treatment, Outcome 17 Triglyceride levels (mmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 17 Triglyceride levels (mmol/L).

17.1 Participants with BMI < 30 kg/m2

3

53

Mean Difference (IV, Fixed, 95% CI)

0.00 [‐0.33, 0.34]

17.2 Participants with BMI ≥ 30 kg/m2

5

256

Mean Difference (IV, Fixed, 95% CI)

0.20 [‐0.02, 0.42]

Open in table viewer
Comparison 2. Metformin combined with clomiphene citrate versus clomiphene citrate alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Live birth rate Show forest plot

9

1079

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.92, 1.59]

Analysis 2.1

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 1 Live birth rate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 1 Live birth rate.

1.1 Participants with BMI < 30 kg/m2 or ≤ 32 kg/m2

5

531

Odds Ratio (M‐H, Fixed, 95% CI)

1.14 [0.78, 1.67]

1.2 Participants with BMI ≥ 30 kg/m2

4

548

Odds Ratio (M‐H, Fixed, 95% CI)

1.28 [0.86, 1.91]

2 Adverse events Show forest plot

3

591

Odds Ratio (M‐H, Fixed, 95% CI)

3.97 [2.59, 6.08]

Analysis 2.2

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 2 Adverse events.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 2 Adverse events.

2.1 Participants with BMI < 30 kg/m2

3

591

Odds Ratio (M‐H, Fixed, 95% CI)

3.97 [2.59, 6.08]

3 Clinical pregnancy rate Show forest plot

16

1529

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.27, 1.99]

Analysis 2.3

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 3 Clinical pregnancy rate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 3 Clinical pregnancy rate.

3.1 Participants with BMI < 30 kg/m2

9

834

Odds Ratio (M‐H, Fixed, 95% CI)

1.46 [1.08, 1.98]

3.2 Participants with BMI ≥ 30 kg/m2

7

695

Odds Ratio (M‐H, Fixed, 95% CI)

1.76 [1.26, 2.47]

4 Ovulation rate Show forest plot

21

1624

Odds Ratio (M‐H, Fixed, 95% CI)

1.57 [1.28, 1.92]

Analysis 2.4

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 4 Ovulation rate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 4 Ovulation rate.

4.1 BMI < 30 kg/m2

11

755

Odds Ratio (M‐H, Fixed, 95% CI)

1.79 [1.32, 2.41]

4.2 BMI ≥ 30 kg/m2

9

814

Odds Ratio (M‐H, Fixed, 95% CI)

1.52 [1.15, 2.01]

4.3 BMI not reported

1

55

Odds Ratio (M‐H, Fixed, 95% CI)

0.43 [0.13, 1.37]

5 Ovulation rate: subgroup analysis by sensitivity to clomiphene citrate Show forest plot

7

271

Odds Ratio (M‐H, Fixed, 95% CI)

4.69 [2.61, 8.44]

Analysis 2.5

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 5 Ovulation rate: subgroup analysis by sensitivity to clomiphene citrate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 5 Ovulation rate: subgroup analysis by sensitivity to clomiphene citrate.

5.1 PCOS and clomiphene‐sensitive

1

56

Odds Ratio (M‐H, Fixed, 95% CI)

3.55 [0.65, 19.37]

5.2 PCOS and clomiphene‐resistant

6

215

Odds Ratio (M‐H, Fixed, 95% CI)

4.89 [2.62, 9.13]

6 Miscarriage rate per woman Show forest plot

9

1096

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.03, 2.46]

Analysis 2.6

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 6 Miscarriage rate per woman.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 6 Miscarriage rate per woman.

6.1 Participants with BMI < 30 kg/m2

5

548

Odds Ratio (M‐H, Fixed, 95% CI)

1.41 [0.76, 2.62]

6.2 Participants with BMI ≥ 30 kg/m2

4

548

Odds Ratio (M‐H, Fixed, 95% CI)

1.79 [0.97, 3.32]

7 Sensitivity analysis: miscarriage rate per pregnancy Show forest plot

8

400

Odds Ratio (M‐H, Fixed, 95% CI)

1.30 [0.80, 2.12]

Analysis 2.7

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.

7.1 Participants with BMI < 30 kg/m2

4

228

Odds Ratio (M‐H, Fixed, 95% CI)

1.27 [0.65, 2.51]

7.2 Participants with BMI ≥ 30 kg/m2

4

172

Odds Ratio (M‐H, Fixed, 95% CI)

1.34 [0.67, 2.68]

8 Multiple pregnancy rate per woman Show forest plot

6

1003

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.18, 1.68]

Analysis 2.8

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 8 Multiple pregnancy rate per woman.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 8 Multiple pregnancy rate per woman.

8.1 Participants with BMI < 30 kg/m2

3

476

Odds Ratio (M‐H, Fixed, 95% CI)

0.50 [0.12, 2.04]

8.2 Participants with BMI ≥ 30kg/m2

3

527

Odds Ratio (M‐H, Fixed, 95% CI)

0.66 [0.11, 4.01]

9 Senstivity analysis: multiple pregnancy rate per pregnancy Show forest plot

6

342

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.15, 1.42]

Analysis 2.9

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 9 Senstivity analysis: multiple pregnancy rate per pregnancy.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 9 Senstivity analysis: multiple pregnancy rate per pregnancy.

9.1 Participants with BMI < 30 kg/m2

3

178

Odds Ratio (M‐H, Fixed, 95% CI)

0.43 [0.10, 1.85]

9.2 Participants with BMI ≥ 30 kg/m2

3

164

Odds Ratio (M‐H, Fixed, 95% CI)

0.50 [0.08, 3.12]

Open in table viewer
Comparison 3. Metformin versus clomiphene citrate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Live birth Show forest plot

5

741

Odds Ratio (M‐H, Fixed, 95% CI)

0.71 [0.49, 1.01]

Analysis 3.1

Comparison 3 Metformin versus clomiphene citrate, Outcome 1 Live birth.

Comparison 3 Metformin versus clomiphene citrate, Outcome 1 Live birth.

1.1 Participants with BMI < 30 kg/m2

3

241

Odds Ratio (M‐H, Fixed, 95% CI)

1.71 [1.00, 2.94]

1.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.30 [0.17, 0.52]

2 Clinical pregnancy rate Show forest plot

7

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Metformin versus clomiphene citrate, Outcome 2 Clinical pregnancy rate.

Comparison 3 Metformin versus clomiphene citrate, Outcome 2 Clinical pregnancy rate.

2.1 Participants with BMI < 30 kg/m2

5

490

Odds Ratio (M‐H, Fixed, 95% CI)

1.56 [1.05, 2.33]

2.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.34 [0.21, 0.55]

3 Ovulation rate Show forest plot

6

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.3

Comparison 3 Metformin versus clomiphene citrate, Outcome 3 Ovulation rate.

Comparison 3 Metformin versus clomiphene citrate, Outcome 3 Ovulation rate.

3.1 Participants with BMI < 30 kg/m2

4

312

Odds Ratio (M‐H, Fixed, 95% CI)

0.81 [0.51, 1.28]

3.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.29 [0.20, 0.43]

4 Miscarriage rate per woman Show forest plot

5

741

Odds Ratio (M‐H, Fixed, 95% CI)

0.92 [0.50, 1.67]

Analysis 3.4

Comparison 3 Metformin versus clomiphene citrate, Outcome 4 Miscarriage rate per woman.

Comparison 3 Metformin versus clomiphene citrate, Outcome 4 Miscarriage rate per woman.

4.1 Participants with BMI < 30 kg/m2

3

241

Odds Ratio (M‐H, Fixed, 95% CI)

1.58 [0.61, 4.09]

4.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.61 [0.27, 1.38]

5 Sensitivity analysis: miscarriage rate per pregnancy Show forest plot

5

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.5

Comparison 3 Metformin versus clomiphene citrate, Outcome 5 Sensitivity analysis: miscarriage rate per pregnancy.

Comparison 3 Metformin versus clomiphene citrate, Outcome 5 Sensitivity analysis: miscarriage rate per pregnancy.

5.1 Participants with BMI < 30 kg/m2

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Participants with BMI ≥ 30 kg/m2

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Multiple pregnancy rate per woman Show forest plot

5

858

Odds Ratio (M‐H, Fixed, 95% CI)

0.29 [0.06, 1.43]

Analysis 3.6

Comparison 3 Metformin versus clomiphene citrate, Outcome 6 Multiple pregnancy rate per woman.

Comparison 3 Metformin versus clomiphene citrate, Outcome 6 Multiple pregnancy rate per woman.

6.1 Participants with BMI < 30 kg/m2

3

358

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.07, 3.16]

6.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.14 [0.01, 2.76]

7 Sensitivity analysis: multiple pregnancy rate per pregnancy Show forest plot

5

201

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.06, 1.68]

Analysis 3.7

Comparison 3 Metformin versus clomiphene citrate, Outcome 7 Sensitivity analysis: multiple pregnancy rate per pregnancy.

Comparison 3 Metformin versus clomiphene citrate, Outcome 7 Sensitivity analysis: multiple pregnancy rate per pregnancy.

7.1 Participants with BMI < 30 kg/m2

3

103

Odds Ratio (M‐H, Fixed, 95% CI)

0.32 [0.05, 2.24]

7.2 Participants with BMI ≥ 30 kg/m2

2

98

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.02, 6.69]

Open in table viewer
Comparison 4. D‐chiro‐inositol versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Ovulation Show forest plot

2

327

Odds Ratio (M‐H, Fixed, 95% CI)

3.57 [1.72, 7.45]

Analysis 4.1

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 1 Ovulation.

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 1 Ovulation.

1.1 Participants with BMI < 30 kg/m2

2

327

Odds Ratio (M‐H, Fixed, 95% CI)

3.57 [1.72, 7.45]

2 Body mass index (kg/m2) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

0.5 [‐0.86, 1.86]

Analysis 4.2

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).

3 Waist‐hip ratio Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.05, 0.03]

Analysis 4.3

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 3 Waist‐hip ratio.

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 3 Waist‐hip ratio.

4 Blood pressure ‐ systolic (mm Hg) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐5.85, 1.85]

Analysis 4.4

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 4 Blood pressure ‐ systolic (mm Hg).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 4 Blood pressure ‐ systolic (mm Hg).

5 Blood pressure ‐ diastolic (mm Hg) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐4.0 [‐7.26, ‐0.74]

Analysis 4.5

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 5 Blood pressure ‐ diastolic (mm Hg).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 5 Blood pressure ‐ diastolic (mm Hg).

6 Serum testosterone (nmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.63 [‐1.37, 0.11]

Analysis 4.6

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 6 Serum testosterone (nmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 6 Serum testosterone (nmol/L).

7 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

69.44 [34.97, 103.91]

Analysis 4.7

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 7 Serum sex hormone‐binding globulin (nmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 7 Serum sex hormone‐binding globulin (nmol/L).

8 Fasting glucose (mmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.28 [‐0.99, 0.43]

Analysis 4.8

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 8 Fasting glucose (mmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 8 Fasting glucose (mmol/L).

9 Fasting insulin (mIU/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐20.0 [‐43.43, 3.43]

Analysis 4.9

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 9 Fasting insulin (mIU/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 9 Fasting insulin (mIU/L).

10 Total cholesterol (mmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.23 [‐0.99, 0.53]

Analysis 4.10

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 10 Total cholesterol (mmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 10 Total cholesterol (mmol/L).

11 Triglyceride levels (mmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐2.20 [‐6.23, 1.83]

Analysis 4.11

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 11 Triglyceride levels (mmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 11 Triglyceride levels (mmol/L).

Open in table viewer
Comparison 5. Rosiglitazone versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Ovulation rate Show forest plot

1

64

Odds Ratio (M‐H, Fixed, 95% CI)

1.91 [0.70, 5.22]

Analysis 5.1

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 1 Ovulation rate.

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 1 Ovulation rate.

1.1 Participants with BMI ≥ 30 kg/m2

1

64

Odds Ratio (M‐H, Fixed, 95% CI)

1.91 [0.70, 5.22]

2 Menstrual frequency Show forest plot

2

100

Odds Ratio (M‐H, Fixed, 95% CI)

5.59 [2.20, 14.19]

Analysis 5.2

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 2 Menstrual frequency.

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 2 Menstrual frequency.

3 Body mass index (kg/m2) Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

0.68 [0.40, 0.96]

Analysis 5.3

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 3 Body mass index (kg/m2).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 3 Body mass index (kg/m2).

4 Waist‐hip ratio Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.02, ‐0.00]

Analysis 5.4

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 4 Waist‐hip ratio.

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 4 Waist‐hip ratio.

5 Blood pressure ‐ systolic (mm Hg) Show forest plot

1

52

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐3.95, ‐0.05]

Analysis 5.5

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 5 Blood pressure ‐ systolic (mm Hg).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 5 Blood pressure ‐ systolic (mm Hg).

6 Blood pressure ‐ diastolic (mm Hg) Show forest plot

1

52

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐1.72, 1.32]

Analysis 5.6

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 6 Blood pressure ‐ diastolic (mm Hg).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 6 Blood pressure ‐ diastolic (mm Hg).

7 Serum testosterone (nmol/L) Show forest plot

1

54

Mean Difference (IV, Fixed, 95% CI)

0.20 [‐0.34, 0.74]

Analysis 5.7

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 7 Serum testosterone (nmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 7 Serum testosterone (nmol/L).

8 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

‐0.69 [‐10.37, 8.98]

Analysis 5.8

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 8 Serum sex hormone‐binding globulin (nmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 8 Serum sex hormone‐binding globulin (nmol/L).

9 Fasting glucose (mmol/L) Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

‐0.21 [‐0.39, ‐0.04]

Analysis 5.9

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 9 Fasting glucose (mmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 9 Fasting glucose (mmol/L).

10 Fasting insulin (mIU/L) Show forest plot

2

80

Mean Difference (IV, Fixed, 95% CI)

‐3.98 [‐9.38, 1.42]

Analysis 5.10

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 10 Fasting insulin (mIU/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 10 Fasting insulin (mIU/L).

11 Total cholesterol (mmol/L) Show forest plot

2

80

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐0.21, ‐0.19]

Analysis 5.11

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 11 Total cholesterol (mmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 11 Total cholesterol (mmol/L).

12 Triglyceride levels (mmol/L) Show forest plot

1

26

Mean Difference (IV, Fixed, 95% CI)

1.0 [0.89, 1.11]

Analysis 5.12

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 12 Triglyceride levels (mmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 12 Triglyceride levels (mmol/L).

Open in table viewer
Comparison 6. Pioglitazone versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Menstrual frequency Show forest plot

2

70

Odds Ratio (M‐H, Fixed, 95% CI)

8.88 [2.35, 33.61]

Analysis 6.1

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 1 Menstrual frequency.

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 1 Menstrual frequency.

1.1 Participants with BMI < 30 kg/m2

1

40

Odds Ratio (M‐H, Fixed, 95% CI)

10.23 [1.12, 93.34]

1.2 Participants with BMI ≥ 30 kg/m2

1

30

Odds Ratio (M‐H, Fixed, 95% CI)

8.0 [1.52, 42.04]

2 Body mass index (kg/m2) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

0.91 [‐1.88, 3.70]

Analysis 6.2

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).

3 Waist‐hip ratio Show forest plot

1

28

Mean Difference (IV, Fixed, 95% CI)

0.02 [‐0.02, 0.06]

Analysis 6.3

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 3 Waist‐hip ratio.

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 3 Waist‐hip ratio.

4 Serum testosterone (nmol/L) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

‐0.12 [‐0.53, 0.29]

Analysis 6.4

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 4 Serum testosterone (nmol/L).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 4 Serum testosterone (nmol/L).

5 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

2.75 [‐5.26, 10.77]

Analysis 6.5

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 5 Serum sex hormone‐binding globulin (nmol/L).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 5 Serum sex hormone‐binding globulin (nmol/L).

6 Fasting insulin (mIU/L) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

‐1.46 [‐3.97, 1.06]

Analysis 6.6

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 6 Fasting insulin (mIU/L).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 6 Fasting insulin (mIU/L).

Study flow diagram since publication
Figuras y tablas -
Figure 1

Study flow diagram since publication

Study flow diagram 2017 update
Figuras y tablas -
Figure 2

Study flow diagram 2017 update

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies
Figuras y tablas -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies

Risk of bias summary: review authors' judgements about each risk of bias item for each included study
Figuras y tablas -
Figure 4

Risk of bias summary: review authors' judgements about each risk of bias item for each included study

Forest plot of comparison: 1 Metformin versus placebo or no treatment, outcome: 1.1 Live birth rate
Figuras y tablas -
Figure 5

Forest plot of comparison: 1 Metformin versus placebo or no treatment, outcome: 1.1 Live birth rate

Forest plot of comparison: 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, outcome: 2.1 Live birth rate
Figuras y tablas -
Figure 6

Forest plot of comparison: 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, outcome: 2.1 Live birth rate

Forest plot of comparison: 3 Metformin versus clomiphene citrate, outcome: 3.1 Live birth.
Figuras y tablas -
Figure 7

Forest plot of comparison: 3 Metformin versus clomiphene citrate, outcome: 3.1 Live birth.

Comparison 1 Metformin versus placebo or no treatment, Outcome 1 Live birth rate.
Figuras y tablas -
Analysis 1.1

Comparison 1 Metformin versus placebo or no treatment, Outcome 1 Live birth rate.

Comparison 1 Metformin versus placebo or no treatment, Outcome 2 Adverse events (gastrointestinal side effects).
Figuras y tablas -
Analysis 1.2

Comparison 1 Metformin versus placebo or no treatment, Outcome 2 Adverse events (gastrointestinal side effects).

Comparison 1 Metformin versus placebo or no treatment, Outcome 3 Clinical pregnancy rate.
Figuras y tablas -
Analysis 1.3

Comparison 1 Metformin versus placebo or no treatment, Outcome 3 Clinical pregnancy rate.

Comparison 1 Metformin versus placebo or no treatment, Outcome 4 Ovulation rate.
Figuras y tablas -
Analysis 1.4

Comparison 1 Metformin versus placebo or no treatment, Outcome 4 Ovulation rate.

Comparison 1 Metformin versus placebo or no treatment, Outcome 5 Menstrual frequency.
Figuras y tablas -
Analysis 1.5

Comparison 1 Metformin versus placebo or no treatment, Outcome 5 Menstrual frequency.

Comparison 1 Metformin versus placebo or no treatment, Outcome 6 Miscarriage rate per woman.
Figuras y tablas -
Analysis 1.6

Comparison 1 Metformin versus placebo or no treatment, Outcome 6 Miscarriage rate per woman.

Comparison 1 Metformin versus placebo or no treatment, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.
Figuras y tablas -
Analysis 1.7

Comparison 1 Metformin versus placebo or no treatment, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.

Comparison 1 Metformin versus placebo or no treatment, Outcome 8 Body mass index (kg/m2).
Figuras y tablas -
Analysis 1.8

Comparison 1 Metformin versus placebo or no treatment, Outcome 8 Body mass index (kg/m2).

Comparison 1 Metformin versus placebo or no treatment, Outcome 9 Waist‐hip ratio.
Figuras y tablas -
Analysis 1.9

Comparison 1 Metformin versus placebo or no treatment, Outcome 9 Waist‐hip ratio.

Comparison 1 Metformin versus placebo or no treatment, Outcome 10 Blood pressure ‐ systolic (mm Hg).
Figuras y tablas -
Analysis 1.10

Comparison 1 Metformin versus placebo or no treatment, Outcome 10 Blood pressure ‐ systolic (mm Hg).

Comparison 1 Metformin versus placebo or no treatment, Outcome 11 Blood pressure ‐ diastolic (mm Hg).
Figuras y tablas -
Analysis 1.11

Comparison 1 Metformin versus placebo or no treatment, Outcome 11 Blood pressure ‐ diastolic (mm Hg).

Comparison 1 Metformin versus placebo or no treatment, Outcome 12 Serum testosterone (nmol/L).
Figuras y tablas -
Analysis 1.12

Comparison 1 Metformin versus placebo or no treatment, Outcome 12 Serum testosterone (nmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 13 Serum sex hormone‐binding globulin (nmol/L).
Figuras y tablas -
Analysis 1.13

Comparison 1 Metformin versus placebo or no treatment, Outcome 13 Serum sex hormone‐binding globulin (nmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 14 Fasting glucose (mmol/L).
Figuras y tablas -
Analysis 1.14

Comparison 1 Metformin versus placebo or no treatment, Outcome 14 Fasting glucose (mmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 15 Fasting insulin (mIU/L).
Figuras y tablas -
Analysis 1.15

Comparison 1 Metformin versus placebo or no treatment, Outcome 15 Fasting insulin (mIU/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 16 Total cholesterol (mmol/L).
Figuras y tablas -
Analysis 1.16

Comparison 1 Metformin versus placebo or no treatment, Outcome 16 Total cholesterol (mmol/L).

Comparison 1 Metformin versus placebo or no treatment, Outcome 17 Triglyceride levels (mmol/L).
Figuras y tablas -
Analysis 1.17

Comparison 1 Metformin versus placebo or no treatment, Outcome 17 Triglyceride levels (mmol/L).

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 1 Live birth rate.
Figuras y tablas -
Analysis 2.1

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 1 Live birth rate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 2 Adverse events.
Figuras y tablas -
Analysis 2.2

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 2 Adverse events.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 3 Clinical pregnancy rate.
Figuras y tablas -
Analysis 2.3

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 3 Clinical pregnancy rate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 4 Ovulation rate.
Figuras y tablas -
Analysis 2.4

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 4 Ovulation rate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 5 Ovulation rate: subgroup analysis by sensitivity to clomiphene citrate.
Figuras y tablas -
Analysis 2.5

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 5 Ovulation rate: subgroup analysis by sensitivity to clomiphene citrate.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 6 Miscarriage rate per woman.
Figuras y tablas -
Analysis 2.6

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 6 Miscarriage rate per woman.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.
Figuras y tablas -
Analysis 2.7

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 7 Sensitivity analysis: miscarriage rate per pregnancy.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 8 Multiple pregnancy rate per woman.
Figuras y tablas -
Analysis 2.8

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 8 Multiple pregnancy rate per woman.

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 9 Senstivity analysis: multiple pregnancy rate per pregnancy.
Figuras y tablas -
Analysis 2.9

Comparison 2 Metformin combined with clomiphene citrate versus clomiphene citrate alone, Outcome 9 Senstivity analysis: multiple pregnancy rate per pregnancy.

Comparison 3 Metformin versus clomiphene citrate, Outcome 1 Live birth.
Figuras y tablas -
Analysis 3.1

Comparison 3 Metformin versus clomiphene citrate, Outcome 1 Live birth.

Comparison 3 Metformin versus clomiphene citrate, Outcome 2 Clinical pregnancy rate.
Figuras y tablas -
Analysis 3.2

Comparison 3 Metformin versus clomiphene citrate, Outcome 2 Clinical pregnancy rate.

Comparison 3 Metformin versus clomiphene citrate, Outcome 3 Ovulation rate.
Figuras y tablas -
Analysis 3.3

Comparison 3 Metformin versus clomiphene citrate, Outcome 3 Ovulation rate.

Comparison 3 Metformin versus clomiphene citrate, Outcome 4 Miscarriage rate per woman.
Figuras y tablas -
Analysis 3.4

Comparison 3 Metformin versus clomiphene citrate, Outcome 4 Miscarriage rate per woman.

Comparison 3 Metformin versus clomiphene citrate, Outcome 5 Sensitivity analysis: miscarriage rate per pregnancy.
Figuras y tablas -
Analysis 3.5

Comparison 3 Metformin versus clomiphene citrate, Outcome 5 Sensitivity analysis: miscarriage rate per pregnancy.

Comparison 3 Metformin versus clomiphene citrate, Outcome 6 Multiple pregnancy rate per woman.
Figuras y tablas -
Analysis 3.6

Comparison 3 Metformin versus clomiphene citrate, Outcome 6 Multiple pregnancy rate per woman.

Comparison 3 Metformin versus clomiphene citrate, Outcome 7 Sensitivity analysis: multiple pregnancy rate per pregnancy.
Figuras y tablas -
Analysis 3.7

Comparison 3 Metformin versus clomiphene citrate, Outcome 7 Sensitivity analysis: multiple pregnancy rate per pregnancy.

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 1 Ovulation.
Figuras y tablas -
Analysis 4.1

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 1 Ovulation.

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).
Figuras y tablas -
Analysis 4.2

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 3 Waist‐hip ratio.
Figuras y tablas -
Analysis 4.3

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 3 Waist‐hip ratio.

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 4 Blood pressure ‐ systolic (mm Hg).
Figuras y tablas -
Analysis 4.4

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 4 Blood pressure ‐ systolic (mm Hg).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 5 Blood pressure ‐ diastolic (mm Hg).
Figuras y tablas -
Analysis 4.5

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 5 Blood pressure ‐ diastolic (mm Hg).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 6 Serum testosterone (nmol/L).
Figuras y tablas -
Analysis 4.6

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 6 Serum testosterone (nmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 7 Serum sex hormone‐binding globulin (nmol/L).
Figuras y tablas -
Analysis 4.7

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 7 Serum sex hormone‐binding globulin (nmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 8 Fasting glucose (mmol/L).
Figuras y tablas -
Analysis 4.8

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 8 Fasting glucose (mmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 9 Fasting insulin (mIU/L).
Figuras y tablas -
Analysis 4.9

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 9 Fasting insulin (mIU/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 10 Total cholesterol (mmol/L).
Figuras y tablas -
Analysis 4.10

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 10 Total cholesterol (mmol/L).

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 11 Triglyceride levels (mmol/L).
Figuras y tablas -
Analysis 4.11

Comparison 4 D‐chiro‐inositol versus placebo or no treatment, Outcome 11 Triglyceride levels (mmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 1 Ovulation rate.
Figuras y tablas -
Analysis 5.1

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 1 Ovulation rate.

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 2 Menstrual frequency.
Figuras y tablas -
Analysis 5.2

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 2 Menstrual frequency.

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 3 Body mass index (kg/m2).
Figuras y tablas -
Analysis 5.3

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 3 Body mass index (kg/m2).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 4 Waist‐hip ratio.
Figuras y tablas -
Analysis 5.4

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 4 Waist‐hip ratio.

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 5 Blood pressure ‐ systolic (mm Hg).
Figuras y tablas -
Analysis 5.5

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 5 Blood pressure ‐ systolic (mm Hg).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 6 Blood pressure ‐ diastolic (mm Hg).
Figuras y tablas -
Analysis 5.6

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 6 Blood pressure ‐ diastolic (mm Hg).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 7 Serum testosterone (nmol/L).
Figuras y tablas -
Analysis 5.7

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 7 Serum testosterone (nmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 8 Serum sex hormone‐binding globulin (nmol/L).
Figuras y tablas -
Analysis 5.8

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 8 Serum sex hormone‐binding globulin (nmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 9 Fasting glucose (mmol/L).
Figuras y tablas -
Analysis 5.9

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 9 Fasting glucose (mmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 10 Fasting insulin (mIU/L).
Figuras y tablas -
Analysis 5.10

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 10 Fasting insulin (mIU/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 11 Total cholesterol (mmol/L).
Figuras y tablas -
Analysis 5.11

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 11 Total cholesterol (mmol/L).

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 12 Triglyceride levels (mmol/L).
Figuras y tablas -
Analysis 5.12

Comparison 5 Rosiglitazone versus placebo or no treatment, Outcome 12 Triglyceride levels (mmol/L).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 1 Menstrual frequency.
Figuras y tablas -
Analysis 6.1

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 1 Menstrual frequency.

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).
Figuras y tablas -
Analysis 6.2

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 2 Body mass index (kg/m2).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 3 Waist‐hip ratio.
Figuras y tablas -
Analysis 6.3

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 3 Waist‐hip ratio.

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 4 Serum testosterone (nmol/L).
Figuras y tablas -
Analysis 6.4

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 4 Serum testosterone (nmol/L).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 5 Serum sex hormone‐binding globulin (nmol/L).
Figuras y tablas -
Analysis 6.5

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 5 Serum sex hormone‐binding globulin (nmol/L).

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 6 Fasting insulin (mIU/L).
Figuras y tablas -
Analysis 6.6

Comparison 6 Pioglitazone versus placebo or no treatment, Outcome 6 Fasting insulin (mIU/L).

Summary of findings for the main comparison. Metformin compared to placebo or no treatment for women with polycystic ovary syndrome

Metformin compared to placebo or no treatment for women with polycystic ovary syndrome, oligo amenorrhoea and subfertility

Patient or population: women with polycystic ovary syndrome, oligo amenorrhoea and subfertility
Settings: outpatient
Intervention: metformin
Comparison: placebo or no treatment

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo or no treatment

Metformin

Live birth rate per woman

141 per 1000

208 per 1000
(141 to 292)

OR 1.59
(1.00 to 2.51)

435
(4 studies)

⊕⊕⊝⊝
lowa,b

Adverse events (gastrointestinal) per woman

106 per 1000

362 per 1000

(267 to 469)

OR 4.76

(3.06 to 7.41)

670

(7 studies)

⊕⊕⊕⊝
moderatea,c

Clinical pregnancy rate per woman

110 per 1000

193 per 1000
(149 to 246)

OR 1.93
(1.42 to 2.64)

1027
(9 studies)

⊕⊕⊕⊝
moderatea

Menstrual frequency per woman

183 per 1000

278 per 1000

(203 to 368)

OR 1.72

(1.14 to 2.61)

427

(7 studies)

⊕⊕⊝⊝
lowa,d

Ovulation rate per woman

200 per 1000

389 per 1000
(312 to 473)

OR
2.55

(1.81 to 3.59)

701
(14 studies)

⊕⊕⊕⊝
moderatea

Miscarriage rate per woman

40 per 1000

43per 1000
(20 to 89)

OR 1.08
(0.50 to 2.35)

748
(4 studies)

⊕⊕⊝⊝
lowa,b

Miscarriage rate per pregnancy OR 0.58, 95% CI 0.25 to 1.34, 200 pregnancies

*The basis for the assumed risk is the median control group risk across studies. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; OR: odds ratio

GRADE Working Group grades of evidence

High quality: we are very confident that the true effect lies close to that of the estimate of the effect

Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of effect, but there is a possibility that it is substantially different

Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect

Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect

aDowngraded one level for serious risk of bias related to failure to report methods of randomisation and/or serious risk of attrition bias in some of the studies.
bDowngraded one level for serious imprecision as the event rate is low and findings are compatible with benefit in one or both groups or with no meaningful difference between the groups.
cModerate inconsistency (I2 = 61%), but not downgraded, as all heterogeneity is attributable to a single small study and the direction of effect largely consistent.
dDowngraded one level for serious inconsistency (I2= 54%); largest study shows no evidence of effect.

Figuras y tablas -
Summary of findings for the main comparison. Metformin compared to placebo or no treatment for women with polycystic ovary syndrome
Summary of findings 2. Metformin combined with clomiphene citrate versus clomiphene citrate alone for women with polycystic ovary syndrome

Metformin combined with clomiphene versus clomiphene alone for women with polycystic ovary syndrome

Population: women with polycystic ovary syndrome
Setting: outpatient
Intervention: metformin combined with ovulation induction agent clomiphene
Comparison: clomiphene alone

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with clomiphene alone

Risk with metformin combined with clomiphene

Live birth rate per woman

257 per 1000

295 per 1000
(241 to 355)

OR 1.21
(0.92 to 1.59)

1079
(9 studies)

⊕⊕⊝⊝
lowa,b

Adverse events (gastrointestinal) per woman

134 per 1000

381 per 1000
(286 to 485)

OR 3.97
(2.59 to 6.08)

591
(3 studies)

⊕⊕⊕⊝
moderatea

Clinical pregnancy rate per woman

243 per 1000

338per 1000
(330 to 436)

OR 1.59
(1.27 to 1.99)

1529
(16 studies)

⊕⊕⊕⊝
moderatea

Menstrual frequency per woman

Not reported by any of the included studies

Ovulation rate per woman

381per 1000

491 per 1000
(441to 542)

OR 1.57

(1.28 to 1.92)

1624
(21 studies)

⊕⊕⊕⊝
moderatea,c

Miscarriage rate per woman

Median rates not calculable as there were no events in the control group in 5/8 studies

OR 1.59
(1.03 to 2.46)

1096
(9 studies)

⊕⊕⊝⊝
lowa,b

Miscarriage rate per pregnancy OR 1.30 95% CI 0.80 to 2.12, 400 pregnancies

*The risk in the intervention group (and its 95% confidence interval) is based on the median risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; OR: odds ratio

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect

aDowngraded one level for serious risk of bias related to failure to describe study methods and/or serious risk of attrition bias in several of the studies.
bDowngraded one level for serious imprecision as findings are compatible with benefit in one or both groups or with no meaningful difference between the group.
cHigh heterogeneity (I2 = 62%), but not downgraded as direction of effect consistent and most inconsistency is due to a single small study.

Figuras y tablas -
Summary of findings 2. Metformin combined with clomiphene citrate versus clomiphene citrate alone for women with polycystic ovary syndrome
Summary of findings 3. Metformin compared to clomiphene citrate for women with polycystic ovary syndrome

Metformin compared to clomiphene citrate for women with polycystic ovary syndrome

Population: women with polycystic ovary syndrome
Setting: outpatient
Intervention: metformin
Comparison: clomiphene citrate

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with clomiphene citrate

Risk with metformin

Live birth rate per woman

Participants with BMI < 30 kg/m2 or ≤ 32 kg/m2a

225 per 1000

171 per 1000

(124 to 227)

OR 1.71 (1.00 to 2.94)

241

(3 studies)

⊕⊝⊝⊝

very lowc,d

High heterogeneity (I2 = 78%)

Live birth rate per woman

Participants with BMI ≥ 30 kg/m2a

198 per 1000

69 per 1000

(40 to 114)

OR 0.30

(0.17 to 0.52)

500

(2 studies)

⊕⊝⊝⊝
very lowc,d

74 events

Adverse events

(gastrointestinal)

Not reported by any of the included studies

Clinical pregnancy rate per woman Participants with BMI < 30 kg/m2 or ≤ 32 kg/m2a

320 per 1000

423 per 1000
(331 to 523)

OR 1.56
(1.05 to 2.33)

490
(5 studies)

⊕⊝⊝⊝
very lowc,d

103 events

Clinical pregnancy rate per woman Participants with BMI ≥ 30 kg/m2a

234 per 1000

94 per 1000
(60 to 144)

OR 0.34
(0.21 to 0.55)

500
(2 studies)

⊕⊝⊝⊝
very lowc,d

98 events

Menstrual frequency

Not reported by any of the included studies

Ovulation rate per woman

Participants with BMI < 30 kg/m2b

625 per 1000

574 per 1000
(459 to 681)

OR 0.81
(0.51 to 1.28)

312
(4 studies)

⊕⊕⊝⊝
lowc

Ovulation rate per woman

Participants with BMI ≥ 30 kg/m2b

534per 1000

250per 1,000
(187 to 330)

OR 0.29
(0.20 to 0.43)

500
(2 studies)

⊕⊕⊝⊝
lowc

Miscarriage rate per woman

29 per 1000

26 per 1000

(15 to 47)

OR 0.92 (0.50 to 1.67)

741
(5 studies)

⊕⊝⊝⊝

very lowc,e

High heterogeneity (I2 = 52%)

*The risk in the intervention group (and its 95% confidence interval) is based on the median risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; OR: odds ratio

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect
Moderate quality: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different
Low quality: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect
Very low quality: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect

aData subgrouped by BMI, as pooling of BMI groups resulted in high heterogeneity (I2 > 85%) with differing directions of effect.
bData subgrouped by BMI, as pooling of BMI groups resulted in high heterogeneity (I2 = 74%), though direction of effect was consistent.
cEvidence downgraded two levels for very serious risk of bias, due to failure to report study methods and/or risk of attrition bias in one or more studies and because findings are based on subgroup analysis.
dEvidence downgraded one level for serious imprecision: low event rate (total 74 events).
eEvidence downgraded for serious inconsistency (where further downgrading feasible).

Figuras y tablas -
Summary of findings 3. Metformin compared to clomiphene citrate for women with polycystic ovary syndrome
Table 1. Abbreviations used

Abbreviation

Definition

BMI

Body mass index

CC

Clomiphene citrate

CI

Confidence interval

CT

Computerised tomography scan

DHEAS

Dehydroepiandrosterone sulphate

FSH

Follicle stimulating hormone

GTT

Glucose tolerance test

HbA1C

Glycosylated haemoglobin

HDL

High‐density lipoprotein cholesterol

IGFBP‐1

Insulin growth factor binding protein 1

LDL

Low‐density lipoprotein cholesterol

LH

Luteinising hormone

NIDDM

Non insulin dependent diabetes mellitus

PAI‐1

Plasminogen activator inhibitor 1

PCO

Polycystic ovary

PCOS

Polycystic ovary syndrome

RCT

Randomised controlled trial

rFSH

Recombinant follicle stimulating hormone

SD

Standard deviation

SE

Standard error of the mean

SHBG

Sex hormone‐binding globulin

VLDL

Very low density lipoprotein cholesterol

vs

Versus

MD

Mean difference

Figuras y tablas -
Table 1. Abbreviations used
Table 2. Conversion factors

Convert from

Convert to

Conversion factor

Cholesterol

mg/dL

mmol/L

0.026

Triglycerides

mg/dL

mmol/L

0.11

Insulin

pmol/L

mIU/L (= microIU/mL)

0.1667

Glucose

mg/dL

mmol/L

0.056

Progesterone

ng/mL

nmol/L

3.18

Testosterone

ng/dL

nmol/L

0.03467

Androstenedione

ng/dL

nmol/L

0.0349

Estradiol

ng/dL

pmol/L

36.71

17‐beta oestradiol

ng/dL

pmol/L

36.71

Dehydroepiandrosterone sulphate

microg/dL

micromol/L

0.02714

Sex hormone‐binding globulin

microg/dL

nmol/L

34.7

Standard deviation

Standard error

Standard deviation

Sqrt n

Confidence intervals

Confidence intervals

Standard error

(upper limit ‐ lower limit)/3.92

Figuras y tablas -
Table 2. Conversion factors
Table 3. Metformin vs placebo: ovulation rate per cycle

Study ID

Metformin

Placebo

P value

Events

Cycles

Events

Cycles

BMI < 30 kg/m2

Baillargeon 2004

27

32

11

32

Carmina 2004

7

12

3

12

Ng 2001

3

9

3

9

Onalan 2005

17

153

20

150

Yarali 2002

6

16

1

16

BMI ≥ 30 kg/m2

Fleming 2002

37

45

30

47

Hoeger 2004

3

9

6

11

Hoeger 2004

4

9

3

9

Jakubowicz 2001

8

28

0

28

Lord 2006

9

22

9

22

Nestler 1998

12

35

1

26

Onalan 2005

5

63

5

51

Otta 2010

7

14

6

15

PCOSMIC 2010

17

32

13

33

Sturrock 2002

0

12

1

14

Vandermolen 2001

1

12

1

15

Figuras y tablas -
Table 3. Metformin vs placebo: ovulation rate per cycle
Table 4. Metformin + clomiphene citrate vs clomiphene citrate: ovulation rate per cycle

Study ID

Metformin +
clomiphene citrate

Clomiphene
citrate alone

P value

Events

Cycles

Events

Cycles

BMI < 30 kg/m2

Ayaz 2013

16

21

8

21

Ben Ayed 2009

10

16

6

16

Boudhraa 2010

17

32

10

31

El‐Biely 2001

35

45

29

45

Hwu 2005

17

40

5

40

Machado 2012

15

21

5

15

Malkawi 2002

11

16

3

12

Moll 2006

84

141

98

168

Ng 2001

4

9

1

9

PCOSMIC 2010

27

35

23

36

BMI ≥ 30 kg/m2

Jakubowicz 2001

26

28

22

28

Khorram 2006

7

16

1

15

Legro 2007

582

964

462

942

Nestler 1998

19

21

2

25

Sahin 2004

38

51

34

55

Siebert 2009

34

52

36

55

Sturrock 2002

5

12

4

14

Vandermolen 2001

9

12

4

15

Zain 2009

38

41

24

41

Figuras y tablas -
Table 4. Metformin + clomiphene citrate vs clomiphene citrate: ovulation rate per cycle
Table 5. Metformin vs clomiphene citrate: ovulation rate per cycle

Metformin

Clomiphene citrate

Study ID

Events

Cycles

Events

Cycles

P value

BMI < 30 kg/m2

Palomba 2005

129

205

148

221

PCOSMIC 2010

23

35

23

36

BMI ≥ 30 kg/m2

Legro 2007

296

1019

462

942

Zain 2009

4

42

7

41

Figuras y tablas -
Table 5. Metformin vs clomiphene citrate: ovulation rate per cycle
Table 6. D‐Chiro‐inositol vs placebo: ovulation rate per cycle

Inositol

Placebo

Study ID

Events

Cycles

Events

Cycles

P value

BMI < 30 kg/m2

Gerli 2003

128

136

130

147

Nestler 1999

19

22

6

22

Figuras y tablas -
Table 6. D‐Chiro‐inositol vs placebo: ovulation rate per cycle
Table 7. Rosiglitazone vs placebo: ovulation rate per cycle

Rosiglitazone

Placebo

Study ID

Events

Cycles

Events

Cycles

P value

BMI ≥ 30 kg/m2

Baillargeon 2004

16

32

11

32

Figuras y tablas -
Table 7. Rosiglitazone vs placebo: ovulation rate per cycle
Comparison 1. Metformin versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Live birth rate Show forest plot

4

435

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.00, 2.51]

1.1 Participants with BMI < 30 kg/m2

3

370

Odds Ratio (M‐H, Fixed, 95% CI)

1.51 [0.94, 2.44]

1.2 Participants with BMI ≥ 30 kg/m2

1

65

Odds Ratio (M‐H, Fixed, 95% CI)

2.87 [0.51, 16.01]

2 Adverse events (gastrointestinal side effects) Show forest plot

7

670

Odds Ratio (M‐H, Fixed, 95% CI)

4.76 [3.06, 7.41]

2.1 Participants with BMI < 30 kg/m2

4

393

Odds Ratio (M‐H, Fixed, 95% CI)

5.61 [2.89, 10.88]

2.2 Participants with BMI ≥ 30 kg/m2

3

277

Odds Ratio (M‐H, Fixed, 95% CI)

4.13 [2.28, 7.49]

3 Clinical pregnancy rate Show forest plot

9

1027

Odds Ratio (M‐H, Fixed, 95% CI)

1.93 [1.42, 2.64]

3.1 Participants with BMI < 30 kg/m2

5

733

Odds Ratio (M‐H, Fixed, 95% CI)

1.89 [1.35, 2.65]

3.2 Participants with BMI ≥ 30 kg/m2

4

294

Odds Ratio (M‐H, Fixed, 95% CI)

2.21 [0.98, 4.98]

4 Ovulation rate Show forest plot

14

701

Odds Ratio (M‐H, Fixed, 95% CI)

2.55 [1.81, 3.59]

4.1 Participants with BMI < 30 kg/m2

5

229

Odds Ratio (M‐H, Fixed, 95% CI)

4.15 [2.31, 7.45]

4.2 Participants with BMI ≥ 30 kg/m2

10

472

Odds Ratio (M‐H, Fixed, 95% CI)

1.96 [1.28, 3.01]

5 Menstrual frequency Show forest plot

7

427

Odds Ratio (M‐H, Fixed, 95% CI)

1.72 [1.14, 2.61]

5.1 Participants with BMI < 30 kg/m2

1

23

Odds Ratio (M‐H, Fixed, 95% CI)

21.15 [1.01, 445.00]

5.2 Participants with BMI ≥ 30 kg/m2

6

404

Odds Ratio (M‐H, Fixed, 95% CI)

1.57 [1.03, 2.41]

6 Miscarriage rate per woman Show forest plot

4

748

Odds Ratio (M‐H, Fixed, 95% CI)

1.08 [0.50, 2.35]

6.1 Participants with BMI < 30 kg/m2

3

683

Odds Ratio (M‐H, Fixed, 95% CI)

1.19 [0.52, 2.71]

6.2 Participants with BMI ≥ 30 kg/m2

1

65

Odds Ratio (M‐H, Fixed, 95% CI)

0.5 [0.04, 5.80]

7 Sensitivity analysis: miscarriage rate per pregnancy Show forest plot

4

200

Odds Ratio (M‐H, Fixed, 95% CI)

0.58 [0.25, 1.34]

7.1 Participants with BMI < 30 kg/m2

3

188

Odds Ratio (M‐H, Fixed, 95% CI)

0.63 [0.26, 1.53]

7.2 Participants with BMI ≥ 30 kg/m2

1

12

Odds Ratio (M‐H, Fixed, 95% CI)

0.25 [0.02, 4.00]

8 Body mass index (kg/m2) Show forest plot

16

827

Mean Difference (IV, Fixed, 95% CI)

‐0.08 [‐0.33, 0.17]

8.1 Participants with BMI < 30 kg/m2

7

419

Mean Difference (IV, Fixed, 95% CI)

‐0.06 [‐0.33, 0.21]

8.2 Participants with BMI ≥ 30 kg/m2

10

408

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐0.92, 0.52]

9 Waist‐hip ratio Show forest plot

11

702

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.01, ‐0.00]

9.1 Participants with BMI < 30 kg/m2

5

389

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.01, ‐0.00]

9.2 Participants with BMI ≥ 30 kg/m2

6

313

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.02, 0.01]

10 Blood pressure ‐ systolic (mm Hg) Show forest plot

7

379

Mean Difference (IV, Fixed, 95% CI)

‐3.59 [‐5.13, ‐2.04]

10.1 Participants with BMI < 30 kg/m2

3

96

Mean Difference (IV, Fixed, 95% CI)

‐3.52 [‐5.29, ‐1.76]

10.2 Participants with BMI ≥ 30 kg/m2

5

283

Mean Difference (IV, Fixed, 95% CI)

‐3.80 [‐5.00, ‐0.60]

11 Blood pressure ‐ diastolic (mm Hg) Show forest plot

6

292

Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐1.35, 1.07]

11.1 Participants with BMI < 30 kg/m2

3

96

Mean Difference (IV, Fixed, 95% CI)

‐0.21 [‐1.55, 1.13]

11.2 Participants with BMI ≥ 30 kg/m2

4

196

Mean Difference (IV, Fixed, 95% CI)

0.18 [‐2.65, 3.02]

12 Serum testosterone (nmol/L) Show forest plot

15

863

Mean Difference (IV, Fixed, 95% CI)

‐0.49 [‐0.59, ‐0.39]

12.1 Participants with BMI < 30 kg/m2

7

419

Mean Difference (IV, Fixed, 95% CI)

‐0.71 [‐0.86, ‐0.56]

12.2 Participants with BMI ≥ 30 kg/m2

9

444

Mean Difference (IV, Fixed, 95% CI)

‐0.29 [‐0.44, ‐0.15]

13 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

15

823

Mean Difference (IV, Fixed, 95% CI)

0.49 [‐1.82, 2.81]

13.1 Participants with BMI < 30 kg/m2

6

387

Mean Difference (IV, Fixed, 95% CI)

‐0.22 [‐6.73, 6.28]

13.2 Participants with BMI ≥ 30 kg/m2

10

436

Mean Difference (IV, Fixed, 95% CI)

0.60 [‐1.88, 3.07]

14 Fasting glucose (mmol/L) Show forest plot

15

849

Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐0.21, ‐0.07]

14.1 Participants with BMI < 30 kg/m2

5

364

Mean Difference (IV, Fixed, 95% CI)

‐0.13 [‐0.22, ‐0.04]

14.2 Participants with BMI ≥ 30 kg/m2

11

485

Mean Difference (IV, Fixed, 95% CI)

‐0.16 [‐0.27, ‐0.05]

15 Fasting insulin (mIU/L) Show forest plot

14

573

Mean Difference (IV, Fixed, 95% CI)

‐4.13 [‐5.67, ‐2.58]

15.1 Participants with BMI < 30 kg/m2

4

85

Mean Difference (IV, Fixed, 95% CI)

‐6.20 [‐8.56, ‐3.84]

15.2 Participants with BMI ≥ 30 kg/m2

11

488

Mean Difference (IV, Fixed, 95% CI)

‐2.57 [‐4.62, ‐0.53]

16 Total cholesterol (mmol/L) Show forest plot

10

562

Mean Difference (IV, Fixed, 95% CI)

‐0.14 [‐0.31, 0.02]

16.1 Participants with BMI < 30 kg/m2

5

276

Mean Difference (IV, Fixed, 95% CI)

‐0.02 [‐0.26, 0.22]

16.2 Participants with BMI ≥ 30 kg/m2

6

286

Mean Difference (IV, Fixed, 95% CI)

‐0.26 [‐0.48, ‐0.03]

17 Triglyceride levels (mmol/L) Show forest plot

7

309

Mean Difference (IV, Fixed, 95% CI)

0.14 [‐0.05, 0.32]

17.1 Participants with BMI < 30 kg/m2

3

53

Mean Difference (IV, Fixed, 95% CI)

0.00 [‐0.33, 0.34]

17.2 Participants with BMI ≥ 30 kg/m2

5

256

Mean Difference (IV, Fixed, 95% CI)

0.20 [‐0.02, 0.42]

Figuras y tablas -
Comparison 1. Metformin versus placebo or no treatment
Comparison 2. Metformin combined with clomiphene citrate versus clomiphene citrate alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Live birth rate Show forest plot

9

1079

Odds Ratio (M‐H, Fixed, 95% CI)

1.21 [0.92, 1.59]

1.1 Participants with BMI < 30 kg/m2 or ≤ 32 kg/m2

5

531

Odds Ratio (M‐H, Fixed, 95% CI)

1.14 [0.78, 1.67]

1.2 Participants with BMI ≥ 30 kg/m2

4

548

Odds Ratio (M‐H, Fixed, 95% CI)

1.28 [0.86, 1.91]

2 Adverse events Show forest plot

3

591

Odds Ratio (M‐H, Fixed, 95% CI)

3.97 [2.59, 6.08]

2.1 Participants with BMI < 30 kg/m2

3

591

Odds Ratio (M‐H, Fixed, 95% CI)

3.97 [2.59, 6.08]

3 Clinical pregnancy rate Show forest plot

16

1529

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.27, 1.99]

3.1 Participants with BMI < 30 kg/m2

9

834

Odds Ratio (M‐H, Fixed, 95% CI)

1.46 [1.08, 1.98]

3.2 Participants with BMI ≥ 30 kg/m2

7

695

Odds Ratio (M‐H, Fixed, 95% CI)

1.76 [1.26, 2.47]

4 Ovulation rate Show forest plot

21

1624

Odds Ratio (M‐H, Fixed, 95% CI)

1.57 [1.28, 1.92]

4.1 BMI < 30 kg/m2

11

755

Odds Ratio (M‐H, Fixed, 95% CI)

1.79 [1.32, 2.41]

4.2 BMI ≥ 30 kg/m2

9

814

Odds Ratio (M‐H, Fixed, 95% CI)

1.52 [1.15, 2.01]

4.3 BMI not reported

1

55

Odds Ratio (M‐H, Fixed, 95% CI)

0.43 [0.13, 1.37]

5 Ovulation rate: subgroup analysis by sensitivity to clomiphene citrate Show forest plot

7

271

Odds Ratio (M‐H, Fixed, 95% CI)

4.69 [2.61, 8.44]

5.1 PCOS and clomiphene‐sensitive

1

56

Odds Ratio (M‐H, Fixed, 95% CI)

3.55 [0.65, 19.37]

5.2 PCOS and clomiphene‐resistant

6

215

Odds Ratio (M‐H, Fixed, 95% CI)

4.89 [2.62, 9.13]

6 Miscarriage rate per woman Show forest plot

9

1096

Odds Ratio (M‐H, Fixed, 95% CI)

1.59 [1.03, 2.46]

6.1 Participants with BMI < 30 kg/m2

5

548

Odds Ratio (M‐H, Fixed, 95% CI)

1.41 [0.76, 2.62]

6.2 Participants with BMI ≥ 30 kg/m2

4

548

Odds Ratio (M‐H, Fixed, 95% CI)

1.79 [0.97, 3.32]

7 Sensitivity analysis: miscarriage rate per pregnancy Show forest plot

8

400

Odds Ratio (M‐H, Fixed, 95% CI)

1.30 [0.80, 2.12]

7.1 Participants with BMI < 30 kg/m2

4

228

Odds Ratio (M‐H, Fixed, 95% CI)

1.27 [0.65, 2.51]

7.2 Participants with BMI ≥ 30 kg/m2

4

172

Odds Ratio (M‐H, Fixed, 95% CI)

1.34 [0.67, 2.68]

8 Multiple pregnancy rate per woman Show forest plot

6

1003

Odds Ratio (M‐H, Fixed, 95% CI)

0.56 [0.18, 1.68]

8.1 Participants with BMI < 30 kg/m2

3

476

Odds Ratio (M‐H, Fixed, 95% CI)

0.50 [0.12, 2.04]

8.2 Participants with BMI ≥ 30kg/m2

3

527

Odds Ratio (M‐H, Fixed, 95% CI)

0.66 [0.11, 4.01]

9 Senstivity analysis: multiple pregnancy rate per pregnancy Show forest plot

6

342

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.15, 1.42]

9.1 Participants with BMI < 30 kg/m2

3

178

Odds Ratio (M‐H, Fixed, 95% CI)

0.43 [0.10, 1.85]

9.2 Participants with BMI ≥ 30 kg/m2

3

164

Odds Ratio (M‐H, Fixed, 95% CI)

0.50 [0.08, 3.12]

Figuras y tablas -
Comparison 2. Metformin combined with clomiphene citrate versus clomiphene citrate alone
Comparison 3. Metformin versus clomiphene citrate

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Live birth Show forest plot

5

741

Odds Ratio (M‐H, Fixed, 95% CI)

0.71 [0.49, 1.01]

1.1 Participants with BMI < 30 kg/m2

3

241

Odds Ratio (M‐H, Fixed, 95% CI)

1.71 [1.00, 2.94]

1.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.30 [0.17, 0.52]

2 Clinical pregnancy rate Show forest plot

7

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 Participants with BMI < 30 kg/m2

5

490

Odds Ratio (M‐H, Fixed, 95% CI)

1.56 [1.05, 2.33]

2.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.34 [0.21, 0.55]

3 Ovulation rate Show forest plot

6

Odds Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Participants with BMI < 30 kg/m2

4

312

Odds Ratio (M‐H, Fixed, 95% CI)

0.81 [0.51, 1.28]

3.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.29 [0.20, 0.43]

4 Miscarriage rate per woman Show forest plot

5

741

Odds Ratio (M‐H, Fixed, 95% CI)

0.92 [0.50, 1.67]

4.1 Participants with BMI < 30 kg/m2

3

241

Odds Ratio (M‐H, Fixed, 95% CI)

1.58 [0.61, 4.09]

4.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.61 [0.27, 1.38]

5 Sensitivity analysis: miscarriage rate per pregnancy Show forest plot

5

Odds Ratio (M‐H, Fixed, 95% CI)

Totals not selected

5.1 Participants with BMI < 30 kg/m2

3

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

5.2 Participants with BMI ≥ 30 kg/m2

2

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

6 Multiple pregnancy rate per woman Show forest plot

5

858

Odds Ratio (M‐H, Fixed, 95% CI)

0.29 [0.06, 1.43]

6.1 Participants with BMI < 30 kg/m2

3

358

Odds Ratio (M‐H, Fixed, 95% CI)

0.46 [0.07, 3.16]

6.2 Participants with BMI ≥ 30 kg/m2

2

500

Odds Ratio (M‐H, Fixed, 95% CI)

0.14 [0.01, 2.76]

7 Sensitivity analysis: multiple pregnancy rate per pregnancy Show forest plot

5

201

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.06, 1.68]

7.1 Participants with BMI < 30 kg/m2

3

103

Odds Ratio (M‐H, Fixed, 95% CI)

0.32 [0.05, 2.24]

7.2 Participants with BMI ≥ 30 kg/m2

2

98

Odds Ratio (M‐H, Fixed, 95% CI)

0.33 [0.02, 6.69]

Figuras y tablas -
Comparison 3. Metformin versus clomiphene citrate
Comparison 4. D‐chiro‐inositol versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Ovulation Show forest plot

2

327

Odds Ratio (M‐H, Fixed, 95% CI)

3.57 [1.72, 7.45]

1.1 Participants with BMI < 30 kg/m2

2

327

Odds Ratio (M‐H, Fixed, 95% CI)

3.57 [1.72, 7.45]

2 Body mass index (kg/m2) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

0.5 [‐0.86, 1.86]

3 Waist‐hip ratio Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.05, 0.03]

4 Blood pressure ‐ systolic (mm Hg) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐5.85, 1.85]

5 Blood pressure ‐ diastolic (mm Hg) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐4.0 [‐7.26, ‐0.74]

6 Serum testosterone (nmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.63 [‐1.37, 0.11]

7 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

69.44 [34.97, 103.91]

8 Fasting glucose (mmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.28 [‐0.99, 0.43]

9 Fasting insulin (mIU/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐20.0 [‐43.43, 3.43]

10 Total cholesterol (mmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐0.23 [‐0.99, 0.53]

11 Triglyceride levels (mmol/L) Show forest plot

1

44

Mean Difference (IV, Fixed, 95% CI)

‐2.20 [‐6.23, 1.83]

Figuras y tablas -
Comparison 4. D‐chiro‐inositol versus placebo or no treatment
Comparison 5. Rosiglitazone versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Ovulation rate Show forest plot

1

64

Odds Ratio (M‐H, Fixed, 95% CI)

1.91 [0.70, 5.22]

1.1 Participants with BMI ≥ 30 kg/m2

1

64

Odds Ratio (M‐H, Fixed, 95% CI)

1.91 [0.70, 5.22]

2 Menstrual frequency Show forest plot

2

100

Odds Ratio (M‐H, Fixed, 95% CI)

5.59 [2.20, 14.19]

3 Body mass index (kg/m2) Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

0.68 [0.40, 0.96]

4 Waist‐hip ratio Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

‐0.01 [‐0.02, ‐0.00]

5 Blood pressure ‐ systolic (mm Hg) Show forest plot

1

52

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐3.95, ‐0.05]

6 Blood pressure ‐ diastolic (mm Hg) Show forest plot

1

52

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐1.72, 1.32]

7 Serum testosterone (nmol/L) Show forest plot

1

54

Mean Difference (IV, Fixed, 95% CI)

0.20 [‐0.34, 0.74]

8 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

‐0.69 [‐10.37, 8.98]

9 Fasting glucose (mmol/L) Show forest plot

3

132

Mean Difference (IV, Fixed, 95% CI)

‐0.21 [‐0.39, ‐0.04]

10 Fasting insulin (mIU/L) Show forest plot

2

80

Mean Difference (IV, Fixed, 95% CI)

‐3.98 [‐9.38, 1.42]

11 Total cholesterol (mmol/L) Show forest plot

2

80

Mean Difference (IV, Fixed, 95% CI)

‐0.20 [‐0.21, ‐0.19]

12 Triglyceride levels (mmol/L) Show forest plot

1

26

Mean Difference (IV, Fixed, 95% CI)

1.0 [0.89, 1.11]

Figuras y tablas -
Comparison 5. Rosiglitazone versus placebo or no treatment
Comparison 6. Pioglitazone versus placebo or no treatment

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Menstrual frequency Show forest plot

2

70

Odds Ratio (M‐H, Fixed, 95% CI)

8.88 [2.35, 33.61]

1.1 Participants with BMI < 30 kg/m2

1

40

Odds Ratio (M‐H, Fixed, 95% CI)

10.23 [1.12, 93.34]

1.2 Participants with BMI ≥ 30 kg/m2

1

30

Odds Ratio (M‐H, Fixed, 95% CI)

8.0 [1.52, 42.04]

2 Body mass index (kg/m2) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

0.91 [‐1.88, 3.70]

3 Waist‐hip ratio Show forest plot

1

28

Mean Difference (IV, Fixed, 95% CI)

0.02 [‐0.02, 0.06]

4 Serum testosterone (nmol/L) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

‐0.12 [‐0.53, 0.29]

5 Serum sex hormone‐binding globulin (nmol/L) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

2.75 [‐5.26, 10.77]

6 Fasting insulin (mIU/L) Show forest plot

2

63

Mean Difference (IV, Fixed, 95% CI)

‐1.46 [‐3.97, 1.06]

Figuras y tablas -
Comparison 6. Pioglitazone versus placebo or no treatment