Scolaris Content Display Scolaris Content Display

Administración de suplementos de vitamina D para embarazadas

Contraer todo Desplegar todo

Referencias

Asemi 2012 {published data only}

Asemi Z, Tabassi Z, Heidarzadeh Z, Khorammian H, Sabihi SS, Samimi M. Effect of calcium‐vitamin D supplementation on metabolic profiles in pregnant women at risk for pre‐eclampsia: a randomized placebo‐controlled trial. Pakistan Journal of Biological Sciences 2012;15(7):316‐24. CENTRAL

Asemi 2013a {published data only}

Asemi Z, Samimi M, Siavashani MA, Mazloomi M, Tabassi Z, Karamali M, et al. Calcium‐vitamin D co‐supplementation affects metabolic profiles, but not pregnancy outcomes, in healthy pregnant women. International Journal of Preventive Medicine 2016;7:49. CENTRAL
Asemi Z, Samimi M, Tabassi Z, Shakeri H, Esmaillzadeh A. Vitamin D supplementation affects serum high‐sensitivity C‐reactive protein, insulin resistance, and biomarkers of oxidative stress in pregnant women. Journal of Nutrition 2013;143(9):1432‐8. CENTRAL

Benson 2009 {published data only}

Benson J. A randomised controlled trial on the effects of antenatal vitamin D supplementation to improve vitamin D levels in the maternal and cord blood at birth in vitamin D deficient pregnant women. anzctr.org.au/Trial/Registration/TrialReview.aspx?id=83375 first received 4 February 2009. CENTRAL
Rodda CP, Benson JE, Vincent AJ, Whitehead CL, Polyakov A, Vollenhoven B. Maternal vitamin D supplementation during pregnancy prevents vitamin D deficiency in the newborn: A randomised controlled trial. Osteoporosis International 2011;Suppl 4:S521‐S559. CENTRAL
Rodda CP, Benson JE, Vincent AJ, Whitehead CL, Polyakov A, Vollenhoven B. Maternal vitamin D supplementation during pregnancy prevents vitamin D deficiency in the newborn: An open label randomised controlled trial. Clinical Endocrinology 2015;83(3):363‐8. CENTRAL

Bhutta 2011 {published data only}

Bhutta ZA. Evaluation of the effectiveness of vitamin D supplementation to pregnant women and their infants in Pakistan. clinicaltrials.gov/ct2/show/NCT01229189 (first received 27 October 2010). CENTRAL
Bhutta ZA. Study of vitamin D supplementation on improvement of gums health. clinicaltrials.gov/ct2/show/NCT01422122 (first received 23 August 2011). CENTRAL
Khan F. A randomized controlled trial of oral vitamin D supplementation in pregnancy to improve maternal periodontal health and birth weight. Journal of International Oral Health 2016;8(6):657‐65. CENTRAL

Brooke 1980 {published data only}

Brooke OG, Brown IR, Bone CD, Carter ND, Cleeve HJ, Maxwell JD, et al. Vitamin D supplements in pregnant Asian women: effects on calcium status and fetal growth. British Medical Journal 1980;1:751‐4. CENTRAL
Brooke OG, Butters F, Wood C. Intrauterine vitamin D nutrition and postnatal growth in Asian infants. British Medical Journal 1981;283:1024. CENTRAL
Brown IR, Brooke OG, Cleeve HJ. Changes in mineral metabolism in the human foetus and newborns associated with maternal vitamin d supplements [proceedings]. Biochemical Society Transactions 1980;8(1):136‐7. CENTRAL
Maxwell JD, Ang L, Brooke OG, Brown IR. Vitamin D supplements enhance weight gain and nutritional status in pregnant Asians. British Journal of Obstetrics and Gynaecology 1981;88:987‐91. CENTRAL

Delvin 1986 {published data only}

Delvin EE, Salle BL, Glorieux FH, Adeleine P, David LS. Vitamin D supplementation during pregnancy: effect on neonatal calcium homeostasis. Journal of Pediatrics 1986;109:328‐34. CENTRAL

Diogenes 2013 {published data only}

Bezerra F. Calcium plus vitamin d supplementation during pregnancy of adolescent mothers: effects on maternal and infant bone mass, calcium and bone metabolism and breast milk composition. clinicaltrials.gov/ct2/show/NCT01732328 (first received 22 October 2012). CENTRAL
Diogenes ME, Bezerra FF, Rezende EP, Donangelo CM. Calcium plus vitamin D supplementation during third trimester of pregnancy in adolescents accustomed to low calcium diets did not affect infant bone mass at early lactation in a randomized controlled trial. Journal of Nutrition 2015;145:1515‐23. CENTRAL
Diogenes ME, Bezerra FF, Rezende EP, Taveira MF, Pinhal I, Donangelo CM. Effect of calcium plus vitamin D supplementation during pregnancy in Brazilian adolescent mothers: a randomized, placebo‐controlled trial. American Journal of Clinical Nutrition 2013;98(1):82‐91. CENTRAL
Normando P, Diogenes ME, Cabello PH, Cabello GM, Donangelo CM, Bezerra FF. Calcium plus vitamin D supplementation during pregnancy interacts with polymorphisms in the promoter region of the VDR gene to affect postpartum bone mass of Brazilian adolescent mothers: a randomized controlled trial. Nutrition 2016;32(10):1068‐74. CENTRAL

Grant 2013 {published data only}

Grant C. Randomised placebo controlled study of vitamin D during pregnancy and infancy. anzctr.org.au/Trial/Registration/TrialReview.aspx?id=320842 (first received 2 February 2010). CENTRAL
Grant C, Milne T, Knight J, Sinclair J, Camargo C. Vitamin D supplementation during pregnancy and infancy reduces food allergen sensitisation and parental‐reported food allergy: a randomised controlled trial. European Journal of Pediatrics 2016;175(11):1438. CENTRAL
Grant C, Stewart A, Scragg R, Milne T, Rowden J, Ekeroma A, et al. What dose of vitamin D supplementation is required in pregnancy and infancy to increase infants' serum 25‐hydroxyvitamin D concentration > 20ng/ml?. Pediatric Academic Societies Annual Meeting; 2013 May 4‐7; Washington DC, USA. 2013. CENTRAL
Grant CC, Crane J, Mitchell EA, Sinclair J, Stewart A, Milne T, et al. Vitamin D supplementation during pregnancy and infancy reduces aeroallergen sensitisation: a randomised controlled trial. Allergy 2016;71(9):1325‐34. CENTRAL
Grant CC, Crane J, Mitchell EA, Sinclair J, Stewart A, Milne T, et al. Vitamin D supplementation during pregnancy and infancy reduces aeroallergen sensitization: A randomized controlled trial. Pediatric Academic Societies Annual Meeting; 2016 April 30‐May 3; Baltimore, USA. 2016:1650.8. CENTRAL
Grant CC, Kaur S, Waymouth E, Mitchell EA, Scragg R, Ekeroma A, et al. Reduced primary care respiratory infection visits following pregnancy and infancy vitamin D supplementation: a randomised‐controlled trial. Acta Paediatrica 2015;104(4):396‐404. CENTRAL
Grant CC, Stewart AW, Scragg R, Milne T, Rowden J, Ekeroma A, et al. Vitamin D during pregnancy and infancy and infant serum 25‐hydroxyvitamin D concentration. Pediatrics 2014;133(1):e143‐53. CENTRAL
Wall CR, Stewart AW, Camargo CA, Scragg R, Mitchell EA, Ekeroma A, et al. Vitamin D activity of breast milk in women randomly assigned to vitamin D3 supplementation during pregnancy. American Journal of Clinical Nutrition 2016;103:382‐8. CENTRAL

Harvey 2012 {published data only}

Barker M, D'Angelo S, Ntani G, Lawrence W, Baird J, Jarman M, et al. The relationship between maternal self‐efficacy, compliance and outcome in a trial of vitamin D supplementation in pregnancy. Osteoporosis International 2017;28(1):77‐84. CENTRAL
Cook E, Curtis EM, Krstic N, D'Angelo S, Crozier SR, Moon RJ, et al. Perinatal DNA methylation at the RXRA promoter is associated with gestational vitamin d supplementation: results from the mavidos trial. Rheumatology 2017;56(Suppl 2):ii112. CENTRAL
Cooper C. A randomised double‐blind placebo‐controlled trial of vitamin D supplements for pregnant women with low levels of vitamin D in early pregnancy. isrctn.com/ISRCTN82927713 (first received 25 February 2008). CENTRAL
Cooper C, Harvey NC, Bishop NJ, Kennedy S, Papageorghiou AT, Schoenmakers I, et al. Maternal gestational vitamin D supplementation and offspring bone health (MAVIDOS): A multicentre, double‐blind, randomised placebo‐controlled trial. Lancet Diabetes and Endocrinology 2016;4(5):393‐402. CENTRAL
Cooper C, Harvey NC, Javaid MK, Bishop NJ, Kennedy S, Papageorghiou AT, et al. Effectiveness of maternal Vitamin D supplementation: A multicentre randomised, double‐blind, placebo controlled trial (MAVIDOS). Osteoporosis International 2015;26(1 Suppl 1):S40. CENTRAL
Curtis E, D'Angelo S, Crozier S, Moon R, Inskip H, Cook E, et al. Dna methylation at the rxra promoter at birth is associated with gestational vitamin d supplementation: results from the mavidos trial. Osteoporosis International 2017;28(Suppl 1):S72‐S73. CENTRAL
Harvey N, Moon R, D'Angelo S, Crozier S, Inskip H, Schoenmakers I, et al. Predictors of maternal 25(OH)‐vitamin D response to antenatal vitamin d supplementation: results from the mavidos trial. Osteoporosis International 2016;27(2 Suppl):S628‐S629. CENTRAL
Harvey NC. Influence of diet and lifestyle during pregnancy on offspring bone mass and body composition. The Power of Programming 2014: International Conference on Developmental Origins of Adiposity and Long‐Term Health; 2014 March 13‐15; Munich, Germany. 2014:27. CENTRAL
Harvey NC, Bishop NJ, Kennedy S, Papageorghiou AT, Schoenmakers I, Fraser R, et al. O16 Maternal gestational vitamin d supplementation results in greater bone mass for offspring born during the winter months: the mavidos multicentre randomized, double‐blind, placebo‐controlled trial. Rheumatology 2016;55(Suppl 1):i41. CENTRAL
Harvey NC, Javaid K, Bishop N, Kennedy S, Papageorghiou AT, Fraser R, et al. MAVIDOS Maternal Vitamin D Osteoporosis Study: Study protocol for a randomized controlled trial. The MAVIDOS Study Group. Trials 2012;13:13. CENTRAL
Harvey NC, Moon RJ, D'Angelo S, Crozier S, Schoenmakers I, Bishop NJ, et al. Determinants of the maternal response to vitamin D supplementation during pregnancy: the MAVIDOS trial. Osteoporosis International 2016;27(Suppl 1):S49‐S50, Abstract no: OC19. CENTRAL
Ioannou C, Cavallaro A, D'Angelo S, Javaid MK, Cooper C, Harvey NC, et al. Antenatal vitamin D supplementation does not affect fetal femur volume: secondary outcome of a randomised controlled trial. BJOG: an international journal of obstetrics and gynaecology 2017;124:28. CENTRAL
Moon RJ, Harvey NC, Cooper C, D'Angelo S, Crozier SR, Inskip HM, et al. Determinants of the maternal 25‐hydroxyvitamin D response to vitamin D supplementation during pregnancy. Journal of Clinical Endocrinology and Metabolism 2016;101(12):5012‐20. CENTRAL
Moon RJ, Harvey NC, Cooper C, D'Angelo S, Curtis EM, Crozier SR, et al. 25‐Hydroxyvitamin D response to gestational cholecalciferol supplementation is associated with common vitamin D related genetic variants: findings from the mavidos trial. Rheumatology 2017;56(Suppl 2):ii153. CENTRAL
Moon RJ, Harvey NC, Cooper C, D'Angelo S, Curtis EM, Crozier SR, et al. 25‐hydroxyvitamin d response to gestational cholecalciferol supplementation is associated with common vitamin d related genetic variants: findings from the mavidos trial. Osteoporosis International 2017;28(Suppl 1):S70‐1. CENTRAL
Moon RJ, Harvey NC, Cooper C, D'Angelo S, Curtis EM, Crozier SR, et al. Response to antenatal cholecalciferol supplementation is associated with common vitamin d related genetic variants. Journal of Clinical Endocrinology and Metabolism 2017;102(8):2941‐9. CENTRAL

Kaur 1991 {published data only}

Kaur J, Marya RK, Rathee S, lal H, Singh GP. Effect of pharmacological doses of vitamin D during pregnancy on placental protein status and birth weight. Nutrition Research 1991;11(9):1077‐81. CENTRAL

Li 2000a {published data only}

Li X, Gou W. Study on prevention of pregnancy induced hypertension and effect of platelet intracellular free ca˜(2+) by calcium supplementation [补钙预防妊高征及对血小板细胞内游离钙浓度的影响]. Journal of Xi'an Medical University 2000;21(1):46‐8. CENTRAL

Mallet 1986 {published data only}

Mallet E, Gugi B, Brunelle P, Henocq A, Basuyau JP, Lemeur H. Vitamin D supplementation in pregnancy: a controlled trial of two methods. Obstetrics & Gynecology 1986;68:300‐4. CENTRAL

Marya 1987 {published data only}

Marya RK, Rathee S, Manrow M. Effect of calcium and vitamin D supplementation on toxaemia of pregnancy. Gynecologic and Obstetric Investigation 1987;24(1):38‐42. CENTRAL

Marya 1988 {published data only}

Marya RK, Rathee S, Dua V, Sangwan K. Effect of vitamin D supplementation during pregnancy on foetal growth. Indian Journal of Medical Research 1988;88:488‐92. CENTRAL

Mazurkevich 2013 {published data only}

Mazurkevich M, Doronin G, Firsova T. State of placental complex during physiological pregnancy during correction of mineral insufficiency. Journal of Perinatal Medicine 2013;41(Suppl 1):Abstract no:1213. CENTRAL

Mirghafourvand 2013 {published data only}

Mansouri A, Mirghafourvand M, Charandabi SM, Najafi M. The effect of vitamin d and calcium plus vitamin d on leg cramps in pregnant women: a randomized controlled trial. Journal of Research in Medical Sciences 2017;22(1):24. CENTRAL
Mirghafourvand M. The effect of vitamin D and calcium plus vitamin D for leg cramps in pregnant women: a randomised controlled trial. en.irct.ir/trial/10789 (first received 4 May 2013). CENTRAL
Mirghafourvand M, Mohammad‐Alizadeh‐Charandabi S, Mansouri A, Najafi M, Khodabande F. The effect of vitamin D and calcium plus vitamin D on sleep quality in pregnant women with leg cramps: A controlled randomized clinical trial. Journal of Isfahan Medical School 2015;32(320):2444‐53. CENTRAL
Mohammad‐Alizadeh‐Charandabi S, Mirghafourvand M, Mansouri A, Najafi M, Khodabande F. The effect of vitamin d and calcium plus vitamin d during pregnancy on pregnancy and birth outcomes: a randomized controlled trial. Journal of Caring Science 2015;4(1):35‐44. CENTRAL

Naghshineh 2016 {published data only}

Naghshineh E, Sheikhaliyan S. Effect of vitamin D supplementation in the reduce risk of preeclampsia in nulliparous women. Advanced Biomedical Research 2016;5:7. CENTRAL

Roth 2010 {published data only}

Akhtar E, Mily A, Haq A, Al‐Mahmud A, El‐Arifeen S, Hel Baqui A, et al. Prenatal high‐dose vitamin D3 supplementation has balanced effects on cord blood Th1 and Th2 responses. Nutrition Journal 2016;15(1):75. CENTRAL
Bailey D, Perumal N, Yazdanpanah M, Al Mahmud A, Baqui AH, Adeli K, et al. Maternal‐fetal‐infant dynamics of the C3‐epimer of 25‐hydroxyvitamin D. Clinical Biochemistry 2014;47(9):816‐22. CENTRAL
Dimitris MC, Perumal N, Craig‐Barnes HA, Leadley M, Mahmud AA, Baqui AH, et al. Effect of weekly high‐dose vitamin D3 supplementation on serum cholecalciferol concentrations in pregnant women. Journal of Steroid Biochemistry and Molecular Biology 2016;158:76‐81. CENTRAL
Harrington J, Perumal N, Al Mahmud A, Baqui A, Roth DE. Vitamin D and fetal‐neonatal calcium homeostasis: findings from a randomized controlled trial of high‐dose antenatal vitamin D supplementation. Pediatric Research 2014;76(3):302‐9. CENTRAL
Perumal N, Al Mahmud A, Baqui A, Raqib R, Roth D. Effect of high‐dose vitamin D supplementation on blood pressure during the third trimester of pregnancy: A randomized controlled trial in Bangladesh. American Journal of Epidemiology 2012;176(1):80, Abstract no: 2. CENTRAL
Perumal N, Al Mahmud A, Baqui AH, Roth DE. Prenatal vitamin d supplementation and infant vitamin d status in Bangladesh. Public Health Nutrition 2017;20(10):1865‐73. CENTRAL
Perumal N, Mahmud A, Baqui A, Roth D. Prenatal vitamin D supplementation and infant vitamin D status in Bangladesh. FASEB Journal 2014;28(1 Suppl 1):[Abstract no. 256.4]. CENTRAL
Raqib R, Ly A, Akhtar E, Mily A, Perumal N, Al‐Mahmud A, et al. Prenatal vitamin D3 supplementation suppresses LL‐37 peptide expression in ex vivo activated neonatal macrophages but not their killing capacity. British Journal of Nutrition 2014;112(6):908‐15. CENTRAL
Roth D. Antenatal vitamin D3 supplementation in Bangladesh: randomized controlled trial (AViDD‐2). clinicaltrials.gov/ct2/show/NCT01126528 (first received 19 May 2010). CENTRAL
Roth DE, Al Mahmud A, Perumal N, Pezzack B, Raqib R, Baqui AH. Effect of prenatal third‐trimester vitamin D3 supplementation (35,000 IU/week) on fetal and infant linear growth in Bangladesh: the AViDD trial. Pediatric Academic Societies Annual Meeting; 2013 May 4‐7; Washington DC, USA. 2013. CENTRAL
Roth DE, Al Mahmud A, Raqib R, Akhtar E, Perumal N, Pezzack B, et al. Randomized placebo‐controlled trial of high‐dose prenatal third‐trimester vitamin D3 supplementation in Bangladesh: the AViDD trial. Nutrition Journal 2013;12:47. CENTRAL
Roth DE, Al Mahmud A, Raqib R, Baqui AH. Effects of high‐dose antenatal 3rd‐trimester vitamin D supplementation (35,000 IU/week) on maternal and newborn vitamin D status: A randomized placebo‐controlled trial in Dhaka, Bangladesh. FASEB Journal 2012;26:[Abstract no. 392.3]. CENTRAL
Roth DE, Al‐Mahmud A, El Arifeen S, Raqib R, Black RE, Baqui AH. Randomized pilot trial of two oral vitamin d3 supplementation regimens during the third trimester of pregnancy in Bangladeshi women: effects on neonatal vitamin d status and safety. Pediatric Academic Societies and Asian Society for Pediatric Research Joint Meeting; 2011 April 30‐May 3; Denver, Colorado, USA. 2011:1411.154. CENTRAL
Roth DE, Perumal N, Al A, Baqui AH. Maternal vitamin D3 supplementation during the third trimester of pregnancy: Effects on infant growth in a longitudinal follow‐up study in Bangladesh. Journal of Pediatrics 2013;163(6):1605‐11. CENTRAL

Roth 2013 {published data only}

Morris SK, Pell LG, Rahman MZ, Dimitris MC, Mahmud A, Islam MM, et al. Maternal vitamin D supplementation during pregnancy and lactation to prevent acute respiratory infections in infancy in Dhaka, Bangladesh (MDARI trial): protocol for a prospective cohort study nested within a randomized controlled trial. BMC Pregnancy and Childbirth 2016;16(1):309. CENTRAL
Roth D. Maternal vitamin D for infant growth (MDIG) trial. https://clinicaltrials.gov/ct2/show/NCT01924013(first received August 2013). CENTRAL
Roth D, Mahmud AA, Morris S, Zlotkin S, Gernand A, Tahmeed A, et al. Maternal vitamin D supplementation during pregnancy and lactation to promote infant growth in Dhaka, Bangladesh (mdig trial): a randomized controlled trial. Annals of Nutrition and Metabolism 2017;71(Suppl 2):571. CENTRAL
Roth DE, Gernand AD, Morris SK, Pezzack B, Islam MM, Dimitris MC, et al. Maternal vitamin D supplementation during pregnancy and lactation to promote infant growth in Dhaka, Bangladesh (MDIG trial): study protocol for a randomized controlled trial. Trials 2015;16:300. CENTRAL
Roth DE, Morris SK, Zlotkin S, Gernand AD, Ahmed T, Shanta SS, et al. Vitamin D supplementation in pregnancy and lactation and infant growth. New England Journal of Medicine 2018;379(6):535‐46. CENTRAL

Sabet 2012 {published data only}

Sabet Z, Ghazi AA, Tohidi M, Oladi B. Vitamin D supplementation in pregnant Iranian women: Effects on maternal and neonatal vitamin D and parathyroid hormone status. Acta Endocrinologica 2012;8(1):59‐66. CENTRAL

Sablok 2015 {published data only}

Sablok A, Batra A, Thariani K, Batra A, Bharti R, Aggarwal AR, et al. Supplementation of vitamin D in pregnancy and its correlation with feto‐maternal outcome. Clinical Endocrinology 2015;83(4):536‐41. CENTRAL

Samimi 2016 {published data only}

Samimi M, IRCT201102135444N3. Effects of calcium and vitamin D co‐supplementation on pre‐eclampsia parameters, metabolic status and biomarkers of oxidative stress in pregnant women at risk for pre‐eclampsia. en.search.irct.ir/view/5288 (first received 31 January 2015). CENTRAL
Samimi M, Kashi M, Foroozanfard F, Karamali M, Bahmani F, Asemi Z, et al. The effects of vitamin D plus calcium supplementation on metabolic profiles, biomarkers of inflammation, oxidative stress and pregnancy outcomes in pregnant women at risk for pre‐eclampsia. Journal of Human Nutrition and Dietetics 2016;29(4):505‐16. CENTRAL

Samimi 2017 {published data only}

Samimi M, Foroozanfard F, Amini F, Sehat M. Effect of vitamin D supplementation on unexplained recurrent spontaneous abortion: a double‐blind randomized controlled trial. Global Journal of Health Science 2017;9(3):95. CENTRAL

Sasan 2017 {published data only}

Sasan SB, Zandvakili F, Soufizadeh N, Baybordi E. The effects of vitamin d supplement on prevention of recurrence of preeclampsia in pregnant women with a history of preeclampsia. Obstetrics and Gynecology International 2017;2017:8249264. CENTRAL

Shahgheibi 2016 {published data only}

Shahgheibi S, Farhadifar F, Pouya B. The effect of vitamin D supplementation on gestational diabetes in high‐risk women: Results from a randomized placebo‐controlled trial. Journal of Research in Medical Sciences 2016;21(1):2. CENTRAL

Singh 2015 {published data only}

Singh J, Hariharan C, Bhaumik D. Role of vitamin D in reducing the risk of preterm labour. International Journal of Reproduction, Contraception, Obstetrics and Gynecology 2015;1:86‐93. CENTRAL

Taherian 2002 {published data only}

Taherian AA, Taherian A, Shirmani A. Prevention of preeclampsia with low‐dose aspirin or calcium supplementation. Archives of Iranian Medicine 2002;5(3):151‐6. CENTRAL

Tehrani 2014 {published data only}

Tehrani HG. Comparison of effectiveness of vitamin D supplementation in decreasing the development of the gestational diabetes mellitus in pregnant women. en.irct.ir/trial/7997 (first received 3 January 2014). CENTRAL
Tehrani HG, Mostajeran F, Banihashemi B. Effect of vitamin d supplementation on the incidence of gestational diabetes. Advanced Biomedical Research 2017;6:79. CENTRAL

Vaziri 2016 {published data only}

Vaziri F, Dabbaghmanesh MH, Samsami A, Nasiri S, Shirazi PT. Vitamin D supplementation during pregnancy on infant anthropometric measurements and bone mass of mother‐infant pairs: A randomized placebo clinical trial. Early Human Development 2016;103:61‐8. CENTRAL
Vaziri F, Nasiri S, Tavana Z, Dabbaghmanesh MH, Sharif F, Jafari P. A randomized controlled trial of vitamin D supplementation on perinatal depression: in Iranian pregnant mothers. BMC Pregnancy and Childbirth 2016;16:239. CENTRAL

Yu 2008 {published data only}

Goldring S. Effects of prenatal vitamin D supplementation on respiratory and allergic phenotypes and bone density in the first three years of life. UKCRN (http://public.ukcrn.org.uk) [accessed 6 April 2011]2010. CENTRAL
Goldring ST, Griffiths CJ, Martineau AR, Robinson S, Yu C, Poulton S, et al. Prenatal vitamin D supplementation and child respiratory health: a randomised controlled trial. PLOS One 2013;8(6):e66627. CENTRAL
Griffiths M, Goldring S, Griffiths C, Shaheen SO, Martineau A, Cross L, et al. Effects of pre‐natal vitamin D supplementation with partial correction of vitamin D deficiency on early life healthcare utilisation: A randomised controlled trial. PLOS One 2015;10(2):e0145303. CENTRAL
Yu C, Newton L, Robinson S, Teoh TG, Sethi M. Vitamin D deficiency and supplementation in pregnant women of four ethnic groups. Archives of Disease in Childhood. Fetal and Neonatal Edition 2008;93(Suppl 1):Fa68. CENTRAL
Yu CK, Sykes L, Sethi M, Teoh TG, Robinson S. Vitamin D deficiency and supplementation during pregnancy. Clinical Endocrinology 2009;70(5):685‐90. CENTRAL

Ala‐Houhala 1986 {published data only}

Ala‐Houhala M, Koskinen T, Terho A, Koivula T, Visakorpi J. Maternal compared with infant vitamin D supplementation. Archives of Disease in Childhood 1986;61:1159‐63. CENTRAL

Asemi 2013b {published data only}

Asemi Z, Hashemi T, Karamali M, Samimi M, Esmaillzadeh A. Effects of vitamin D supplementation on glucose metabolism, lipid concentrations, inflammation, and oxidative stress in gestational diabetes: a double‐blind randomized controlled clinical trial. American Journal of Clinical Nutrition 2013;98(6):1425‐32. CENTRAL
Asemi Z, Karamali M, Esmaillzadeh A. Favorable effects of vitamin d supplementation on pregnancy outcomes in gestational diabetes: a double blind randomized controlled clinical trial. Hormone and Metabolic Research 2015;47(8):565‐70. CENTRAL

Asemi 2014 {published data only}

Asemi Z, Karamali M, Esmaillzadeh A. Effects of calcium‐vitamin D co‐supplementation on glycaemic control, inflammation and oxidative stress in gestational diabetes: a randomised placebo‐controlled trial. Diabetologia 2014;57(9):1798‐806. CENTRAL

Asemi 2015 {published data only}

Asemi Z, Esmaillzadeh A. The effect of multi mineral‐vitamin D supplementation on pregnancy outcomes in pregnant women at risk for pre‐eclampsia. International Journal of Preventive Medicine 2015;6:62. CENTRAL

Atkinson 2010 {published data only}

Atkinson SA, Moore C. Bone Health in Pregnancy (B‐Hip). clinicaltrials.gov/ct2/show/NCT01693510 (first received 26 September 2012). CENTRAL

Azami 2017 {published data only}

Azami M, Azadi T, Farhang S, Rahmati S, Pourtaghi K. The effects of multi mineral‐vitamin D and vitamins (C+E) supplementation in the prevention of preeclampsia: An RCT. International Journal of Reproductive Biomedicine (Yazd, Iran) 2017;15(5):273‐8. CENTRAL
Azami M, IRCT2016062528617N1. The effects of multi minerals (Zn, Mg and Ca) and vitamins (C and E) supplementation in the prevention of preeclampsia. en.search.irct.ir/view/31125 (first received 24 September 2016). CENTRAL

Baqui 2009 {published data only}

Baqui A. Antenatal vitamin d supplementation to improve neonatal health outcomes in Dhaka, Bangladesh: preliminary dose‐finding and safety study. clinicaltrials.gov/ct2/show/NCT00938600 (first received 14 July 2009). [NCT00938600]CENTRAL
Roth DE, Al A, Raqib R, Akhtar E, Black RE, Baqui AH. Pharmacokinetics of high‐dose weekly oral vitamin D3 supplementation during the third trimester of pregnancy in Dhaka, Bangladesh. Nutrients 2013;5(3):788‐810. CENTRAL
Roth DE, Al Mahmud A, Raqib R, Black RE, Baqui AH. Pharmacokinetics of a single oral dose of vitamin D3 (70,000 IU) in pregnant and non‐pregnant women. Nutrition Journal 2012;11(114):doi: 10.1186/1475‐2891‐11‐114. CENTRAL
Roth DE, Al‐Mahmud A, El S, Raqib R, Black RE, Baqui AH. Randomized open‐label trial of two weekly oral vitamin D3 supplementation regimens during the third trimester of pregnancy in Bangladeshi women: Effects on maternal vitamin D status and safety. FASEB Journal 2011;25(Suppl):236.6. CENTRAL

Bhatia 2010 {published data only}

Bhatia V. A study of the effect of vitamin D supplementation in pregnant women, on the growth and biochemical features of the newborn baby and infant. ctri.nic.in/Clinicaltrials/pdf_generate.php?trialid=1674&EncHid=&modid=&compid= (first received 19 May 2010). [CTRI/2010/091/000499]CENTRAL
Kalra P, Das V, Agarwal A, Kumar M, Ramesh V, Bhatia E, et al. Effect of vitamin D supplementation during pregnancy on neonatal mineral homeostasis and anthropometry of the newborn and infant. British Journal of Nutrition 2012;108(6):1052‐8. CENTRAL
Sahu M, Das V, Aggarwal A, Rawat V, Saxena P, Bhatia V. Vitamin D replacement in pregnant women in rural north India: a pilot study. European Journal of Clinical Nutrition 2009;63(9):1157‐9. CENTRAL

Bhatia 2012 {published data only}

Bhatia V. Vitamin D supplementation in pregnancy: regimens and long term effects on offspring. Clinical Trials Registry ‐ India CTRI/2012/02/0023952012. [CTRI/2012/02/002395]CENTRAL
Bhatia V, Katam K, Agarwal A, Das V, Ramesh V. Vitamin D supplementation in pregnancy and its effect on cord blood 25 hydroxycholecalciferol and anthropometry of the newborn. Hormone Research in Paediatrics 2013;80(Suppl 1):220. CENTRAL
Sahoo SK, Katam KK, Das V, Agarwal A, Bhatia V. Maternal vitamin d supplementation in pregnancy and offspring outcomes: a double‐blind randomized placebo‐controlled trial. Journal of Bone and Mineral Metabolism 2017;35(4):464‐71. CENTRAL

Bisgaard 2009 {published data only}

Bisgaard H. Vitamin D supplementation during pregnancy for prevention of asthma in childhood (ABCvitaminD). clinicaltrials.gov/ct2/show/NCT00856947 (first received 6 March 2009). CENTRAL
Chawes B, Bønnelykke K, Stokholm J, Heickendorff L, Brix S, Rasmussen M, et al. Effect of vitamin D3 supplementation during pregnancy on risk of persistent wheeze in the offspring: a randomised clinical trial. Clinical and Translational Allergy 2016;6:6 [OP10]. CENTRAL
Chawes BL, Bonnelykke K, Stokholm J, Vissing NH, Bjarnadottir E, Schoos AM, et al. Effect of vitamin D3 supplementation during pregnancy on risk of persistent wheeze in the offspring: a randomized clinical trial. JAMA 2016;315(4):353‐61. CENTRAL

Chandy 2016 {published data only}

Chandy DD, Kare J, Singh SN, Agarwal A, Das V, Singh U, et al. Effect of vitamin D supplementation, directly or via breast milk for term infants, on serum 25 hydroxyvitamin D and related biochemistry, and propensity to infection: a randomised placebo‐controlled trial. British Journal of Nutrition 2016;116(1):52‐8. CENTRAL

Cockburn 1980 {published data only}

Cockburn F, Belton NR, Purvis RJ, Giles MM, Brown JK, Turner TL, et al. Maternal vitamin D intake and mineral metabolism in mothers and their newborn infants. British Medical Journal 1980;281(6232):11‐4. CENTRAL

Czech‐Kowalska 2013 {published data only}

Czech‐Kowalska J, Latka‐Grot J, Bulsiewicz D, Jaworski M, Pludowski P, Chazan B, et al. Influence of vitamin D supplementation on vitamin D status and bone mass during lactation ‐ double blinded randomized control trial. Annals of Nutrition & Metabolism 2013;63(Suppl 1):796, Abstract no: PO1128. CENTRAL

Das 2010 {published data only}

Das V. Vitamin D and calcium nutrition in pregnancy‐evaluation of optimal supplementation dose of vitamin D during antenatal period. ctri.nic.in/Clinicaltrials/pmaindet2.php?trialid=1553 (first received 2 June 2010). [CTRI/2010/091/000352]CENTRAL

Dawodu 2013 {published data only}

Dawodu A, Saadi HF, Bekdache G, Javed Y, Altaye M, Hollis BW. Randomized controlled trial (RCT) of vitamin D supplementation in pregnancy in a population with endemic vitamin D deficiency. Journal of Clinical Endocrinology & Metabolism 2013;98(6):2337‐46. CENTRAL

de Menibus 1984 {published data only}

de Menibus CH, Mallet E, Henocq A, Lemeur H. [Should vitamin d supplements be given to pregnant women?]. Bulletin De L'academie Nationale De Medecine 1984;168(7‐8):909‐16. CENTRAL

Etemadifar 2015 {published data only}

Etemadifar M, Janghorbani M. Efficacy of high‐dose vitamin D3 supplementation in vitamin D deficient pregnant women with multiple sclerosis: Preliminary findings of a randomized‐controlled trial. Iranian Journal of Neurology 2015;14(2):67‐73. CENTRAL

Gerais 2015 {published data only}

Gerais AS, Hussein S. Vitamin D deficiency in pregnancy and pregnancy outcome. International Journal of Gynecology and Obstetrics 2015;131(Suppl 5):E352. CENTRAL

Hashemipour 2014 {published data only}

Abotorabi S, Hashemi poor S, Esmailzadehha N, Ziaee A, Khoeiniha MH. Effect of treatment with Vitamin D on maternal and neonatal indices in pregnant women with hypocalcemia: a randomized controlled trial. International Journal of Pediatrics 2017;5(9):5733‐9. CENTRAL
Hashemipour S. Effect of treatment of vitamin D deficiency during pregnancy on hypocalcemia. clinicaltrials.gov/ct2/show/NCT02021864 (first received 27 December 2013). CENTRAL
Hashemipour S, Ziaee A, Javadi A, Movahed F, Elmizadeh K, Javadi EH, et al. Effect of treatment of vitamin D deficiency and insufficiency during pregnancy on fetal growth indices and maternal weight gain: a randomized clinical trial. European Journal of Obstetrics & Gynecology and Reproductive Biology 2014;172:15‐9. CENTRAL

Hossain 2012 {published data only}

Hossain N, Kanani F, Khanani R, Ayaz S, Pal L. Effect of maternal supplementation with vitamin D during pregnancy on neonatal serum vitamin D levels and anthropometric measurements. International Journal of Gynecology and Obstetrics 2012;119(Suppl 3):S372. CENTRAL
Hossain N, Kanani FH, Ramzan S, Kausar R, Ayaz S, Khanani R, et al. Obstetric and neonatal outcomes of maternal vitamin D supplementation: results of an open‐label, randomised controlled trial of antenatal vitamin D supplementation in Pakistani women. Journal of Clinical Endocrinology and Metabolism 2013;99(7):2448‐55. CENTRAL

Ito 1994 {published data only}

Ito M, Koyama H, Ohshige A, Maeda T, Yoshimura T, Okamura H. Prevention of preeclampsia with calcium supplementation and vitamin D3 in an antenatal protocol. International Journal of Gynecology & Obstetrics 1994;47(2):115‐20. CENTRAL

Jamilian 2016 {published data only}

Jamilian M, Karamali M, Taghizadeh M, Sharifi N, Jafari Z, Memarzadeh MR, et al. Vitamin D and evening primrose oil administration improve glycemia and lipid profiles in women with gestational diabetes. Lipids 2016;51(3):349‐56. CENTRAL

Jamilian 2017 {published data only}

Jamilian M, Samimi M, Ebrahimi FA, Hashemi T, Taghizadeh M, Razavi M, et al. The effects of vitamin D and omega‐3 fatty acid co‐supplementation on glycemic control and lipid concentrations in patients with gestational diabetes. Journal of Clinical Lipidology 2017;11(2):459‐68. CENTRAL

Kachhawa 2014 {published data only}

Gupta T, Kachhawa G, Sharma H, Bajpai J, Kulshreshtha V, Khadgawat R, et al. A randomized double blind controlled trial to investigate the effects of vitamin D supplementation on maternal and new‐born baby's vitamin D status in Asian‐Indian subjects. Indian Journal of Endocrinology and Metabolism 2017;21(8 Suppl 2):S44‐S45. CENTRAL
Kachhawa G. A randomized controlled trial to investigate the effects of vitamin D supplementation on maternal and new‐born baby's vitamin D status in Asian‐Indian subjects ‐ VIDIMAN. http://ctri.nic.in/Clinicaltrials/pdf_generate.php?trialid=8544&EncHid=&modid=&compid=%27,%278544det%27 (first received 13 June 2014). CENTRAL

Karamali 2014 {published data only}

Karamali M, Asemi Z, Ahmadi‐Dastjerdi M, Esmaillzadeh A. Calcium plus vitamin D supplementation affects pregnancy outcomes in gestational diabetes: randomized, double‐blind, placebo‐controlled trial. Public Health Nutrition 2016;19(1):156‐63. CENTRAL
Karamali M, IRCT201407115623N23. Effect of vitamin D plus calcium co‐supplementation on pregnancy outcomes in gestational diabetes. en.search.irct.ir/view/19118 (first received 21 July 2014). CENTRAL

Karamali 2015 {published data only}

Karamali M, Beihaghi E, Mohammadi AA, Asemi Z. Effects of high‐dose vitamin D supplementation on metabolic status and pregnancy outcomes in pregnant women at risk for pre‐eclampsia. Hormone and Metabolic Research 2015;47(12):867‐82. CENTRAL

Kermack 2017 {published data only}

Kermack AJ, Calder PC, Macklon NS, Fisk HL, Houghton FD, Lowen PK, et al. Prepare trial: a randomised double blinded controlled trial of a preconception omega 3 and vitamin d rich dietary supplement in couples undergoing assisted reproduction treatment. Human Reproduction 2017;32:i290. CENTRAL

Kiely 2015 {published data only}

Kiely M. Randomized controlled trial to determine the nutritional requirement for vitamin d for prevention of deficiency during pregnancy and in the early neonatal period (D‐MAT). clinicaltrials.gov/ct2/show/NCT02506439 (first received first received 23 July 2015). [NCT02506439]CENTRAL
O'Callaghan K, Hennessy A, Dowling KG, Hull G, Kenny LC, Cashman KD, et al. Estimation of the dietary requirement for vitamin D in pregnancy: a dose‐response, double‐blind, randomized placebo‐controlled trial. Annals of Nutrition and Metabolism 2017;71(Suppl 2):624. CENTRAL
O'Callaghan K, Hennessy A, Kiely M, Kenny LC. Habitual calcium and vitamin D intakes in pregnant Irish women; preliminary data from the DMAT randomised controlled trial. Proceedings of the Nutrition Society 2016;75(OCE3):E191. CENTRAL
O'Callaghan KM, Hennessy A, Hull GL, Healy K, Ritz C, Kenny LC, et al. Estimation of the maternal vitamin D intake that maintains circulating 25‐hydroxyvitamin D in late gestation at a concentration sufficient to keep umbilical cord sera >/=25‐30 nmol/L: a dose‐response, double‐blind, randomized placebo‐controlled trial in pregnant women at northern latitude. American Journal of Clinical Nutrition 2018;108(1):77‐91. CENTRAL

Lalooha 2012 {published data only}

Lalooha F. The effect of vitamin D supplementation during pregnancy on newborn's anthropometric index. en.irct.ir/trial/10079IRCT (first received 16 August 2012). CENTRAL

Li 2016 {published data only}

Li Q, Xing B. Vitamin D3‐supplemented yogurt drink improves insulin resistance and lipid profiles in women with gestational diabetes mellitus: a randomized double blinded clinical trial. Annals of Nutrition & Metabolism 2016;68(4):285‐90. CENTRAL

MacDonald 1986 {published data only}

MacDonald HN. Fetal and maternal benefits from calcium and vitamin D supplementation of pregnant Asians. Personal communication1986. CENTRAL

March 2010 {published data only}

Anon. Erratum for March et al. Maternal vitamin D3 supplementation at 50 μg/d protects against low serum 25‐hydroxyvitamin D in infants at 8 wk of age: a randomised controlled trial of 3 doses of vitamin D beginning in gestation and continued in lactation. Am J Clin Nutr 2015;102:402–10. American Journal of Clinical Nutrition 2016;104(5):1491. CENTRAL
Chen NN, March K, Innis SM, Shand A, von Dadelszen P, Lyon P, et al. The effect of vitamin D supplementation during pregnancy and lactation on maternal and infant 25‐hydroxyvitamin D (25OHD) concentration. FASEB Journal2013; Vol. 27, issue Suppl 1. CENTRAL
March K. Vitamin D dose‐response study throughout pregnancy and lactation. clinicaltrials.gov/ct2/show/NCT01112891 (first received 29 April 2010). CENTRAL
March KM, Chen NN, Karakochuk CD, Shand AW, Innis SM, von Dadelszen P, et al. Maternal vitamin D3 supplementation at 50 ug/d protects against low serum 25‐hydroxyvitamin D in infants at 8 wk of age: a randomized controlled trial of 3 doses of vitamin D beginning in gestation and continued in lactation. American Journal of Clinical Nutrition 2015;102(2):402‐10. CENTRAL

Marya 1981 {published data only}

Marya RK, Rathee S, Lata V, Mudgil S. Effects of vitamin D supplementation in pregnancy. Gynecologic and Obstetric Investigation 1981;12:155‐61. CENTRAL

McLean 2012 {published data only}

McLean M. Effect of high‐dose versus low‐dose vitamin D supplementation in pregnancy on maternal glucose metabolism and the risk of gestational diabetes. anzctr.org.au/Trial/Registration/TrialReview.aspx?id=308236 (first received 30 October 2012). CENTRAL

Mojibian 2015 {published data only}

Mojibian M. The effect of 50000 IU Vitamin D supplement administered two weekly on neonatal and pregnant women outcome. en.irct.ir/trial/7525 (first received 27 February 2012). [IRCT201107237097N1]CENTRAL
Mojibian M, Soheilykhah S, Fallah Zadeh MA, Jannati Moghadam M. The effects of vitamin D supplementation on maternal and neonatal outcome: a randomized clinical trial. Iranian Journal of Reproductive Medicine 2015;13(11):687‐96. CENTRAL

Mozzafari 2010 {published data only}

Hosseinzadeh‐Shamsi‐Anar M, Mozaffari‐Khosravi H, Salami MA, Hadinedoushan H, Mozayan MR. The efficacy and safety of a high dose of vitamin d in mothers with gestational diabetes mellitus: A randomized controlled clinical trial. Iranian Journal of Medical Sciences 2012;37(3):159‐65. CENTRAL
Mozaffari H. Effect of vitamin D supplementation on glucose status, lipid profiles and inflammatory factors in mothers with a history of gestational diabetes. en.irct.ir/trial/3967 (first received 21 May 2010). CENTRAL
Mozaffari‐Khosravi H, Hosseinzadeh‐Shamsi‐Anar M, Salami MA, Hadinedoushan H, Mozayan MR. Effects of a single post‐partum injection of a high dose of vitamin D on glucose tolerance and insulin resistance in mothers with first‐time gestational diabetes mellitus. Diabetic Medicine 2012;29(1):36‐42. CENTRAL

Mutlu 2013 {published data only}

Mutlu GY, Ozsu E, Kalaca S, Yuksel A, Cizmecioglu FM, Hatun S. The evaluation of vitamin D supplementation dose during pregnancy in a high‐risk population. Hormone Research in Paediatrics 2013;80(Suppl 1):92. CENTRAL
Mutlu GY, Ozsu E, Kalaca S, Yuksel A, Pehlevan Y, Cizmecioglu F, et al. Evaluation of vitamin D supplementation doses during pregnancy in a population at high risk for deficiency. Hormone Research in Paediatrics 2014;81(6):402‐8. CENTRAL

Nausheen 2014 {published data only}

Nausheen S. Assessment of dose effectiveness of vitamin D supplementation during pregnancy. clinicaltrials.gov/ct2/show/NCT02215213 (first received 13 August 2014). CENTRAL

Niramitmahapanya 2017 {published data only}

Niramitmahapanya S, Kaoiean S, Sangtawesin V, Bordeerat NK, Deerochanawong C. Correlation of 25‐hydroxyvitamin d levels in serum vs breastmilk in vitamin d‐supplementation breastfeeding women during lactation: randomized double blinded control trial. Endocrine Reviews2017; Vol. 38, issue 3. CENTRAL

Pandey 2015 {published data only}

Pandey U, Divakar H. Role of vitamin D in iron metabolism, a pilot study. International Journal of Reproduction, Contraception, Obstetrics and Gynecology 2015;4(5):1494‐8. CENTRAL

Qian 2015 {published data only}

Qian L, Wang H, Wu F, Li M, Chen W, Lv L. Vitamin D3 alters toll‐like receptor 4 signaling in monocytes of pregnant women at risk for preeclampsia. International Journal of Clinical and Experimental Medicine 2015;8(10):18041‐9. CENTRAL

Razavi 2017 {published data only}

Razavi M, Jamilian M, Samimi M, Afshar Ebrahimi F, Taghizadeh M, Bekhradi R, et al. The effects of vitamin d and omega‐3 fatty acids co‐supplementation on biomarkers of inflammation, oxidative stress and pregnancy outcomes in patients with gestational diabetes. Nutrition & Metabolism 2017;14:80. CENTRAL

Rostami 2018 {published data only}

Rostami M, Ramezani Tehrani F, Simbar M, Bidhendi Yarandi R, Minooee S, Hollis BW, et al. Effectiveness of prenatal vitamin D deficiency screening and treatment program: a stratified randomized field trial. Journal of Clinical Endocrinology and Metabolism2018; Vol. 103, issue 8:2936–48. CENTRAL
Rostami M, Ramezani Tehrani F, Simbar M, Hosseinpanah F, Alavi Majd H. Rationale and design of Khuzestan vitamin D deficiency screening program in pregnancy: a stratified randomized vitamin D supplementation controlled trial. JMIR Research Protocols 2017;6(4):e54. CENTRAL

Shakiba 2013 {published data only}

Shakiba M, Iranmanesh MR. Vitamin D requirement in pregnancy to prevent deficiency in neonates: A randomised trial. Singapore Medical Journal2013; Vol. 54, issue 5:285‐8. CENTRAL

Shi 2017 {published data only}

Shi DD, Wang Y, Guo JJ, Zhou L, Wang N. Vitamin d enhances efficacy of oral nifedipine in treating preeclampsia with severe features: a double blinded, placebo‐controlled and randomized clinical trial. Frontiers in Pharmacology 2017;8:865. CENTRAL

Simsek 2011 {published data only}

Simsek S. Vitamin D supplementation in gestational diabetes mellitus. trialregister.nl/trialreg/admin/rctview.asp?TC=3158 (first received 21 November 2011). CENTRAL

Soheilykhah 2013 {published data only}

Soheilykhah S. Effect of different doses of vitamin D on insulin resistance in pregnant women attending in Shahid Sadoughi and Mojibian prenatal clinics. en.irct.ir/trial/3385 (first received 23 May 2011). [IRCT138811203312N1]CENTRAL
Soheilykhah S, Mojibian M, Moghadam MJ, Shojaoddiny‐Ardekani A. The effect of different doses of vitamin D supplementation on insulin resistance during pregnancy. Gynecological Endocrinology 2013;29(4):396‐9. CENTRAL

Stephensen 2011 {published data only}

Stephensen CB. Effects of vitamin D supplementation during pregnancy on clinical outcomes and immune function. clinicaltrials.gov/ct2/show/NCT01417351(first received August 2011). CENTRAL
Zerofsky M, Jacoby B, Pedersen TL, Stephensen CB. Effects of a randomized, controlled trial of daily vitamin D3 supplementation during pregnancy on regulatory immunity and inflammation. FASEB Journal 2016;30(Suppl 1):296.7. CENTRAL
Zerofsky M, Jacoby B, Stephensen C. A randomized controlled trial of vitamin D supplementation in pregnancy: Effects on vitamin D status and clinical outcomes. FASEB Journal 2014;28(1 Suppl 1):[Abstract no. 1041.5]. CENTRAL
Zerofsky MS, Jacoby BN, Pedersen TL, Stephensen CB. Daily cholecalciferol supplementation during pregnancy alters markers of regulatory immunity, inflammation, and clinical outcomes in a randomized controlled trial. Journal of Nutrition 2016;146(11):2388‐97. CENTRAL

Sudfeld 2017 {published data only}

Sudfeld CR, Manji KP, Duggan CP, Aboud S, Muhihi A, Sando DM, et al. Effect of maternal vitamin d3 supplementation on maternal health, birth outcomes, and infant growth among hiv‐infected tanzanian pregnant women: study protocol for a randomized controlled trial. Trials 2017;18(1):411. CENTRAL

Taheri 2014 {published data only}

Taheri M, Baheiraei A, Rahimi A, Modarres M. Resolving vitamin d deficiency in the preconception period among high‐risk reproductive women: A randomized controlled trial. Iranian Red Crescent Medical Journal 2014;16(1):e11175. CENTRAL

Thiele 2014 {published data only}

Anderson CM, Thiele DK, Ralph JL, Perley D, OHM JE. Vitamin D supplementation and DNA methylation patterns during pregnancy in and lactation in mothers and infants. FASEB Journal 2016;30(Suppl 1):Abstract no: 1028.3. CENTRAL
Keesling Thiele D. The Impact of Continuous Prenatal and Early Postpartum Maternal Vitamin D Supplementation on the Vitamin D status of Exclusively Breastfed Infants [thesis]. University of North Dakota, 2013. CENTRAL
Thiele D, Anderson CM. CS_018. Maternal vitamin D supplementation increases infant vitamin D status at birth but the impact diminishes during breastfeeding. 17th Conference of the International Society for Research in Human Milk and Lactation (ISRHML); 2014 Oct 23‐27; Kiawah Island, South Carolina, USA. 2014:122. CENTRAL
Thiele DK, Ralph J, El‐Masri M, Anderson CM. Vitamin d3 supplementation during pregnancy and lactation improves vitamin d status of the mother‐infant dyad. Journal of Obstetric, Gynecologic, and Neonatal Nursing : Jognn 2017;46:135‐47. CENTRAL

Valizadeh 2016 {published data only}

Valizadeh M, Piri Z, Mohammadian F, Kamali K, Amir Moghadami HR. The impact of vitamin D supplementation on post‐partum glucose tolerance and insulin resistance in gestational diabetes: A randomized controlled trial. International Journal of Endocrinology and Metabolism 2016;14(2):e34312. CENTRAL

von Hurst 2009 {published data only}

von Hurst PR. The role of vitamin D in metabolism and bone health. A thesis presented in partial fulfilment of the requirements for the Degree of Doctor of Philosophy in Nutritional Science. Vol. 1, Albany: Massey University, 2009. CENTRAL
von Hurst PR, Stonehouse W, Coad J. Vitamin D supplementation reduces insulin resistance in South Asian women living in New Zealand who are insulin resistant and vitamin D deficient ‐ a randomised, placebo‐controlled trial. British Journal of Nutrition 2010;103(4):549‐55. CENTRAL
von Hurst PR, Stonehouse W, Matthys C, Conlon C, Kruger MC, Coad J. Study protocol‐‐metabolic syndrome, vitamin D and bone status in South Asian women living in Auckland, New Zealand: a randomised, placebo‐controlled, double‐blind vitamin D intervention. BMC Public Health 2008;8:267. CENTRAL

Wagner 2006a {published data only}

Appelgren KE, Nietert PJ, Hulsey TC, Hollis BW, Wagner CL. Analyzing adherence to prenatal supplement: does pill count measure up?. International Journal of Endocrinology 2010;2010:1‐8. CENTRAL
Hollis BW, Johnson D, Hulsey TC, Ebeling M, Wagner CL. Vitamin D supplementation during pregnancy: Double‐blind, randomized clinical trial of safety and effectiveness. Journal of Bone and Mineral Research 2011;26(10):2341‐57. CENTRAL
Wagner CL. Evaluation of vitamin D requirements during pregnancy (ongoing trial). clinicaltrials.gov/ct2/show/NCT00292591 (first received 16 February 2006). [NCT00292591]CENTRAL
Wagner CL, Johnson D, Hulsey TC, Ebeling M, Shary J, Smith PG, et al. Vitamin D supplementation during pregnancy part 2 NICHD/CTSA randomized clinical trial (RCT): outcomes. Pediatric Academic Societies' 2010 Annual Meeting; 2010 May 1‐4; Vancouver, Canada. 2010. CENTRAL
Wagner CL, Johnson D, Hulsey TC, Ebeling M, Shary J,  Smith PG, et al. Vitamin D supplementation during pregnancy Part I NICHD/CTSA randomized clinical trial (RCT): safety consideration. Pediatric Academic Societies Annual Meeting; 2010 May 1‐4; Vancouver, Canada. 2010. CENTRAL
Wei W, Shary JR, Garrett‐Mayer E, Anderson B, Forestieri NE, Hollis BW, et al. Bone mineral density during pregnancy in women participating in a randomized controlled trial of vitamin d supplementation. American Journal of Clinical Nutrition 2017;106(6):1422‐30. CENTRAL

Wagner 2006b {published data only}

Wagner CL. Vitamin D status of pregnant women and their children in Eau Claire, South Carolina: a prevalence and supplementation model for community health care centers in the U.S. clinicaltrials.gov/ct2/show/NCT00412087 (first received 15 December 2006). CENTRAL
Wagner CL, McNeil R, Hamilton SA, Davis DJ, Prudgen C, Winkler J, et al. Vitamin D (vitD) supplementation during pregnancy: Thrasher Research Fund RCT in SC community center networks. Pediatric Academic Societies 2010 Annual Meeting; 2010 May 1‐4; Vancouver, Canada. 2010. CENTRAL
Wagner CL, McNeil R, Hamilton SA, Winkler J, Rodriguez Cook C, Warner G, et al. A randomized trial of vitamin D supplementation in 2 community health center networks in South Carolina. American Journal of Obstetrics and Gynecology 2013;208(2):137.e1‐137.e13. CENTRAL
Wagner CL, McNeil RB, Johnson DD, Hulsey TC, Ebeling M, Robinson C, et al. Health characteristics and outcomes of two randomized vitamin D supplementation trials during pregnancy: A combined analysis. Journal of Steroid Biochemistry & Molecular Biology 2013;136:313‐20. CENTRAL
Wagner CL, Sen S, Ebeling M, Hollis BW. BMI and vitamin D status are inversely related in breastfeeding mothers through 7 months. Breastfeeding Medicine 2016;11(2):A‐75, Abstract no:P‐121. CENTRAL

Wagner 2013 {published data only}

Moore RS, Mulligan JK, Harmon H, Hollis BW, Wagner CL. Immune mediators and vitamin D status in the development of comorbidities of pregnancy. Pediatric Academic Societies Annual Meeting; 2016 April 30‐May 3; Baltimore, USA. 2016:4109.85. CENTRAL
Schulz EV, Wagner CL, Cruze L, Wei W, Gehris J. Maternal vitamin d sufficiency and reduced placental gene expression in angiogenic biomarkers related to comorbidities of pregnancy. Journal of Steroid Biochemistry and Molecular Biology 2017;173:273‐9. CENTRAL
Wagner CL. Preventing health disparities during pregnancy through vitamin D supplementation. clinicaltrials.gov/ct2/show/NCT01932788 (first received 30 August 2013). CENTRAL

Weiss 2009 {published data only}

Al‐Garawi A, Carey VJ, Chhabra D, Mirzakhani H, Morrow J, Lasky‐Su J, et al. The role of vitamin D in the transcriptional program of human pregnancy. PLOS One 2016;11(10):e0163832. CENTRAL
Al‐Garawi A, Carey VJ, Chhabra D, Morrow J, Lasky‐Su J, Koh A, et al. Differentially expressed genes during the course of pregnancy and their correlation with maternal vitamin d levels. American Journal of Respiratory and Critical Care Medicine 2015;191:A5994. CENTRAL
Al‐Garawi A, Carey VJ, Qiu W, Mirzakhani H, Litonjua AA, Weiss ST. Cord blood vitamin D levels and gene expression profiles at birth are associated with wheezing in the first year of life: results from the vitamin D antenatal asthma reduction trial (vdaart). American Journal of Respiratory and Critical Care Medicine 2016;193:A3168. CENTRAL
Bhaskaran K, Smeeth L, Evans S. Prenatal vitamin d and offspring wheezing. JAMA 2016;315(24):2730. CENTRAL
Blighe K, Chawes BL, Kelly RS, Mirzakhani H, McGeachie M, Litonjua AA, et al. Vitamin d prenatal programming of childhood metabolomics profiles at age 3 y. American Journal of Clinical Nutrition 2017;106(4):1092‐9. CENTRAL
Grotegut CA. Prevention of preeclampsia. Journal of Clinical Investigation 2016;126(12):4396‐8. CENTRAL
Hornsby E, Pfeffer PE, Hawrylowicz C, Laranjo N, Litonjua AA, Weiss ST, et al. Vitamin d supplementation during pregnancy: effect on the neonatal immune system in a randomized controlled trial. Journal of Allergy and Clinical Immunology 2018;141(1):269. CENTRAL
Litonjua A, Weiss T. Prenatal vitamin d and offspring wheezing‐‐reply. JAMA 2016;315(24):2731. CENTRAL
Litonjua AA, Carey VJ, Laranjo N, Harshfield BJ, McElrath TF, O'Connor GT, et al. Effect of prenatal supplementation with vitamin D on asthma or recurrent wheezing in offspring by age 3 years: the VDAART randomized clinical trial. JAMA 2016;315(4):362‐70. CENTRAL
Litonjua AA, Lange NE, Carey VJ, Brown S, Laranjo N, O'Connor GT, et al. The Vitamin D Antenatal Asthma Reduction Trial (VDAART): Rationale, design, and methods of a randomized, controlled trial of vitamin D supplementation in pregnancy for the primary prevention of asthma and allergies in children. Contemporary Clinical Trials 2014;38:37‐50. CENTRAL
Merenstein D, D'Amico F. Prenatal vitamin d and offspring wheezing. JAMA 2016;315(24):2731. CENTRAL
Mirzakhani H, O'Connor G, Bacharier LB, Zeiger RS, Schatz MX, Weiss ST, et al. Asthma control status in pregnancy, body mass index, and maternal vitamin D levels. Journal of Allergy and Clinical Immunology 2017;140(5):1453‐6.e7. CENTRAL
Mirzakhani H, O'Connor GT, Bacharier L, Zeiger RS, Schatz MX, Weiss S, et al. Asthma control status during pregnancy and its relationship with vitamin D level: an observation from the vitamin D antenatal asthma reduction trial (VDAART). American Journal of Respiratory and Critical Care Medicine2017; Vol. 195, issue no pagination. CENTRAL
Sordillo JE, McGeachie MJ, Ziniti J, Lange N, Laranjo N, Carey V, et al. Factors influencing the infant gut microbiome at age 3‐6 months: findings from the ethnically diverse Vitamin D Antenatal Asthma Reduction Trial (VDAART). Journal of Allergy and Clinical Immunology 2017;139(2):482‐91. CENTRAL
Weiss NCT. Randomized trial: maternal vitamin d supplementation to prevent childhood asthma (vdaart). clinicaltrials.gov/ct2/show/NCT00920621 (first received 15 June 2009). CENTRAL
Wolsk HM, Harshfield BJ, Laranjo N, Carey VJ, O'Connor GT, Sandel M, et al. Vitamin d supplementation in pregnant women of different races and the risk of asthma/recurrent wheeze in the child: findings from the vitamin d antenatal asthma reduction trial (vdaart). American Journal of Respiratory and Critical Care Medicine 2016;193:A3169. CENTRAL
Wolsk HM, Harshfield BJ, Laranjo N, Carey VJ, Weiss ST, Litonjua AA, et al. Vitamin d supplementation in pregnancy, prenatal 25(oh)d levels, race, and subsequent asthma or recurrent wheeze in offspring: secondary analyses from the vitamin d antenatal asthma reduction trial. Journal of Allergy and Clinical Immunology 2017;140(5):1423. CENTRAL

Yap 2014 {published data only}

Yap C, Cheung NW, Gunton JE, Athayde N, Munns CF, Duke A, et al. Erratum. Vitamin d supplementation and the effects on glucose metabolism during pregnancy: a randomized controlled trial. Diabetes care 2014;37:1837‐1844. Diabetes Care 2015;38(10):1992. CENTRAL
Yap C, Cheung NW, Gunton JE, Athayde N, Munns CF, Duke A, et al. Vitamin D supplementation and the effects on glucose metabolism during pregnancy: a randomized controlled trial. Diabetes Care 2014;37(7):1837‐44. CENTRAL

Yazdchi 2016 {published data only}

Yazdchi R, Gargari BP, Asghari‐Jafarabadi M, Sahhaf F. Effects of vitamin D supplementation on metabolic indices and hs‐CRP levels in gestational diabetes mellitus patients: a randomized, double‐blinded, placebo‐controlled clinical trial. Nutrition Research and Practice 2016;10(3):328‐35. CENTRAL

Zhang 2016 {published data only}

Zhang Q, Cheng Y, He M, Li T, Ma Z, Cheng H. Effect of various doses of vitamin D supplementation on pregnant women with gestational diabetes mellitus: a randomized controlled trial. Experimental and Therapeutic Medicine 2016;12(3):1889‐95. CENTRAL

Referencias de los estudios en espera de evaluación

Bimson 2017 {published data only}

Bimson B, Brustman L, Al Ibraheemi Z, Herrera K, Rosenn B. Can weekly vitamin D dosing adequately treat vitamin D deficiency in pregnancy?. American Journal of Obstetrics and Gynecology 2017;216(1):S380‐S381. CENTRAL

Das 2009 {published data only}

Das V, Agarwal A, Bhatia V, Pandey A, Agarwal S, Saxena P, et al. Evaluation of vitamin d status and need for supplementation in pregnant women of a rural area of North India (abstract 0205). International Journal of Gynecology & Obstetrics 2009;107 (Suppl 2):S151. CENTRAL

Baird 2016 {published data only}

Baird J, Barker M, Harvey NC, Lawrence W, Vogel C, Jarman M, et al. Southampton PRegnancy Intervention for the Next Generation (SPRING): protocol for a randomised controlled trial. Trials [Electronic Resource] 2016;17(1):493. CENTRAL

Jelsma 2013 {published data only}

Jelsma JG, van Poppel MN, Galjaard S, Desoye G, Corcoy R, Devlieger R, et al. DALI: Vitamin D and lifestyle intervention for gestational diabetes mellitus (GDM) prevention: an European multicentre, randomised trial ‐ study protocol. BMC Pregnancy and Childbirth 2013;13:142. CENTRAL

Judkins 2010 {published data only}

Judkins A. A randomised double blinded interventional trial to determine the effect of 50,000 IU vitamin D supplementation monthly or twice monthly from 20 weeks gestation. anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12610001044011 23 November 2010. CENTRAL

Lindqvist 2010 {published data only}

Lindqvist P. Vitamin D supplementation for prevention of placenta mediated pregnancy complications. ‐ Prevention of pregnancy complications with vitamin D. https://www.clinicaltrialsregister.eu/ctr‐search/search?query=eudract_number:2010‐019483‐37 (first received 4 June 2010). CENTRAL

Mosalanejad 2016 {published data only}

Mosalanejad N, IRCT2016121430612N2. Compare the effect of vitamin D and calcium plus vitamin D on pregnancy outcomes in pregnant women. en.search.irct.ir/view/34642 (first received 26 December 2016). CENTRAL

Rasmussen 2009 {published data only}

Rasmussen G. Additional information on registered trial. Effects of vitamin D supplement before, during and after pregnancy on complications, birth weight and bone mineral density during lactation (gravita). Personal communication2011. CENTRAL
Rasmussen GB. Effects of vitamin D supplement before and during pregnancy on birth weight (gravita). clinicaltrials.gov/ct2/show/NCT01038453 (first received 24 December 2009). CENTRAL

ACOG 2015

American College of Obstetricians and Gynecologists. Vitamin D: screening and supplementation during pregnancy. Committee Opinion No. 495. Obstetrics and Gynecology 2011 (Reaffirmed 2015) 2015;118:197–8.

Aghajafari 2013

Aghajafari F, Nagulesapillai T, Ronksley PE, Tough SC, O'Beirne M, Rabi DM. Association between maternal serum 25‐hydroxyvitamin D level and pregnancy and neonatal outcomes: systematic review and meta‐analysis of observational studies. BMJ 2013;346:f1169.

Akcakus 2006

Akcakus M, Koklu E, Budak N, Kula M, Kurtoglu S, Koklu S. The relationship between birthweight, 25‐hydroxyvitamin D concentrations and bone mineral status in neonates. Annals of Tropical Paediatrics 2006;26(4):267‐75.

Ariyuki 1987

Ariyuki F. Growth retardation induced in rat fetuses by maternal fasting and massive doses of ergocalciferol. Journal of Nutrition 1987;117(2):342‐8.

Armas 2004

Armas LA, Hollis BW, Heaney RP. Vitamin D2 is much less effective than vitamin D3 in humans. Journal of Clinical Endocrinology and Metabolism 2004;89(11):5387‐91.

Arunabh 2003

Arunabh S, Pollack S, Yeh J, Aloia JF. Body fat content and 25‐hydroxyvitamin D levels in healthy women. Journal of Clinical Endocrinology and Metabolism 2003;88(1):157‐61.

August 1992

August P, Marcaccio B, Gertner JM, Druzin ML, Resnick LM, Laragh JH. Abnormal 1,25‐dihydroxyvitamin D metabolism in preeclampsia. American Journal of Obstetrics and Gynecology 1992;166(4):1295‐9.

Bandeira 2006

Bandeira F, Griz L, Dreyer P, Eufrazino C, Bandeira C, Freese E. Vitamin D deficiency: a global perspective [Deficiência de vitamina D: uma perspectiva global]. Arquivos Brasileiros de Endocrinologia e Metabologia 2006;50(4):640‐6.

Bener 2009

Bener A, Alsaied A, Al‐Ali M, Al‐Kubaisi A, Basha B, Abraham A, et al. High prevalence of vitamin D deficiency in type 1 diabetes mellitus and healthy children. Acta Diabetologica 2009;4(2):183‐9.

Bodnar 2007

Bodnar LM, Catov JM, Simhan HN, Holick MF, Powers RW, Roberts JM. Maternal vitamin D deficiency increases the risk of preeclampsia. Journal of Clinical Endocrinology and Metabolism 2007;92(9):3517‐22.

Bodnar 2010

Bodnar LM,  Catov JM,  Zmuda JM,  Cooper ME,  Parrott MS,  Roberts JM,  et al. Maternal serum 25‐hydroxyvitamin D concentrations are associated with small‐for‐gestational age births in white women. Journal of Nutrition 2010;140(9):999‐1006.

Butte 2002

Butte NF, Lopez‐Alarcon MG, Garza C. Nutrient Adequacy of Exclusive Breastfeeding of the Term Infant During the First Six Months of life. Geneva: World Health Organization, 2002.

Canadian Paediatric Society 2007

Canadian Paediatric Society, First Nations, Inuit and Métis Health Committee. Vitamin D supplementation: recommendations for Canadian mothers and infants. Paediatrics & Child Health 2007;12(7):583‐98.

Cantorna 2008

Cantorna MT, Yu S, Bruce D. The paradoxical effects of vitamin D on type 1 mediated immunity. Molecular Aspects of Medicine 2008;29:369‐75.

Cardus 2006

Cardus A, Parisi E, Gallego C, Aldea M, Fernandez E, Valdivielso JM. 1,25‐Dihydroxyvitamin D3 stimulates vascular smooth muscle cell proliferation through a VEGF‐mediated pathway. Kidney International 2006;69:1377‐84.

Chan 1979

Chan GM, Buchino JJ, Mehlhorn D, Bove KE, Steichen JJ, Tsang RC. Effect of vitamin D on pregnant rabbits and their offspring. Pediatric Research 1979;13(2):121‐6.

Chocano‐Bedoya 2009

Chocano‐Bedoya P, Ronnenberg AG. Vitamin D and tuberculosis. Nutrition Reviews 2009;67(5):289‐93.

Christian 2003

Christian P, Khatry SK, Katz J, Pradhan EK, LeClerq SC, Shrestha SR, et al. Effects of alternative maternal micronutrient supplements on low birth weight in rural Nepal: double blind randomised community trial. BMJ 2003;326(7389):571.

Clemens 1982

Clemens TL, Adams JS, Henderson SL, Holick MF. Increased skin pigment reduces the capacity of skin to synthesise vitamin D3. Lancet 1982;319:74‐6.

Clifton‐Bligh 2008

Clifton‐Bligh RJ, McElduff P, McElduff A. Maternal vitamin D deficiency, ethnicity and gestational diabetes. Diabetic Medicine 2008;25(6):678‐84.

Cope 2016

Committee on Publication Ethics (COPE). What to do if you suspect plagiarism flowchart. https://publicationethics.org/files/Full%20set%20of%20English%20flowcharts_9Nov2016.pdf2016.

Dawodu 2005

Dawodu A, Agarwal M, Sankarankutty M, Hardy D, Kochiyil J, Badrinath P. Higher prevalence of vitamin D deficiency in mothers of rachitic than nonrachitic children. Journal of Pediatrics 2005;147(1):109‐11.

Dawodu 2011

Dawodu A,  Nath R. High prevalence of moderately severe vitamin D deficiency in pre‐term infants. Pediatrics International2011; Vol. 53, issue 2:207‐10.

Dawson‐Hughes 2005

Dawson‐Hughes B, Heaney RP, Holick MF, Lips P, Meunier PJ, Vieth R. Estimates of optimal vitamin D status. Osteoporosis International 2005;16:713‐6.

Dawson‐Hughes 2008

Dawson‐Hughes B. Serum 25‐hydroxyvitamin D and functional outcomes in the elderly. American Journal of Clinical Nutrition 2008;88(2):527S‐540S.

DeLuca 2004

DeLuca HF. Overview of general physiologic features and functions of vitamin D. American Journal of Clinical Nutrition 2004;80(6 Suppl):1689S‐1696S.

Devereux 2007

Devereux G, Litonjua AA, Turner SW, Craig LC, McNeill G, Martindale S, et al. Maternal vitamin D intake during pregnancy and early childhood wheezing. American Journal of Clinical Nutrition 2007;85(3):853‐9.

Diaz 2002

Diaz L, Arranz C, Avila E, Halhai A, Vilchis F, Larrae F. Expression and activity of 25‐hydroxyvitamin D‐1 alpha‐hydroxylase are restricted in cultures of human syncytiotrophoblast cells from preeclamptic pregnancies. Journal of Clinical Endocrinology and Metabolism 2002;87(8):3876‐82.

Drincic 2012

Drincic AT, Armas LA, Van Diest EE, Heaney RP. Volumetric dilution, rather than sequestration best explains the low vitamin D status of obesity. Obesity (Silver Spring, Md.) 2012;20(7):1444‐8.

Eckard 2013

Eckard AR, Leong T, Avery A, Castillo MD, Bonilla H, Storer N, et al. Short communication: high prevalence of vitamin D deficiency in HIV‐infected and HIV‐uninfected pregnant women. AIDS Research and Human Retroviruses 2013;29(9):1224‐8.

EFSA 2016

European Food Safety Authority (EFSA). Scientific Opinion on Dietary Reference Values for vitamin D. European Food Safety Authority Journal2016.

Egan 2008

Egan KM, Signorello LB, Munro HM, Hargreaves MK, Hollis BW, Blot WJ. Vitamin D insufficiency among African‐Americans in the southeastern United States: implications for cancer disparities (United States). Cancer Causes and Control 2008;19(5):527‐35.

El Koumi 2013

El Koumi MA, Ali YF, Abd El Rahman RN. Impact of maternal vitamin D status during pregnancy on neonatal vitamin D status. Turkish Journal of Pediatrics 2013;55(4):371‐7.

Evans 2004

Evans KN, Bulmer JN, Kilby MD, Hewison M. Vitamin D and placental‐decidual function. Journal of the Society for Gynecologic Investigation 2004;11(5):263‐71.

Farrant 2009

Farrant HJ,  Krishnaveni GV,  Hill JC,  Boucher BJ,  Fisher DJ,  Noonan K,  et al. Vitamin D insufficiency is common in Indian mothers but is not associated with gestational diabetes or variation in newborn size. European Journal of Clinical Nutrition 2009;63(5):646‐52.

Fitzpatrick 1988

Fitzpatrick TB. The validity and practicality of sun‐reactive skin types I through VI. Archives of Dermatology 1988;124(6):869‐73.

Ford 1973

Ford JA, Davidson DC, McIntosh WB, Fyfe WM, Dunnigan MG. Neonatal rickets in Asian immigrant population. BMJ 1973;3(5873):211‐2.

Frenkel 1991

Frenkel Y, Barkai G, Mashiach S, Dolev E, Zimlichmann R, Weiss M. Hypocalciuria of preeclampsia is independent of parathyroid hormone level. Obstetrics & Gynecology 1991;77:822‐5.

Friedman 1969

Friedman WF, Mills LF. The relationship between vitamin D and the craniofacial and dental anomalies of the supravalvular aortic stenosis syndrome. Pediatrics 1969;43(1):12‐8.

Gale 2008

Gale CR, Robinson SM, Harvey NC, Javaid MK, Jiang B, Martyn CN, et al. Maternal vitamin D status during pregnancy and child outcomes. European Journal of Clinical Nutrition 2008;62(1):68‐77.

Ganji 2012

Ganji V, Zhang X, Tangpricha V. Serum 25‐hydroxyvitamin D concentrations and prevalence estimates of hypovitaminosis D in the U.S. population based on assay‐adjusted data. Journal of Nutrition 2012;142(3):498‐507.

Gilchrest 2008

Gilchrest BA. Sun exposure and vitamin D sufficiency. American Journal of Clinical Nutrition 2008;88:570S‐577S.

Glerup 2000

Glerup H, Mikkelsen K, Poulsen L, Hass E, Overbeck S, Andersen H, et al. Hypovitaminosis D myopathy without biochemical signs of osteomalacic bone involvement. Calcified Tissue International 2000;66(6):419‐24.

GRADE Handbook

Schünemann H, Brożek J, Guyatt G, Oxman A, editors. Handbook for grading the quality of evidence and the strength of recommendations using the GRADE approach (updated October 2013). GRADE Working Group, 2013. Available from gdt.guidelinedevelopment.org/app/handbook/handbook.html.

GRADEpro 2015 [Computer program]

McMaster University (developed by Evidence Prime). GRADEpro GDT. Version Accessed 12 July 2018. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015.

Halhali 1995

Halhali A, Bourgues H, Carrillo A, Garabedian M. Lower circulating insulin‐like growth factor I and 1,25‐dihydroxyvitamin D levels in preeclampsia. Revista de Investigacion Clinica 1995;47(4):259‐66.

Halhali 2000

Halhali A, Tovar AR, Torres N, Bourgues H, Garabedian M, Larrae F. Preeclampsia is associated with low circulating levels of insulin‐like growth factor I and 1,25‐dihydroxyvitamin D in maternal and umbilical cord compartments. Journal of Clinical Endocrinology and Metabolism 2000;85(5):1828‐33.

Hall 2010

Hall WB, Sparks AA, Aris RM. Vitamin D deficiency in cystic fibrosis. International Journal of Endocrinology 2010;2010:218691.

Harvey 2014

Harvey NC, Holroyd C, Ntani G, Javaid K, Cooper P, Moon R, et al. Vitamin D supplementation in pregnancy: a systematic review. Health Technology Assessment 2014;18(45):1‐190.

Hathcock 2007

Hathcock JN, Shao A, Vieth R, Heaney R. Risk assessment for vitamin D. American Journal of Clinical Nutrition 2007;85(1):6‐18.

Heaney 2003

Heaney RP, Davies KM, Chen TC, Holick MF, Barger‐Lux MJ. Human serum 25‐hydroxycholecalciferol response to extended oral dosing with cholecalciferol. American Journal of Clinical Nutrition 2003;77(1):204‐10.

Heaney 2008

Heaney RP. Vitamin D: criteria for safety and efficacy. Nutrition Reviews 2008;66(10 Suppl 2):S178‐S181.

Hewison 1992

Hewison M. Vitamin D and the immune system. Journal of Endocrinology 1992;132(2):173‐6.

Higgins 2011

Higgins JP, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org.

Holick 2002

Holick MF. Too little vitamin D in premenopausal women: why should we care?. American Society for Clinical Nutrition 2002;76:1, 3‐4.

Holick 2005

Holick MF, Siris ES, Binkley N, Beard MK, Khan A, Katzer JT, et al. Prevalence of Vitamin D inadequacy among postmenopausal North American women receiving osteoporosis therapy. Journal of Clinical Endocrinology and Metabolism 2005;90(6):3215‐24.

Holick 2007a

Holick MF. Vitamin D deficiency. New England Journal of Medicine 2007;357(3):266‐81.

Holick 2007b

Holick MF, Chen TC, Lu Z, Sauter E. Vitamin D and skin physiology: a D‐lightful story. Journal of Bone and Mineral Research 2007;22 Suppl 2:V28‐V33.

Holick 2008

Holick MF. Vitamin D deficiency: a worldwide problem with health consequences. American Journal of Clinical Nutrition 2008;87(4):1080S‐1086S.

Hollick 2009

Holick MF. Vitamin D status: measurement, interpretation and clinical application. Annals of Epidemiology 2009;19:73‐8.

Hollis 2004

Hollis BW, Wagner CL. Vitamin D requirements during lactation: high‐dose maternal supplementation as therapy to prevent hypovitaminosis D for both the mother and the nursing infant. American Journal of Clinical Nutrition 2004;80(6 Suppl):1752S‐178S.

Hollis 2007

Hollis B. Vitamin D requirement during pregnancy and lactation. Journal of Bone and Mineral Research 2007;22 Suppl 2:V39‐V44.

Hyppönen 2013

Hyppönen E, Cavadino A, Williams D, Fraser A, Vereczkey A, Fraser WD, et al. Vitamin D and pre‐eclampsia: original data, systematic review and meta‐analysis. Annals of Nutrition & Metabolism 2013;63(4):331‐40.

Ioannou 2012

Ioannou C, Javaid MK, Mahon P, Yaqub MK, Harvey NC, Godfrey KM, et al. The effect of maternal vitamin D concentration on fetal bone. Journal of Clinical Endocrinology and Metabolism 2012;97(11):E2070‐7.

IOM 2011

Institute of Medicine (IOM). Dietary Reference Intakes for Calcium and Vitamin D. 2nd Edition. Washington DC: National Academy Press, 2011.

Javaid 2006

Javaid MK, Crozier SR, Harvey NC, Gale CR, Dennison EM, Boucher BJ, et al. Princess Anne Hospital Study Group. Maternal vitamin D status during pregnancy and childhood bone mass at age 9 years: a longitudinal study. Lancet 2006;367:36‐43.

Jones 2008

Jones G. Pharmacokinetics of vitamin D toxicity. American Journal of Clinical Nutrition 2008;88(2):582S‐586S.

Karras 2018

Karras SN, Wagner CL, Castracane VD. Understanding vitamin D metabolism in pregnancy: From physiology to pathophysiology and clinical outcomes. Metabolism 2018;86:112‐23.

Lankes 2015

Lankes U, Elder PA, Lewis JG, George P. Differential extraction of endogenous and exogenous 25‐OH‐vitamin D from serum makes the accurate quantification in liquid chromatography‐tandem mass spectrometry assays challenging. Annals of Clinical Biochemistry 2015;52(Pt 1):151‐60.

Levis 2005

Levis S, Gomez A, Jimenez C, Veras L, Ma F, Lai S, et al. Vitamin D deficiency and seasonal variation in an adult south Florida population. Journal of Clinical Endocrinology and Metabolism 2005;90(3):1557‐62.

Li 2000b

Li DK, Wi S. Maternal pre‐eclampsia/eclampsia and the risk of sudden infant death syndrome in offspring. Paediatric and Perinatal Epidemiology 2000;14(2):141‐4.

Li 2002

Li Y, Kong J, Wei M, Chen ZF, Liu S, Cao LP. 1,25‐dihydroxyvitamin D3 is a negative endocrine regulator of the renin‐angiotensin system. Journal of Clinical Investigation 2002;110(2):229‐39.

Lips 2001

Lips P. Vitamin D deficiency and secondary hyperparathyroidism in the elderly: consequences for bone loss and fractures and therapeutic implications. Endocrine Reviews 2001;22(4):477‐501.

Litonjua 2009

Litonjua AA. Childhood asthma may be a consequence of vitamin D deficiency. Current Opinion in Allergy and Clinical Immunology 2009;9(3):202‐7.

Logan 2013

Logan VF, Gray AR, Peddie MC, Harper MJ, Houghton LA. Long‐term vitamin D3 supplementation is more effective than vitamin D2 in maintaining serum 25‐hydroxyvitamin D status over the winter months. British Journal of Nutrition 2013;109(6):1082‐8.

MacKay 2001

MacKay AP, Berg CJ, Atrash HK. Pregnancy‐related mortality from preeclampsia and eclampsia. Obstetrics & Gynecology 2001;97(4):533‐8.

Maghbooli 2007

Maghbooli Z, Hossein‐Nezhad A, Shafaei AR, Karimi F, Madani FS, Larijani B. Vitamin D status in mothers and their newborns in Iran. BMC Pregnancy and Childbith 2007;7:1.

Maghbooli 2008

Maghbooli Z, Hossein‐Nezhad A, Karimi F, Shafaei AR, Larijani B. Correlation between vitamin D3 deficiency and insulin resistance in pregnancy. Diabetes/Metabolism Research and Reviews 2008;24(1):27‐32.

Mahon 2010

Mahon P, Harvey N, Crozier S, Inskip H, Robinson S, Arden N, et al. Low maternal vitamin D status and fetal bone development: cohort study. Journal of Bone and Mineral Research2010; Vol. 25, issue 1:14‐9.

Matsuoka 1991

Matsuoka LY, Wortsman J, Haddad JG, Kolm P, Hollis BW. Racial pigmentation and the cutaneous synthesis of vitamin D. Archives of Dermatology 1991;127(4):536‐8.

Mave 2012

Mave V, Shere D, Gupte N, Suryavanshi N, Kulkarni V, Patil S, et al. SWEN India and Byramjee‐Jeejeebhoy Medical College Clinical Trials Unit Study Team. Vitamin D deficiency is common among HIV‐infected breastfeeding mothers in Pune, India, but is not associated with mother‐to‐child HIV transmission. HIV Clinical Trials 2012;13(5):278‐83.

Maxwell 1981

Maxwell JD, Ang L, Brooke OG, Brown IR. Vitamin D supplements enhance weight gain and nutritional status in pregnant Asians. British Journal of Obstetrics and Gynaecology 1981;88(10):987‐91.

McCullough 2007

McCullough  M. Vitamin D deficiency in pregnancy: bringing the issues to light. Journal of Nutrition 2007;137:305‐6.

McGrath 2001

McGrath J. Does 'imprinting' with low prenatal vitamin D contribute to the risk of various adult disorders?. Medical Hypotheses 2001;56(3):367‐71.

Mehta 2009

Mehta S, Hunter DJ, Mugusi FM, Spiegelman D, Manji KP, Giovannucci EL, et al. Perinatal outcomes, including mother‐to‐child transmission of HIV, and child mortality and their association with maternal vitamin D status in Tanzania. Journal of Infectious Diseases2009; Vol. 200, issue 7:1022‐30.

Merewood 2009

Merewood A, Mehta SD, Chen TC, Bauchner H, Holick MF. Association between vitamin D deficiency and primary cesarean section. Journal of Clinical Endocrinology and Metabolism 2009;94(3):940‐5.

Miller 2010

Miller J, Gallo RL. Vitamin D and innate immunity. Dermatologic Therapy 2010;23(1):13‐22.

Moller 2013

Møller UK, Streym S, Mosekilde L, Heickendorff L, Flyvbjerg A, Frystyk J, et al. Changes in calcitropic hormones, bone markers and insulin‐like growth factor I (IGF‐I) during pregnancy and postpartum: a controlled cohort study. Osteoporosis International 2013;24(4):1307‐20.

Morley 2006

Morley R, Carlin JB, Pasco JA, Wark JD. Maternal 25‐hydroxyvitamin D and parathyroid hormone concentrations and offspring birth size. Journal of Clinical Endocrinology and Metabolism 2006;91(3):906‐12.

Namgunga 2003

Namgunga R, Tsang RC. Bone in the pregnant mother and newborn at birth. Clinica Chimica Acta 2003;333(1):1‐11.

Nesby‐O'Dell 2002

Nesby‐O'Dell S, Scanlon KS, Cogswell ME, Gillespie C, Hollis BW, Looker AC, et al. Hypovitaminosis D prevalence and determinants among African American and white women of reproductive age: third National Health and Nutrition Examination Survey, 1988‐1994. American Journal of Clinical Nutrition 2002;76(1):187‐92.

Nicolaidou 2006

Nicolaidou P, Hatzistamatiou Z, Papadopoulou A, Kaleyias J, Floropoulou E, Lagona E, et al. Low vitamin D status in mother‐newborn pairs in Greece. Calcified Tissue International 2006;78(6):337‐42.

Nnoaham 2008

Nnoaham KE, Clarke A. Low serum vitamin D levels and tuberculosis: a systematic review and meta‐analysis. International Journal of Epidemiology 2008;37(1):113‐9.

O'Riordan 2008

O'Riordan MN, Kiely M, Higgins JR, Cashman KD. Prevalence of suboptimal vitamin D status during pregnancy. Irish Medical Journal 2008;101(8):240, 242‐3.

Ohta 2009

Ohta H, Kuroda T, Onoe Y, Orito S, Ohara M, Kume M, et al. The impact of lifestyle factors on serum 25‐hydroxyvitamin D levels: across‐sectional study in Japanese women aged 19‐25 years. Journal of Bone and Mineral Metabolism 2009;27(6):682‐8.

Ornoy 1968

Ornoy A, Menczel J, Nebel L. Alterations in the mineral composition and metabolism of rat fetuses and their placentas induced by maternal hypervitaminosis D2. Israel Journal of Medical Sciences 1968;4(4):827‐32.

Ornoy 1969

Ornoy A, Nebel L, Menczel Y. Impaired osteogenesis of fetal long bones.Induced by maternal hypervitaminosis D2. Archives of Pathology 1969;87(6):563‐71.

Palacios 2014

Palacios C, Gonzalez L. Is vitamin D deficiency a major global public health problem?. Journal of Steroid Biochemistry and Molecular Biology 2014;144(Pt A):138‐45.

Palacios 2018

Palacios C, Lips P, Martinez RX, Lopez‐Perez L, Peña‐Rosas JP, Trak‐Fellermeier MA. Regimens of vitamin D supplementation for women during pregnancy. Prospero CRD42018103763 https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD420181037632018. [CRD42018103763]

Palomer 2008

Palomer X, González‐Clemente JM, Blanco‐Vaca F, Mauricio D. Role of vitamin D in the pathogenesis of type 2 diabetes mellitus. Diabetes, Obesity and Metabolism 2008;10(3):185‐97.

Perez‐Lopez 2015

Pérez‐López FR, Pasupuleti V, Mezones‐Holguin E, Benites‐Zapata VA, Thota P, Deshpande A, et al. Effect of vitamin D supplementation during pregnancy on maternal and neonatal outcomes: a systematic review and meta‐analysis of randomized controlled trials. Fertility and Sterility 2015;103(5):1278‐88.

Pierrot‐Deseilligny 2010

Pierrot‐Deseilligny C, Souberbielle JC. Is hypovitaminosis D one of the environmental risk factors for multiple sclerosis?. Brain 2010;133(Pt 7):1869‐88.

Pludowski 2013a

Pludowski P, Holick MF, Pilz S, Wagner CL, Hollis BW, Grant WB, et al. Vitamin D effects on musculoskeletal health, immunity, autoimmunity, cardiovascular disease, cancer, fertility, pregnancy, dementia and mortality‐a review of recent evidence. Autoimmunity Reviews 2013;12(10):976‐89.

Pludowski 2013b

Płudowski P, Karczmarewicz E, Bayer M, Carter G, Chlebna‐Sokół D, Czech‐Kowalska J, et al. Practical guidelines for the supplementation of vitamin D and the treatment of deficits in Central Europe — recommended vitamin D intakes in the general population and groups at risk of vitamin D deficiency. Endokrynologia Polska 2013;64(4):319‐27.

RCOG 2014

Royal College of Obstetricians and Gynaecologists. Vitamin D in Pregnancy. Scientific Impact Paper https://www.rcog.org.uk/globalassets/documents/guidelines/scientific‐impact‐papers/vitamin_d_sip43_june14.pdf2014; Vol. No. 43:1‐11.

RevMan 2014 [Computer program]

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

Rockell 2005

Rockell JE, Green TJ, Skeaff CM, Whiting SJ, Taylor RW, Williams SM, et al. Season and ethnicity are determinants of serum 25‐hydroxyvitamin D concentrations in New Zealand children aged 5–14 y. Journal of Nutrition 2005;135:2602‐8.

Roth 2011a

Roth DE. Vitamin D supplementation during pregnancy: safety considerations in the design and interpretation of clinical trials. Journal of Perinatology 2011;31(7):449‐59.

Roth 2017

Roth DE, Leung M, Mesfin E, Qamar H, Watterworth J, Papp E. Vitamin D supplementation during pregnancy: state of the evidence from a systematic review of randomised trials. BMJ 2017;359:j5237.

Roth 2018

Roth DE, Abrams SA, Aloia J, Bergeron G, Bourassa MW, Brown KH, et al. Global prevalence and disease burden of vitamin D deficiency: a roadmap for action in low‐ and middle‐income countries. Annals of the New York Academy of Sciences 2018;1430(1):44‐79.

Sachan 2005

Sachan A, Gupta R, Das V, Agarwal A, Awasthi PK, Bhatia V. High prevalence of vitamin D deficiency among pregnant women and their newborns in northern India. American Journal of Clinical Nutrition 2005;81(5):1060‐4.

Sahu 2009

Sahu M, Das V, Aggarwal A, Rawat V, Saxena P, Bhatia V. Vitamin D replacement in pregnant women in rural north India: a pilot study. European Journal of Clinical Nutrition 2009;63(9):1157‐9.

Scholl 2012

Scholl TO, Chen X, Stein P. Maternal vitamin D status and delivery by cesarean. Nutrients 2012;4(4):319‐30.

Sloka 2009

Sloka S, Stokes J, Randell E, Newhook LA. Seasonal variation of maternal serum vitamin D in Newfoundland and Labrador. Journal of Obstetrics and Gynaecology Canada: JOGC 2009;31(4):313‐21.

Tabesh 2013

Tabesh M, Salehi‐Abargouei A, Tabesh M, Esmaillzadeh A. Maternal vitamin D status and risk of pre‐eclampsia: a systematic review and meta‐analysis. Journal of Clinical Endocrinology and Metabolism 2013;98(8):3165‐73.

Theodoratou 2014

Theodoratou E, Tzoulaki I, Zgaga L, Ioannidis JP. Vitamin D and multiple health outcomes: umbrella review of systematic reviews and meta‐analyses of observational studies and randomised trials. BMJ 2014;348(g2035):1‐19.

Theodoropoulos 2003

Theodoropoulos C, Demers C, Delvin E, Ménard D, Gascon‐Barré M. Calcitriol regulates the expression of the genes encoding the three key vitamin D3 hydroxylases and the drug‐metabolizing enzyme CYP3A4 in the human fetal intestine. Clinical Endocrinology 2003;58(4):489‐99.

Thorne‐Lyman 2012

Thorne‐Lyman A, Fawzi WW. Vitamin D during pregnancy and maternal, neonatal and infant health outcomes: a systematic review and meta‐analysis. Paediatric and Perinatal Epidemiology 2012;26(Suppl 1):75‐90.

Tolaymat 1994

Tolaymat A, Sanchez‐Ramos L, Yergey AL, Vieira NE, Abrams SA, Edelstein P. Pathophysiology of hypocalciuria in preeclampsia: measurement of intestinal calcium absorption. International Journal of Gynecology & Obstetrics 1994;83(2):283.

UK Department of Health 2009

UK Department of Health. Maternal nutrition: Vitamin D. UK Department of Health (http://www.dh.gov.uk/en/Healthcare/Children/Maternity/Maternalandinfantnutrition/Maternalnutrition/index.htm)(accessed 2009).

Utiger 1998

Utiger RD. The need for more vitamin D. New England Journal of Medicine 1998;338(12):828‐9.

van der Pligt 2018

van der Pligt P, Willcox J, Szymlek‐Gay EA, Murray E, Worsley A, Daly RM. Associations of maternal vitamin D deficiency with pregnancy and neonatal complications in developing countries: a systematic review. Nutrients 2018;10(5):640.

van Schoor 2011

van Schoor NM, Lips P. Worldwide vitamin D status. Best Practice & Research. Clinical Endocrinology & Metabolism 2011;25(4):671‐80.

Vieth 1999

Vieth R. Vitamin D supplementation, 25‐hydroxyvitamin D concentrations, and safety. American Journal of Clinical Nutrition 1999;69(5):842‐56.

Vieth 2001

Vieth R, Chan PC, MacFarlane GD. Efficacy and safety of vitamin D3 intake exceeding the lowest observed adverse effect level. American Journal of Clinical Nutrition 2001;73(2):288‐94.

Vilarrasa 2007

Vilarrasa N, Maravall J, Estepa A, Sánchez R, Masdevall C, Navarro MA, et al. Low 25‐hydroxyvitamin D concentrations in obese women: their clinical significance and relationship with anthropometric and body composition variables. Journal of Endocrinological Investigation 2007;30(8):653‐8.

Viljakainen 2010

Viljakainen HT, Saarnio E, Hytinantti T, Miettinen M, Surcel H, Makitie O, et al. Maternal vitamin D status determines bone variables in the newborn. Journal of Clinical Endocrinology and Metabolism 2010;Epub:1391.

Vimaleswaran 2013

Vimaleswaran KS, Berry DJ, Lu C, Tikkanen E, Pilz S, Hiraki LT, et al. Causal relationship between obesity and vitamin D status: bi‐directional Mendelian randomisation analysis of multiple cohorts. PLOS Medicine 2013;10(2):e1001383.

Wagner 2008

Wagner CL, Greer FR, American Academy of Pediatrics Section on breastfeeding and American Academy of Pediatrics Committee on Nutrition. Prevention of rickets and vitamin D deficiency in infants, children, and adolescents. Pediatrics 2008;122(5):1142‐52.

Walker 2009

Walker VP, Modlin RL. The vitamin D connection to pediatric infections and immune function. Pediatrics Research 2009;65(5 Pt 2):106R‐113R.

Wang 2004

Wang TT, Nestel FP, Bourdeau V, Nagai Y, Wang Q, Liao J, et al. Cutting edge: 1,25‐dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. Journal of Immunology 2004;173:2909‐12.

Wei 2013

Wei SQ, Qi HP, Luo ZC, Fraser WD. Maternal vitamin D status and adverse pregnancy outcomes: a systematic review and meta‐analysis. Journal of Maternal‐fetal & Neonatal Medicine 2013;26(9):889‐99.

WHO 2004

World Health Organization and Food and Agriculture Organization. Vitamin and Mineral Requirements in Human Nutrition. 2nd Edition. Geneva: WHO, 2004.

WHO 2016

World Health Organization. WHO Recommendations on Antenatal Care for a Positive Pregnancy Experience. Geneva: World Health Organization, 2016.

Williams 2008

Williams B, Williams AJ, Anderson ST. Vitamin D deficiency and insufficiency in children with tuberculosis. Pediatric Infectious Disease Journal 2008;27(10):941‐2.

Wortsman 2000

Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF. Decreased bioavailability of vitamin D in obesity. American Journal of Clinical Nutrition 2000;72(3):690‐3.

Xiong 1999

Xiong X, Mayes D, Demianczuk N, Olson DM, Davidge ST, Newburn‐Cook C, et al. Impact of pregnancy‐induced hypertension on fetal growth. American Journal of Obstetrics and Gynecology 1999;180(1 Pt 1):207‐13.

Xuan 2013

Xuan Y, Zhao HY, Liu JM. Vitamin D and type 2 diabetes mellitus (D2). Journal of Diabetes 2013;5(3):261‐7.

Yu 2009

Yu CK, Sykes L, Sethi M, Teoh TG, Robinson S. Vitamin D deficiency and supplementation during pregnancy. Clinical Endocrinology 2009;70(5):685‐90.

Zehnder 2002

Zehnder D, Evans KN, Kilby MD, Bulmer JN, Innes BA, Stewart PM, et al. The ontogeny of 25‐hydroxyvitamin D(3) 1alpha‐hydroxylase expression in human placenta and decidua. American Journal of Pathology 2002;161(1):105‐14.

Zhang 2008

Zhang C, Qiu C, Hu FB, David RM, Van Dam RM, Bralley A, et al. Maternal plasma 25‐hydroxyvitamin D concentrations and the risk for gestational diabetes mellitus. PLOS One 2008;3(11):e3753.

Referencias de otras versiones publicadas de esta revisión

Ansary 2010

Ansary A, Palacios C, De‐Regil LM, Peña‐Rosas JP. Vitamin D supplementation for women during pregnancy. Cochrane Database of Systematic Reviews 2010, Issue 12. [DOI: 10.1002/14651858.CD008873]

De‐Regil 2012

De‐Regil LM, Palacios C, Ansary A, Kulier R, Peña‐Rosas JP. Vitamin D supplementation for women during pregnancy. Cochrane Database of Systematic Reviews 2012, Issue 2. [DOI: 10.1002/14651858.CD008873.pub2]

De‐Regil 2016

De‐Regil LM, Palacios C, Lombardo LK, Peña‐Rosas JP. Vitamin D supplementation for women during pregnancy. Cochrane Database of Systematic Reviews 2016, Issue 1. [DOI: 10.1002/14651858.CD008873.pub3]

Mahomed 1999

Mahomed K, Gülmezoglu AM. Vitamin D supplementation in pregnancy. Cochrane Database of Systematic Reviews 1999, Issue 1. [DOI: 10.1002/14651858.CD000228]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Asemi 2012

Methods

Randomised single‐blinded controlled trial with 2 arms: vitamin D plus calcium and placebo.

Participants

54 pregnant women at risk for pre‐eclampsia, primigravida, aged 18 to 35 years old carrying singleton pregnancy at their third trimester attending maternity clinics affiliated to Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran (latitude: 33.9889° N, 51.4772° E).

Exclusion criteria: maternal severe pre‐eclampsia, IUFD, placenta abortion, preterm delivery and GDM.

Interventions

Participants were randomly allocated to 1 of 2 groups: group 1 (n = 27): women received 500 mg of carbonate calcium plus 200 IU of vitamin D (cholecalciferol‐D3) daily for 9 weeks; group 2 (n = 27): women received placebo. The intervention lasted 9 weeks overall, starting at 25 weeks of pregnancy until week 34. Participants were asked not to alter their routine PA or usual diets and not to consume any supplement other than the one provided to them by the investigators.

Health worker cadre: the trial was carried out in maternity clinics affiliated to Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran and the investigators provided the supplements to the participants.

Outcomes

Maternal: body weight and height, BMI, fasting plasma glucose levels, serum total cholesterol, triglycerol concentrations, serum HDL‐cholesterol, serum LDL‐cholesterol levels, dietary intakes, total HDL: cholesterol ratio, gestational diabetes, severe pre‐eclampsia, preterm delivery.

Laboratory method used for assessment of vitamin D concentrations: serum 25‐hydroxyvitamin D concentrations were measured using a commercial ELISA kit (Immuno Diagnostic Systems). The inter‐ and intra‐assay coefficient of variation for serum 25(OH)D assays ranged from 5% to 7.5%.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU vitamin D or less;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): overweight;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: mixed/unknown.

Source of funding: study was funded by research grant from the Vice‐Chancellor for research, KUMS, and Iran.

Dates of the study and location: April 2011 to February 2012, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Trial reported randomisation performed by the use of computer‐generated random numbers.

Allocation concealment (selection bias)

Low risk

Trial reported that the appearance of the placebo capsules, such as colour, shape, size, and packaging, was identical to the vitamin D3 capsules.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial reported that it was single‐blinded. Participants were blinded to the interventions so it is assumed that the research staff were not blinded.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Trial is reported as single‐blinded and the methods for concealment of the intervention were described for participants. Therefore, it is assumed that it was not blinded to research staff.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Lost to follow‐up of 3 women in the vitamin D group due to preterm delivery (n = 1), IUFD (n = 1), and placental abruption (n = 1). 3 women in the placebo group were also excluded for the following reasons: gestational diabetes (n = 1), preterm delivery (n = 1), and severe pre‐eclampsia (n = 1).

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Asemi 2013a

Methods

Randomised, double‐blind, placebo‐controlled clinical trial with 2 arms: vitamin D and placebo, during March 2012 to September 2012.

Participants

48 healthy pregnant women, primigravida, aged 18–40 years old at 25 weeks of gestation and a singleton pregnancy attending maternity clinics affiliated with Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran. Women with pre‐eclampsia, hypertension, GDM, IUFD, or those with a history of rheumatoid arthritis, hepatic or renal failure, metabolic bone disease and malabsorption, or thyroid, parathyroid, or adrenal diseases were excluded from the analysis. Also, smokers and those taking medications including nonsteroidal antiinflammatory drugs and aspirin were excluded.

Interventions

Participants were randomly assigned to receive 1 of 2 groups: group 1 (n = 24) received 400 IU vitamin D (cholecalciferol‐D3) supplements daily; and group 2 (n = 24) received placebo for 9 weeks.

Additionally, all participants also consumed 400 mcg (0.4 mg) folic acid daily from the beginning of pregnancy and 60 mg elemental iron (as ferrous sulphate) daily from the second trimester.

Health worker cadre: the trial was carried out in maternity clinics affiliated to Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran and the investigators provided the supplements to the participants. A trained midwife at the maternity clinic performed anthropometric measurements at study baseline and at 6 weeks after the intervention.

Outcomes

Maternal: weight, height, BMI, systolic blood pressure and diastolic blood pressure, serum calcium concentrations, serum 25‐hydroxyvitamin D [25(OH)D], serum hs‐C‐reactive protein, fasting plasma glucose, serum cholesterol, LDL‐cholesterol, HDL‐cholesterol concentrations, serum insulin, quantitative Insulin sensitivity check index (QUICKI) score, plasma total antioxidant capacity, plasma total glutathione, GDM, preterm delivery, IUFD, placental abruption, severe pre‐eclampsia.

Laboratory method used for assessment of vitamin D concentrations: serum 25‐hydroxyvitamin D concentrations were measured using a commercial ELISA kit (Immuno Diagnostic Systems).

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU vitamin D or less;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): overweight (25 or higher);

  • supplementation scheme/regimen: daily in a 9‐week period;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: spring‐summer period.

Source of funding: study was funded by research grant from the Vice‐chancellor for Research, Kashan University of Medical Sciences, Kashan, Iran.

Dates of the study and location: March 2012 to September 2012, Kashan, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): there are no conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random assignment was performed by the use of computer‐generated random numbers.

Allocation concealment (selection bias)

Low risk

A trained midwife at the maternity clinic performed the randomised allocation sequence and assigned participants to the groups. Placebo pills contained microcrystalline cellulose and were packed in identical tablets and coded by the producer to guarantee blinding.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and investigators were blind to the interventions.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Measurements of laboratory were performed in a blinded fashion, in duplicate, in pairs (before/after intervention) at the same time, in the same analytical run, and in random order to reduce systematic error and inter assay variability.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

3 in each group were lost to follow‐up.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Benson 2009

Methods

Randomised controlled trial.

Participants

78 pregnant women between 14 to 18 weeks' gestation at risk, defined as: dark skinned, veiled; with vitamin D deficiency that has not commenced treatment prior to recruitment. Exclusion criteria: women taking barbiturates or anticonvulsants (decreased vitamin D absorption) and severe renal failure.

Interventions

Participants were individually randomised to 1 of 2 groups: group 1 (n = 38): 2000 IU of cholecalciferol orally daily commencing between 14 and 18 weeks' gestation (if still deficient at 28 weeks the dose was doubled to 4000 IU orally daily until birth); group 2 (n = 40): no treatment during pregnancy. The mother received 300,000 IU cholecalciferol orally immediately and the baby 150,000 IU cholecalciferol orally immediately after birth.

Health worker cadre: in order to facilitate compliance, encouragement was given from midwifery/medical staff at each 2–4 weekly antenatal visit with additional intervening telephone calls to women with poor compliance. Pill counts were not performed.

Outcomes

Maternal: vitamin D level.

Infant: vitamin D level.

Laboratory method used for assessment of vitamin D concentrations: serum 25‐OH vit D concentrations were determined by direct competitive chemiluminescence immunoassay for quantitative determination of total serum 25‐OH vit D (LIAISON®) Diasorin 25‐OH vitamin D assay (Stillwater, MN,USA).

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 20000 IU vitamin D;

  • start of supplementation: 14 to 18 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): mixed/unknown;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: south of Tropic of Capricorn;

  • season at the start of pregnancy: all year round.

Source of funding: J.E. Benson was a recipient of the Luke Proposch Perinatal Research Scholarship from the Australian and New Zealand College of Obstetrics and Gynaecology Research Foundation enabling her to undertake this research. Study was funded by research grant.

Dates of the study and location: between 2008 and 2009, Melbourne, Australia.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors have no conflict of interest to disclose.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomly allocated (envelopes in a tamper proof box, ratio 1:1).

Allocation concealment (selection bias)

Low risk

Envelopes in a tamper‐proof box, ratio 1:1.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial reported that it was single‐blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Single‐blinded study but authors did not specify if staff performing assessments were blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

57.9% intervention and 57.5% control data reported.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Bhutta 2011

Methods

Randomised, parallel assignment, double‐blind trial.

Participants

115 pregnant females from 12 to 20 weeks of gestation who agreed to participate in the study with presence of at least 20 natural teeth in mouth excluding third molars. For controls: non pregnant, healthy females matched with pregnant women with respect to age and education. Exclusion criteria: pregnant females with high vitamin D levels, women with metabolic diseases such as diabetes (type 1 or 2), presence of acute dental or periodontal disease, presence of systemic disease and/or medication affecting the periodontium; receipt of systemic antibiotic treatment or dental prophylaxis in the previous 3 months and those who do not provide informed consent.

Interventions

Participants were individually randomised to 1 of 2 groups: group 1 (n = 36): vitamin D3 4000 mg per day (given as 1 tablespoon syrup per day); and group 2 n = (49): placebo (given as 1 table spoon syrup per day,) for approximately 6 months.

Health worker cadre: CHWs were responsible for the delivery of supplementation to the study participants. The CHWs were assigned to visit study participants, on a fortnightly basis. The first supplementation was provided by the physician at the time of recruitment; later on, the CHWs continued to replenish the supply fortnightly.

Outcomes

Maternal: Periodontal Probing Depth, Interleukin 6 (IL‐6), IL‐2, IL‐4, IL‐10, TNF, IFN‐ɣ and IL‐17 levels.

Laboratory method used for assessment of vitamin D concentrations: vitamin D levels were analysed on DiaSorin‐LIASON Inc, kit.

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 200,000 IU;

  • start of supplementation: 12 to 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: unknown.

Source of funding: the study was supported with a research grant from Pakistan Initiative for Mothers and Newborns (PAIMAN).

Dates of the study and location: launched in 2004, Jhelum, Pakistan.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The study participants were randomised in blocks.

Allocation concealment (selection bias)

Low risk

Allocation codes for vitamin D and placebo were kept in a sealed envelope in a locked cabinet at the Aga Khan University until the completion of the study.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The investigators, study staff, and the participants were blinded about the group allocation.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Allocation codes for vitamin D and placebo were kept in a sealed envelope in a locked cabinet at the Aga Khan University until the completion of the study.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only recorded birthweight from 63/85 (74.1%) participants.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Brooke 1980

Methods

Randomised double‐blind controlled trial; 2‐arm design with individual randomisation.

Participants

126 Asian pregnant women 28 to 32 weeks of gestation attending the antenatal clinic at St George's Hospital, London, UK (latitude: 51°30'N, north of Tropic of Cancer). All pregnant women were first‐generation immigrants mostly from India, Pakistan, Bangladesh, Sri Lanka, Mauritius and east Africa.

Exclusion and elimination criteria: preterm deliveries, congenital malformations and maternal illnesses likely to affect fetal growth (such as diabetes) although these data are not presented.

Interventions

Participants were randomly allocated to 1 of 2 groups: group 1 (n = 59) received daily 1000 IU vitamin D (ergocalciferol‐D2) daily until term (estimated total dose: 56,000 to 84,000 IU); and group 2 (n = 67) received a placebo until term.

Start of supplementation: 28 to 32 weeks gestation.

Length of the intervention/follow‐up: 8 to 12 weeks from supplementation to term.

Health worker cadre: St George's Hospital Medical School, London, UK. Medical doctors that were part of the team conducted the measurements and provided the supplements.

Outcomes

Maternal: maternal weight gain, dietary vitamin D intake, 25‐hydroxyvitamin D (25‐OHD) concentrations in cord blood and at term. Plasma calcium (adjusted for albumin concentration), inorganic phosphate, bilirubin, albumin concentrations and total alkaline phosphatase activity, alanine transaminase and ʏ‐glutamyl transferase activities, vitamin D binding globulin concentration, compliance.

Infant: weight, crown‐heel length, crown‐rump length, rump‐heel length, occipitofrontal head circumference, forearm length, lower leg length, triceps and subscapular skinfold thickness, fontanelle area, plasma cholecalciferol at day 3 and day 6. weight, length and head circumference at 3, 6, 9 and 12 months.

Laboratory method used for assessment of vitamin D concentrations: Serum 25‐hydroxyvitamin D was measured by competitive protein binding assay after chromatographic purification of lipid extracts of serum.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 5 more than 56,000 to 200,000 IU;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: authors report that to avoid distortion of the results due to seasonal variation in sunlight hours the trial was carried out during autumn and winter 1977, the whole of 1978 and spring and summer 1979.

Source of funding: the pathological research fund, St George's Hospital Medical School, and the South‐west Thames Regional Health Authority. This study was funded by a combination of a research grant and non governmental organisations.

Dates of the study and location: autumn and winter 1977, the whole of 1978 and spring and summer 1979, London, UK.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Trial reported random allocation to the groups, although the method of sequence generation was not described.

Allocation concealment (selection bias)

Unclear risk

The trial reported that it was double‐blinded but the method of concealment was not described.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial reported that it was double‐blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial reported that it was double‐blinded but they did not specify if those performing the assessments were blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Unclear number of randomised participants. Preterm deliveries, congenital malformations, and maternal illnesses likely to affect fetal growth (such as diabetes) were eliminated from the trial. There is not complete documentation of the exclusions.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias. There were no significant baseline differences between the groups in maternal age, parity, height, vegetarian: non‐vegetarian ratio or the distribution of the various countries of origin.

Delvin 1986

Methods

Randomised trial; 2‐arm design with individual randomisation.

Participants

40 pregnant women attending their compulsory visit during the third month of pregnancy at the Obstetrical Unit of the Hopital Edouard Herriot, Lyon, France (latitude: 45° 45' 0" N north of Tropic of Cancer). Inclusion criterion: singleton pregnancy at term and uneventful vaginal deliveries. Pre‐gestational BMI and skin pigmentation not reported.

Interventions

Participants were randomly assigned to 1 of 2 groups at the time of the compulsory visit: group 1 (n = 20): women received daily 1000 IU vitamin D (cholecalciferol‐D3) (estimated total dose: 55,000 IU) and group 2 (n = 20): women received no supplement, during the last trimester of pregnancy for 12 weeks from start of supplementation to term.

Health worker cadre: compliance was verified by a weekly visit by a midwife.

Outcomes

Maternal: serum (during last trimester of pregnancy) and cord blood immunoreactive PTH, 25‐hydroxyvitamin D (25‐OHD), 1‐alfa,25‐dihydroxyvitamin D (1,25(OH)2D), total calcium, ionised calcium, magnesium, inorganic phosphate.

Infant: immunoreactive PTH, 25‐hydroxyvitamin D (25‐OHD), 1‐alfa,25‐dihydroxyvitamin D (1,25(OH)2D), total calcium, ionised calcium, magnesium, inorganic phosphate at 4 days of age.

Laboratory method used for assessment of vitamin D concentrations: Serum 25‐hydroxyvitamin D and 1,25‐dihydroxyvitamin D levels were measured by radioligand assays with slight modifications. With sample volumes of 0.75 mL to 1.5 mL, the inter assay variation coefficient for the 2 assays were 8% and 10%, respectively.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU vitamin D or less;

  • start of supplementation: 20 weeks of pregnancy, or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: winter‐spring. All selections were performed in December, and all deliveries occurred in June.

Source of funding: Shriners of North America, the France‐Quebec Exchange Program, and INSERM Grant 121023. This study was funded by a combination of research grant and non governmental organisations.

Dates of the study and location: not reported dates, Lyon, France.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Trial reported as randomised but the method of sequence generation was not described.

Allocation concealment (selection bias)

Unclear risk

The method of concealment was not described.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial reported that women were assigned, by a blind randomisation process, to 1 of 2 groups at the compulsory visit in the third month of pregnancy. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

There is insufficient information to permit judgement.

Incomplete outcome data (attrition bias)
All outcomes

High risk

1 participant from the control group (5%) and 5 (25%) from the vitamin D supplemented group were lost. Laboratory methods reported for 25 to 30 participants (depending on the outcome) out of 40 originally randomised.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Diogenes 2013

Methods

Randomised, placebo‐controlled trial; 2‐arm design with individual randomisation.

Participants

84 pregnant adolescents (13 to 19 years of age) primigravidae (pregnant for the first time) with singleton pregnancies and 23 to 29 weeks of gestation attending prenatal care at the Maternidade Escola, Universidade Federal do Rio de Janeiro, Brazil (latitude: 22.9083° S, 43.1964° W) from September 2009 to June 2011 and intending to exclusively or predominantly breast feed.

Women with chronic health problems, pregnancy complications, smokers, users of nutritional supplements besides iron plus folate supplements provided during standard prenatal care, and mothers who decided not to breast feed were excluded from the study.

Interventions

Participants were randomly assigned to: 1 of 2 groups: group 1 (n = 43) received a commercially available supplement (Rexall Sundown®) containing 600 mg calcium (as calcium carbonate) plus 200 IU vitamin D (cholecalciferol‐D3) daily and group 2 (n = 41) received placebo (capsules of microcrystalline cellulose and corn starch; Quintessencia) daily. The protocol allowed pregnant women to continue with their iron and folate supplements, as part of their standard prenatal care. The use and composition of these supplements was not provided.

Health worker cadre: capsules of calcium plus vitamin D or placebo were provided monthly to participants by a member of the research team during prenatal visits. Compliance was controlled by counting the remaining capsules at each visit and by telephone reminders. Calcium and vitamin D dietary intake was assessed by at least 3 24‐hour dietary recall questionnaires applied by a trained nutritionist. Standing height and body weight were measured by using a stadiometer (Seca) and a calibrated electronic scale (Filizola), respectively. The same operator performed all scanning and calibration.

Outcomes

Maternal: 1 measurement at 5 and 20 weeks postpartum, serum 25(OH)D, PTH, insulin‐like growth factor (IGF‐I), lumbar spine PA, bone mineral content, serum prolactin and oestradiol.

Laboratory method used for assessment of vitamin D concentrations: serum 25‐hydroxyvitamin D, intact PTH, and IGF‐I were analysed by using a chemiluminescent enzyme‐labelled immunometric assay.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU vitamin D or less;

  • start of supplementation: 20 weeks of pregnancy or more. The supplementation started from 26 weeks of pregnancy (baseline) until parturition;

  • pre‐gestational BMI (kg/m2): normal weight;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: the city lies on the Tropic of Capricorn;

  • season at the start of pregnancy: all year round.

Source of funding: Conselho Nacional de Desenvolvimento Cientıfico e Tecnologico [grant 471872/2008‐3 (to CMD) and a doctoral fellowship (to MELD)] and the Fundacao Carlos Chagas Filho de Amparo a` Pesquisa do Estado do Rio de Janeiro (grant E‐26/102.759/2008; to CMD), Brazil. This study was funded by a combination of Government programmes and non‐governmental organisations (NGOs).

Dates of the study and location: September 2009 to June 2011, Rio de Janeiro, Brazil.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none of the authors had a conflict of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Random assignment was done by a member of the research team in a 1:1 ratio within permuted blocks of size 10.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the method of concealment.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial reported that it was single‐blinded, only participants were blinded to the assigned groups. It is assumed that the assessment team was not blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Insufficient information to permit judgment.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Out of 43 women in the intervention group, 13 did not complete the study. Out of 41 women in the placebo group, 14 did not complete the study.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Unclear risk

The study appears to be free of other sources of bias.

Grant 2013

Methods

Randomised, double‐blind, placebo‐controlled multi‐arm parallel study.

Participants

260 pregnant women 26 to 30 weeks' gestation, with a singleton pregnancy attending community based primary care maternity clinic in Auckland, New Zealand (latitude 36°S) from April 2010 to July 2011 and then their infants, from birth to age 6 months.

Women already taking vitamin D supplementation 200 IU per day, a history of renal stones or hypercalcaemia, or any serious pregnancy complication at enrolment were excluded from the study.

Interventions

Participants were randomly assigned to 1 of 3 mother/infant groups: group 1 (n = 87) women received placebo from 26 to 30 weeks of pregnancy until parturition and their infants also received placebo from 0‐6 months of age; group 2 (n = 87) women received 1000 IU vitamin D (cholecalciferol‐D3) from 26 to 30 weeks of pregnancy until parturition and their infants received 400 IU vitamin D from 0 to 6 moths of age; group 3 (n = 86) women received 2000 IU vitamin D (cholecalciferol‐D3) from 26 to 30 weeks of pregnancy until parturition and their infants received 800 IU from birth to 6 months of age. Data from groups 2 and 3 were combined for our analysis.

Health worker cadre: the study was conducted by the research team but it is not reported who provided the supplements or measured the outcomes.

Outcomes

Maternal: serum 25(OH)D concentration.

Infant: serum 25(OH)D concentration.

Laboratory method used for assessment of vitamin D concentrations: serum 25‐hydroxyvitamin D concentration was measured using isotope‐dilution liquid chromatography–tandem mass spectrometry in a Vitamin D External Quality Assurance Scheme–certified laboratory.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU vitamin D or less;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude:between Tropics of Cancer and Capricorn;

  • season at the start of pregnancy: all year round.

Source of funding: Health Research Council of New Zealand, grant number 09/215R. Dr Mitchell is supported by Cure Kids. Study medicine was prepared by the Ddrops Company (Woodbridge, Ontario, Canada). This study was funded by a combination of government programmes and non‐governmental organisations (NGOs).

Dates of the study and location: April 2010 to July 2011, Auckland, New Zealand.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors have indicated they have no potential conflicts of interest to disclose.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Trial reported computer‐generated randomisation list.

Allocation concealment (selection bias)

Low risk

The allocation sequence was concealed from research staff involved in recruitment. Trial reported randomly allocated treatment to each participant and labelled identical study medicine bottles such that study staff and participants were unaware of the treatment status.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The study statistician randomly allocated a treatment to each participant and labelled identical study medicine bottles such that study staff and participants were unaware of the treatment status.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The study staff and participants were unaware of the treatment status.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Reported compliance did not differ between groups. In the placebo group, 6 did not complete the study; in the lower dose vitamin D group, 6 did not complete the study. In the higher vitamin D dose group, 6 did not complete the study.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Harvey 2012

Methods

Randomised, double‐blind, placebo‐controlled trial.

Participants

1200 pregnant women living in the UK, aged 18 years old and older, with a singleton pregnancy with less than 17 weeks' gestation at first assessment (based on last menstrual period and dating scan), aiming to give birth at local maternity hospital, and with serum 25‐hydroxyvitamin D is 25 to 100 nmol/L at nuchal fold/dating scan (10 to 17 weeks' gestation).

Interventions

Participants were randomly assigned to 1 of 2 groups: group 1 (n = 565): received 1000 IU cholecalciferol orally daily and group 2 (n = 569): received placebo, starting from 14 weeks' gestation until delivery.

Health worker cadre: the medication was blister packed in a single box for each woman for the duration of pregnancy. Study medication (active/placebo) was supplied to the local pharmacy pre‐randomised by the manufacturer (1:1, unstratified by centre) and sequentially numbered for storage and dispensing. Code break envelopes were supplied to the lead pharmacist, but were not available to the investigative team. Emergency code break access was available through the local principal investigator and on call pharmacist. A single pack for each participant was issued sequentially (containing all pills for duration of the study). Each pack was individually prescribed for each participant. The trials pharmacist allocated a pack to that prescription, documenting both the pack number and the MAVIDOS participant ID; these were checked again by the research nurse on collection, and documented in the participant’s notes; the medication pack came with a tear‐off adhesive label, which was placed in the participant’s notes as an added safeguard against errors in pack allocation. The research nurse collected the medication pack for all participants attending to the clinic that day and issued to the participants directly.

Outcomes

Infant: whole body bone mineral content of the neonate adjusted for gestational age and age at neonatal DXA scan, whole body bone area, bone mineral density, and size corrected bone mineral density (BMC adjusted for BA, length and weight), body composition adjusted for gestational age and age at DXA scan.

Laboratory method used for assessment of vitamin D concentrations: A blood sample was taken and plasma was stored at ‐80°C for measurement of 25(OH)‐vitamin D, vitamin D binding protein (DBP), calcium, bone specific alkaline phosphatase and albumin centrally (MRC Human Nutrition Research, Cambridge, UK) at the end of the study.

Notes

  • Total dose of supplementary vitamin D during pregnancy: less than 200,000 IU;

  • start of supplementation: less than 20 weeks of pregnancy;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of Tropic of Cancer;

  • season at the start of pregnancy: all year round.

Source of funding: Arthritis Research UK, Medical Research Council, Bupa Foundation, and National Institute for Health Research. Study was funded by a combination of research grants, government programmes, non‐governmental organizations (NGOs).

Dates of the study and location: October 2008 to February 2014, Southampton, Sheffield, Oxford, the UK.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): CC reports personal fees, consultancy, lecture fees, and honoraria from Alliance for Better Bone Health, Amgen, Eli Lilly, GlaxoSmithKline, Medtronic, Merck, Novartis, Pfizer, Roche, Servier, and Takeda, outside the submitted work. NCH reports personal fees, consultancy, lecture fees, and honoraria from Alliance for Better Bone Health, AMGen, MSD, Eli Lilly, Servier, Shire, Consilient Healthcare, and Internis Pharma, outside the submitted work. NJB reports remuneration from Internis Pharmaceuticals, outside the submitted work. ATP reports grants from the Arthritis Research Council, during the conduct of the study. NKA has received honoraria, held advisory board positions (which involved receipt of fees), and received consortium research grants from Merck, grants from Roche, Bioiberica, and Novartis, personal fees from Smith & Nephew, Nicox, Flexion, Bioventus, and Freshfields, outside the submitted work. KMG reports reimbursement for speaking at Nestle Nutrition Institute conferences, and grants from Abbott Nutrition and Nestec, outside the submitted work. KMG also has a patent pending for phenotype prediction, a patent pending for predictive use of CpG methylation, and a patent pending for maternal nutrition composition, not directly related to this work. HMI reports grants from the Medical Research Council (MRC), Arthritis Research UK, and European Union’s Seventh Framework Programme, during the conduct of the study; and while not directly receiving funding from other bodies, members of her team have received funding from the following companies from other work: Danone, Nestec, and Abbott Nutrition. RE reports grants and personal fees from Amgen and Alexion; grants from the Department of Health, AstraZeneca, ARUK/MRC Centre for Excellence in Musculoskeletal Ageing Research, National Institute for Health Research, MRC/AZ Mechanisms of Diseases Call, and the MRC; grants, personal fees, and non‐financial support from Immunodiagnostic Systems; grants and membership of a clinical and scientific committee from the National Osteoporosis Society; grants, personal fees, and advisory board membership from Roche; personal fees from Otsuka, Novartis, Merck, Bayer, Johnson & Johnson, Fonterra Brands, Janssen Research, Ono Pharma, Alere (Unipath), Chronos, Teijin Pharma Limited, D‐STAR, and GSK Nutrition; personal fees and advisory board membership from Eli Lilly, and CL Biosystems; and advisory board membership from the European Calcifi ed Tissue Society, IOF CSA, and the American Society for Bone and Mineral Research, outside the submitted work. MKJ reports personal fees from Stirling Anglia, Consilient Health, and Internis, outside the submitted work. All other authors declare no competing interests.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated sequence in randomly permuted blocks of 10.   

Allocation concealment (selection bias)

Low risk

The treatments were blister packed in a single box for each woman for the duration of pregnancy and supplied to the local pharmacy pre‐randomised by the manufacturer (1:1, unstratified by centre) and sequentially numbered for storage and dispensing. The lead pharmacist was the only one with access to the code break envelopes.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blinded, matched pills, only lead pharmacist knew about pills.     

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All assessments were double‐blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

94% of sample had vitD assessment at term.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

High risk

Participants were allowed to continue taking their own multivitamin with 400 IU/d of vitamin D but this was not recorded.

Kaur 1991

Methods

Randomised controlled trial.

Participants

50 pregnant women with similar socioeconomic conditions in India.

Interventions

Participants were randomised into 2 groups: group 1 (n = 25) received orally 2 pharmacological doses of vitamin D˜ (60,000 IU each) in 6th and 7th month of pregnancy; group 2 (n = 25) did not receive any vitamin supplement and served as controls.

Health worker cadre: not specified.

Outcomes

Infant: mean birthweight, placental weight and DNA content, total protein and RNA, protein/DNA and RNA/DNA ratios.

Laboratory method used for assessment of vitamin D concentrations: not applicable.

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 56,000 to 200,000 IU;

  • start of supplementation: 24 and 28 weeks of pregnancy;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: monthly;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of Tropic of Cancer;

  • season at the start of pregnancy: unknown.

Source of funding: unknown/unreported.

Dates of the study and location: not reported dates, India.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): not reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Authors only mentioned that women were randomly selected for trial initially.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the method of concealment. It is assumed that no method was used as one of the groups did not receive any supplementation.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report if it was blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

There is insufficient information to permit judgement.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

The number lost to follow‐up was not reported in results.

Selective reporting (reporting bias)

Unclear risk

The total number of participants that completed the study was not specified in results.

Other bias

Low risk

The study appears to be free of other sources of bias.

Li 2000a

Methods

Clinical controlled trial with 3 arms.

Participants

88 pregnant women with a predisposition to pregnancy‐induced hypertension, at 20 to 24 weeks' gestation, a BMI index of lower than 24, and an arterial pressure of < 11.3 kPa attending an outpatient clinic and labour ward of the First Afilliated Hospital of Xi’an Medical University, Xi’an, China.

Interventions

Participants were divided into 3 groups: group 1 (n = 29) received a daily dose of a tablet containing 600 mg of calcium and 200 IU of vitamin D (Caltrate‐D) daily from 20 to 24 weeks until deliver; group 2 (n = 29) received 1200 mg of calcium and 400 IU vitamin D (Caltrate‐D) daily from 20 to 24 weeks until deliver; group 3 (n = 30) received no intervention from 20 to 24 weeks until delivery.

Health worker cadre: not specified.

Outcomes

Maternal: blood pressure, ionised calcium and platelet intracellular calcium, incidence rates of pregnancy‐induced hypertension.

Laboratory method used for assessment of vitamin D concentrations: not applicable.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU vitamin D or less;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude:north of Tropic of Cancer;

  • season at the start of pregnancy: all year round.

Source of funding: unknown/unreported.

Dates of the study and location: August 1996 to December 1998, China.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

The trial did not report if participants were randomly allocated to the treatment groups. It is unclear if it was random or not.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the method of concealment. It is assumed that it was not conceal as one of the groups did not receive any supplementation

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report if it was blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not mention if the study was single or double blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Loss to follow‐up not reported.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to make a judgement.

Other bias

High risk

The report is very short, with most details of the methods not available.

Mallet 1986

Methods

Randomised controlled trial; 3‐arm design with individual randomisation.

Participants

77 white pregnant women 18 to 36 years of age in the last trimester of pregnancy living in Northwest of France (latitude: 49° 26' 0" N north of Tropic of Cancer). Pre‐gestational BMI not reported.

Interventions

Participants were randomly assigned to 1 of 3 groups: group 1 (n = 21) women received daily 1000 IU of vitamin D (ergocalciferol‐D2) for the last 3 months of pregnancy (estimated total dose throughout pregnancy: 90,000 IU); group 2 (n = 27) women received a single dose of 200,000 IU (5 mg) vitamin D at the 7th month of pregnancy; group 3 (n = 29) women received no supplement and served as controls.

Length of the intervention/follow‐up: 12 weeks from start of supplementation to term.

Health worker cadre: the study was conducted by the research team at the maternity of Balvedere, Rouen, France but the roles are not described. It is unclear who provided the supplements and measured the outcomes.

Outcomes

Maternal: 24‐hour urinary calcium excretion after 6 weeks supplementation, calcium, 25‐hydroxyvitamin D (25‐OHD) and1‐alfa,25‐dihydroxyvitamin D (1,25(OH)2D) metabolites of vitamin D from serum and cord during labour and delivery.

Infant: serum calcium levels at days 2 and 6 of life, birthweight.

Laboratory method used for assessment of vitamin D concentrations: for 25‐hydroxyvitamin D and 1,25‐dihydroxyvitamin D determinations the following techniques were used: extraction with chloroform‐methanol‐water according to Preece, double step purification, first on a Sephadex LH 20 column with chloroform hexan 45 to 55 vol/vol as solvent, then on a high‐pressure liquid pression system according to Shepard. Plasma metabolites were measured by competitive assay using rat protein for 25 OHD and chicken intestine cytosol for 1,25 (OH)2 D according to Jongen. Assay sensitivity for 1,25 (OH)2 D was 5 pmol/tube and for 25 OHD was 25 pmol/tube.

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 56,000 to 200,000 IU;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: single/daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): unknown/mixed;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: winter pregnancy. Infants born during February and March.

Source of funding: unknown/unreported.

Dates of the study and location: January 1979 to December 1982, France.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was conducted by random numbers table.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the method of concealment. It is assumed that it was not conceal as one of the groups did not receive any supplementation.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report if it was blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not report if it was blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

It is unclear if there was attrition, but given the uneven number of participants reported, it is likely that there were losses to follow‐up.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

High risk

Groups are reported with notorious different sample sizes. It is unclear whether the numbers reflect the participants who finished the trial (unclear and uneven losses to follow‐up); a non randomised process; or a selection bias in which randomised participants did not receive the intervention.

Marya 1987

Methods

Randomised controlled trial; 2‐arm design with randomisation at individual level.

Participants

400 pregnant women 20 to 35 years of age, attending the antenatal clinic of Medical College Hospital in Rohtak, India (latitude: 76° 34' 0' north of Tropic of Cancer). Pre‐gestational BMI and skin pigmentation not reported.

Interventions

Participants were allocated to 1 of 2 groups: group 1 (n = 200) received a daily supplement containing 1200 IU vitamin D and 375 mg calcium (estimated total dose from week 20 to 24 of gestation to term:134,400‐168,000 IU); group 2 (n = 200) received no supplement from 20 to 24 weeks of pregnancy until delivery and served as controls.

Length of the intervention/follow‐up: 20 to 24 weeks from start of supplementation to term.

Health worker cadre: not specified.

Outcomes

Maternal: pre‐eclampsia (defined as blood pressure of 140 mmHg or higher systolic and/or 90 mmHg diastolic along with proteinuria higher than 300 mg/24 hours); systolic and diastolic blood pressure at 24, 28, 32 and 36 weeks of gestation. Serum calcium and creatinine.

Laboratory method used for assessment of vitamin D concentrations: not applicable.

Notes

Biochemical analyses were made for those who developed pre‐eclampsia (n = 12) and also in a group of women with no pre‐eclampsia (n = 25) and a control group of non pregnant women. The results of the stratified analysis are not reported in this review.

  • Total dose of supplementary vitamin D during pregnancy: more than 56,000 to 200,000 IU;

  • start of supplementation: 20 weeks of pregnancy, or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: mixed/unknown.

Source of funding: unknown/unreported.

Dates of the study and location: not reported dates, India.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial reported that participants were randomly allocated to the intervention groups but they did not report the method of sequence generation.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the method of concealment. It is assumed that they did not conceal the allocation as one of the groups did not receive any supplementation.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report if the study was blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not report if the research staff was blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

Only data on biochemical were reported for those who developed pre‐eclampsia and some of those with no pre‐eclampsia and a group of non pregnant controls.

Selective reporting (reporting bias)

High risk

Outcomes reported for some subgroups only.

Other bias

Low risk

The study appears to be free of other sources of bias.

Marya 1988

Methods

Randomised clinical trial; 2‐arm design with individual randomisation.

Participants

200 pregnant women, aged 22 to 35 years old, attending the antenatal clinic of the Medical College Hospital, Rohtak, India (latitude: 76° 34' 0' north of Tropic of Cancer). Inclusion criterion: uncomplicated single pregnancy. Exclusion criteria: pre‐eclampsia, antepartum haemorrhage, premature delivery. Pre‐gestational BMI and skin pigmentation not reported.

Interventions

Participants were allocated to 1 of the following groups: group 1 (n = 100) women received 2 doses of 600,000 IU (each dose at 7th and 8th month of pregnancy (estimated total dose: 1,200,000 IU); group 2 (n = 100) women received no intervention and served as controls.

Length of the intervention/follow‐up: 12 weeks from start of supplementation to term.

Health worker cadre: not specified.

Outcomes

Maternal: venous and cord serum calcium, serum proteins, inorganic phosphate, alkaline phosphatase, weight. Radiological examination on women with abnormal biochemistry or osteomalacia symptomatology. Side effects: back age, leg‐pains, general weakness, cramps.

Infant: birthweight, LBW, crown‐heel length, head circumference, mid‐arm circumference within 24 hours after birth. Skinfold thickness (triceps and infrascapular).

Laboratory method used for assessment of vitamin D concentrations: not applicable.

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 200,000 IU of vitamin D;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: 2 single doses were provided at 7th and 8th month of pregnancy;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: mixed/unknown.

Source of funding: unknown/unreported.

Dates of the study and location: not reported dates, India.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial reported that participants were randomly allocated to the intervention groups but they did not report the method of sequence generation.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the method of concealment. It is assumed that they did not conceal the allocation as one of the groups did not receive any supplementation.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report if the study was blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not report if the research staff was blinded.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

Losses to follow‐up are not documented although exclusions included pregnancy complications. Results tables mention that each arm was comprised of 100 women, a number that corresponds to that described for the treatment allocation.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Mazurkevich 2013

Methods

Randomised control trial.

Participants

72 pregnant women with physiological pregnancy aged 18 to 35 years with low alimentary consumption of calcium (< 600 mg/day) who attended to Moscow State University of medicine and dentistry, department of obstetrics and gynaecology. (Latitude: 55.7500° N, 37.6167° E).

Interventions

Participants were randomly assigned to 1 of 2 groups: group 1 (n = 43) received 1250 mg of calcium carbonate and 200 IU of vitamin D (cholecalciferol‐D3) from the second pregnancy trimester until term, in 2 takes a day; group 2 (n = 29) did not receive any treatment and served as controls.

Health worker cadre: not specified.

Outcomes

Maternal: resistance of uterine arteries, resistance of umbilical arteries, uterine‐placental circulation.

Infant: fetal‐placental circulation, intrauterine growth retardation, assessed by dopplerometry.

Laboratory method used for assessment of vitamin D concentrations: not applicable.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU or less IU;

  • start of supplementation: 20 weeks of pregnancy, or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: mixed/unknown.

Source of funding: unknown/unreported.

Dates of the study and location: not reported dates, Moscow, Russia.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial reported that participants were randomly allocated to the intervention groups but they did not report the method of sequence generation.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the method of concealment. It is assumed that they did not conceal the allocation as one of the groups did not receive any supplementation.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report if the study was blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not report if research staff was blinded.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

There is insufficient information to permit judgement.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Mirghafourvand 2013

Methods

Triple‐blind randomised controlled clinical trial.

Participants

126 pregnant women, aged 18 to 39 years with gestational age of 25 to 30 weeks referring to Tabriz health centres, Iran in 2013 to 2014.

Interventions

Participants were allocated to 3 groups using a randomised block design with block sizes of 3 and 6 with the allocation ratio 1:1:1: group 1 (n = 40) Calcium‐vitamin D group (300 mg carbonate calcium plus 1000 units of vitamin D supplements; group 2 (n = 42) vitamin D group (1000 units of vitamin D supplements; and group 3 (n = 42) received placebo.

To hide the allocation, each participant received 2 small envelopes, each with enough medicine for 3 weeks, inside a large matte‐coloured envelope of the same shape that were serially numbered. Each participant received 1 pill every day for 42 days. All pills were of the same shape, size, and weight.

Health worker cadre: not specified.

Outcomes

Maternal: gestational age, mode of delivery based on gestational age. food consumption, in terms of calcium and vitamin D content, pre‐pregnancy BMI, BMI during pregnancy

Infant: weight, height, and head circumference, birthweight, height, head circumference.

Laboratory method used for assessment of vitamin D concentrations: not specified.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 to 200,000 IU;

  • Start of supplementation: 25 weeks of pregnancy or more;

  • Pre‐gestational BMI (kg/m2): most were overweight;

  • Supplementation scheme/regimen: daily;

  • Skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • Latitude: north of the Tropic of Cancer;

  • Season at the start of pregnancy: all year round.

Source of funding: this study was funded by a research grant of Tabriz University of Medical Sciences (Project number: 388).

Dates of the study and location: July 2013 to April 2014, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): no conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were allocated to 3 groups using a randomised block design with block sizes of 3 and 6 with the allocation ratio 1:1:1.

Allocation concealment (selection bias)

Unclear risk

To hide the allocation, each participant received 2 small envelopes, each with enough medicine for 3 weeks, inside a large matte‐coloured envelope of the same shape that were serially numbered. Each participant received 1 pill every day for 42 days. Pills were of the same shape, size, and weight. However, authors did not mention how this was concealed from study staff.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants were blinded to the study treatments.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial reported that it was double‐blinded but they did not specify if those performing the assessments were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Losses to follow‐up were documented. No missing data and no participant was eliminated from the analysis.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Naghshineh 2016

Methods

Double‐blind randomised controlled trial.

Participants

140 nulliparous pregnant women who had been referred to “Shahid Beheshti” hospital in Isfahan, Iran. Pregnant women at less than 16 weeks' gestation from outpatient clinics at “Shahid Beheshti” hospital were eligible if they did not have any sign of vitamin D deficiency, did not using aspirin and had no diagnosis of chronic hypertension, gestational diabetes, renal disease or systemic lupus erythematous.

Interventions

Subjects were randomly divided into 2 groups: Group 1 (n = 70) received supplementation with 600 IU daily of vitamin D at 16 weeks' gestation until labour; Group 2 (n = 70) received daily supplementation free of vitamin D and followed until labour (placebo group). Women were unaware of the treatment allocation.

Health worker cadre: women were followed up monthly by a doctor who was blinded to the study groups.

Outcomes

Maternal: age and gestational age at delivery, pre‐eclampsia

Infant: birthweight

Laboratory method used for assessment of vitamin D concentrations: not applicable.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU or less;

  • start of supplementation: less than 20 weeks of pregnancy;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: From May 2012 until January 2012.

Source of funding: this study was funded by a research grant. Financial support was provided by the Isfahan University of Medical Sciences (Grant 392004); Isfahan, Iran.

Dates of the study and location: May 2012 to January 2012, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Using random‐maker software “Random Allocation”, participants were randomly divided into 2 groups: intervention and placebo.

Allocation concealment (selection bias)

Unclear risk

Women were unaware of the treatment allocation but no other information was provided.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Research staff was unaware of the treatment allocation.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Follow‐up visits was done by blinded study personnel.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 participants for the intervention group were missing, but it was explained in the results section that they did not want to continue in the study. Total of 138 participants (68 cases and 70 controls) were analysed and described

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Roth 2010

Methods

Randomised placebo‐controlled trial (AViDD‐2 trial)

Participants

160 pregnant women aged 18 < 35 years old, attending to the International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh (latitude: 23.7000° N, 90.3750° E, north of the Tropic of Cancer). Inclusion criteria: women with residence in Dhaka, with plans to have the delivery performed at the Shimantik maternity centre, and to stay in Dhaka throughout the pregnancy and 1 month past the delivery, with gestational age of 26th to 29th (inclusive), estimated based on the first day of the last menstrual period. Exclusion criteria: use of any dietary supplement containing more than 400 IU/day (10 mcg/day) of vitamin D within the month prior to enrolment, or refusal to stop taking supplemental vitamin D at any dose after enrolment, current use of anti‐convulsant or anti‐mycobacterial (tuberculosis) medications, severe anaemia (haemoglobin concentration < 70 g/L), complicated medical or obstetric history: cardiovascular disease, uterine haemorrhage, placenta praevia, threatened abortion, hypertension, pre‐eclampsia, preterm labour, or multiple gestation), prior history of delivery of an infant with a major congenital anomaly, birth asphyxia, or perinatal death (stillbirth or death within first week of life).

Interventions

Participants were randomly assigned to 1 of 2 groups: group 1 (n = 80): women received vitamin D (cholecalciferol‐D3) 35,000 IU per week, started at 26 to 29 weeks' gestation, until delivery; group 2 (n = 80): women received placebo control administered weekly from 26 to 29 weeks' gestation until delivery.

Health worker cadre: supplement doses were measured in disposable plastic syringes and orally administered by study personnel.

Outcomes

Maternal: serum 25‐hydroxyvitamin D concentration, serum calcium concentration, urine Ca:Cr ratio.

Infant: immune function, infant growth, postnatal vitamin D status, serum calcium.

Laboratory method used for assessment of vitamin D concentrations: Serum 25‐hydroxyvitamin D was quantified by high‐performance liquid chromatography tandem mass spectroscopy (LCMS/MS) in the Department of Pathology and Laboratory Medicine at the Hospital for Sick Children.

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 200,000 IU;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: summer.

Source of funding: this study was funded by a Non‐governmental organization (NGO). The Thrasher Research Fund, Salt Lake City, USA.

Dates of the study and location: August 2010 to January 2011, Dhaka, Bangladesh.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors declare that they have no competing interests.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Trial reported computer‐generated randomisation list for the randomisation procedures.

Allocation concealment (selection bias)

Low risk

The allocation sequence was prepared by International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh personnel not otherwise involved in the study, and was concealed from investigators.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Trial reported that participants were blinded to allocation.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Trial reported that research staff (including lab personnel) were blinded to allocation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Of the 160 participants recruited and randomly assigned to intervention or placebo, 13 were lost to follow‐up prior to delivery (6 in the placebo group and 7 in the vitamin D group), all because of having left the Dhaka area.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Roth 2013

Methods

Randomised, double‐blind, placebo‐controlled trial (MDIG trial).

Participants

1300 generally‐healthy pregnant women between 17 and 24 weeks of gestation.

Interventions

Participants were randomly assigned at enrolment to 1 of 5 groups: group 1 (n = 260) received placebo throughout the prenatal period and for 26 weeks postpartum; group 2 (n = 260) received 4200 IU per week prenatally and no supplementation postpartum; group 3 (n = 260) received 16,800 IU per week prenatally and no supplementation postpartum; group 4 (n = 260) received 28,000 IU per week prenatally and no supplementation postpartum; and group 5 (n = 260) received 28,000 IU per week prenatally and during the postpartum for 26 weeks. Data from groups 2‐5 were combined into the intervention group for this analysis. All participants received calcium (500 mg per day), iron (66 mg per day), and folic acid (350 mcg per day) throughout the intervention phase.

Health worker cadre: trial personnel contacted participants weekly from enrolment until 26 weeks postpartum, and infants were further assessed at 9 months and 12 months of age. Visits were conducted in the home or at a clinic and included the use of standardized questionnaires, point‐of‐care tests, anthropometric measurements, and specimen collection.

Outcomes

Maternal: maternal serum 25‐hydroxyvitamin D and calcium concentration, urinary calcium excretion, and maternal PTH concentrations.

Infant: length‐for‐age, birth outcomes, morbidity and serum 25‐hydroxyvitamin D and calcium concentrations.

Laboratory method used for assessment of vitamin D concentrations: point‐of‐care tests.

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 56,000 IU;

  • start of supplementation: 17‐24 weeks of pregnancy;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: weekly;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: unknown.

Source of funding: this study was funded by the Bill & Melinda Gates Foundation.

Dates of the study and location: March 2014 to September 2015, Dhaka, Bangladesh.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): no potential conflict of interest was reported.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was conducted by computer‐generated, simple randomisation scheme created independently by the trial statistician.

Allocation concealment (selection bias)

Low risk

Concealment of trial‐group assignments was ensured with the use of pre‐labelled and sequentially numbered but otherwise identical supplement vials.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial reported that a master list linking participants to supplementation groups was not accessible to trial personnel until final group assignments were revealed.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The trial reported that a master list linking participants to supplementation groups was not accessible to trial personnel until final group assignments were revealed.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Among 1164 infants assessed at 1 year of age (89.5% of 1300 pregnancies), < 5% of participants withdrew or were excluded after randomisation until birth.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

There is no any evidence of other bias.

Sabet 2012

Methods

Randomised, double‐blind trial

Participants

50 pregnant women, in their third trimester, who were scheduled to deliver at Mahdieh Hospital in Tehran.

Interventions

Participants were randomly allocated to 1 of 2 groups: Group 1 (n = 25) received oral vitamin D3 100,000 IU monthly, 3 times; Group 2 (n = 25) control (placebo); until term.

Health worker cadre: not specified.

Outcomes

Maternal: the final maternal 25(OH) serum concentrations at delivery, cord 25(OH) vitamin D concentration serum 25(OH), maternal serum iPTH and cord blood iPTH concentration mean PTH concentration

Infant: serum vitamin D lower than30 ng/mL in newborn infants

Laboratory method used for assessment of vitamin D concentrations: Serum 25 (OH) D concentrations were measured by EIA using the 25(OH) Vit D kit (Immune diagnostic system Ltd, Bolden, UK).

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 200,000 IU;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: monthly;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: unknown.

Source of funding: this study was funded by a research grant from the Research Institute of Endocrine Sciences, Shahid Beheshti University of Medical Sciences.

Dates of the study and location: 2009 to 2010, Tehran, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): no conflict of interest declared

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Participants were randomly allocated to the treatment or placebo but methods describing this process were not reported.

Allocation concealment (selection bias)

Unclear risk

The trial reported that it was double‐blinded, but no methods of describing the process were reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial reported that it was double‐blinded, but no methods of describing the process were reported.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial reported that it was double‐blinded but they did not specify if those performing the assessments were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data reported for all 50 participants.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Sablok 2015

Methods

Randomised controlled trial with 2 arms, with randomisation at the individual level from years 2010 to 2012.

Participants

180 primigravidae women with singleton pregnancy at 14 to 20 weeks in the Department of Obstetrics and Gynaecology in Safdarjung Hospital, New Delhi, India (28°38′08″ N, 77°13′28″ E north of Tropic of Cancer).
Pregnant women with pre‐existing osteomalacia, known hyperparathyroidism, renal, liver dysfunction, tuberculosis, sarcoidosis and women not willing to comply to the study protocol were excluded.

Interventions

Participants were randomly assigned to 1 of 2 groups: group 1 (n = 60) did not receive any supplementation of vitamin D; group 2 (n = 120) received vitamin D (cholecalciferol‐D3) supplementation in dosages depending upon the level of serum 25(OH)‐D levels estimated at entry into the study. Participants from this second group with sufficient levels of vitamin D (serum 25(OH)‐D levels > 50 nmol/L), received only 1 dose of 60,000 IU vitamin D (cholecalciferol‐D3) at 20 weeks; participants with insufficient levels of vitamin D (serum 25(OH)‐D levels 25–50 nmol/L) received 2 doses of 120,000 IU vitamin D (cholecalciferol‐D3) at 20 weeks and 24 weeks; and participants with deficient levels of vitamin D status (serum 25(OH)‐D levels < 25 nmol/L) received 4 doses of 120,000 IU vitamin D cholecalciferol‐D3) at 20, 24, 28 and 32 weeks. Independently of the dose, all participants in group 2 were grouped and compared to group 1 for this analysis.

Health worker cadre: unclear what the roles of the researchers and other workers in the health worker cadre.

Outcomes

Maternal: preterm labour, pre‐eclampsia, gestational diabetes, serum 25(OH)‐D concentration, serum calcium, phosphorus and serum ALP levels.
Infants: Apgar score, birthweight, LBW, 25(OH)‐D concentration in cord blood, small‐for‐gestational age; appropriate for gestational age.

Laboratory method used for assessment of vitamin D concentrations: serum 25‐hydroxyvitamin D was quantified by sandwich ELISA.

Notes

  • By total dose of supplementary vitamin D during pregnancy: more than 56,000 to 200,000 IU to more than 200,000 IU of vitamin D;

  • by start of supplementation: 20 weeks of pregnancy, or more;

  • by pre‐gestational BMI (kg/m2): healthy weight;

  • by supplementation scheme/regimen: single given at different weeks of gestation in the supplemented group;

  • by skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • by latitude: north of the Tropic of Cancer;

  • by season at the start of pregnancy: all year round. Authors report that sufficient levels of vitamin D were seen in 93.3% women who had more than 4 hours of sun exposure every day as compared to 18.5% in those with less than 1 hour of daily sun exposure.

Source of funding: self‐funded.

Dates of the study and location: 2010 to 2012, India.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): all the authors have nothing to disclose

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed using computer‐generated random number tables.

Allocation concealment (selection bias)

High risk

As participants were assigned to either no intervention or intervention and the intervention dosage depended on the vitamin D status, there was a selection bias based on status of vitamin D at baseline.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report if the study was blinded. It is assumed that it was not blinded to participants as one of the groups did not receive any supplementation and the other groups received different doses of vitamin D at different times.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

At the time of delivery, both the groups underwent clinical evaluations and complete anthropometric assessment of the neonate, but it was not reported if staff was blinded to the intervention groups.

Incomplete outcome data (attrition bias)
All outcomes

High risk

The level of attrition was different in groups 1 and 2: 3/60 (5%) participants in group 1 and 12/120 (10%) participants in group 2 were lost to follow‐up.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other evident sources of bias.

Samimi 2016

Methods

Prospective, double‐blind, placebo‐controlled trial.

Participants

60 primigravida pregnant women, aged 18–40 years old, who were at risk for pre‐eclampsia, and lived approximately 20 km or less from the clinic and hospital. Women ‘at‐risk’ for pre‐eclampsia were recognised by laboratory tests including free β‐human chorionic gonadotrophin, inhibin a dimeric, unconjugated oestriol and maternal serum α‐foetoprotein, and haemodynamic assessment of uterine artery Doppler wave form at 16 to 20 weeks of gestation.

Interventions

Participants were randomly allocated into 2 groups: Group 1 (n = 30) received 50,000 IU vitamin D3 every 2 weeks plus 1000 mg day 1 calcium supplements (as calcium carbonate); Group 2 (n = 30) received placebos at the same times; from 20 to 32 weeks of gestation.

Health worker cadre: an investigator with no clinical involvement in the present study packed cholecalciferol, calcium supplements and placebos into numbered bottles based on the random list. Anthropometric measurements of pregnant women at maternity clinic were measured by a trained midwife at baseline and then after 12 weeks of intervention.

Outcomes

Maternal: serum 25(OH)D concentrations, fasting plasma glucose, serum insulin concentrations, homeostasis model assessment (HOMA)‐B, inQUICKI score, serum HDL‐cholesterol, plasma GSH concentrations, systolic blood pressure, diastolic blood pressure, lipid profiles and inflammatory markers, pre‐eclampsia.

Infant: LBW (< 2500 g), newborn’s birth size (newborn’s weight, length and head circumference) and prevalence of preterm delivery (< 37 weeks).

Laboratory method used for assessment of vitamin D concentrations: Serum 25‐hydroxyvitamin D concentrations was determined using a commercial enzyme‐linked immunosorbent assay (ELISA) kit (IDS, Boldon, UK) with inter‐ and intra‐assay coefficients of variation (CVs) of 4.5–7.0%, respectively.

Notes

  • By total dose of supplementary vitamin D during pregnancy: more than 200,000 IU of vitamin D;

  • by start of supplementation: 20 weeks of pregnancy, or more;

  • by pre‐gestational BMI (kg/m2): mixed;

  • by supplementation scheme/regimen: bi weekly;

  • by skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • by latitude: north of the Tropic of Cancer;

  • by season at the start of pregnancy: unknown.

Source of funding: the study was supported by a research grant from Kashan University of Medical Sciences.

Dates of the study and location: September 2014 to February 2015, Kashan, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors declare that there are no conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Parallel, balanced randomisation (1:1).

Allocation concealment (selection bias)

Low risk

Randomisation and allocation were concealed from both researchers and participants until the statistical analysis was completed.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Only the person responsible for the distribution of drugs knew how the women were allocated to the treatment groups.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial reported that it was double‐blinded but they did not specify if those performing the assessments were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data were reported for all participants.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Samimi 2017

Methods

Double‐blind randomised and controlled clinical trial.

Participants

80 women aged 18 to 35 years were examined from November 2013 to March 2015 at the Shabihkhani Maternity Hospital in Kashan, Iran.

Interventions

Participants were randomised into 2 groups: Group 1 (n = 40) received a pill of vitamin D3 400 IU/day and Group 2 (n = 40) received a placebo tablet that was similar to vitamin D3, with no active ingredient as a placebo. Both groups received standard treatment with vaginal progesterone (Behvarzan, Iran) at a dose of 400 mg per day. The serum levels of vitamin D3 were evaluated in the tenth and twentieth weeks to prevent any possible poisoning. If so, the patient was excluded from the study.

Health worker cadre: all participants in the study received antenatal care and were given folic acid and ferrous sulphate at least 1 month prior to pregnancy, under the supervision of a gynaecologist. They were checked by monitoring serum β‐hCG level levels and abdominal ultrasound until the confirmation of pregnancy, after which the mothers were divided into 2 groups of intervention and control using permuted block randomisation with twenty blocks of size 4. Only the person responsible for the distribution of drugs knew how the women were allocated to the treatment groups.

Outcomes

Maternal: the serum level of vitamin D3, serum level of IL‐23, serum levels of vitamin D3 and IL‐23, spontaneous abortions.

Laboratory method used for assessment of vitamin D concentrations: not specified.

Notes

  • By total dose of supplementary vitamin D during pregnancy: more than 200,000 IU of vitamin D;

  • by start of supplementation: less than 20 weeks of pregnancy;

  • by pre‐gestational BMI (kg/m2): unknown;

  • by supplementation scheme/regimen: daily;

  • by skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • by latitude: north of the Tropic of Cancer;

  • by season at the start of pregnancy: unknown.

Source of funding: this study was funded by a research grant of the Kashan University of Medical Sciences.

Dates of the study and location: November 2013 to March 2015, Kashan, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors declare that there is no conflict of interests regarding the publication of this paper.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were divided into 2 groups of intervention and control using permuted block randomisation with 20 blocks of size 4.

Allocation concealment (selection bias)

Low risk

Only the person responsible for the distribution of drugs knew how the women were allocated to the treatment groups.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Only the person responsible for the distribution of drugs knew how the women were allocated to the treatment groups.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial reported that it was double blinded but they did not specify if those performing the assessments were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data were reported for all participants.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other sources of bias.

Sasan 2017

Methods

Randomised controlled clinical trial.

Participants

142 women who were referred to the obstetrical clinic in Besat Hospital of Sanandaj City, Kurdistan Province, Iran, who were receiving prenatal care and had a history of pre‐eclampsia in previous pregnancies.

Interventions

The participants were randomly placed into 2 groups: Group 1 (n = 70) received 50,000 IU pearl vitamin D3 once every 2 weeks; and Group 2 (n = 72) received placebo. Vitamin D or placebo was given until the 36th week of pregnancy.

Health worker cadre: not specified.

Outcomes

Maternal: level of vitamin D, pre‐eclampsia.

Laboratory method used for assessment of vitamin D concentrations: level of vitamin D was determined through Liebermann–Burchard method.

Notes

  • By total dose of supplementary vitamin D during pregnancy: more than 56,000 and less than 20,0000 IU of vitamin D;

  • by start of supplementation: 10 weeks of pregnancy, or more;

  • by pre‐gestational BMI (kg/m2): mixed;

  • by supplementation scheme/regimen: once every 2 weeks;

  • by skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • by latitude: north of the Tropic of Cancer;

  • by season at the start of pregnancy: unknown.

Source of funding: unknown/unreported.

Dates of the study: not reported dates, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors announce that there are no conflicts of interest between different individuals and organisations involved in the study.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

140 pockets of drug and placebo were randomly (by using table of random numbers).

Allocation concealment (selection bias)

Unclear risk

140 pockets of drug and placebo were randomly offered and both study staff and participants did not know about administration of the treatments.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both study staff and participants did not know about administration of the treatments.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Both study staff and participants did not know about administration of the treatments.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data reported for all participants.

Selective reporting (reporting bias)

Unclear risk

Although participant's level of 25‐hydroxy vitamin D was used to determine study eligibility, serum levels of vitamin D were not reported.

Other bias

Low risk

The study appears to be free of other sources of bias.

Shahgheibi 2016

Methods

Randomised double‐blind placebo‐controlled trial.

Participants

90 pregnant women with at least 1 risk factor for gestational diabetes including BMI (BMI; kg/m2) more than 25, history of macrosomic neonate, positive family history for diabetes and gestational diabetes, history of gestational diabetes in previous pregnancies, and glycosuria.

Interventions

Participants were randomised into 1 of 2 groups: group 1 (n = 46) received 5000 units of vitamin D weekly; and group 2 (n = 44) received placebo. Both groups were treated until the 26th week of pregnancy. Then the glucose challenge test (GCT) and the glucose tolerance test (GTT) were performed to evaluate GDM.

Health worker cadre: not specified.

Outcomes

Maternal: vitamin D levels and GCT, incidence of diabetes.

Laboratory method used for assessment of vitamin D concentrations: vitamin D level was determined in a laboratory by the Liebermann–Burchard method, in which the patient should fast for 12 hours and not have a fatty dinner.

Notes

  • Total dose of supplementary vitamin D during pregnancy: less than 56,000 IU;

  • start of supplementation: enrol until 26 weeks of pregnancy;

  • pre‐gestational BMI (kg/m2): > 25;

  • supplementation scheme/regimen: weekly;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: North of the Tropic of Cancer;

  • season at the start of pregnancy: unknown.

Source of funding: no funding.

Dates of the study and location: 2013, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors have no conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Women were included and randomly divided into 2 groups using permuted block randomisation of size 2.

Allocation concealment (selection bias)

Low risk

Blinding was carried out in which drugs and placebos were prepared to be completely similar in appearance and taste and put in numbered pockets based on a randomisation.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both study staff and participants did not know about administration of the treatments.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Both study staff and participants did not know about administration of the treatments.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Data reported for all participants.

Selective reporting (reporting bias)

Unclear risk

Apgar score was taken but is not in results.

Other bias

High risk

Intervention described in abstract does not match intervention described in methods. In abstract: participants in the intervention group took 5000 units of vitamin D daily. In methods: participants in the intervention group took 5000 units of vitamin D weekly.

Singh 2015

Methods

Randomised controlled trial

Participants

100 healthy, pregnant women, primigravida with a singleton pregnancy, gestational age: 12‐16 weeks in Sawangi, Meghe, Wardha.

Interventions

Participants were randomised into 2 groups: Group 1 (n = 50): received 2000 IU of vitamin D3 per day from 12‐16 weeks of gestation of pregnancy; and Group 2 (n = 50) received no supplementation and served as controls. 25‐hydroxyvitamin D [25(OH)D]in maternal blood was measured by chemiluminescence immunoassay, at recruitment and at the time of delivery and a serum 25(OH)D level lower than30 nmol/L was defined as deficiency.

Health worker cadre: not specified.

Outcomes

Maternal: deficiency of vitamin D, mean gestational age, preterm birth.

Laboratory method used for assessment of vitamin D concentrations: Mean serum 25(OH)D levels were measured by Roche diagnostic ELECSYS (Electrochemiluminescence immunoassay) 2010 Cobase E 411 Analyser Immunoassay System Germany.

Notes

  • Total dose of supplementary vitamin D during pregnancy: 56,000 IU;

  • start of supplementation: 12 to 16 weeks of pregnancy;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: North of the Tropic of Cancer;

  • season at the start of pregnancy: all year, from October 2012‐September 2014.

Source of funding: no funding sources reported.

Dates of the study and location: October 2012 to September 2014, India.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial reported that participants were randomly allocated to the treatment or no supplementation but methods describing this process were not reported.

Allocation concealment (selection bias)

Unclear risk

The trial did not report this; however, since one group did not receive any supplementation, it is assumed that the intervention was not concealed to participants.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not report this; however, it is assumed that participants were not blinded as one group did not receive any supplementation.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not report if study staff was blinded.

Incomplete outcome data (attrition bias)
All outcomes

High risk

It is assumed that each group had 50 participants as stated in the abstract but the trial did not report final number of participants that completed the trial or the sample size in any of the tables.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other evident sources of bias.

Taherian 2002

Methods

Randomised controlled study with 3 arms.

Participants

990 nulliparous women attending antenatal outpatient clinics of Isfahan Health Centers (32.6333° N, 51.6500° E north of Tropic of Cancer) between April 1998 and March 2001, with singleton pregnancies, first prenatal visit before 20 weeks of gestation, systolic/diastolic blood pressure lower than 130/80 mmHg, and no proteinuria detectable by a dipstick.

Women with history of cardiovascular, renal or endocrinologic problems, medical or obstetric complications and those with known hazardous condition (multifetal gestation, hydatidiform mole) were excluded.

Interventions

Participants were randomly assigned to 1 of 3 groups: Group 1 (n = 330) received 75 mg aspirin each day from 20th week of gestation until delivery; Group 2 (n = 330) received a tablet containing 500 mg calcium carbonate + 200 IU vitamin D (cholecalciferol‐D3) daily from 20th week of gestation until delivery; and group 3 received no intervention (n = 330). All cases received standard prenatal care.

Health worker cadre: the women were examined by trained staff every 4 weeks through the 28 weeks of gestation, and every 2 weeks through the 36th week and weekly thereafter. Blood pressure was measured by a certified examiner.

Outcomes

Maternal: blood pressure, bodyweight, BMI, maternal height, urine protein measurements, maternal weight gain, duration of gestation.

Infant: neonatal weight at birth, the presence of respiratory distress syndrome, sepsis, jaundice and intrauterine growth retardation, fetal or neonatal death.

Laboratory method used for assessment of vitamin D concentrations: not applicable.

Notes

  • Total dose of supplementary vitamin D during pregnancy: less than 56,000 IU;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): normal weight (18.5 to 24.9);

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: April 1998 to March 2001.

Source of funding: this study was funded by a research grant of the Research Deputy of Isfahan University of Medical Sciences grant (No: 76085).

Dates of the study and location: April 1998 to March 2001, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised by table of random numbers.

Allocation concealment (selection bias)

Unclear risk

The trial did not report the methods for allocation concealment. It is unclear whether intervention allocation could have been foreseen in advance of, or during recruitment, or changed after assignment. It is assumed that it was not conceal as one group did not receive any treatment.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial did not mention if it was blinded. It is assumed that it was not blinded to participants as one group did not receive any treatment.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not mention if it was blinded to those conducting the assessments.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

No attrition reported.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other evident sources of bias.

Tehrani 2014

Methods

Single‐arm study, not blinded.

Participants

210 pregnant women referring to obstetric clinic of Shahid Beheshti and Alzahra hospital in Esfahan city in 2012. Inclusion criteria: patient satisfaction; normal BMI; gestational age below 16 weeks; no history of diabetes mellitus type 2 or GDM; no family history of diabetes mellitus type 1 in first degree relatives. Exclusion criteria: patient dissatisfaction; incorrect consumption of vitamin D supplementation; follow‐up discontinuation.

Interventions

Participants will be individually randomised to 1 of 2 groups: Group 1 (n = 70) received vitamin D supplementation with dose of 50,000 unit every 2 weeks for 10 weeks; Group 2 received a placebo. Pregnant women with levels of above 25 nmol/L were selected as the normal healthy control group and were the ones who received placebo.

Health worker cadre: not specified.

Outcomes

Maternal: gestational blood sugar level, serum vitamin D level.

Laboratory method used for assessment of vitamin D concentrations: not specified.

Notes

  • Total dose of supplementary vitamin D during pregnancy: more than 200,000 IU;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): normal BMI;

  • supplementation scheme/regimen: bi weekly;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: unknown.

Source of funding: this study was funded by a non‐governmental organization, Sponsor: Isfahan University of Medical Sciences.

Dates of the study and location: January 2013 to January 2014, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): there are no conflicts of interest.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised by table of random numbers.

Allocation concealment (selection bias)

Low risk

Researcher, the distributors of the drug and the women, did not know which group was taken the vitamin D or placebo.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Researcher, the distributors of the drug and the women, did not know which group was taken the vitamin D or placebo.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Researcher, the distributors of the drug and the women, did not know which group was taken the vitamin D or placebo.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All data reported.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other evident sources of bias.

Vaziri 2016

Methods

Randomised clinical trial.

Participants

169 pregnant women, both nulliparous and multiparous, aged 18 years or older, no history of mental illness and internal diseases such as hyper/hypothyroidism, no addiction to any kind of narcotic drugs or alcohol, not divorced or widowed, no pregnancy complications such as pre‐eclampsia, gestational diabetes, ruptured membranes and suspicion of preterm birth, no previous caesarean sections, with a live fetus singleton pregnancy, and gestational age of 26 to 28 weeks based on ultrasound results. who were under prenatal care in Hafez teaching hospital in Shiraz, Iran.

Interventions

Participants were assigned to 2 groups through block randomisation design: Group 1 (n = 78) received 2000 IU of vitamin D3 per day; and Group 2 (n = 75) received placebo. Both groups received their assigned treatments from 26 to 28 weeks of gestation until childbirth. Maternal serum 25‐hydroxyvitamin D concentrations were measured at baseline and childbirth. Besides, depression scores were evaluated 4 times: at 26 to 28 and 38 to 40 weeks of gestation, and finally at 4 and 8 weeks after birth. Participants were allowed to use prescribed supplementation outside this study’s protocol. Usually, pregnant women in Iran are prescribed iron and folic acid, which may be consumed as part of the multivitamin supplementation with 200–400 IU of vitamin D. They reported that 55 participants in the intervention and 69 in the control used other supplements but they did not provide the composition of the supplements.

Health worker cadre: at first, a research team member who was responsible for data collection visited the prenatal care clinic of the hospital daily and based on the inclusion criteria, invited the mothers to participate in the study. The consumption of pills were assessed in later prenatal care visits and over the phone.

Outcomes

Maternal: baseline 25‐hydroxy vitamin D concentrations and at childbirth, depression score.

Infant: vitamin D concentrations, anthropometric measurements of their infants at birth, 4th and 8th weeks of birth.

Laboratory method used for assessment of vitamin D concentrations: Serum 25‐hydroxyvitamin D was measured with the Chemiluminescence immunoassay (CLIA) method.

Notes

  • Total dose of supplementary vitamin D during pregnancy: less than 56,000 IU;

  • start of supplementation: 26 to 28 weeks of pregnancy;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: North of the Tropic of Cancer;

  • season at the start of pregnancy: all year, from November 2014 to October 2015.

Source of funding: the study was financially supported by a research grant from the Research Vice‐chancellor of Shiraz University of Medical Sciences.

Dates of the study and location: November 2014 to October 2015, Iran.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): the authors declare that they have no competing interests.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised using a block randomisation design. 

Allocation concealment (selection bias)

Unclear risk

The trial reported that it was a single‐blinded study, but it is not clear who was blinded and they did not report the allocation concealment. It is assumed that participants were blinded as the control group also received 2 pills, similar to the intervention group.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial reported that it was a single‐blinded study. However, data collection related to depression was done by a trained midwife outside the research team, who was blinded to group allocations but the rest of the assessments were not clear if they were performed by a blinded staff.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial did not report if study staff conducting the assessments were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

143 women began study and 136 finished. Serum vit D checked for 130 women, but does not specify how many women were in each group.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Low risk

The study appears to be free of other evident sources of bias.

Yu 2008

Methods

Randomised controlled trial; 4 x 3 block design with randomisation at individual level.

Participants

180 pregnant women (45 Indian Asians, 45 Middle Eastern, 45 Black and 45 Caucasian) women at 27 weeks' gestation attending the routine antenatal clinic at St Mary’s Hospital, London, the UK (latitude: 51°30'N north of tropic of Cancer). Exclusion criteria: pre‐existing sarcoidosis, osteomalacia, renal dysfunction and tuberculosis. Pre‐gestational BMI and skin pigmentation (in addition to ethnicity) not reported. The study took place between April 2007 and November 2007. As well, a follow‐up trial of the infants of these trial participants. All of the offspring of the 180 mothers recruited in this trial were eligible and were invited to participate in a follow‐up study when their children were 3 years of age.

Interventions

Participants were randomised in blocks of 15 within each of the 4 ethnic groups to 3 groups; Group 1 (n = 60) received a daily dose of vitamin D (ergocalciferol D2) at 800 IU; Group 2 (n = 60) received a one dose of 200,000 IU of calciferol; Group 3 (n = 60) received no treatment and served as controls. All groups received the intervention for 13 weeks, from start of supplementation to term. Data from groups 1 and 2 were collapsed for this analysis.

Health worker cadre: each woman collected her tablets directly from the hospital pharmacy department or her local pharmacy.

Outcomes

Maternal: maternal and cord 25‐hydroxyvitamin D levels at delivery, maternal PTH and corrected calcium levels at delivery, adverse events.

Infant: small‐for‐gestational age was defined as birthweight less than the 10th percentile after adjustments for gestation at delivery, infant sex, maternal ethnicity, parity, height and weight. Wheezing episode in the first 3 years of life, measured at 36 to 48 months, use of inhaled bronchodilators in the last 12 months, doctor‐diagnosed rhinitis, any wheezing episode in the preceding 12 months, doctor‐diagnosed asthma, doctor‐diagnosed eczema, doctor‐diagnosed food allergy, positive skin prick test responses, 25‐hydroxyvitamin D levels, bronchodilator responsiveness, exhaled nitric oxide level (in parts per billion), nasal secretions for inflammatory mediators, pulmonary airflow resistance and reactance at a range of frequencies using impulse oscillometry, total number of all wheezing episodes since birth and total number of upper and lower respiratory tract infections since birth, at 36 to 48 months.

Laboratory method used for assessment of vitamin D concentrations: not specified.

Notes

Women who did not speak English were only included if a health advocate was able to interpret and a leaflet was provided in their language;

  • Total dose of supplementary vitamin D during pregnancy: more than 56,000 to 200,000 IU;

  • start of supplementation: 20 weeks of pregnancy or more;

  • pre‐gestational BMI (kg/m2): unknown/mixed;

  • supplementation scheme/regimen: single and daily;

  • skin pigmentation based on Fitzpatrick skin tone chart (Fitzpatrick 1988): mixed/unknown;

  • latitude: north of the Tropic of Cancer;

  • season at the start of pregnancy: summer. April to November 2007; summer.

Source of funding: this study was funded by a research grant from the Institute of Obstetrics and Gynaecology Trust, Wolfson and Weston Research Centre for Family Health, Imperial College, Du Cane Road, Hammersmith Hospital, London W12 0NN, UK.

Dates of the study and location: April 2007 to November 2007, London, UK.

Declarations of interest among primary researchers (or state where this information is not reported by the trial authors): none declared.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Computer‐generated random number lists were drawn up by an independent researcher, with randomisation in blocks of 15.

Allocation concealment (selection bias)

Low risk

The research staff allocating participants used the next available number on entry to the trial, and each woman collected her tablets directly from the hospital pharmacy department or her local pharmacy.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Study personnel and participants were not blinded to treatment assignment.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Study personnel was not blinded to treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Only 1 loss to follow‐up on group 3.

Selective reporting (reporting bias)

Unclear risk

There is insufficient information to permit judgement.

Other bias

Unclear risk

Women were randomised within each ethnic group. It is not clear if the ethnicity can be clearly established as it was self reported. Women who did not speak English were included only if a health advocate was able to interpret and a leaflet was provided in their language (English, Arabic, Bengali and Farsi) although the ability to read was not clearly established.

β‐hCG: beta human chorionic gonadotropin
BMI: body mass index
CHW: community health workers
ELISA: enzyme‐linked immunosorbent assay
GDM: gestational diabetes mellitus
GSH: glutathione
HDL: high‐density lipoprotein
IGF‐I: insulin‐like growth factor
IU: international units
IUFD: intrauterine fetal death
LBW: low birthweight
LDL: low‐density lipoprotein
mcg: microgram
PA: physical activity
PTH: parathyroid hormone
25 (OH)D: 25‐hydroxycholecalciferol

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Ala‐Houhala 1986

49 healthy, well‐nourished mothers delivering in January 1984 in the maternity wards and outpatient clinic of the Department of Paediatrics of the University Central Hospital of Tampere, Finland (latitude 61°N) and exclusively breastfeeding their infants, were divided in succession into 3 groups: group 1 (n = 17): mothers were given 2000 IU vitamin D3 a day, infants not supplemented; group 2 (n = 16): mothers were given 1000 IU vitamin D3 a day, infants not supplemented; group 3 (n = 16): mothers were not supplemented, and their breast fed infants were given 400 IU of vitamin D2 a day.

This is not a randomised trial and the intervention includes mothers at postpartum and their infants.

Asemi 2013b

54 pregnant women aged 18 to 40 years diagnosed with GDM by a 100‐g oral glucose‐tolerance test at 24 to 28 weeks' gestation attending maternity clinics affiliated with Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran. Participants were randomly assigned to 1 of 2 groups: group 1 (n = 27), women received capsules containing 50,000 IU vitamin D (cholecalciferol‐D3) (D‐Vitin 50000; Zahravi Pharm Co) 2 times during the study (at baseline and at day 21 of the intervention): group 2 (n = 27), women received 2 placebos (Barij Essence Co) at the same times. All pregnant women in the study had a diagnosis of gestational diabetes. The type of participant is outside the scope of this review.

Asemi 2014

56 pregnant women 18 to 40 years of age with gestational diabetes and 24 to 28 weeks' gestation attending prenatal care at maternity clinics affiliated to Kashan University of Medical Sciences, Kashan, Iran were randomly assigned to 1 of 2 groups: group 1 (n = 28) received 1000 mg calcium per day and a 50,000 U vitamin D (cholecalciferol‐D3) pearl twice during the study (at study baseline and on day 21 of the intervention); group 2 (n = 28) received 2 placebos at the same times. Participants were pregnant women with diagnosis of gestational diabetes. The type of participant is outside the scope of this review.

Asemi 2015

46 women at risk for pre‐eclampsia at 27 weeks' gestation with positive roll‐over test were randomly assigned to receive either the multi mineral‐vitamin D supplements (n = 23) or the placebo (n = 23) for 9 weeks. Study was conducted in Kashan, Iran, during November 2013 to May 2014. Multi mineral‐vitamin D supplements were containing 800 mg calcium, 200 mg magnesium, 8 mg zinc, and 400 IU vitamin D3. Fasting blood samples were taken at baseline and after 9‐week intervention to measure related factors. Newborn's outcomes were determined. This type of intervention is outside the scope of this review.

Atkinson 2010

120 African American or Caucasian primigravidae women 19 to 40 years of age in their first trimester of pregnancy in Children’s Hospital & Research Center Oakland, California, USA were included in this study. Women who were smokers, had a pre‐pregnancy BMI higher than 30, had a medical condition that affected bone or taking a medication that affected bone were excluded. Participants were randomly assigned to 1 of 3 groups: group 1: 1000 mg of calcium; group 2: 2000 IU vitamin D; group 3: placebo. The intervention started at week 16 of pregnancy until delivery. The type of intervention is outside the scope of this review.

Azami 2017

90 pregnant women,with least 1 of the risk factors for PE were randomly divided into 3 groups according to randomised selection: Group A received 1 ferrous sulphate tablet (Rooz daru©, Iran) + 1 Claci‐care multimineral‐vitamin D tablet [(VitanePharma©, Germany) contained 800 mg Ca, 200 mg Mg, 8 mg Zn and 400 IU vitamin D3)]per day; Group B received 1 ferrous sulphate tablet (Rooz daru©, Iran,) + 250 mg vitamin C and 55 mg vitamin E, and control group only received Ferrous sulphate daily. This type of intervention is outside the scope of this review.

Baqui 2009

28 pregnant women were enrolled at a maternal health clinic in inner‐city Dhaka, Bangladesh Aged 18 to 34 years; at 27 to 30 weeks of pregnancy. Participants were randomly assigned to 1 of 2 groups: group 1 (N = 14) were assigned to receive a single dose of vitamin D3 70,000 IU (1.75 mg, where 1 mg = 40,000 IU) on day 0 followed by vitamin D3 35,000 IU (0.875 mg) per week starting on day 7 and continuing until delivery); Group 2 (N = 14), were assigned to receive vitamin D3 14,000 IU (0.350 mg) per week starting on day 0 and continuing until delivery. All participants received vitamin D supplementation in different regimens. The type of intervention is outside the scope of this review.

Bhatia 2010

150 consecutive pregnant women pregnant women during their second trimester from 6 villages of a poor socio‐economic region in district Barabanki (latitude 26.8 ºN), Uttar Pradesh, north India.  The participants were initially randomised to receive either no dose or 1 dose of 60,000 IU cholecalciferol under observation in the 5th gestational month. This is not a randomised trial and the comparisons are outside the scope of this review.

Bhatia 2012

299 pregnant women with 12 and 24 weeks of gestation of lower‐middle and middle socio‐economic groups attending the antenatal clinic in Queen Mary Hospital, Chhatrapati Sahuji Maharaj, India, were randomly assigned to 1 of 2 groups, group 1: received 1500 mcg cholecalciferol at induction into the study, or group 2 3000 mcg cholecalciferol at induction as well as at 28 weeks of gestation. All were prescribed 1 g of elemental calcium daily as calcium carbonate without vitamin D. This type of intervention is outside the scope of our review.

Bisgaard 2009

623 women were recruited at 24 weeks of pregnancy. Women were randomised 1:1 to a daily dose of 2400 IU vitamin D3 supplementation or matching placebo tablets (Camette A/S) from pregnancy week 24 to 1 week postpartum. In addition, all women were instructed to continue supplementation of 400 IU of vitamin D3 during pregnancy as recommended by the Danish National Board of Health; thus, the study is a dose comparison of 2800 IU/day vs 400 IU/day of vitamin D3 supplementation. Both groups received vitamin D. This type of intervention is outside the scope of our review.

Chandy 2016

230 infants and mothers giving birth in 2 maternity units of the institution, who intended to continue exclusive breast‐feeding until the first 6 months and come to the hospital of birth for immunisation, were eligible, from September 2012 and June 2014 at the King George Medical University, and Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow (latitude 26°N). After maternal blood sample was collected for serum 25(OH)D 2 to 4 days after delivery, mother–infant pairs were randomly assigned at birth to 1 of 3 treatment regimens described below, to be followed for 9 months.

Intervention was postpartum.

Cockburn 1980

1139 pregnant women were assigned to 1 of 2 wards: group 1 (n = 506) Caucasian pregnant women assigned to 1 ward of the Simpson Memorial Maternity Pavilion, Edinburgh, UK during the 9 months from September to May, were given a daily dietary supplement of 400 IU of vitamin D2 from about the 12th week of pregnancy until delivery; group 2 women (n = 633) were assigned to another ward over the same period and were given a placebo containing no vitamin D. This is not a randomised trial.

Czech‐Kowalska 2013

174 healthy postpartum women who had delivered babies at term in Poland, were randomised to 1 of 2 groups: group 1 (n = 70) received 1200 IU/day vitamin D (cholecalciferol‐D3 as 800 IU/day alone + 400 IU/day from a multiple micronutrient supplements; group 2 (n = 67) received 400 IU/day vitamin D (cholecalciferol‐D3 as placebo + 400 IU/day from multiple micronutrient supplements) during 6 months of lactation. Participants from both groups received vitamin D supplements. The participants were postpartum women. The type of participant and the type of interventions are outside the scope of this review.

Das 2010

200 consecutive pregnant women attending antenatal clinic of at Queen Mary Hospital in CSMMU (former KGMC) were enrolled into the study after taking informed consent and randomly allocated to 1 of 3 groups: (1) Intervention group 1 vitamin D 1,20,000IU in 3 doses each 8 weeks apart + calcium carbonate containing 500 mg elemental calcium with 250 IU vitamin D twice a day, daily throughout pregnancy; (2) Intervention group 2 vitamin D 60,000IU in 3 doses each 8 weeks apart + calcium carbonate containing 500 mg elemental calcium with 250 IU vitamin D twice a day, daily throughout pregnancy; (3) Comparator agent group 3 calcium carbonate containing 500 mg elemental calcium with 250 IU vitamin D twice a day, daily throughout pregnancy.

This is a registry for an ongoing study (open to recruitment).

The type of intervention is outside the scope of this review.

Dawodu 2013

192 Arab women between 12–16 weeks of gestation after their last menstrual period or by ultrasound assessment who had a singleton pregnancy; and planned to receive prenatal and delivery care in primary healthcare clinics affiliated with Tawam Hospital, Al Ain, United Arab Emirates. All participants received vitamin D supplementation in different regimens. The type of intervention is outside the scope of this review.

de Menibus 1984

77 mother‐child couples were divided into 3 groups, according to whether women were not receiving vitamin D (29 couples) during the last 3 months of pregnancy ending in winter or taking 1000 units of vitamin D in the form of uvesterol (21 pairs) or a single dose of 200,000 units of vitamin D at 7 months (27 pairs).

This type of intervention, with no placebo group, is outside the scope of this review.

Etemadifar 2015

45 pregnant women with confirmed multiple sclerosis who attended an outpatient clinic in Isfahan University of Medical Sciences, Iran aged 20 to 40 years with low serum 25‐hydroxyvitamin D (25(OH)D) levels were randomly allocated to 2 groups in an open‐label randomised, controlled clinical Phase I/II pilot study. 1 group received 50,000 IU/week vitamin D3 (n = 21) or routine care (n = 22) from 12 to 16 weeks of gestation till delivery. This type of participant is outside the scope of this review.

Gerais 2015

88 women were recruited at different gestational age, the incidence of vitamin D deficiency about 66%. A single daily dose ranging from 1000 IU to 2000 IU according to the level of deficiency were given to the patient for 6 weeks. In our study 34.1% of women had a level below 10 ng/mL This type of intervention, with no placebo group, is outside the scope of this review.

Hashemipour 2014

160 pregnant women (24 to 26 weeks of gestation) who attended an obstetric clinic in Qazvin, Iran, from December 2011 to March 2012 were randomised, and included in 2 arms. Women in the control group received a multivitamin containing 400 IU vitamin D3 plus 200 mg elemental calcium each day until delivery. Women in the intervention group received a weekly dose of 50,000 IU oral vitamin D3 for 8 weeks (from 26 to 28 weeks of pregnancy) as well as the drug regimen (multivitamin and elemental calcium) given to the control group. Both groups received vitamin D and calcium. This type of intervention is outside the scope of our review.

Hossain 2012

200 pregnant women who attended the Department of Obstetrics & Gynaecology unit 3, Dow University and Civil Hospital Karachi, Pakistan aged between 18 and 40 years were randomised, and included in 2 arms. Participants were allocated to 1 of 2 groups: group 1 (n = 100) received along with ferrous sulphate, 4000 IU of vitamin D3; group 2 (n = 100) received routine antenatal care (ferrous sulphate and calcium). Both groups received above medications from 20 weeks of pregnancy until delivery.

This type of intervention is outside the scope of our review.

Ito 1994

876 singleton pregnant women with blood pressure lower than 140/90 mmHg at 20 weeks’ gestation, and no evidence of proteinuria, who were attending the obstetric clinic of Kumamoto University Hospital, Japan were divided into 2 groups: group 1 (n = 666) women received conventional antenatal care; group 2 (n = 210 women) were managed under a protocol for the prediction of pre‐eclampsia with an angiotensin sensitivity test and prevention of the condition by calcium supplementation. This is not a randomised trial and the type of intervention is outside the scope of this review.

Jamilian 2016

60 participants with GDM were divided into 2 groups of either 1000

IU vitamin D3 and 1000 mg EPO or placebo for 6 weeks. At the beginning and end of the study, fasting blood samples were obtained from the participants to measure related variables.

The type of participant is outside the scope of this review.

Jamilian 2017

140 GDM patients. Participants were randomly divided into 4 groups to receive: (1) 1000 mg omega‐3 fatty acids containing 360 mg eicosapentaenoic acid and 240 mg docosahexaenoic acid (DHA) twice a day + vitamin D placebo (n = 35); (2) 50,000 IU vitamin D every 2 weeks + omega‐3 fatty acids placebo (n = 35); (3) 50,000 IU vitamin D every 2 weeks + 1000 mg omega‐3 fatty acids twice a day (n = 35), and (4) vitamin D placebo + omega‐3 fatty acids placebo (n = 35) for 6 weeks. The type of participant is outside the scope of this review.

Kachhawa 2014

243 pregnant patients attending the antenatal clinic, between 18 to 40 years old and with gestational age between 12 to 16 weeks were randomised into 4 groups in a ratio of 1:1:1:1. Group 1 active control group received 600 units of vitamin D per day, group 2: 1000 units/day, group 3: 2000 units/day and group 4: 4000 units per day. This type of intervention is outside the scope of our review.

Karamali 2014

60 women in Arak, Iran, with GDM were divided into 2 groups to receive Ca + vitamin D supplements or placebo. Individuals in the Ca + vitamin D group (n 30) received 1000 mg Ca/day and 2 pearls containing 1250 μg (50 000 IU) of cholecalciferol (vitamin D3) during the intervention (one at study baseline and another at day 21 of the intervention); those in the placebo group (n 30) received 2 placebos of vitamin D at the mentioned times and placebos of Ca every day for 6 weeks. The type of participant is outside the scope of this review.

Karamali 2015

60 pregnant women Arak, Iran at risk for pre‐eclampsia according to abnormal uterine artery Doppler waveform were randomly divided into 2 groups to receive 50,000 IU vitamin D supplements (n = 30) or receive placebo (n = 30) every 2 weeks from 20 to 32 weeks of gestation. All pregnant women were also taking 400 μg/day folic acid from the start of pregnancy, 60 mg/day ferrous sulphate from the second trimester, and a multivitamin mineral capsule (containing 400 IU vitamin D) from the second half of pregnancy. The type of participant is outside the scope of this review.

Kermack 2017

111 couples were recruited for a 6‐week intervention prior to oocyte retrieval consisted of a daily drink, containing 2 g of DHA plus EPA and 10 mcg of vitamin D, and olive oil and olive oil spreads, all in unmarked containers. The control group received a placebo drink and sun flower oil and spreads, again in unmarked containers. 55 couples were randomised to the treatment group and 56 to placebo. Following IVF, embryos were cultured in an Embryoscope and validated morphokinetic markers of embryo quality were recorded; day 3 and 5 KIDScores (Known Implantation Data Score) were calculated for individual embryos. This type of intervention is outside the scope of our review.

Kiely 2015

144 pregnant women aged older than 18 years of age, with no more than 18 weeks' gestation, in good general health, with low‐risk pregnancy and not consuming > 10 mcg/day vitamin D from supplements were randomised in a 3‐arm, parallel, double‐blind, placebo‐controlled dose‐response intervention study with vitamin D. Group 1 received 10 mcg (400 IU) vitamin D3 once daily taken from baseline visit (approximately 15 weeks' gestation) until endpoint (delivery). Group 2 received 20 mcg (800 IU) vitamin D3 once daily from baseline visit (approximately 15 weeks' gestation) until endpoint (delivery). Group 3 (placebo) received a placebo capsule containing 0 mcg (0 IU) of vitamin D3 taken from baseline visit (approximately 15 weeks' gestation) until endpoint (delivery). The primary outcome was serum 25‐hydroxyvitamin D in pregnant women and cord blood. Women were permitted to continue with self‐administration of antenatal supplements containing ≤ 10 μg vitamin D per day. This type of intervention is outside the scope of our review.

Lalooha 2012

Participants were randomly assigned to 1 of 2 groups: group 1: vitamin D capsule 50,000 U weekly for 8 weeks from 28 gestational age and multivitamin tablet include 400 IU vitamin D daily till termination; group 2: multivitamin tab include 400 IU vitamin D daily till termination. This type of intervention is outside the scope of our review.

Li 2016

103 pregnant GDM women were eligible to participate, using a permuted block randomisation method stratified according to their baseline 1‐hour OGTT results, all participants in their second trimester were randomly assigned to consume 2 servings (100 g per serving) of either plain yogurt (PY) drink (‘PY’ without any vitamin D3 supplement, from Mengniu Dairy, Hohhot, China) or VDY drink (‘PY’ supplemented with 500 IU vitamin D3, from Mengniu Dairy, Hohhot, China), with 1 serving at breakfast and the other 1 at dinner, on a daily basis for a period of 16 weeks. The type of participant is outside the scope of this review.

MacDonald 1986

This trial was registered in 1986 on the Oxford Database of Perinatal Trials and reports the recruitment and follow‐up completed in 1979. The registration form reports a randomised controlled trial to assess the efficacy of calcium and vitamin D supplementation versus placebo in the prevention of maternal and fetal hypocalcaemia. The reports indicates that the sample size was 55 Asian women with morbidity and laboratory results as primary outcomes but no further information is available.

March 2010

226 healthy pregnant women from Greater Vancouver, British Columbia, Canada, from 13 to 24 weeks of gestation were randomly allocated to 10, 25, or 50 mg vitamin D/d from 13 to 24 weeks of gestation until 8 weeks postpartum, with no infant supplementation. Mother and infant blood was collected at 8 weeks postpartum (n = 76, n = 76, and n = 74, respectively). The overall study retention rate from beginning to end was 76% (n = 172). This type of intervention, with no placebo group, is outside the scope of this review.

Marya 1981

45 Hindu pregnant women were randomly assigned to 1 of 2 groups: group 1 (n = 25) received tablets containing 1200 IU vitamin D and 375 mg calcium daily throughout the 3rd trimester; group 2 (n = 20) received oral single dose of 600,000 IU vitamin D2 once during 7th month and 8th month (total 2 doses). This group was compared with group 3 (n = 75) who had not received vitamin D supplements during pregnancy. The results were also compared with data from 25 non pregnant, non‐lactating healthy women. The randomised study compares 2 doses of vitamin D supplementation. The type of study, type of participants and types of interventions are outside the scope of this review.

McLean 2012

Pregnant women, aged 18 years or more, with less than 20 weeks’ gestation at recruitment. Participants will be randomly assigned to 1 of 2 groups: group 1: received high‐dose vitamin D supplementation (5000 IU/day); group 2: standard dose pregnancy vitamin supplementation (400 IU vitamin D daily), administered as an oral capsule, from the time of the first antenatal clinic visit (around 12 weeks’ gestation) until delivery.

This type of intervention is outside the scope of this review.

Mojibian 2015

500 women with gestational age 12 to 16 weeks and serum 25 hydroxy vitamin D (25 (OH) D ) less than 30 ng/mL randomly categorized in 2 groups: group A received 400 IU vitamin D daily and group B 50,000 IU vitamin D every 2 weeks orally until delivery. Maternal and neonatal outcomes were assessed in 2 groups.

This intervention had no placebo group.

Mozzafari 2010

Women between 20 to 45 years old with gestational diabetes at their recent pregnancy, from the list of GDM Diabetes Research Center of Yazd University, and without thyroid disease, kidney disease, bone disease, PCO, liver disease and not using anti‐epilepsy drugs, glucocorticoids, and statins. Exclusion criteria: risk of any illness that requires medication and lack of any willingness to co‐operate, were randomly assigned to 1 of 2 groups: group 1: intramuscular injection of vitamin D with 300,000 IU dose; group 2: control: not receive any intervention. The type of participant is outside the scope of this review.

Mutlu 2013

91 pregnant women aged 16 to 42 years were admitted to Kocaeli Maternity and Children Hospital between April 2011 and April 2012. The participants were randomly divided into 3 groups: 600 IU/d (control group; n = 31); 1,200 IU/d (n = 31), and 2,000 IU/d (n = 32) of vitamin D. All groups received vitamin D supplements. This type of comparison is outside the scope of our review.

Nausheen 2014

315 pregnant women aged 15 to 45 years with less than 16 weeks of gestation in a hospital in Pakistan. Pregnant women with pre‐existing type 1 or type II diabetes, pre‐existing hypertension, multiple fetuses,babies (twins, triplets) or with a diagnosis of pregnancy with a fetal anomaly in scan will be excluded. Participants were randomly assigned to 1 of 3 groups: groups 1 received a dose of 400 IU/day till the time of delivery; group 2 received 2000 IU/day till the time of delivery; group 3 received 4000 IU/day till the time of delivery. This type of intervention is outside the scope of this review.

Niramitmahapanya 2017

76 Thai lactating mothers and their breast‐fed infants were studied with maternal 25 hydroxyvitamin D 25 (OH) D levels of 10‐30 ng/mL determined using Liquid Chromatography Mass Spectrometry Tandem (LC‐MS/MS). 1 group received vitamin D3 1800 IU/day supplementation for 6 weeks, and members of the other group were given a placebo. 25 (OH) D level of colostrum and 6‐week serum from breast‐fed milk were measured by High Performance Liquid Chromatography (HPLC). The data from the 2 groups were analysed and compared. The type of participant is outside the scope of this review.

Pandey 2015

20 women with 25 (OH) D < 20 ng/mL & Hb = 8‐10 g/dL were randomised into groups using a computerised program (8 patients in iron alone group and 12 pregnant mothers in iron + vitamin D group). Recruited pregnant mothers received group iron + vitamin D: tablets containing fixed dose combination of vitamin D (1000 IU) + ferrous ascorbate (100 mg of elemental iron) + folic acid (1 mg) + vit B12 (7.5 mcg) (1 tablet/day) for 12 weeks. Group iron alone: tablets containing fixed dose combination of ferrous ascorbate (100 mg of elemental iron) + folic acid (1.1 mg) (1 tablet/day) for 12 weeks.

This type of intervention is outside the scope of this review.

Qian 2015

60 pregnant women at risk for pre‐eclampsia, experiencing their first pregnancy, aged between 20 and 32 years, between 18 and 20 weeks' gestation, and pregnant with a single fetus were eligible. Each pregnant woman selected for the study showed the following abnormalities on uterine artery Doppler, qualifying them as high‐risk: average resistance index (RI) > 0.67, pulsatility index (PI) > 1.65, and incisura at early diastole phase. Pregnant women were randomised into 2 groups to take either cholecalciferol supplements (n = 30) or placebo (n = 30). This study was retracted due to having data very similar to another paper.

Razavi 2017

120 women with GDM were randomly divided into 4 groups to receive: 1) 1000 mg omega‐3 fatty acids containing 180 mg eicosapentaenoicacid (EPA) and 120 mg docosahexaenoic acid (DHA) twice a day + vitamin D placebo (n = 30); 2) 50,000 IU vitamin D every 2 weeks + omega‐3 fatty acids placebo (n = 30); 3) 50,000 IU vitamin D every 2 weeks + 1000 mg omega‐3 fatty acids twice a day (n = 30) and 4) vitamin D placebo + omega‐3 fatty acids placebo (n = 30) for 6 weeks. The type of participant is outside the scope of this review.

Rostami 2018

1600 and 900 first trimester pregnant women, aged 18 to 40 years, with gestational age < 14 weeks, with singleton pregnancy in Masjed‐Soleiman, Khuzestan province,Iran were randomised by levels of vitamin D in serum to:

Moderate deficiency

  • I1: 50,000 IU of oral D3 weekly for a total duration of 6 weeks

  • I2: 50,000 IU of oral D3 weekly for a total duration of 6 weeks and then a monthly maintenance dose of 50,000 IU of D3 until delivery

  • I3: a single dose of intramuscular administration of 300,000 IU of D3

  • I4: a single dose of intramuscular administration of 300,000 IU of D3 and then a monthly maintenance dose of 50,000 IU of D3 until delivery

Severe deficiency

  • I5: 50,000 IU of oral D3 weekly for a total duration of 12 weeks

  • I6: 50,000 IU of oral D3 weekly for a total duration of 12 weeks and then a monthly maintenance dose of 50,000 IU of D3 until delivery

  • I7: intramuscular administration of 300,000 IU of D3; 2 doses for 6 weeks

  • I8: intramuscular administration of 300,000 IU of D3; 2 doses for 6 weeks, followed by a monthly maintenance dose of 50,000 IU of D3 until delivery

Pregnant women with normal vitamin D status were the controls. All women were allowed to consumed multivitamins containing no more than 400 IU per day of vitamin D3. This type of intervention is outside the scope of this review.

Shakiba 2013

51 healthy pregnant women from the beginning of their second trimester of pregnancy during the autumn and winter of 2009 in recruited from 2 primary care clinics in Yazd (31°53’50”N/54°22’04”E), Iran. Participants were distributed in 3 groups according to their serum 25(OH)D at the beginning of the second trimester of pregnancy. Participants with low concentrations (25(OH)D levels < 20 ng/mL) (n = 17) were treated with 200,000 IU (50,000 IU/week for 4 weeks) of vitamin D (as (cholecalciferol‐D3), followed by supplementation with 50,000 IU/month vitamin D (cholecalciferol‐D3). The other 34 participants were randomly assigned to 1 of 2 groups: group 1 received 50,000 IU/month vitamin D (cholecalciferol‐D3); group 2 received 100,000 IU/month vitamin D (50,000 IU every 2 weeks) of vitamin D (cholecalciferol‐D3) supplementation. All participants received vitamin D supplements. The type of study design and the type of intervention are outside the scope of this review.

Shi 2017

602 women with singleton pregnancy who were diagnosed of pre‐eclampsia in Cangzhou Central Hospital participated in the present trial and were divided into 2 groups using stratified permuted‐block randomisation method with diastolic blood pressure as a factor: (1) nifedipine + VD group (n = 298), given 1 capsule containing nifedipine (10 mg per capsule) and VD (200 IU per capsule) every 15 min orally, up to 4 doses, until blood pressure was equal to or below 150/100 mmHg; (2) nifedipine + placebo group (n = 304), given 1 capsule containing nifedipine (10 mg per capsule) plus glucose (20 mg per capsule) as placebo every 15 minutes orally, up to 4 doses, until blood pressure was equal to or below 150/100 mmHg. The type of participant is outside the scope of this review.

Simsek 2011

Women with gestational diabetes, defined by the WHO criteria: fasting glucose ≥ 7.0 mmol/L or; oral glucose tolerance test: 75 g glucose, 2‐hour glucose ≥ 7.8 mmol/L recognised during pregnancy with a written informed consent, aged between 18 to 42 years. Participants will be randomly assigned to 1 of 2 groups: group 1: cholecalciferol 15,000 IU once a week during pregnancy; group 2: placebo. Per communication with the author, this study was not completed due to low recruitment.

Soheilykhah 2013

120 women with gestational age less than 12 weeks without gestational diabetes, history of PCO, BMI less than 30 kg/m2 before pregnancy, no vitamin D supplementation in the past 6 months were randomised into 2 groups: supplementation with 50,000 IU of vitamin D monthly (2000 IU daily) or 50,000 IU every 2 weeks (4000 IU daily).

Maternal and neonatal outcomes were assessed in 2 groups.

This intervention had no placebo group.

Stephensen 2011

Pregnant women less than 20 weeks' gestation and over 18 years of age with no use of medications known to affect vitamin D metabolism, diagnosis of type 1 diabetes, history of thyroid, renal, or liver disease, problems with digestion or absorption participated in the study at USDA Western Human Nutrition Research Center and clinicians at UC Davis Medical Center. They were distributed into 2 groups, receiving: either 400 IU or 2000 IU of vitamin D per day for the duration of their pregnancy.

Both groups received vitamin D supplements. This type of intervention is outside the scope of our review.

Sudfeld 2017

2300 HIV‐infected pregnant women receiving triple‐drug ART under Option B+ in Dar es Salaam Tanzania. HIV‐infected pregnant women of 12 to 27 weeks' gestation were randomised to either: 1) 3000 IU vitamin D3 taken daily from randomisation in pregnancy until trial discharge at 12 months postpartum; or 2) a matching placebo regimen. Maternal participants are followed‐up at monthly clinic visits during pregnancy, at delivery, and then with their children at monthly postpartum clinic visits.

The type of participant is outside the scope of this review.

Taheri 2014

229 women 18 to 35 years old, who were confirmed to be vitamin D deficient (vitamin D < 75 nmol/L), were randomised into the intervention, and control groups and after 15 weeks consumption of the supplement (2000 IU/day oral vitamin D) and placebo. The study was conducted among reproductive women in a high‐risk population for vitamin D deficiency.

The participants of the study were not pregnant women. The type of participant is outside the scope of this review.

Thiele 2014

16 pregnant women at 24 to 28 weeks' gestation were enrolled. The control group (N = 8) received a prenatal vitamin containing 400 IU vitamin D daily, plus a placebo capsule. The experimental group (N = 8) received the same prenatal vitamin with an additional capsule containing 3400 IU vitamin D, for a total of 3800 IU daily.

This study had no placebo group, therefore, this type of intervention is outside the scope of this review.

Valizadeh 2016

96 women with GDM at weeks 12 to 32 of gestation, age > 16 years, singleton pregnancy were randomly assigned to either the intervention (n = 48) or control group (n = 48). Patients were referred from primary health centres affiliated with Zanjan University of Medical Sciences, as well as private obstetric clinics throughout the city. The type of participant is outside the scope of this review.

von Hurst 2009

235 South Asian women, aged 23 to 68 years, living in Auckland, New Zealand were recruited for the study and those who were insulin resistant ‐ homeostasis model assessment 1 (HOMA1) > 1.93 and had serum 25‐hydroxyvitamin D concentration < 50 nmol/L were randomly assigned to 1 of 2 groups: group 1 (n = 42) received 100 μg (4000 IU) vitamin D(3); group 2 (n = 39) received a placebo daily for 6 months. The study participants were non‐pregnant women. The type of participant is outside the scope of this review.

Wagner 2006a

494 apparently healthy pregnant women (16 to 45 years of age) with 12 to 16 weeks' gestation of singletons attending prenatal care in Medical University of South Carolina, Charleston, United States were randomised into 1 of 3 groups stratified by race: group 1 received 400 IU vitamin D (cholecalciferol‐D3)/day; group 2 received 2000 IU vitamin D (cholecalciferol‐D3)/day; and group 3 received 4000 IU vitamin D (cholecalciferol‐D3)/day until delivery. All women received daily multiple micronutrients supplements. All women received vitamin D supplementation at different doses. The type of intervention is outside the scope of this review.

Wagner 2006b

This is an analysis of data from 2 randomised controlled trials by the same research group (Wagner 2006a; Wagner 2010a). In Wagner 2006a, women were randomised to 400, 2000, or 4000 IU vitamin D (cholecalciferol‐D3)/day, stratified by race. In Wagner 2010a, participants were randomised to 2000 or 4000 IU vitamin D (cholecalciferol‐D3)/day.

Wagner 2013

258 women, exclusively breastfeeding (n = 201) and formula‐feeding (n = 57) women participating in a prospective, randomised controlled trial of vitamin D supplementation were compared at baseline 1 month postpartum (V1), at 4 months (V4), and 7 months postpartum (V7) on the basis of vitamin D status (measured by total circulating 25(OH)D concentration) and BMI.

The type of intervention is outside the scope of this review.

Weiss 2009

881 pregnant women with either a personal history of asthma or allergies or a similar history in the spouse or partner, between 18 and 40 years of age and at an estimated gestational age between 10 and 18 weeks, were recruited at a scheduled obstetrical prenatal visit at 3 clinical centres: Boston Medical Center (Boston, MA), Washington University at Saint Louis (St. Louis, MO), and Kaiser Permanente Southern California Region (San Diego, CA). Participants were randomised to either vitamin D (cholecalciferol, 4000 IU/day; equivalent to 100 μg/day) or placebo. All pregnant mother participants received prenatal vitamins containing 400 IU (10 μg/day) of cholecalciferol; thus, the vitamin D arm received a total of 4400 IU/day (110 μg/day) and the placebo arm received 400 IU/day (10 μg/day). Both groups received vitamin D supplements. This type of intervention is outside the scope of our review.

Yap 2014

179 pregnant women 18 years of age or older, with singleton pregnancy, with plasma 25‐hydroxivitamin D (25OHD) concentrations lower than 32 ng/mL, less than 20 weeks of gestation were randomly assigned to 1 of 2 groups: group 1 (n = 89) received 5000 IU/d of vitamin D (cholecalciferol‐D3) until delivery; group 2 (n = 90) received 400 IU/d of vitamin D (cholecalciferol‐D3) until delivery. Outcomes included glycaemia and glucose tolerance, gestational diabetes at 26‐28 weeks of gestation; neonatal 25OHD, maternal hypertension, mode of delivery, prematurity, birthweight, crown‐heel length, occipitofrontal head circumference. All participants received vitamin D supplements at different doses. The type of intervention is outside the scope of this review.

Yazdchi 2016

76 pregnant women without a prior diagnosis of glucose intolerance in the first trimester were asked to participate in a 75‐g OGTT at 24‐28 weeks of gestation. Diagnosis of GDM was based on the International Association of Diabetes and Pregnancy Study Groups criteria. Patients were recruited from Al‐Zahra Hospital, the academic outpatient centre of the Tabriz University of Medical Sciences, Tabriz, Iran. Participants in the vitamin D group received oral capsules containing 50,000 IU vitamin D 3 (D‐Vitin50000; Zahravi Pharm Co., Iran), once every 2 weeks for 2 months, for a total of 4 capsules. Those in the placebo group received placebos composed of paraffin oil (Dana Pharm Co., Iran) using the same schedule, for a total of 4 placebos. The type of participant is outside the scope of this review.

Zhang 2016

133 pregnant women with GDM during weeks 24 to 28 of pregnancy. The patients were randomly divided into 4 groups. The control group (n = 20) received a placebo (sucrose; 1 granule/day), the low‐dosage group (n = 38) received the daily recommended intake of 200 IU vitamin D (calciferol) daily, the medium‐dosage group (n = 38) received 50,000 IU monthly (2000 IU daily for 25 days) and the high‐dosage group (n = 37) received 50,000 IU every 2 weeks (4000 IU daily for 12.5 days). The general characteristics and dietary intakes of the patients with GDM were similar between each group. High‐dose vitamin D supplementation (50,000 IU every 2 weeks) significantly improved insulin resistance in pregnant women with GDM. The type of participant is outside the scope of this review.

BMI: body mass index
Ca: calcium
DHA: docosahexaenoic
DRI: dietary references intakes
EPA: eicosapentaenoic acid
FPG: fasting plasma glucose
GDM: gestational diabetes mellitus
Hb: haemoglobin
IU: international units
IVF: in vitro fertilisation
mcg: microgram
OGTT: oral glucose tolerance test
PCO: polycystic ovary syndrome
PTH: parathyroid hormone
25OHD: 25‐hydroxycholecalciferol

Characteristics of studies awaiting assessment [ordered by study ID]

Bimson 2017

Methods

Randomised, double‐blind, placebo‐controlled study.

Participants

67 pregnant women with vitamin D (25(OH)D) level lower than 25 ng/mL, English or Spanish literacy, age 18 years and gestational age less than 20 weeks.

Interventions

Women were randomised to: group 1 (n = 36) received 50,000 IU vitamin D3 or group 2 (n = 31) received placebo, once a week for 8 weeks in addition to a prenatal vitamin with 400 IU vitamin D3.

Outcomes

After 8 weeks of treatment, VD levels were 48.1 19.6 ng/mL and 21.1 5.9 ng/mL (P <.0001) with 84.4% and 3.9% achieving VD sufficiency (> 30 ng/mL) in the TG and PG, respectively (P <.0001). At delivery, VD levels were 28.8 9.8 ng/mL and 20.58.9 ng/mL in the TG and PG, respectively (P < 0.0027).

Notes

Mount Sinai, West Hospital, New York, NY, USA

Das 2009

Methods

Randomised controlled trial

Participants

150 pregnant women in 2nd trimester from 6 villages of North India

Interventions

All women were educated regarding sunshine exposure & were given 1 g calcium carbonate daily. They were randomised to 3 groups no vitamin D, 60,000 IU vitamin D and 2,40,000 IU vitamin D supplementation.

Outcomes

Maternal: serum 25 OH D levels at baseline and post delivery

Notes

This is an abstract. No information on registry or funding source.

IU: international units
PG: placebo group
TG: treatment group
VD: vitamin D
25OHD: 25‐hydroxycholecalciferol

Characteristics of ongoing studies [ordered by study ID]

Baird 2016

Trial name or title

Southampton PRegnancy Intervention for the Next Generation (SPRING): protocol for a randomised controlled trial

Methods

Randomised controlled trial that uses a 2‐by‐2 factorial design

Participants

600 women, with less than 17 weeks' gestation at recruitment based on LMP, aged over 18 years, with a singleton pregnancy and aiming to give birth at local maternity (Princess Anne) hospital

Interventions

Healthy conversation skills support plus vitamin D supplementation (1000 IU cholecalciferol) (n = 150); healthy conversation skills support plus placebo (n = 150); usual care plus vitamin D supplementation (n = 150); usual care plus placebo (n = 150)

Outcomes

This trial is evaluating 2 approaches to improving maternal diet: a behaviour change intervention and vitamin D supplementation. The factorial design of this trial has the advantage of enabling each intervention be tested separately as well as allowing exploration of the synergistic effect of both interventions on women’s diets and vitamin D levels.

Starting date

Registered on 13 September 2013

Contact information

Janis Baird: MRC Lifecourse Epidemiology Unit, University of Southampton,Southampton SO16 6YD, UK. [email protected]

Notes

Sponsor: Medical Research Council,NIHR Southampton Nutrition Biomedical Research Centre and Danone Nutricia Early Life Nutrition.

Jelsma 2013

Trial name or title

DALI: vitamin D and lifestyle intervention for gestational diabetes mellitus (GDM) prevention.

Methods

Randomised controlled trial with a factorial design.

Participants

Pregnant women with gestational age at recruitment < 12 weeks, and more than 18 years of age.

Inclusion criteria: pre‐pregnancy BMI (self‐reported weight, measured height) is >= 29 kg/m2), sufficiently fluent in major language of the country of recruitment, being able to be moderately physically active, giving written informed consent, agree to give birth in 1 of the participating hospitals. Exclusion criteria: pre‐existing diabetes, diagnosed with (gestational) diabetes mellitus before randomisation, defined as fasting glucose ≥ 5.1 mmol/L and/or 1‐hour glucose ≥ 10 mmol/L and/or 2‐hour glucose ≥ 8.5 mmol/L at baseline, not able to walk at least 100 metres safely, requirement for complex diets, advanced chronic conditions (e.g. valvular heart disease), significant psychiatric disease, unable to speak major language of the country of recruitment fluently, known abnormal calcium metabolism (hypo/hyperparathyroidism, nephrolithiasis, hypercalciuria) or hypercalciuria detected at screening (0.6 mmol/mmol creatinine in spot morning urine) and twin pregnancy.

Interventions

The design is that of 2 trials with a factorial design: PA, diet, PA & diet, control, vitamin D, PA & diet and placebo, vitamin D & PA & diet, placebo; to compare the impact of increased PA, enhanced nutrition and vitamin D supplementation either alone or in combination on maternal glucose tolerance, maternal weight gain and insulin sensitivity.

The doses of vitamin D that will be tested in the dosing study are 500, 1000 and 1500 IU/day. One of these doses will be used in the trial.

Outcomes

Maternal: weight gain during pregnancy, fasting plasma glucose, HbA1c, fasting C peptide, leptin, triglycerides, free fatty acids, high density lipoprotein cholesterol (HDL‐C) and low density lipoprotein cholesterol (LDL‐C), adiponectin 2. 3 beta‐hydroxybutyrate, blood pressure, C‐reactive protein.

Infant: neonatal growth, adiposity, adipo‐insular axis, glucose‐insulin axis, electrolyte concentrations, clinical outcomes and hypoxia exposure at birth, biparietal diameter, head circumference, abdominal circumference, femur length and determinants of fetal body composition variables (lean body mass and fat body mass, C‐peptide, glucose, leptin, triglycerides, 3‐ beta‐hydroxybutyric acid, pH and erythropoietin, jaundice, hypocalcaemia, hypomagnesaemia.

Starting date

21/11/2011.

Contact information

Dr David Simmons

Addenbrooke's Treatment Centre
Hills Road, Cambridge CB2 0QQ

UK

Notes

Sponsor: European Union (EU) (Belgium) ‐ Seventh Framework Programme (FP7).

Judkins 2010

Trial name or title

A randomised double‐blinded interventional trial to determine the effect of 50,000 IU vitamin D supplementation monthly or twice monthly from 20 weeks' gestation.

Methods

Randomised double‐masked clinical trial with randomisation at the individual level. Method of sequence generation: serial tossing of a coin. Allocation will be not concealed.

Participants

Pregnant women seeking maternity care with midwifery services involved in the study. Exclusion criteria: antenatal vitamin D level is > 75 nmol/L when enrolling in study.

Interventions

Participants will be assigned to 1 of 2 groups: group 1: will receive 50,000 IU tablets twice monthly, 2 weeks apart; group 2: will receive 50,000 IU monthly and a placebo monthly, 2 weeks apart from 20 weeks' gestation until delivery of baby.

The placebo tablet contains lactose monohydrate, acacia, calcium carbonate, castor oil, maize starch, povidone, sucrose, purified talc, hydrated silica, powdered cellulose, magnesium stearate, shellac, gelatin, beeswax white, titanium dioxide and prepared theobroma.

Outcomes

Infant: vitamin D levels taken from the cord blood samples at delivery. If emergencies at delivery prevent a cord blood sample being taken then a maternal venous blood sample will be taken for analysis.

Starting date

Status: not yet recruiting participants.

Contact information

Dr Annie Judkins

Newtown Union Health Service
14 Hall Ave, Newtown,. Wellington 6021, New Zealand

Email: [email protected]

Notes

Sponsors: Royal New Zealand College of GP's, New Zealand and Wellington Medical Research Foundation, New Zealand.

ACTR Number: ACTRN12610001044011.

Lindqvist 2010

Trial name or title

Vitamin D supplementation for prevention of placenta mediated pregnancy complications.

Methods

Randomised, controlled trial.

Participants

Pregnant women > 18 years of age, from 3 maternal healthcare units who agree to participate in the study.

Exclusion criteria: < 18 years of age, hyperparathyroidism and sarcoidosis.

Interventions

Participants will be randomly assigned to 1 of 2 groups: group 1: will receive vitamin D, oral drops; group 2: will receive placebo.

Outcomes

Maternal: pre‐eclampsia, blood loss at delivery.

Infant: blood flow in umbilical artery, growth restriction, and prematurity.

Starting date

06/04/2011.

Contact information

N/A

Notes

Sponsor: Karolinska University Hospital.

EudraCT Number: 2010‐019483‐37.

Mosalanejad 2016

Trial name or title

Compare the effect of vitamin D and calcium plus vitamin D on pregnancy outcomes in pregnant women

Methods

Randomised single‐blind controlled clinical trial

Participants

460 pregnant women between 20 to 40 ages, with gestational age less than 10 weeks of gestation; with no history of diabetes; HTN; a history of PCO syndrome; lack of family history of diabetes in first‐degree relatives; no family history of high blood pressure in first‐degree relatives; tend the sick; the vitamin D < 30 ng/L in women 10 to 16 weeks of pregnancy; BMI between 19 to 26; lack of vitamin D during the last 6 months; singleton pregnancy; gestational first to third who referred to women clinic of Shariati Hospital of Bandarabas, Iran from 2015‐2016

Interventions

In intervention group is used vitamin D3 1000 unit oral daily since 16 weeks until the end of pregnancy. They also prescribed as routine prenatal care multivitamin that has 400 units vitamin D‐ca since 16 weeks of pregnancy. The control group is used multivitamin that has 400 unit vitamin D daily until the end of pregnancy

Outcomes

Vitamin D concentration in the serum, diabetes, pre‐eclampsia, preterm delivery

Starting date

Registration date: 2016‐12‐26, 1395/10/06

Contact information

Najmehsadat Mosalanejad, email: [email protected]

Notes

Sponsor: Hormozgan University of Medical Sciences

Rasmussen 2009

Trial name or title

Effects of vitamin D supplement before and during and after pregnancy on complications, birthweight and bone mineral density during lactation.

Methods

Double‐blind, randomised, placebo‐controlled trial.

Participants

400 apparently healthy women 30 to 35 years of age, all with concentrations of P‐25‐hydroxyvitamin D‐ lower than 50 nmol/L. All women included attempts to get pregnant. Visits take place at Clinic of Osteoporosis, Department of Endocrinology, at Aarhus University Hospital, Aarhus, Denmark.

Women with infertility, an intake of 400 IU or more vitamin D/day, cancer, history of alcohol or drug abuse, calcium metabolic disturbances or spontaneous abortion within last 6 months will be excluded.

Interventions

Participants will be randomly assigned to 1 of 3 groups: group 1: will receive 35 mcg per day cholecalciferol; group 2: will receive 70 mcg per day cholecalciferol; group 3: will receive placebo. All women will receive 2 tablets daily from baseline until 16 weeks after delivery.

Intervention with cholecalciferol or placebo starts before pregnancy is achieved and continues until 4 months after the women has given birth.  

Outcomes

Primary:

Infant: birthweight.

Maternal: none.

Secondary:

Infant: weight, crown‐heel length and head circumference, and infections within 16 weeks after birth. Concentration of 25‐hydroxyvitamin D in umbilical cord and venous sample 16 weeks after birth.

Maternal: postpartum effects of vitamin D supplement on maternal bone mineral density, concentration of 25‐hydroxyvitamin D in mothers milk, incidence of pre‐eclampsia and abortions.

Starting date

Date of start: 12/2009.

Status: recruiting participants.

Estimated study completion date: December 2011.

Contact information

Gitte Bloch Rasmussen, MD

Department of Endocrinology, Aarhus University Hospital

University of Aarhus

Tel: +45 89 4976 81

Email: [email protected]

Notes

Sponsor: University of Aarhus, Denmark.

BMI: body mass index
DRI: dietary references intakes
GDM: gestational diabetes mellitus
HTN: hypertension
IU: international units
LMP: last menstrual period
mcg: microgram
PA: physical activity
PCO: polycystic ovary

Data and analyses

Open in table viewer
Comparison 1. Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pre‐eclampsia (ALL) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

Analysis 1.1

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 1 Pre‐eclampsia (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 1 Pre‐eclampsia (ALL).

2 Gestational diabetes (ALL) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

Analysis 1.2

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 2 Gestational diabetes (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 2 Gestational diabetes (ALL).

3 Maternal adverse events Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 3 Maternal adverse events.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 3 Maternal adverse events.

3.1 Severe postpartum haemorrhage

1

1134

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.51, 0.91]

3.2 Nephritic syndrome

1

135

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.01, 4.06]

3.3 Hypercalcaemia

1

1134

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Preterm birth (less than 37 weeks' gestation) (ALL) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

Analysis 1.4

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 4 Preterm birth (less than 37 weeks' gestation) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 4 Preterm birth (less than 37 weeks' gestation) (ALL).

5 Low birthweight (less than 2500 g) (ALL) Show forest plot

5

697

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.35, 0.87]

Analysis 1.5

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 5 Low birthweight (less than 2500 g) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 5 Low birthweight (less than 2500 g) (ALL).

6 Pre‐eclampsia (by start of supplementation) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

Analysis 1.6

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 6 Pre‐eclampsia (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 6 Pre‐eclampsia (by start of supplementation).

6.1 Less than 20 weeks of pregnancy

1

138

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.06, 1.37]

6.2 20 weeks of pregnancy or more

2

219

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.21, 1.24]

6.3 Unknown/unreported/mixed

1

142

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.98]

7 Pre‐eclampsia (by pre‐gestational BMI) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

Analysis 1.7

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 7 Pre‐eclampsia (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 7 Pre‐eclampsia (by pre‐gestational BMI).

7.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

7.2 Normal weight (18.5 to 24.9)

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.21, 1.33]

7.3 Overweight (25 or higher)

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

7.4 Unknown/unreported/mixed

2

280

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.86]

8 Pre‐eclampsia (by supplementation scheme/regimen) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

Analysis 1.8

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 8 Pre‐eclampsia (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 8 Pre‐eclampsia (by supplementation scheme/regimen).

8.1 Single dose

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.21, 1.33]

8.2 Daily

2

192

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.08, 1.20]

8.3 Weekly/monthly

1

142

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.98]

9 Pre‐eclampsia (by skin pigmentation based on Fitzpatrick skin tone chart) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

Analysis 1.9

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 9 Pre‐eclampsia (by skin pigmentation based on Fitzpatrick skin tone chart).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 9 Pre‐eclampsia (by skin pigmentation based on Fitzpatrick skin tone chart).

9.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

9.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

9.3 Unknown/unreported/mixed

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

10 Pre‐eclampsia (by latitude) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

Analysis 1.10

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 10 Pre‐eclampsia (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 10 Pre‐eclampsia (by latitude).

10.1 Between Tropics of Cancer and Capricorn

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

10.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

10.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

11 Pre‐eclampsia (by season at the start of pregnancy) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

Analysis 1.11

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 11 Pre‐eclampsia (by season at the start of pregnancy).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 11 Pre‐eclampsia (by season at the start of pregnancy).

11.1 Summer

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

11.2 Fall

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

11.3 Winter

1

138

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.06, 1.37]

11.4 Mixed/unknown

2

307

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.31, 0.88]

12 Gestational diabetes (by start of supplementation) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

Analysis 1.12

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 12 Gestational diabetes (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 12 Gestational diabetes (by start of supplementation).

12.1 Less than 20 weeks of pregnancy

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.34, 2.28]

12.2 20 weeks of pregnancy or more

2

219

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.05, 3.45]

12.3 Unknown/unreported/mixed

1

87

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.13, 0.82]

13 Gestational diabetes (by pre‐gestational BMI) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

Analysis 1.13

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 13 Gestational diabetes (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 13 Gestational diabetes (by pre‐gestational BMI).

13.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13.2 Normal weight (18.5 to 24.9)

2

305

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.33, 2.05]

13.3 Overweight (25 or higher)

2

141

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.13, 0.79]

13.4 Unknown/unreported/mixed

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14 Gestational diabetes (by supplementation scheme/regimen) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

Analysis 1.14

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 14 Gestational diabetes (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 14 Gestational diabetes (by supplementation scheme/regimen).

14.1 Single dose

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.03, 8.28]

14.2 Daily

2

141

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.13, 0.79]

14.3 Weekly/monthly

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.34, 2.28]

15 Gestational diabetes (by skin pigmentation based on Fitzpatrick skin tone chart) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

Analysis 1.15

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 15 Gestational diabetes (by skin pigmentation based on Fitzpatrick skin tone chart).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 15 Gestational diabetes (by skin pigmentation based on Fitzpatrick skin tone chart).

15.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

15.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

15.3 Unknown/unreported/mixed

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

16 Gestational diabetes (by latitude) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

Analysis 1.16

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 16 Gestational diabetes (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 16 Gestational diabetes (by latitude).

16.1 Between Tropics of Cancer and Capricorn

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

16.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

16.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

17 Gestational diabetes (by season at the start of supplementation) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

Analysis 1.17

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 17 Gestational diabetes (by season at the start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 17 Gestational diabetes (by season at the start of supplementation).

17.1 Summer

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

17.2 Fall

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

17.3 Winter

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.34, 2.28]

17.4 Mixed/unknown

2

252

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.14, 0.82]

18 Preterm birth (less than 37 weeks' gestation) (by start of supplementation) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

Analysis 1.18

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 18 Preterm birth (less than 37 weeks' gestation) (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 18 Preterm birth (less than 37 weeks' gestation) (by start of supplementation).

18.1 Less than 20 weeks of pregnancy

3

1149

Risk Ratio (M‐H, Random, 95% CI)

0.73 [0.26, 2.04]

18.2 20 weeks of pregnancy or more

4

491

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.13, 1.87]

18.3 Unknown/unreported/mixed

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

19 Preterm birth (less than 37 weeks' gestation) (by pre‐gestational BMI) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

Analysis 1.19

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 19 Preterm birth (less than 37 weeks' gestation) (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 19 Preterm birth (less than 37 weeks' gestation) (by pre‐gestational BMI).

19.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

19.2 Normal weight (18.5 to 24.9)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

19.3 Overweight (25 or higher)

2

138

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.15, 2.46]

19.4 Unknown/unreported/mixed

5

1502

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.27, 1.54]

20 Preterm birth (less than 37 weeks' gestation) (by supplementation scheme/regimen) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

Analysis 1.20

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 20 Preterm birth (less than 37 weeks' gestation) (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 20 Preterm birth (less than 37 weeks' gestation) (by supplementation scheme/regimen).

20.1 Single dose

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

20.2 Daily

6

1495

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.23, 1.58]

20.3 Weekly/monthly

1

145

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.32, 1.54]

21 Preterm birth (less than 37 weeks' gestation) (by skin pigmentation based on Fitzpatrick skin tone chart) Show forest plot

9

1943

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.30, 0.91]

Analysis 1.21

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 21 Preterm birth (less than 37 weeks' gestation) (by skin pigmentation based on Fitzpatrick skin tone chart).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 21 Preterm birth (less than 37 weeks' gestation) (by skin pigmentation based on Fitzpatrick skin tone chart).

21.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

21.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

21.3 Unknown/unreported/mixed

9

1943

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.30, 0.91]

22 Preterm birth (less than 37 weeks' gestation) (by latitude) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

Analysis 1.22

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 22 Preterm birth (less than 37 weeks' gestation) (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 22 Preterm birth (less than 37 weeks' gestation) (by latitude).

22.1 Between Tropics of Cancer and Capricorn

2

358

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.05, 1.34]

22.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

5

1282

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.59, 1.66]

22.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

23 Preterm birth (less than 37 weeks' gestation) (by season at the start of supplementation) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

Analysis 1.23

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 23 Preterm birth (less than 37 weeks' gestation) (by season at the start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 23 Preterm birth (less than 37 weeks' gestation) (by season at the start of supplementation).

23.1 Summer

2

199

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.34, 1.53]

23.2 Winter

1

34

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

23.3 Mixed/unknown

4

1407

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.19, 1.66]

24 Low birthweight (less than 2500 g) (by start of supplementation) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

Analysis 1.24

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 24 Low birthweight (less than 2500 g) (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 24 Low birthweight (less than 2500 g) (by start of supplementation).

24.1 Less than 20 weeks of pregnancy

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

24.2 20 weeks of pregnancy or more

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

24.3 Unknown/unreported/mixed

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25 Low birthweight (less than 2500 g) (by pre‐gestational BMI) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

Analysis 1.25

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 25 Low birthweight (less than 2500 g) (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 25 Low birthweight (less than 2500 g) (by pre‐gestational BMI).

25.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25.2 Normal weight (18.5 to 24.9)

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.19, 0.98]

25.3 Overweight (25 or higher)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25.4 Unknown/unreported/mixed

2

326

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.14, 0.88]

26 Low birthweight (less than 2500 g) (by supplementation scheme/regimen) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

Analysis 1.26

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 26 Low birthweight (less than 2500 g) (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 26 Low birthweight (less than 2500 g) (by supplementation scheme/regimen).

26.1 Single dose

2

365

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.16, 0.65]

26.2 Daily

1

126

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.23, 1.21]

26.3 Weekly/monthly

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

27 Low birthweight (less than 2500 g) (by skin pigmentation based on Fitzpatrick skin to Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

Analysis 1.27

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 27 Low birthweight (less than 2500 g) (by skin pigmentation based on Fitzpatrick skin to.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 27 Low birthweight (less than 2500 g) (by skin pigmentation based on Fitzpatrick skin to.

27.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

27.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

27.3 Unknown/unreported/mixed

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

28 Low birthweight (less than 2500 g) (by latitude) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

Analysis 1.28

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 28 Low birthweight (less than 2500 g) (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 28 Low birthweight (less than 2500 g) (by latitude).

28.1 Between Tropics of Cancer and Capricorn

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

28.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

28.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29 Low birthweight (less than 2500 g) (by season at the start of pregnancy) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

Analysis 1.29

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 29 Low birthweight (less than 2500 g) (by season at the start of pregnancy).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 29 Low birthweight (less than 2500 g) (by season at the start of pregnancy).

29.1 Summer

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29.2 Fall

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29.3 Winter

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29.4 Mixed/unknown

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

30 Caesarean section Show forest plot

10

1104

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.80, 1.21]

Analysis 1.30

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 30 Caesarean section.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 30 Caesarean section.

31 Gestational hypertension Show forest plot

2

1130

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.41, 1.49]

Analysis 1.31

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 31 Gestational hypertension.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 31 Gestational hypertension.

32 Maternal death (death while pregnant or within 42 days of termination of pregnancy) (ALL) Show forest plot

1

180

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

Analysis 1.32

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 32 Maternal death (death while pregnant or within 42 days of termination of pregnancy) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 32 Maternal death (death while pregnant or within 42 days of termination of pregnancy) (ALL).

33 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL) Show forest plot

14

2470

Mean Difference (IV, Random, 95% CI)

35.66 [24.19, 47.13]

Analysis 1.33

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 33 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 33 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

34 Birth length (cm) Show forest plot

8

931

Mean Difference (IV, Random, 95% CI)

0.57 [0.19, 0.95]

Analysis 1.34

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 34 Birth length (cm).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 34 Birth length (cm).

35 Head circumference at birth (cm) Show forest plot

8

1841

Mean Difference (IV, Random, 95% CI)

0.11 [‐0.21, 0.44]

Analysis 1.35

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 35 Head circumference at birth (cm).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 35 Head circumference at birth (cm).

36 Birthweight (g) Show forest plot

17

2828

Mean Difference (IV, Random, 95% CI)

80.30 [‐14.40, 175.00]

Analysis 1.36

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 36 Birthweight (g).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 36 Birthweight (g).

37 Stillbirth Show forest plot

3

584

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.06, 1.98]

Analysis 1.37

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 37 Stillbirth.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 37 Stillbirth.

38 Neonatal death Show forest plot

2

326

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.04, 1.67]

Analysis 1.38

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 38 Neonatal death.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 38 Neonatal death.

39 Apgar score less than seven at five minutes Show forest plot

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.11, 2.53]

Analysis 1.39

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 39 Apgar score less than seven at five minutes.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 39 Apgar score less than seven at five minutes.

Open in table viewer
Comparison 2. Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pre‐eclampsia (ALL) Show forest plot

4

1174

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.32, 0.78]

Analysis 2.1

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 1 Pre‐eclampsia (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 1 Pre‐eclampsia (ALL).

2 Gestational diabetes (ALL) Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

Analysis 2.2

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 2 Gestational diabetes (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 2 Gestational diabetes (ALL).

3 Preterm birth (less than 37 weeks' gestation) (ALL) Show forest plot

5

942

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.01, 2.28]

Analysis 2.3

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).

4 Low birthweight (less than 2500 g) (ALL) Show forest plot

2

110

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.10, 4.55]

Analysis 2.4

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 4 Low birthweight (less than 2500 g) (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 4 Low birthweight (less than 2500 g) (ALL).

5 Caesarean section Show forest plot

2

146

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.87, 1.54]

Analysis 2.5

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 5 Caesarean section.

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 5 Caesarean section.

6 Gestational hypertension Show forest plot

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.06, 1.12]

Analysis 2.6

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 6 Gestational hypertension.

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 6 Gestational hypertension.

7 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

12.5 [3.80, 21.20]

Analysis 2.7

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 7 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 7 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

8 Birth length (cm) Show forest plot

3

194

Mean Difference (IV, Random, 95% CI)

‐0.07 [‐0.67, 0.52]

Analysis 2.8

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 8 Birth length (cm).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 8 Birth length (cm).

9 Head circumference at birth (cm) Show forest plot

3

198

Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.39, 0.33]

Analysis 2.9

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 9 Head circumference at birth (cm).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 9 Head circumference at birth (cm).

10 Birthweight (g) Show forest plot

3

194

Mean Difference (IV, Random, 95% CI)

42.39 [‐86.96, 171.74]

Analysis 2.10

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 10 Birthweight (g).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 10 Birthweight (g).

11 Neonatal death Show forest plot

1

660

Risk Ratio (M‐H, Random, 95% CI)

0.2 [0.01, 4.15]

Analysis 2.11

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 11 Neonatal death.

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 11 Neonatal death.

Open in table viewer
Comparison 3. Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gestational diabetes (ALL) Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.10, 1.73]

Analysis 3.1

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 1 Gestational diabetes (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 1 Gestational diabetes (ALL).

2 Maternal adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 2 Maternal adverse events.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 2 Maternal adverse events.

2.1 Hypercalcaemia

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Hypercalciuria

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.02, 3.97]

3 Preterm birth (less than 37 weeks' gestation) (ALL) Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.68, 1.59]

Analysis 3.3

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).

4 Low birthweight (less than 2500 g) (ALL) Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.82, 1.51]

Analysis 3.4

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 4 Low birthweight (less than 2500 g) (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 4 Low birthweight (less than 2500 g) (ALL).

5 Caesarean section Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.95, 1.27]

Analysis 3.5

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 5 Caesarean section.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 5 Caesarean section.

6 Gestational hypertension Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.31, 2.79]

Analysis 3.6

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 6 Gestational hypertension.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 6 Gestational hypertension.

7 Maternal death (death while pregnant or within 42 days of termination of pregnancy) Show forest plot

1

1300

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.02, 3.98]

Analysis 3.7

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 7 Maternal death (death while pregnant or within 42 days of termination of pregnancy).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 7 Maternal death (death while pregnant or within 42 days of termination of pregnancy).

8 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL) Show forest plot

1

635

Mean Difference (IV, Random, 95% CI)

75.17 [71.97, 78.37]

Analysis 3.8

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 8 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 8 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

9 Birth length (cm) Show forest plot

1

1297

Mean Difference (IV, Fixed, 95% CI)

0.0 [‐0.28, 0.28]

Analysis 3.9

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 9 Birth length (cm).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 9 Birth length (cm).

10 Head circumference at birth (cm) Show forest plot

1

1297

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.17, 0.17]

Analysis 3.10

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 10 Head circumference at birth (cm).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 10 Head circumference at birth (cm).

11 Birthweight (g) Show forest plot

1

1297

Mean Difference (IV, Random, 95% CI)

‐7.0 [‐55.95, 41.95]

Analysis 3.11

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 11 Birthweight (g).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 11 Birthweight (g).

12 Stillbirth Show forest plot

1

1300

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.29, 1.46]

Analysis 3.12

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 12 Stillbirth.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 12 Stillbirth.

13 Neonatal death Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.22, 2.14]

Analysis 3.13

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 13 Neonatal death.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 13 Neonatal death.

Study flow diagram for this update
Figuras y tablas -
Figure 1

Study flow diagram for this 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 2

'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 3

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.

Funnel plot of comparison: 1 Vitamin D alone versus no treatment/placebo (no vitamins or minerals), outcome: 1.15 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).
Figuras y tablas -
Figure 4

Funnel plot of comparison: 1 Vitamin D alone versus no treatment/placebo (no vitamins or minerals), outcome: 1.15 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 1 Pre‐eclampsia (ALL).
Figuras y tablas -
Analysis 1.1

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 1 Pre‐eclampsia (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 2 Gestational diabetes (ALL).
Figuras y tablas -
Analysis 1.2

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 2 Gestational diabetes (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 3 Maternal adverse events.
Figuras y tablas -
Analysis 1.3

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 3 Maternal adverse events.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 4 Preterm birth (less than 37 weeks' gestation) (ALL).
Figuras y tablas -
Analysis 1.4

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 4 Preterm birth (less than 37 weeks' gestation) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 5 Low birthweight (less than 2500 g) (ALL).
Figuras y tablas -
Analysis 1.5

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 5 Low birthweight (less than 2500 g) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 6 Pre‐eclampsia (by start of supplementation).
Figuras y tablas -
Analysis 1.6

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 6 Pre‐eclampsia (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 7 Pre‐eclampsia (by pre‐gestational BMI).
Figuras y tablas -
Analysis 1.7

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 7 Pre‐eclampsia (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 8 Pre‐eclampsia (by supplementation scheme/regimen).
Figuras y tablas -
Analysis 1.8

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 8 Pre‐eclampsia (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 9 Pre‐eclampsia (by skin pigmentation based on Fitzpatrick skin tone chart).
Figuras y tablas -
Analysis 1.9

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 9 Pre‐eclampsia (by skin pigmentation based on Fitzpatrick skin tone chart).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 10 Pre‐eclampsia (by latitude).
Figuras y tablas -
Analysis 1.10

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 10 Pre‐eclampsia (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 11 Pre‐eclampsia (by season at the start of pregnancy).
Figuras y tablas -
Analysis 1.11

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 11 Pre‐eclampsia (by season at the start of pregnancy).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 12 Gestational diabetes (by start of supplementation).
Figuras y tablas -
Analysis 1.12

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 12 Gestational diabetes (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 13 Gestational diabetes (by pre‐gestational BMI).
Figuras y tablas -
Analysis 1.13

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 13 Gestational diabetes (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 14 Gestational diabetes (by supplementation scheme/regimen).
Figuras y tablas -
Analysis 1.14

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 14 Gestational diabetes (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 15 Gestational diabetes (by skin pigmentation based on Fitzpatrick skin tone chart).
Figuras y tablas -
Analysis 1.15

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 15 Gestational diabetes (by skin pigmentation based on Fitzpatrick skin tone chart).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 16 Gestational diabetes (by latitude).
Figuras y tablas -
Analysis 1.16

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 16 Gestational diabetes (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 17 Gestational diabetes (by season at the start of supplementation).
Figuras y tablas -
Analysis 1.17

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 17 Gestational diabetes (by season at the start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 18 Preterm birth (less than 37 weeks' gestation) (by start of supplementation).
Figuras y tablas -
Analysis 1.18

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 18 Preterm birth (less than 37 weeks' gestation) (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 19 Preterm birth (less than 37 weeks' gestation) (by pre‐gestational BMI).
Figuras y tablas -
Analysis 1.19

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 19 Preterm birth (less than 37 weeks' gestation) (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 20 Preterm birth (less than 37 weeks' gestation) (by supplementation scheme/regimen).
Figuras y tablas -
Analysis 1.20

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 20 Preterm birth (less than 37 weeks' gestation) (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 21 Preterm birth (less than 37 weeks' gestation) (by skin pigmentation based on Fitzpatrick skin tone chart).
Figuras y tablas -
Analysis 1.21

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 21 Preterm birth (less than 37 weeks' gestation) (by skin pigmentation based on Fitzpatrick skin tone chart).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 22 Preterm birth (less than 37 weeks' gestation) (by latitude).
Figuras y tablas -
Analysis 1.22

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 22 Preterm birth (less than 37 weeks' gestation) (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 23 Preterm birth (less than 37 weeks' gestation) (by season at the start of supplementation).
Figuras y tablas -
Analysis 1.23

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 23 Preterm birth (less than 37 weeks' gestation) (by season at the start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 24 Low birthweight (less than 2500 g) (by start of supplementation).
Figuras y tablas -
Analysis 1.24

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 24 Low birthweight (less than 2500 g) (by start of supplementation).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 25 Low birthweight (less than 2500 g) (by pre‐gestational BMI).
Figuras y tablas -
Analysis 1.25

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 25 Low birthweight (less than 2500 g) (by pre‐gestational BMI).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 26 Low birthweight (less than 2500 g) (by supplementation scheme/regimen).
Figuras y tablas -
Analysis 1.26

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 26 Low birthweight (less than 2500 g) (by supplementation scheme/regimen).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 27 Low birthweight (less than 2500 g) (by skin pigmentation based on Fitzpatrick skin to.
Figuras y tablas -
Analysis 1.27

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 27 Low birthweight (less than 2500 g) (by skin pigmentation based on Fitzpatrick skin to.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 28 Low birthweight (less than 2500 g) (by latitude).
Figuras y tablas -
Analysis 1.28

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 28 Low birthweight (less than 2500 g) (by latitude).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 29 Low birthweight (less than 2500 g) (by season at the start of pregnancy).
Figuras y tablas -
Analysis 1.29

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 29 Low birthweight (less than 2500 g) (by season at the start of pregnancy).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 30 Caesarean section.
Figuras y tablas -
Analysis 1.30

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 30 Caesarean section.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 31 Gestational hypertension.
Figuras y tablas -
Analysis 1.31

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 31 Gestational hypertension.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 32 Maternal death (death while pregnant or within 42 days of termination of pregnancy) (ALL).
Figuras y tablas -
Analysis 1.32

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 32 Maternal death (death while pregnant or within 42 days of termination of pregnancy) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 33 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).
Figuras y tablas -
Analysis 1.33

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 33 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 34 Birth length (cm).
Figuras y tablas -
Analysis 1.34

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 34 Birth length (cm).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 35 Head circumference at birth (cm).
Figuras y tablas -
Analysis 1.35

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 35 Head circumference at birth (cm).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 36 Birthweight (g).
Figuras y tablas -
Analysis 1.36

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 36 Birthweight (g).

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 37 Stillbirth.
Figuras y tablas -
Analysis 1.37

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 37 Stillbirth.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 38 Neonatal death.
Figuras y tablas -
Analysis 1.38

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 38 Neonatal death.

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 39 Apgar score less than seven at five minutes.
Figuras y tablas -
Analysis 1.39

Comparison 1 Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals), Outcome 39 Apgar score less than seven at five minutes.

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 1 Pre‐eclampsia (ALL).
Figuras y tablas -
Analysis 2.1

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 1 Pre‐eclampsia (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 2 Gestational diabetes (ALL).
Figuras y tablas -
Analysis 2.2

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 2 Gestational diabetes (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).
Figuras y tablas -
Analysis 2.3

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 4 Low birthweight (less than 2500 g) (ALL).
Figuras y tablas -
Analysis 2.4

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 4 Low birthweight (less than 2500 g) (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 5 Caesarean section.
Figuras y tablas -
Analysis 2.5

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 5 Caesarean section.

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 6 Gestational hypertension.
Figuras y tablas -
Analysis 2.6

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 6 Gestational hypertension.

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 7 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).
Figuras y tablas -
Analysis 2.7

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 7 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 8 Birth length (cm).
Figuras y tablas -
Analysis 2.8

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 8 Birth length (cm).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 9 Head circumference at birth (cm).
Figuras y tablas -
Analysis 2.9

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 9 Head circumference at birth (cm).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 10 Birthweight (g).
Figuras y tablas -
Analysis 2.10

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 10 Birthweight (g).

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 11 Neonatal death.
Figuras y tablas -
Analysis 2.11

Comparison 2 Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals), Outcome 11 Neonatal death.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 1 Gestational diabetes (ALL).
Figuras y tablas -
Analysis 3.1

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 1 Gestational diabetes (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 2 Maternal adverse events.
Figuras y tablas -
Analysis 3.2

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 2 Maternal adverse events.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).
Figuras y tablas -
Analysis 3.3

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 3 Preterm birth (less than 37 weeks' gestation) (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 4 Low birthweight (less than 2500 g) (ALL).
Figuras y tablas -
Analysis 3.4

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 4 Low birthweight (less than 2500 g) (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 5 Caesarean section.
Figuras y tablas -
Analysis 3.5

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 5 Caesarean section.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 6 Gestational hypertension.
Figuras y tablas -
Analysis 3.6

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 6 Gestational hypertension.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 7 Maternal death (death while pregnant or within 42 days of termination of pregnancy).
Figuras y tablas -
Analysis 3.7

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 7 Maternal death (death while pregnant or within 42 days of termination of pregnancy).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 8 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).
Figuras y tablas -
Analysis 3.8

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 8 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 9 Birth length (cm).
Figuras y tablas -
Analysis 3.9

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 9 Birth length (cm).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 10 Head circumference at birth (cm).
Figuras y tablas -
Analysis 3.10

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 10 Head circumference at birth (cm).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 11 Birthweight (g).
Figuras y tablas -
Analysis 3.11

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 11 Birthweight (g).

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 12 Stillbirth.
Figuras y tablas -
Analysis 3.12

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 12 Stillbirth.

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 13 Neonatal death.
Figuras y tablas -
Analysis 3.13

Comparison 3 Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D), Outcome 13 Neonatal death.

Summary of findings for the main comparison. Vitamin D supplementation compared to placebo or no intervention for pregnancy and neonatal health outcomes

Vitamin D supplementation compared to placebo/control for pregnancy and neonatal health outcomes

Patient or population: pregnant women and their infants.
Setting: trials were carried from 1980s to 2015 in countries from Bangladesh, India, Iran, New Zealand and UK. Most trials were conducted outside the tropics and in different seasons.
Intervention: vitamin D supplementation.
Comparison: placebo or no intervention.

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with placebo/control

Risk with vitamin D supplementation

Pre‐eclampsia

Study population

RR 0.48 (0.30, 0.79)

499
(4 RCTs)

⊕⊕⊕⊝
MODERATE1

Included trials: Asemi 2013a; Naghshineh 2016; Sablok 2015; Sasan 2017

168 per 1000

79 per 1000
(49 to 131)

Gestational diabetes

Study population

RR 0.51
(0.27 to 0.97)

446
(4 RCTs)

⊕⊕⊕⊝
MODERATE 2

Included trials: Asemi 2013a; Sablok 2015; Shahgheibi 2016; Tehrani 2014

127 per 1000

65 per 1000
(34 to 123)

Maternal adverse events: severe postpartum haemorrhage

Study population

RR 0.68
(0.51 to 0.91)

1134
(1 RCT)

⊕⊕⊝⊝
LOW 3

Included trial: Harvey 2012

158 per 1000

106 per 1000
(79 to 142)

Maternal adverse event: nephritic syndrome

Study population

RR 0.17 (0.01 to 4.06)

135

(1 RCT)

⊕⊝⊝⊝
VERY LOW 4,5

Included trial: Yu 2008

22 per 1000

4 per 1000 (0 to 90)

Maternal adverse event: hypercalcaemia

Study population

Not estimable

1134
(1 RCT)

⊕⊕⊝⊝
LOW 3,6

Included trial: Harvey 2012

0 per 1000

0 per 1000

Preterm birth (less than 37 weeks' gestation)

Study population

RR 0.66 (0.34 to 1.30)

1640
(7 RCTs)

⊕⊕⊝⊝
LOW 7,8

Included trials: Asemi 2013a; Delvin 1986; Grant 2013; Harvey 2012; Mirghafourvand 2013; Roth 2010; Singh 2015

87 per 1000

57 per 1000
(29 to 113)

Low birthweight (less than 2500 g)

Study population

RR 0.55
(0.35 to 0.87)

697
(5 RCTs)

⊕⊕⊕⊝
MODERATE 9

Included trials: Brooke 1980; Bhutta 2011; Marya 1988; Roth 2010; Sablok 2015

136 per 1000

75 per 1000
(48 to 118)

*The risk in the intervention group (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; RCT: randomised controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 We downgraded (1) level for serious limitations in study design due to one trial being assessed as high risk of bias for several domains and two trials having unclear allocation concealment.

2 We downgraded (1) level for serious limitations in study design due to one trial being assessed as high risk of bias for several domains.

3 We downgraded (2) levels for very serious limitations in study design due to one study being assessed as high risk of other bias because we do not know the impact of the participants who were allowed to continue taking their own multivitamin with 400 IU/d of vitamin D as this was not recorded.

4 We downgraded (1) level for serious limitations in study design due to one study being assessed as high risk of bias for performance and detection bias.

5 We downgraded (2) levels for very serious limitations in imprecision as only one small study, with a small number of events and wide 95% confidence intervals (CI) contributed data.

6 We downgraded (1) level for serious limitations in imprecision due to a single study with zero events contributing data.

7 We downgraded (1) level for serious limitations in study design due to two studies being at unclear risk of selection bias and one study being at high risk of other bias.

8 We downgraded (1) level for serious limitations in imprecision as the 95% confidence interval (CI) was wide and crossed the line of no effect.

9 We downgraded (1) level for serious limitations in study design due to two studies being at unclear risk of selection bias, one study being at high risk of bias for allocation concealment, and three studies being at high risk of attrition bias.

Figuras y tablas -
Summary of findings for the main comparison. Vitamin D supplementation compared to placebo or no intervention for pregnancy and neonatal health outcomes
Summary of findings 2. Vitamin D + calcium supplementation compared to placebo or no intervention for pregnancy and neonatal health outcomes

Vitamin D + calcium supplementation compared to placebo/control for pregnancy and neonatal health outcomes

Patient or population: pregnant women and their infants..
Setting: trials were carried from 1980s to 2015 in countries from Iran, India, and Brazil. Only the study in Brazil was within the tropics. Most did not report the season in which it was carried out or it was mixed.
Intervention: vitamin D + calcium supplementation.
Comparison: placebo/control.

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with placebo/control

Risk with vitamin D + calcium supplementation

Pre‐eclampsia

Study population

RR 0.50
(0.32 to 0.78)

1174
(4 RCTs)

⊕⊕⊕⊝
MODERATE1

Included trials: Asemi 2012; Marya 1987; Samimi 2016; Taherian 2002

94 per 1000

47 per 1000
(30 to 73)

Gestational diabetes

Study population

RR 0.33
(0.01 to 7.84)

54
(1 RCT)

⊕⊝⊝⊝
VERY LOW2,3

Included trial: Asemi 2012

37 per 1000

12 per 1000
(0 to 290)

Maternal adverse events

No trials reported on this outcome

Preterm birth (less than 37 weeks' gestation)

Study population

RR 1.52
(1.01 to 2.28)

942
(5 RCTs)

⊕⊕⊝⊝
LOW4,5

Included trials: Asemi 2012; Diogenes 2013, Mirghafourvand 2013, Samimi 2016; Taherian 2002;

72 per 1000

110 per 1000
(73 to 165)

Low birthweight (less than 2500 g)

Study population

RR 0.68
(0.10 to 4.55)

110
(2 RCTs)

⊕⊝⊝⊝
VERY LOW6,7

Included trials: Diogenes 2013; Samimi 2016

59 per 1000

40 per 1000
(6 to 268)

*The risk in the intervention group (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; RCT: randomised controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 We downgraded (1) level for serious limitations in study design due to one study being at high risk of attrition and selection bias and three studies being at high risk of performance and detection bias.

2 We downgraded (1) level for serious limitations in study design due to one study being at high risk of performance and detection bias.

3 We downgraded (2) levels for very serious limitations in imprecision due to one small study, with a single event and wide 95% confidence intervals (CI) crossing the line of no effect contributing data.

4 We downgraded (1) level for serious limitations in study design due to three studies being at unclear risk of allocation concealment and three studies being at high risk of performance and detection bias.

5 We downgraded (1) level for serious limitations in imprecision due to wide 95% confidence intervals (CI).

6 We downgraded (1) level for serious limitations in study design due to one study being at unclear risk of allocation concealment and one study being at high risk of attrition bias.

7 We downgraded (2) levels for very serious limitations in imprecision due two small studies, with very few events and wide 95% confidence intervals (CI) crossing the line of no effect contributing data.

Figuras y tablas -
Summary of findings 2. Vitamin D + calcium supplementation compared to placebo or no intervention for pregnancy and neonatal health outcomes
Summary of findings 3. Vitamin D + calcium + other vitamins and minerals compared to calcium + other vitamins and minerals (but no vitamin D) for pregnancy and neonatal health outcomes

Vitamin D + calcium + other vitamins and minerals compared to calcium + other vitamins and minerals (but no vitamin D) for pregnancy and neonatal health outcomes

Patient or population: pregnant women and their infants..
Setting: the only study included in this comparison was conducted in Bangladesh, which is located outside the tropics and it was conducted in different seasons of the year.
Intervention: vitamin D + calcium + other vitamins and minerals.
Comparison: calcium + other vitamins and minerals (but no vitamin D).

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with calcium + other vitamins and minerals (but no vitamin D)

Risk with vitamin D + calcium + other vitamins and minerals

Pre‐eclampsia

Study population

(0 trials)

No trials reported on this outcome

see comment

see comment

Gestational diabetes

Study population

RR 0.42
(0.10 to 1.73)

1298
(1 RCT)

⊕⊝⊝⊝
VERY LOW 1,2

Included trial: Roth 2013

12 per 1000

5 per 1000
(1 to 20)

Maternal adverse event: hypercalcaemia

Study population

1298
(1 RCT)

⊕⊝⊝⊝
VERY LOW 2,3

Included trial: Roth 2013

23 per 1000

64 per 1000
(28 to 147)

Maternal adverse event: hypercalciuria

Study population

0.25 (0.02 to 3.97)

1298
(1 RCT)

⊕⊝⊝⊝
VERY LOW1,2

Included trial: Roth 2013

4 per 1000

1 per 1000 (0 to 15)

Preterm birth (less than 37 weeks' gestation)

Study population

RR 1.04
(0.68 to 1.59)

1298
(1 RCT)

⊕⊕⊝⊝
LOW 2,3

Included trial: Roth 2013

93 per 1000

96 per 1000
(63 to 147)

Low birthweight (less than 2500 g)

Study population

RR 1.12
(0.82 to 1.51)

1298
(1 RCT)

⊕⊕⊝⊝
LOW 2,3

Included trial: Roth 2013

162 per 1000

182 per 1000
(133 to 245)

*The risk in the intervention group (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; RCT: randomised controlled trial; RR: Risk ratio

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 We downgraded (2) levels for very serious limitations in imprecision with only one trial, with few events, and wide 95% confidence intervals (CI) crossing the line of no effect contributing data.

2 We downgraded (1) level for serious indirectness as there were multiple nutrient interventions in addition to vitamin D.

2 We downgraded (2) levels for very serious limitations in imprecision with only one trial, with zero events, and wide 95% confidence intervals (CI) crossing the line of no effect contributing data.

3 We downgraded (1) level for serious limitations in imprecision due to only one trial with wide 95% confidence intervals (CI) crossing the line of no effect contributing data.

Figuras y tablas -
Summary of findings 3. Vitamin D + calcium + other vitamins and minerals compared to calcium + other vitamins and minerals (but no vitamin D) for pregnancy and neonatal health outcomes
Comparison 1. Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pre‐eclampsia (ALL) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

2 Gestational diabetes (ALL) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

3 Maternal adverse events Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 Severe postpartum haemorrhage

1

1134

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.51, 0.91]

3.2 Nephritic syndrome

1

135

Risk Ratio (M‐H, Random, 95% CI)

0.17 [0.01, 4.06]

3.3 Hypercalcaemia

1

1134

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

4 Preterm birth (less than 37 weeks' gestation) (ALL) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

5 Low birthweight (less than 2500 g) (ALL) Show forest plot

5

697

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.35, 0.87]

6 Pre‐eclampsia (by start of supplementation) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

6.1 Less than 20 weeks of pregnancy

1

138

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.06, 1.37]

6.2 20 weeks of pregnancy or more

2

219

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.21, 1.24]

6.3 Unknown/unreported/mixed

1

142

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.98]

7 Pre‐eclampsia (by pre‐gestational BMI) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

7.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

7.2 Normal weight (18.5 to 24.9)

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.21, 1.33]

7.3 Overweight (25 or higher)

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

7.4 Unknown/unreported/mixed

2

280

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.86]

8 Pre‐eclampsia (by supplementation scheme/regimen) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

8.1 Single dose

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.21, 1.33]

8.2 Daily

2

192

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.08, 1.20]

8.3 Weekly/monthly

1

142

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.98]

9 Pre‐eclampsia (by skin pigmentation based on Fitzpatrick skin tone chart) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

9.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

9.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

9.3 Unknown/unreported/mixed

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

10 Pre‐eclampsia (by latitude) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

10.1 Between Tropics of Cancer and Capricorn

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

10.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

10.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

11 Pre‐eclampsia (by season at the start of pregnancy) Show forest plot

4

499

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.30, 0.79]

11.1 Summer

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

11.2 Fall

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

11.3 Winter

1

138

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.06, 1.37]

11.4 Mixed/unknown

2

307

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.31, 0.88]

12 Gestational diabetes (by start of supplementation) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

12.1 Less than 20 weeks of pregnancy

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.34, 2.28]

12.2 20 weeks of pregnancy or more

2

219

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.05, 3.45]

12.3 Unknown/unreported/mixed

1

87

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.13, 0.82]

13 Gestational diabetes (by pre‐gestational BMI) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

13.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

13.2 Normal weight (18.5 to 24.9)

2

305

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.33, 2.05]

13.3 Overweight (25 or higher)

2

141

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.13, 0.79]

13.4 Unknown/unreported/mixed

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

14 Gestational diabetes (by supplementation scheme/regimen) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

14.1 Single dose

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.03, 8.28]

14.2 Daily

2

141

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.13, 0.79]

14.3 Weekly/monthly

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.34, 2.28]

15 Gestational diabetes (by skin pigmentation based on Fitzpatrick skin tone chart) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

15.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

15.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

15.3 Unknown/unreported/mixed

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

16 Gestational diabetes (by latitude) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

16.1 Between Tropics of Cancer and Capricorn

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

16.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

16.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

17 Gestational diabetes (by season at the start of supplementation) Show forest plot

4

446

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.27, 0.97]

17.1 Summer

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

17.2 Fall

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

17.3 Winter

1

140

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.34, 2.28]

17.4 Mixed/unknown

2

252

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.14, 0.82]

18 Preterm birth (less than 37 weeks' gestation) (by start of supplementation) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

18.1 Less than 20 weeks of pregnancy

3

1149

Risk Ratio (M‐H, Random, 95% CI)

0.73 [0.26, 2.04]

18.2 20 weeks of pregnancy or more

4

491

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.13, 1.87]

18.3 Unknown/unreported/mixed

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

19 Preterm birth (less than 37 weeks' gestation) (by pre‐gestational BMI) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

19.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

19.2 Normal weight (18.5 to 24.9)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

19.3 Overweight (25 or higher)

2

138

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.15, 2.46]

19.4 Unknown/unreported/mixed

5

1502

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.27, 1.54]

20 Preterm birth (less than 37 weeks' gestation) (by supplementation scheme/regimen) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

20.1 Single dose

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

20.2 Daily

6

1495

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.23, 1.58]

20.3 Weekly/monthly

1

145

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.32, 1.54]

21 Preterm birth (less than 37 weeks' gestation) (by skin pigmentation based on Fitzpatrick skin tone chart) Show forest plot

9

1943

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.30, 0.91]

21.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

21.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

21.3 Unknown/unreported/mixed

9

1943

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.30, 0.91]

22 Preterm birth (less than 37 weeks' gestation) (by latitude) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

22.1 Between Tropics of Cancer and Capricorn

2

358

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.05, 1.34]

22.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

5

1282

Risk Ratio (M‐H, Random, 95% CI)

0.99 [0.59, 1.66]

22.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

23 Preterm birth (less than 37 weeks' gestation) (by season at the start of supplementation) Show forest plot

7

1640

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.34, 1.30]

23.1 Summer

2

199

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.34, 1.53]

23.2 Winter

1

34

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

23.3 Mixed/unknown

4

1407

Risk Ratio (M‐H, Random, 95% CI)

0.55 [0.19, 1.66]

24 Low birthweight (less than 2500 g) (by start of supplementation) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

24.1 Less than 20 weeks of pregnancy

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

24.2 20 weeks of pregnancy or more

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

24.3 Unknown/unreported/mixed

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25 Low birthweight (less than 2500 g) (by pre‐gestational BMI) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

25.1 Underweight (lower than 18.5)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25.2 Normal weight (18.5 to 24.9)

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.19, 0.98]

25.3 Overweight (25 or higher)

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

25.4 Unknown/unreported/mixed

2

326

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.14, 0.88]

26 Low birthweight (less than 2500 g) (by supplementation scheme/regimen) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

26.1 Single dose

2

365

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.16, 0.65]

26.2 Daily

1

126

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.23, 1.21]

26.3 Weekly/monthly

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

27 Low birthweight (less than 2500 g) (by skin pigmentation based on Fitzpatrick skin to Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

27.1 Three or less

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

27.2 Four or more

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

27.3 Unknown/unreported/mixed

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

28 Low birthweight (less than 2500 g) (by latitude) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

28.1 Between Tropics of Cancer and Capricorn

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

28.2 North of the Tropic of Cancer or South of the Tropic of Capricorn

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

28.3 Unknown/unreported

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29 Low birthweight (less than 2500 g) (by season at the start of pregnancy) Show forest plot

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

29.1 Summer

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29.2 Fall

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29.3 Winter

0

0

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

29.4 Mixed/unknown

3

491

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.24, 0.65]

30 Caesarean section Show forest plot

10

1104

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.80, 1.21]

31 Gestational hypertension Show forest plot

2

1130

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.41, 1.49]

32 Maternal death (death while pregnant or within 42 days of termination of pregnancy) (ALL) Show forest plot

1

180

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

33 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL) Show forest plot

14

2470

Mean Difference (IV, Random, 95% CI)

35.66 [24.19, 47.13]

34 Birth length (cm) Show forest plot

8

931

Mean Difference (IV, Random, 95% CI)

0.57 [0.19, 0.95]

35 Head circumference at birth (cm) Show forest plot

8

1841

Mean Difference (IV, Random, 95% CI)

0.11 [‐0.21, 0.44]

36 Birthweight (g) Show forest plot

17

2828

Mean Difference (IV, Random, 95% CI)

80.30 [‐14.40, 175.00]

37 Stillbirth Show forest plot

3

584

Risk Ratio (M‐H, Random, 95% CI)

0.35 [0.06, 1.98]

38 Neonatal death Show forest plot

2

326

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.04, 1.67]

39 Apgar score less than seven at five minutes Show forest plot

1

165

Risk Ratio (M‐H, Random, 95% CI)

0.53 [0.11, 2.53]

Figuras y tablas -
Comparison 1. Supplementation with vitamin D alone versus placebo or no intervention (no vitamins or minerals)
Comparison 2. Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pre‐eclampsia (ALL) Show forest plot

4

1174

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.32, 0.78]

2 Gestational diabetes (ALL) Show forest plot

1

54

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 7.84]

3 Preterm birth (less than 37 weeks' gestation) (ALL) Show forest plot

5

942

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.01, 2.28]

4 Low birthweight (less than 2500 g) (ALL) Show forest plot

2

110

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.10, 4.55]

5 Caesarean section Show forest plot

2

146

Risk Ratio (M‐H, Random, 95% CI)

1.16 [0.87, 1.54]

6 Gestational hypertension Show forest plot

1

59

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.06, 1.12]

7 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL) Show forest plot

1

60

Mean Difference (IV, Random, 95% CI)

12.5 [3.80, 21.20]

8 Birth length (cm) Show forest plot

3

194

Mean Difference (IV, Random, 95% CI)

‐0.07 [‐0.67, 0.52]

9 Head circumference at birth (cm) Show forest plot

3

198

Mean Difference (IV, Random, 95% CI)

‐0.03 [‐0.39, 0.33]

10 Birthweight (g) Show forest plot

3

194

Mean Difference (IV, Random, 95% CI)

42.39 [‐86.96, 171.74]

11 Neonatal death Show forest plot

1

660

Risk Ratio (M‐H, Random, 95% CI)

0.2 [0.01, 4.15]

Figuras y tablas -
Comparison 2. Supplementation with vitamin D + calcium versus placebo or no intervention (no vitamin or minerals)
Comparison 3. Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Gestational diabetes (ALL) Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.10, 1.73]

2 Maternal adverse events Show forest plot

1

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Hypercalcaemia

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

2.2 Hypercalciuria

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.02, 3.97]

3 Preterm birth (less than 37 weeks' gestation) (ALL) Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.68, 1.59]

4 Low birthweight (less than 2500 g) (ALL) Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.82, 1.51]

5 Caesarean section Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.95, 1.27]

6 Gestational hypertension Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.31, 2.79]

7 Maternal death (death while pregnant or within 42 days of termination of pregnancy) Show forest plot

1

1300

Risk Ratio (M‐H, Random, 95% CI)

0.25 [0.02, 3.98]

8 Maternal vitamin D concentration at term (25‐hydroxyvitamin D) (nmol/L) (ALL) Show forest plot

1

635

Mean Difference (IV, Random, 95% CI)

75.17 [71.97, 78.37]

9 Birth length (cm) Show forest plot

1

1297

Mean Difference (IV, Fixed, 95% CI)

0.0 [‐0.28, 0.28]

10 Head circumference at birth (cm) Show forest plot

1

1297

Mean Difference (IV, Random, 95% CI)

0.0 [‐0.17, 0.17]

11 Birthweight (g) Show forest plot

1

1297

Mean Difference (IV, Random, 95% CI)

‐7.0 [‐55.95, 41.95]

12 Stillbirth Show forest plot

1

1300

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.29, 1.46]

13 Neonatal death Show forest plot

1

1298

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.22, 2.14]

Figuras y tablas -
Comparison 3. Supplementation with vitamin D + calcium + other vitamins and minerals versus calcium + other vitamins and minerals (but no vitamin D)