Sie sind auf Seite 1von 10

Articles

Effect of metformin in addition to dietary and lifestyle


advice for pregnant women who are overweight or obese:
the GRoW randomised, double-blind, placebo-controlled
trial
Jodie M Dodd, Jennie Louise, Andrea R Deussen, Rosalie M Grivell, Gustaaf Dekker, Andrew J McPhee, William Hague

Summary
Background Maternal overweight and obesity are associated with well recognised pregnancy complications. Antenatal Lancet Diabetes Endocrinol 2018
dietary and lifestyle interventions have a modest effect on gestational weight gain without affecting pregnancy Published Online
outcomes. We aimed to assess the effects on maternal and infant outcomes of antenatal metformin given in addition December 4, 2018
http://dx.doi.org/10.1016/
to dietary and lifestyle advice among overweight and obese pregnant women.
S2213-8587(18)30310-3
See Online/Comment
Methods GRoW was a multicentre, randomised, double-blind, placebo-controlled trial in which pregnant women at http://dx.doi.org/10.1016/
10–20 weeks’ gestation with a BMI of 25 kg/m² or higher were recruited from three public maternity units in Adelaide, S2213-8587(18)30337-1
SA, Australia. Women were randomly assigned (1:1) via a computer-generated schedule to receive either metformin Discipline of Obstetrics &
(to a maximum dose of 2000 mg per day) or matching placebo. Participants, their antenatal care providers, and Gynaecology and Robinson
Research Institute
research staff (including outcome assessors) were masked to treatment allocation. All women received an antenatal
(Prof J M Dodd PhD, J Louise PhD,
dietary and lifestyle intervention. The primary outcome was the proportion of infants with birthweight greater than A R Deussen BHSc,
4000 g. Secondary outcomes included measures of maternal weight gain, maternal diet and physical activity, maternal Prof G Dekker MD,
pregnancy and birth outcomes, maternal quality of life and emotional wellbeing, and infant birth outcomes. Outcomes Prof W Hague MD) and School
of Public Health (J Louise),
were analysed on an intention-to-treat basis (including all randomly assigned women who did not withdraw consent
University of Adelaide,
to use their data, and who did not have a miscarriage or termination of pregnancy before 20 weeks’ gestation, or a Adelaide, SA, Australia;
stillbirth). The trial is registered with the Australian New Zealand Clinical Trials Registry, number Department of Perinatal
ACTRN12612001277831. Medicine (J M Dodd, W Hague)
and Department of Neonatal
Medicine (A J McPhee MD),
Findings Of 524 women who were randomly assigned between May, 28 2013 and April 26, 2016, 514 were included in Women’s and Children’s
outcome analyses (256 in the metformin group and 258 in the placebo group). Median gestational age at trial entry Hospital, North Adelaide, SA,
was 16·29 weeks (IQR 14·43–18·00) and median BMI was 32·32 kg/m² (28·90–37·10); 167 (32%) participants were Australia; Department of
Obstetrics and Gynaecology,
overweight and 347 (68%) were obese. There was no significant difference in the proportion of infants with birthweight
Flinders University, Bedford
greater than 4000 g (40 [16%] with metformin vs 37 [14%] with placebo; adjusted risk ratio [aRR] 0·97, 95% CI Park, SA, Australia
0·65 to 1·47; p=0·899). Women receiving metformin had lower average weekly gestational weight gain (adjusted (R M Grivell PhD); and Lyell
mean difference –0·08 kg, 95% CI –0·14 to –0·02; p=0·007) and were more likely to have gestational weight gain McEwin Hospital, Elizabeth
Vale, SA, Australia (G Dekker)
below recommendations (aRR 1·46, 95% CI 1·10 to 1·94; p=0·008). Total gestational weight gain, pregnancy and
Correspondence to:
birth outcomes, maternal diet and physical activity, and maternal quality of life and emotional wellbeing did not differ
Prof Jodie M Dodd, Discipline of
significantly between groups. Similar numbers of women in both treatment groups (76% [159/208] in the metformin Obstetrics & Gynaecology and
group and 73% [144/196] in the placebo group) reported side-effects including nausea, diarrhoea, and vomiting. Two Robinson Institute, University of
stillbirths (placebo group) and one neonatal death (metformin group) occurred; none of the perinatal deaths were Adelaide, and Women’s &
Children’s Hospital, North
determined to be attributable to participation in the trial.
Adelaide, SA 5006, Australia
jodie.dodd@adelaide.edu.au
Interpretation For pregnant women who are overweight or obese, metformin given in addition to dietary and lifestyle
advice initiated at 10–20 weeks’ gestation does not improve pregnancy and birth outcomes.

Funding Australian National Health and Medical Research Council.

Copyright © 2018 Elsevier Ltd. All rights reserved.

Introduction associated with infant birthweights greater than 4000 g,


The worldwide prevalence of overweight and obesity which in turn is recognised as an independent predictor
continues to grow.1 In high-income countries, roughly of childhood obesity, associated with a more than two-
50% of women enter pregnancy with a BMI greater than times increased risk.4
25 kg/m²,2 placing both the woman and her infant at risk There has been considerable research and clinical
of a range of well documented adverse pregnancy and interest in the provision of antenatal dietary and lifestyle
birth outcomes.3 Furthermore, high maternal BMI is interventions for pregnant women, particularly women

www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3 1


Articles

Research in context
Evidence before this study gestational weight gain or pre-eclampsia. In the MOP trial,
We searched the Cochrane Pregnancy and Childbirth Group’s investigators reported no difference in neonatal birthweight
Trials Register by contacting the trials search coordinator (April but decreased gestational weight gain and reduced incidence of
13, 2016). Briefly, the Cochrane Pregnancy and Childbirth pre-eclampsia in the group assigned to metformin.
Group’s Trials Register is maintained by the trials search Importantly, women participating in these previous
coordinator and contains trials identified from: monthly randomised trials were not provided with a dietary and lifestyle
searches of the Cochrane Central Register of Controlled Trials intervention. Additionally, these previous trials have only
(CENTRAL); weekly searches of MEDLINE (Ovid); weekly included women who were obese, whereas there might be
searches of Embase (Ovid); monthly searches of CINAHL clinical benefit in providing treatment for women who are
(EBSCO); manual searches of 30 journals and the proceedings of overweight as well as for those who are obese.
major conferences; weekly current awareness alerts for an
Added value of this study
additional 44 journals and monthly BioMed Central email
To our knowledge, this is the first trial to assess the effect of
alerts. Search terms from this comprehensive register include
metformin given in addition to an antenatal dietary and
“metformin”, “randomiz(s)ed trial”, and “pregnancy”. Antenatal
lifestyle intervention, and to include women who are
dietary and lifestyle interventions for pregnant women,
overweight as well as those who are obese. The use of
particularly those who are overweight or obese, have been
metformin in addition to an antenatal dietary and lifestyle
investigated as a strategy to limit gestational weight gain, but
intervention in women who are overweight or obese did not
have shown only a modest effect on weight gain in pregnancy
affect the proportion of infants with a birthweight greater than
and very little effect on clinical outcomes. Metformin has been
4000 g. There was no effect of metformin on total gestational
proposed as a possible drug treatment for use among obese
weight gain or on clinical pregnancy and birth outcomes.
pregnant women, although recent studies have reported
conflicting findings with regards to the effect on gestational Implications of all the available evidence
weight gain and some pregnancy outcomes. In the EMPOWaR The use of metformin in this clinical setting should not be
trial, the investigators reported no difference between advocated. Future research strategies should focus on
metformin and placebo groups in birthweight centile and no improving women’s health and diet to encourage weight loss
differences in any of the secondary outcomes including before conception.

who are overweight or obese, as a strategy to limit since the risk of adverse pregnancy outcomes increases
gestational weight gain and thereby improve preg­ with increasing maternal BMI,3 there might be clinical
nancy, birth, and infant outcomes. In an individual
­ benefit in providing treatment for women who are
participant data meta-analysis incorporating data from overweight as well as for those who are obese.
36 randomised trials and more than 12 500 pregnant The aim of the GRoW (metformin and dietary advice to
women who received an antenatal dietary or lifestyle improve insulin sensitivity and promote Gestational
intervention,2 a modest effect on gestational weight gain Restriction of Weight in pregnant women who are
(mean difference –0·7 kg) was identified, but with very overweight or obese) randomised trial was to assess the
little effect on clinical pregnancy outcomes, consistent effects on maternal and infant outcomes of antenatal
with other reports.5 These findings suggest the need for metformin in addition to dietary and lifestyle advice
assessment of additional strategies in this population. among overweight and obese pregnant women.
Metformin has been considered for use in pregnant
women who are overweight or obese, in view of the Methods
associations between high maternal BMI and gestational Study design and participants
diabetes, and a similar intrauterine milieu of insulin resis­ GRoW was a multicentre, randomised, double-blind,
tance, hyperglycaemia, hyperlipidaemia, and chro­ nic placebo-controlled trial in which women were recruited
inflammation.6 Metformin has insulin-sensitising from the three major public maternity hospitals in
properties, reducing hepatic glucose production and metropolitan Adelaide, SA, Australia (Women’s and
increasing peripheral glucose utilisation,7 and is used Children’s Hospital, Lyell McEwin Hospital, and Flinders
increasingly in the treatment of women with gestational Medical Centre). Eligible women had a live singleton
diabetes.8 pregnancy between 10 and 20 weeks’ gestation and
Previous trials9,10 assessing the use of metformin in were overweight (BMI 25·0–29·9 kg/m²) or obese
obese pregnant women have had conflicting findings with (BMI ≥30·0 kg/m²) at their first antenatal visit. Women
respect to effects on gestational weight gain and some with a multiple pregnancy, type 1 or 2 diabetes diagnosed
pregnancy outcomes. Importantly, women partici­pating prior to pregnancy, or with significant renal or hepatic
in these previous randomised trials were not provided impairment such that metformin therapy was
with a dietary and lifestyle intervention. Furthermore, contraindicated, were excluded.

2 www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3


Articles

The study protocol (appendix)11 was approved by the women were provided with written information, an See Online for appendix
Women’s and Children’s Health Network Human individual diet and physical activity plan, recipe book,
Research Ethics Committee, with local institutional and example menu plans. Participants were encouraged
approval at each site. Participating women provided to set achievable goals for dietary and exercise change
written informed consent. and were supported to make these lifestyle changes and
to self-monitor their progress using a SMART (Specific,
Randomisation and masking Measurable, Achievable, Relevant, and Timely) goals
All women presenting for antenatal care at the partici­ approach. These principles were reinforced at
pating centres had their height and weight measured subsequent face-to-face visits with the dietitian and
and BMI calculated at their first antenatal appointment. research assistant, and during the telephone contacts.
Eligible women were then counselled by a research All participants completed a food frequency question­
assistant. Those women who consented to participate naire, exercise diary, and quality-of-life and emotional
were randomly assigned (1:1) to metformin or placebo, wellbeing assessments at trial entry and at 28 and
by use of a central online randomisation service. The 36 weeks’ gestation. Consistent with state-wide clinical
computer-generated randomisation schedule util­ ised practice guidelines for South Australia, all women were
blocks of four and was prepared by an investigator not screened for gestational diabetes at about 28 weeks’
involved with recruitment or clinical care. Stratifi­cation gestation.15 During the course of the trial, diagnostic
occurred for parity (0 vs 1 or more), BMI at ante­natal criteria for gestational diabetes changed across the state
booking visit (25·0–29·9 kg/m² vs ≥30·0 kg/m²), and from a positive 75 g oral glucose tolerance test with a
collaborating centre. fasting blood glucose concentration of 5·5 mmol/L or
Metformin tablets (500 mg) and placebo tablets above, or a 2 h blood glucose concentration of
identical in appearance were packaged by an independent 7·8 mmol/L above, to a fasting blood glucose of
pharmaceutical packaging company (Pharmaceutical 5·1 mmol/L or above, a 1 h blood glucose concentration
Packaging Professionals, Port Melbourne, VIC, of 10·0 mmol/L or above, or a 2 h blood glucose con­
Australia) and coded in accordance with the centration of 8·5 mmol/L or above.15 Women diagnosed
randomisation schedule. Participants, their antenatal with gestational diabetes remained in the study and
care providers, and research staff (including outcome were offered treatment with further dietary modification
assessors) were masked to treatment allocation. and metformin or insulin added as required to maintain
appropriate glycaemic control.15 Women who required
Procedures metformin for the treatment of gestational diabetes
All participants received a 16-week supply of tablets were prescribed this medication in addition to their
(metformin 500 mg or identical placebo) and a further study medication. All other care during pregnancy and
12-week supply at 28 weeks’ gestation. Women com­ birth was done in accordance with local hospital
menced tablets from randomisation, starting with one practices.15
tablet per day for 1 week, and increasing to a maximum
of two tablets twice per day (maximum dose metformin Outcomes
2000 mg per day) over 4 weeks as tolerated, and then The primary outcome was the proportion of infants with
continuing until birth. At 36 weeks’ gestation, women birthweight greater than 4000 g.
completed a questionnaire to ascertain com­pliance with Several categories of secondary outcomes were also
medication and the occurrence of any side-effects. assessed. First, maternal weight gain outcomes assessed
All participating women received dietary and lifestyle were total gestational weight gain (defined as the
advice delivered over three face-to-face sessions (two difference between pregnancy weight obtained at
with a dietitian, shortly after trial entry and at 28 weeks, 36 weeks’ gestation or nearest to birth and early
and one with a research assistant at 36 weeks) and pregnancy weight); average weekly gestational weight
three telephone calls from the research assistant at 20, gain (calculated as total gestational weight gain divided
24, and 32 weeks. Dietary advice was consistent with by actual time in weeks between measurements); and
Australian standards.12,13 Women were advised to main­ gestational weight gain below, within, or above the US
tain a balance of carbohydrates, fat, and protein, while Institute of Medicine (IoM) recom­mendations according
specifically encouraging a reduction in the intake of to early pregnancy BMI (defined as 7·0 to 11·5 kg for
energy-dense and non-core foods high in refined women who were overweight and 5·0 to 9·0 kg for
carbohydrates and saturated fats, increased intake of women who were obese).16 Second, maternal diet and
fibre, and consumption of two servings of fruit, physical activity, as measured by questionnaires
five servings of vegetables, and three servings of dairy completed at trial entry and at 28 and 36 weeks’ gestation
each day.12,13 (using the Harvard Semi-Quantitative Food Frequency
Tailoring of the intervention was informed by stage Questionnaire17 and the Short Questionnaire to Assess
theories of health decision making.14 Initially, there was Health-Enhancing Physical Activity18). Third, maternal
a planning session with a research dietitian, in which pregnancy and birth outcomes assessed were

www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3 3


Articles

hypertension (defined as a systolic blood pressure Statistical analysis


≥140 mm Hg or diastolic blood pressure ≥90 mm Hg on The primary clinical outcome was the proportion of
two occasions 4 h or more apart) and pre-eclampsia infants born with birthweight greater than 4000 g, which
(based on criteria from the Australasian Society for the was estimated to occur in 15·5% of offspring of women
Study of Hypertension in Pregnancy19); clinical diagnosis eligible for the trial.12 To detect a change from
of gestational diabetes;15 antepartum haemorrhage 15·5% to 7·35% (ie, a reduction of about 47%; alpha
requiring admission; preterm prelabour ruptured 0·05, power 80%),12 and accounting for a 5% rate of
membranes; chorioamnionitis; induction of labour; attrition (based on our experience with the LIMIT
caesarean section; postpartum haemorrhage (defined as study12), we estimated that a sample size of 524 women
blood loss >600 mL); 3rd or 4th degree perineal trauma; would be required.
wound infection; endo­ metritis; thromboembolic All analyses followed a prespecified statistical analysis
disease; antenatal admission; antibiotic use during plan (appendix). Baseline characteristics of all randomly
delivery; and maternal death. Fourth, maternal quality of assigned women were examined descriptively. Primary
life and emotional wellbeing, as measured by question­ and secondary outcomes were analysed on an intention-
naires completed by participants at trial entry and at to-treat basis (including all randomly assigned women
28 and 36 weeks’ gestation relating to quality of life who did not withdraw consent to use their data, and who
(SF12 Health Survey Questionnaire20), anxiety (Short did not have a miscarriage or termination of pregnancy
Form Spielberger State Trait Inventory21), and depression before 20 weeks’ gestation, or a stillbirth), according to
(Edinburgh Postnatal Depression Scale22). the treatment allocated (metformin or placebo) at
Finally, the secondary outcomes also included random­isation. Continuous outcomes were analysed by
additional infant birth outcomes. These infant outcomes use of linear regression and binary outcomes were
were preterm birth before 37 weeks; perinatal mortality analysed by use of log binomial regression. Outcomes
(defined as an intrauterine fetal death after trial entry measured at multiple timepoints included a time-by-
and after 20 weeks’ gestation but prior to birth, or the treatment interaction term, with generalised estimating
death of a liveborn infant before hospital discharge equations used to account for correlation between
[excluding lethal congenital anomalies]); infant birth­ repeated measures. Results for continuous outcomes are
weight (inclu­ding weight <2500 g, and weight >4500 g); reported as adjusted differences in means (metformin
large for gestational age (defined as weight greater than minus placebo) and results for binary outcomes are
the 90th centile for gestational age and infant sex); small reported as adjusted risk ratios (aRRs; metformin vs
for gestational age (defined as weight less than the placebo). No adjustments were made for multiple
10th centile for gestational age and infant sex); hypo­ comparisons (secondary outcomes were considered
glycaemia requiring treatment; infant admis­sion to the exploratory rather than confirmatory).
neonatal intensive care unit or special care baby unit; As specified in the statistical analysis plan (appendix),
hyperbilirubinaemia requiring photo­ therapy; nerve the primary analyses were adjusted analyses based on
palsy; fracture; birth trauma; shoulder dystocia; and imputed data. Unadjusted analyses, and analyses on
new­ born anthropometric measures (including birth unimputed data (not presented), were also done as
length; biceps, triceps, abdominal, supra­ iliac, sub­ secondary sensitivity analyses. Adjusted models included
scapular, and thigh skinfold thicknesses; head, chest, the stratification variables (centre, maternal BMI, and
abdominal, and right upper arm circum­ferences; and parity) as well as smoking status, socioeconomic status
z scores for birthweight, birth length, and circumference (as defined by the Australian Bureau of Statistics’ 2011
measures) taken within the first few days (median 2·0 Socio-Economic Index for Areas—Index of Relative
days [IQR 1·0–4·0]) after birth in accordance with a Socio-Economic Disadvantage quintile24), and maternal
specifically developed protocol.23 age at trial entry as covariates.
The study protocol (appendix) also specified a health There were no missing values for the primary outcome
economic analysis of health-care costs to determine the or for other infant birthweight outcomes; many other
cost of the intervention per live birth. Another secondary outcomes (including infant anthropometry and infant
outcome, fetal growth and adiposity measures deter­ and maternal delivery data) had less than 1% missing
mined by ultrasound at 28 and 36 weeks’ gestation, data; however, infant skinfold thickness measurement
will be reported elsewhere. Infant follow-up at 6 and and other maternal antenatal measures had between
18 months, including weight, length, and circumference 20% and 46% missing data across the study cohort
and skinfold measurements will also be reported (appendix). Multiple imputation by the fully conditional
elsewhere. specification (chained equations) method was used to
Safety was assessed with a questionnaire completed at create 100 complete datasets for analysis.25 The
36 weeks asking about compliance with study medication imputation model included all outcomes, all stratification
and side-effects experienced. Additionally, a multi­discip­ variables, mater­ nal baseline height, weight, and
lin­ary adverse events committee masked to treat­ment gestational age, and maternal weight at 36 weeks’
allocation reviewed all serious adverse events. gestation. Estimates were derived in the standard manner

4 www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3


Articles

by combining the estimates from each imputation using onset fetal growth restriction and pre-eclampsia). There
Rubin’s rules.25 As there were no missing values for the was one neonatal death in the metformin group following
primary outcome, no missing-not-at-random sensitivity extremely preterm birth (before 22 weeks). None of the
analyses were done. perinatal deaths were determined to be attributable to
Secondary analyses for the primary outcome and participation in the trial.
secondary infant anthropometric outcomes were The baseline characteristics of women at trial entry
prespecified to test for evidence of modification of the were similar in the two treatment groups (table 1). Across
effect of treatment by maternal BMI category (overweight the whole study cohort, median gestational age at trial
vs obese). entry was 16·29 weeks (IQR 14·43–18·00) and median
Statistical analyses were done with SAS version 9.4 or BMI was 32·32 kg/m² (28·90–37·10); 167 (32%)
Stata version 14. participants were overweight and 347 (68%) were obese.
We also did a post-hoc meta-analysis incorporating the Adequate data were available for all 514 infants for the
findings of the GRoW trial into our previously reported primary outcome of birthweight greater than 4000 g.
Cochrane systematic review,26 which was done in 404 (79%) of 514 participants provided data for
accordance with standard Cochrane methods, to assess experience of adverse effect, with no differences
the impact of metformin on gestational weight gain, identified between treatment groups (table 2). Of 354
birth­weight, and risk of pre-eclampsia. Briefly, (69%) participants who provided data for medication
randomised and quasi-randomised trials assessing compliance, 296 (84%) took the maximum of four tablets
metformin use (compared with placebo or no metformin) per day (appendix).
in pregnant women who were overweight or obese, There was no significant difference in the proportion
defined as an early pregnancy or pre-pregnancy BMI of of infants with birthweight greater than 4000 g between
25·0 kg/m² or higher, were identified from a search of the metformin and placebo groups (40 [16%] of 256 in the
the Cochrane Pregnancy and Childbirth’s Trials Register.
Two review authors independently assessed risk of bias 3546 eligible women approached to participate
for each study, with statistical analyses done with Review
Manager 5. A fixed-effect meta-analysis was used to
combine data where trials were judged to be sufficiently 3022 declined to participate
259 did not want to
similar, and a random-effects meta-analysis was used to participate in research
produce an overall summary if substantial statistical 481 did not want to take
study medication
heterogeneity was detected. For the present report, we 547 too busy or unable
incorporated the data from the present trial into the to attend for study
visits
existing systematic review and meta-analysis using 531 other reasons*
Review Manager 5. For dichotomous data (risk of pre- 1204 were not interested,
eclampsia), results are presented as summary risk ratio did not give a reason,
or could not be
with 95% CIs; for continuous data (gestational weight contacted
gain and birthweight), results are presented as mean
difference with 95% CIs.
524 consented and randomly assigned
The GRoW trial is registered with the Australian New
Zealand Clinical Trials Registry, number
ACTRN12612001277831.
261 assigned to metformin 263 assigned to placebo
Role of the funding source
The funder had no role in the study design, data
0 withdrew consent 1 withdrew consent
collection, analysis, interpretation, or writing of the 5 had miscarriage before 2 had miscarriage before
report. The corresponding author had full access to the 20 weeks or termination of 20 weeks or termination of
pregnancy pregnancy
data and final responsibility for the decision to submit 0 had stillbirth after 20 weeks 2 had stillbirth after 20 weeks
for publication. 256 liveborn infants 258 liveborn infants
1 neonatal death 0 neonatal death
0 maternal death 0 maternal death
Results
Between May 28, 2013 and April 26, 2016, 3546 eligible
women were approached to participate; 524 women were 256 woman and infant pairs 258 woman and infant pairs
included in the analysis included in the analysis
randomly assigned, 261 to metformin and 263 to placebo
(figure). 514 women and infant pairs were included in
the analyses (256 in the metformin group and 258 in the Figure: Trial profile
*Other reasons includes any reason not covered by the other categories,
placebo group). No maternal deaths occurred. There including partner or family against participation, did not think they were
were two stillborn infants, both in the placebo group (one overweight, were already participating in another research project, and did not
secondary to acute chorioamnionitis and one due to early think that they needed lifestyle intervention.

www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3 5


Articles

Metformin (n=256) Placebo (n=258) Metformin Placebo


(n=208*) (n=196†)
Maternal age (years) 29·87 (5·54) 30·17 (5·37)
Study centre Any reported side-effect 159 (76%) 144 (73%)

Women’s and Children’s Hospital 110 (43%) 112 (43%) Nausea 74 (36%) 65 (33%)

Lyell McEwin Hospital 117 (46%) 118 (46%) Vomiting 49 (24%) 46 (23%)

Flinders Medical Centre 29 (11%) 28 (11%) Abdominal pain 50 (24%) 48 (24%)

Gestational age at entry (weeks) 16·29 (14·43–18·00) 16·29 (14·57–18·14) Diarrhoea 62 (30%) 44 (22%)

BMI (kg/m )2
32·50 (28·71–37·54) 32·05 (29·10–36·80) Fatigue 92 (44%) 98 (50%)

BMI category Weakness 33 (16%) 30 (15%)

25·0–29·9 kg/m 2
83 (32%) 84 (33%) Skin rash 14 (7%) 19 (10%)

≥30·0 kg/m 2
173 (68%) 174 (67%) Loss of appetite 47 (23%) 40 (20%)

Public patient 252 (98%) 254 (98%) Data are the number (%) of women who reported at least one specified adverse
event during the study period up to completion of the questionnaire at 36 weeks.
Weight (kg) 92·89 (19·76) 91·80 (19·79)
Data are for all randomly assigned women who did not withdraw consent to use
Height (cm) 165·27 (6·76) 164·87 (6·79) their data, and who did not have a miscarriage or termination of pregnancy
Race before 20 weeks’ gestation, or stillbirth. *Excludes 48 (19%) of 256 participants
White 210 (82%) 221 (86%) who did not complete the questionnaire assessing compliance with medication
and experience of adverse effects. †Excludes 62 (24%) of 258 participants who did
East Asian 5 (2%) 7 (3%) not complete the questionnaire assessing compliance with medication and
Indian 12 (5%) 6 (2%) experience of adverse effects.
Other 24 (9%) 19 (7%)
Table 2: Adverse effects
Unknown 5 (2%) 5 (2%)
Smoker 24 (9%) 43 (17%)
Nulliparous 88 (34%) 92 (36%) (table 4), including diagnosis of gestational diabetes,
Previous preterm birth 13 (5%) 13 (5%) irrespective of the diagnostic criteria used (appendix).
Previous pre-eclampsia 13 (5%) 10 (4%) Furthermore, following diagnosis of gestational diabetes,
Previous stillbirth or neonatal death 1 (<1%) 6 (2%)
there were no significant differences between the
Previous caesarean section 48 (19%) 67 (26%)
treatment groups with respect to use of either metformin
Family history of diabetes 83 (32%) 78 (30%)
or insulin therapy (appendix).
Family history of hypertension 105 (41%) 104 (40%)
Overall the risks of maternal labour complications did
not differ significantly between the treatment groups
Family history of heart disease 58 (23%) 61 (24%)
(table 5). Although there was a significant reduction in
Index of socioeconomic disadvantage*
risk of caesarean birth among women in the metformin
Quintile 1 (most disadvantaged) 76 (30%) 95 (37%)
group compared with those in the placebo group, this
Quintile 2 78 (30%) 74 (29%)
finding was partly the result of a difference in the number
Quintile 3 31 (12%) 30 (12%)
of women undergoing an elective repeat caesarean birth
Quintile 4 52 (20%) 43 (17%)
(30 women in the metformin group vs 44 women in the
Quintile 5 (least disadvantaged) 19 (7%) 16 (6%)
placebo group). Self-reported maternal quality of life and
Data are mean (SD), median (IQR), or n (%). Data are for all randomly assigned women who did not withdraw consent emotional wellbeing did not differ between groups
to use their data, and who did not have a miscarriage or termination of pregnancy before 20 weeks’ gestation, or a
stillbirth. *Socioeconomic index of relative social disadvantage (as defined by the Australian Bureau of Statistics’
(appendix).
2011 Socio-Economic Index for Areas—Index of Relative Socio-Economic Disadvantage quintile24). Mean gestational age at birth was similar in the
two treat­ment groups, as was mean infant birthweight
Table 1: Baseline characteristics
(table 3). There were no significant differences between
the two groups with respect to other infant outcomes
metformin group vs 37 (14%) of 258 in the placebo group; (table 3) or newborn anthropometric measures
aRR 0·97, 95% CI 0·65 to 1·47; p=0·899; table 3). (appendix) apart from infant abdominal circumference at
Compared with the placebo group, women in the birth, which was 0·5 cm smaller in the metformin group
metformin group had lower average weekly gestational (p=0·049; appendix), although this could be a chance
weight gain and were more likely to have weight gain finding and the small difference is of questionable
below the IoM recommendations; total gestational clinical significance.
weight gain was not significantly different between the A prespecified secondary analysis identified some
treatment groups (table 4). Women in both groups were evidence of effect modification by maternal BMI category,
successful in improving the quality of their dietary with the use of metformin associated with a significant
intake, but dietary patterns and physical activity did not reduction in the proportion of infants with birthweight
differ between the two treatment groups at trial entry or greater 4000 g and reductions in total gestational weight
over the course of pregnancy (appendix). There were no gain and weight gain above IoM recommendations
significant differences between the two treatment groups among overweight women but not among obese women
with respect to maternal risk of pregnancy complications (appendix).

6 www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3


Articles

Metformin Placebo (n=258) Unadjusted Unadjusted Adjusted treatment Adjusted


(n=256) treatment effect p value effect (95% CI) p value
(95% CI)
Birthweight >4000 g 40 (15·63%) 37 (14·34%) 1·09 (0·72 to 1·65) 0·684 0·97 (0·65 to 1·47) 0·899
Birthweight (g) 3487·80 3471·80 16·00 0·739 –13·01 0·785
(531·37) (556·67) (–78·10 to 110·09) (–106·45 to 80·44)
Gestational age at birth (weeks) 39·12 (1·64) 38·93 (1·85) 0·19 (–0·11 to 0·49) 0·222 0·10 (–0·20 to 0·40) 0·532
Large for gestational age 50 (19·53%) 56 (21·71%) 0·90 (0·64 to 1·26) 0·543 0·87 (0·62 to 1·23) 0·428
Small for gestational age 10 (3·91%) 8 (3·10%) 1·26 (0·51 to 3·14) 0·620 1·37 (0·55 to 3·45) 0·502
Birthweight >4500 g 5 (1·95%) 7 (2·71%) 0·72 (0·23 to 2·24) 0·570 0·64 (0·21 to 1·97) 0·433
Birthweight <2500 g 4 (1·56%) 11 (4·26%) 0·37 (0·12 to 1·14) 0·082 0·42 (0·13 to 1·31) 0·135
Stillbirth* 0 2 (0·77%) ·· ·· ·· 0·499
Neonatal death* 1 (0·39%) 0 ·· ·· ·· 0·496
Admission to NICU or SCBU 32 (12·55%) 43 (16·67%) 0·75 (0·49 to 1·15) 0·189 0·78 (0·51 to 1·20) 0·260
Hypoglycaemia requiring treatment 19 (7·45%) 24 (9·30%) 0·80 (0·45 to 1·43) 0·451 0·87 (0·49 to 1·55) 0·632
Hyperbilirubinaemia requiring 16 (6·27%) 20 (7·75%) 0·81 (0·43 to 1·53) 0·513 0·76 (0·41 to 1·40) 0·379
phototherapy†
Nerve palsy‡ 0 0 ·· ·· ·· ··
Fracture* 1 (0·39%) 0 ·· ·· ·· 0·497
Birth trauma* 3 (1·18%) 0 ·· ·· ·· 0·122
Shoulder dystocia 7 (2·73%) 7 (2·71%) 1·01 (0·36 to 2·83) 0·988 0·96 (0·34 to 2·66) 0·935
Results presented are based on imputed data, unless otherwise specified. Birthweight and gestational age at birth are continuous outcomes, for which data are mean (SD)
and treatment effect estimates are differences in means (metformin minus placebo); all other outcomes are binary, for which data are n (%; with numbers calculated from
proportions averaged across imputations) and treatment effect estimates are risk ratios (metformin vs placebo). Data are for all randomly assigned women who did not
withdraw consent to use their data, and who did not have a miscarriage or termination of pregnancy before 20 weeks’ gestation, or stillbirth. Adjusted analyses included
stratification variables (centre, parity, BMI category), maternal smoking status, age at consent, and socioeconomic status quintile as covariates. NICU=neonatal intensive care
unit. SCBU=special care baby unit. *Insufficient events for analysis or imputation; p value from Fisher’s exact test on un-imputed data. †Poisson model with robust variance
estimation used for adjusted analysis because binomial model did not converge. ‡No events in either group so no modelling was possible.

Table 3: Infant outcomes

Metformin Placebo Unadjusted treatment Unadjusted Adjusted treatment Adjusted


(n=256) (n=258) effect (95% CI) p value effect (95% CI) p value
Total gestational weight gain (kg) 7·48 (6·95) 8·72 (6·91) –1·23 (–2·45 to –0·01) 0·048 –1·18 (–2·37 to 0·01) 0·053
Average weekly gestational weight 0·38 (0·34) 0·47 (0·35) –0·08 (–0·14 to –0·02) 0·006 –0·08 (–0·14 to –0·02) 0·007
gain (kg)
Gestational weight gain below IoM 100 (39·20%) 70 (27·00%) 1·45 (1·09 to 1·93) 0·010 1·46 (1·10 to 1·94) 0·008
recommendations16*
Gestational weight gain above IoM 83 (32·27%) 101 (39·10%) 0·83 (0·64 to 1·07) 0·145 0·84 (0·65 to 1·09) 0·185
recommendations16*
Preterm birth before 37 weeks’ gestation 13 (5·08%) 18 (6·98%) 0·73 (0·36 to 1·45) 0·368 0·79 (0·40 to 1·58) 0·504
Hypertension 19 (7·42%) 16 (6·23%) 1·19 (0·63 to 2·26) 0·592 1·25 (0·66 to 2·35) 0·496
Pre-eclampsia 13 (5·08%) 11 (4·28%) 1·19 (0·54 to 2·60) 0·667 1·22 (0·56 to 2·66) 0·618
Gestational diabetes* 72 (27·93%) 62 (23·95%) 1·17 (0·85 to 1·60) 0·335 1·19 (0·88 to 1·62) 0·253
Antenatal admission 48 (18·75%) 62 (24·03%) 0·78 (0·56 to 1·09) 0·146 0·82 (0·59 to 1·15) 0·254
Antepartum haemorrhage requiring 1 (0·39%) 7 (2·69%) ·· ·· ·· 0·068
admission†
Preterm prelabour ruptured membranes† 5 (1·95%) 3 (1·15%) ·· ·· ·· 0·502
Results presented are based on imputed data, unless otherwise specified. Total gestational weight gain and average weekly gestational weight gain are continuous outcomes,
for which data are mean (SD) and treatment effect estimates are differences in means (metformin minus placebo); all other outcomes are binary, for which data are n (%;
with numbers calculated from proportions averaged across imputations) and treatment effect estimates are risk ratios (metformin vs placebo). Data are for all randomly
assigned women who did not withdraw consent to use their data, and who did not have a miscarriage or termination of pregnancy before 20 weeks’ gestation, or stillbirth.
Adjusted analyses included stratification variables (centre, parity, BMI category), maternal smoking status, age at consent, and socioeconomic status quintile as covariates.
IoM=US Institute of Medicine. *Poisson model with robust variance estimation used for adjusted analysis because binomial model did not converge. †Insufficient events for
analysis or imputation; p value from Fisher’s exact test on un-imputed data.

Table 4: Maternal antepartum outcomes

www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3 7


Articles

Metformin Placebo (n=258) Unadjusted risk Unadjusted Adjusted risk ratio Adjusted
(n=256) ratio (95% CI) p value (95% CI) p value
Chorioamnionitis* 3 (2·22%) 5 (3·40%) ·· ·· ·· 0·725
Induction of labour 104 (40·63%) 87 (33·72%) 1·20 (0·96 to 1·51) 0·107 1·19 (0·95 to 1·49) 0·139
Antibiotic use during delivery† 135 (52·73%) 147 (56·98%) 0·93 (0·79 to 1·08) 0·334 0·95 (0·81 to 1·11) 0·493
Caesarean section (all)† 87 (33·98%) 111 (43·02%) 0·79 (0·63 to 0·99) 0·037 0·80 (0·64 to 1·00) 0·049
Elective caesarean section‡ 44 (17·19%) 54 (20·93%) 0·82 (0·57 to 1·18) 0·281 0·72 (0·51 to 1·00) 0·053
Emergency caesarean section 43 (16·80%) 57 (22·09%) 0·76 (0·53 to 1·09) 0·131 0·79 (0·56 to 1·11) 0·168
Postpartum haemorrhage 66 (25·79%) 58 (22·55%) 1·14 (0·84 to 1·56) 0·393 1·15 (0·85 to 1·57) 0·367
>600 mL
3rd or 4th degree perineal 6 (2·34%) 3 (1·15%) ·· ·· ·· 0·336
trauma*
Wound infection* 3 (1·17%) 3 (1·16%) ·· ·· ·· >0·999
Endometritis* 3 (1·17%) 3 (1·16%) ·· ·· ·· >0·999
Thromboembolic disease* 1 (0·39%) 0 ·· ·· ·· 0·497
Results presented are based on imputed data, unless otherwise specified. Data are n (%; with numbers calculated from proportions averaged across imputations), unless
otherwise indicated. Data are for all randomly assigned women who did not withdraw consent to use their data, and who did not have a miscarriage or termination of
pregnancy before 20 weeks’ gestation, or stillbirth. Adjusted analyses included stratification variables (centre, parity, BMI category), maternal smoking status, age at consent,
and socioeconomic status quintile as covariates. *Insufficient events for analysis or imputation; p value from Fisher’s exact test on un-imputed data. †Poisson model with
robust variance estimation used for adjusted analysis because binomial model did not converge. ‡Includes 30 women in the metformin group and 44 women in the placebo
group who had an elective repeat caesrean section.

Table 5: Maternal labour and birth outcomes

The planned health economic cost analysis was not participants, central randomisation, and appropriate
done because the intervention had no significant impact masking of participants, clinicians, and outcome asses­
on clinical outcomes in the overall study population. sors. Furthermore, we pre­specified outcomes of clinical
In a post-hoc meta-analysis incorporating the findings relevance, followed a prespecified analysis plan, and had
of the GRoW trial into the previously reported Cochrane complete follow-up data for the primary outcome.
systematic review,26 metformin was identified to have GRoW is the first trial to assess the effect of metformin
only a modest impact on gestational weight gain among given in addition to an antenatal dietary and lifestyle
pregnant women who were overweight or obese (mean intervention. Following dietary intervention over the
difference –2·27 kg, 95% CI –4·45 to –0·08 [ four studies, course of pregnancy, women across both treatment
1278 women]). There was no evidence of an impact on groups were successful in modifying their dietary intake,
birthweight (mean difference 11·22 g, –52·87 to 75·31 specifically reducing their overall energy intake and
[three studies; 1348 infants]) or the risk of pre-eclampsia improving their healthy eating index score as an
(risk ratio 0·82, 95% CI 0·25 to 2·68 [three studies; indicator of diet quality. These findings are consistent
1355 women]). with those reported previously from the LIMIT trial,12,27
sub­stantiating the reproducibility of the intervention
Discussion among overweight and obese pregnant women in
In the GRoW randomised trial, use of metformin in producing dietary change. Together, these findings also
addition to dietary and lifestyle advice did not affect the suggest that many of the complex determinants of
proportion of infants with a birthweight greater than gestational weight gain might not be readily modifiable
4000 g compared with placebo and dietary and lifestyle by changes to dietary intake and physical activity.
advice. Although there were no significant differences in Our trial population was predominantly white and
total gestational weight gain, women who received from areas with high levels of socioeconomic dis­
metformin were more likely to have weight gain below advantage. Additionally, 85% of eligible women declined
IoM recommendations, and had lower average weekly to participate—among the most common reasons cited
gestational weight gain. However, these differences in were time constraints and lack of willingness to take
gestational weight gain were not accompanied by medication during pregnancy. These factors might limit
significant effects on maternal and infant pregnancy or the generalisability of our findings to some extent.
birth outcomes. However, such rates of uptake of trial participation (15%)
Our trial has several strengths. The trial is the first of are within the range described by similar trials in the
its kind to include both overweight and obese women, scientific literature, which have varied from 13% to 47%.9,10
which is important in view of the potential for benefit Our measure of compliance suggests that 84% of
from interventions at different levels of BMI above the respondents took the maximum of four tablets per day,
normal range. We used robust methods, including with no differences identified between the treatment
prospectively measured height, weight, and BMI in all groups. These figures are difficult to directly compare

8 www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3


Articles

with other studies, which have used varying measures to –4·45 to –0·08 [ four studies; 1278 women]). Although
assess treatment compliance,9,10 although our measure is there are inherent variations in the design of the
similar to the overall compliance of 80% reported by randomised trials done to date,9,10,29 including the BMI of
Syngelaki and colleagues10 and 66% reported by Chiswick women recruited (overweight and obese in the current
and colleagues.9 trial vs obese only9,10,29), the dose of metformin assessed
Although metformin is increasingly used in pregnancy (1000 mg29 up to 3000 mg10 per day), and the inclusion of
for the treatment of gestational diabetes, and there is a dietary and lifestyle intervention for all women in the
evidence to support its safety,8 longer-term childhood current trial, these differences seem insufficient to
follow-up of the offspring of women who participated in explain the overall observations of a limited reduction in
this trial will be important. The primary findings from gestational weight gain and lack of effect on clinical
the MIG trial did not identify any differences in infant outcomes.
birthweights for women with gestational diabetes treated Such a limited effect of metformin is, however,
with insulin or metformin.8 However, follow-up of child consistent more broadly with the evidence relating to
participants at age 7–9 years suggested that children ex­ prenatal dietary and lifestyle interventions2 for women
posed in utero to metformin, as compared with insulin, who are overweight or obese, and raises important
had higher body­weight and BMI, with some suggestion questions concerning their modest at best reduction in
of increased adipose tissue deposition as measured by gestational weight gain, and little evidence of effect on
dual-energy x-ray absorptiometry.28 clinical outcomes. Importantly, the IoM recom­ men­
During the course of the GRoW trial there was a dations16 for gestational weight gain are based on
change in the diagnostic criteria for gestational diabetes extensive observational evidence identifying associations
across South Australia.15 However, the criteria were between gestational weight gain and pregnancy out­
applied equally across the two treatment groups, and we comes, with an underlying assumption that the observed
demonstrated no significant differences in diagnosis or associations are causal in nature. Furthermore, it has
requirements for metformin or insulin between the two been assumed that gestational weight gain can be
treatment groups. The change in criteria might have modified either through use of metformin, or via
resulted in an increase in the number of women being changes to diet and lifestyle during pregnancy, and that
diagnosed with gestational diabetes; however, discussion weight gain within the recommended range leads to
of the relative merits and evidence base to support a optimal maternal and infant outcomes.
range of different testing approaches and criteria is These prevailing assumptions need to be questioned
beyond the scope of this report. in view of the mounting available clinical evidence from
Our findings do not suggest that metformin impacts randomised trials. For pregnant women who are
clinical pregnancy and birth outcomes—this conclusion over­­weight or obese, intervention during pregnancy
is broadly consistent with the existing scientific (whether through metformin, dietary or lifestyle
literature.9,10,29 Syngelaki and colleagues reported a signi­ modification, or a combination of the two) might be too
fi­cant reduction in the risk of pre-eclampsia with met­ little, too late, emphasising the need to target women,
formin use.10 However, this effect was not evident when particularly those who are overweight or obese, before
the current trial data were incorporated into the available conception to improve their diet and lifestyle and to
published meta-analysis,26 raising the possibility that this encourage weight loss.30 Notwithstanding the logistical
was a spurious finding from a secondary outcome in implications of inter­vention before conception, there is
Syngelaki and colleagues’ trial. currently little evidence to support improved pregnancy
We did not identify a significant reduction in total outcomes following preconception weight loss, and
gestational weight gain, although there was some robust evalu­ ation of such an approach is urgently
evidence of effect modification, with a possible benefit needed.
from metformin in reducing the risk of infant birth­ In conclusion, our findings suggest that the use of
weight greater than 4000 g and reducing total gestational metformin in addition to a dietary and lifestyle inter­
weight gain among overweight women, but not among vention among pregnant women who are overweight or
obese women. Although this secondary analysis was obese was associated with some evidence of reduced
prespecified, these findings should be interpreted with gestational weight gain measures, but did not affect
caution, and warrant confirmation by others. clinical pregnancy and birth outcomes. Use of metformin
Our overall findings in relation to gestational weight in this clinical setting should not be advocated on the
gain are consistent with those of Chiswick and colleagues basis of these data.
(EMPOWaR trial),9 but not with those of Syngelaki and Contributors
colleagues (MOP trial)10 and Abd El Fattah.29 All authors were involved in the development and design of the trial, the
Incorporation of our data into the Cochrane systematic conduct of the trial, drafting of the report, and revision for intellectual
content. JL and JMD did the statistical analyses.
review and meta-analysis26 suggests that, overall,
metformin has a very modest effect on gestational Declaration of interests
We declare no competing interests.
weight gain (mean difference –2·27 kg, 95% CI

www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3 9


Articles

Data sharing statement 14 Bennett P, Murphy S. Psychology and health promotion.


Additional trial-related documents and requests for de-identified data Buckingham: Open University Press, 1997.
(aggregate or individual participant level) may be requested by written 15 Department of Health, Government of South Australia. South
application to the corresponding author and will be considered on an Australian perinatal practice guidelines: diabetes mellitus and
individual basis by the trial steering committee. gestational diabetes. 2015. https://www.sahealth.sa.gov.au/wps/
Acknowledgments wcm/connect/146238004ee2144cb404bdd150ce4f37/Diabetes+Mellit
us+and+Gestational+Diabetes_July2015.pdf?MOD=AJPERES&CAC
This study was funded by an Australian National Health and Medical
HEID=ROOTWORKSPACE-146238004ee2144cb404bdd150ce4f37-
Research Council (NHMRC) project grant (ID 1043181). JMD is funded mhbYUex (accessed Nov 22, 2018).
by an NHMRC practitioner fellowship (ID 1078980). We thank the
16 US Institute of Medicine. Weight gain during pregnancy:
women and their infants who participated in the trial. We also thank reexamining the guidelines. Institute of Medicine: Washington, DC,
John Svigos and Michael Stark (both at Women’s & Children’s Hospital, 2009.
North Adelaide, SA, Australia and University of Adelaide, Adelaide, SA, 17 Willett WC, Reynolds RD, Cottrell-Hoehner S, Sampson L,
Australia) for their role in the trial adverse events committee. Additional Browne ML. Validation of a semi-quantitative food frequency
acknowledgments are in appendix. questionnaire: comparison with a 1-year diet record. J Am Diet Assoc
References 1987; 87: 43–47.
1 Ng M, Fleming T, Robinson M, et al. Global, regional, and national 18 Wendel-Vos GC, Schuit AJ, Saris WH, Kromhout D. Reproducibility
prevalence of overweight and obesity in children and adults during and relative validity of the short questionnaire to assess
1980–2013: a systematic analysis for the Global Burden of Disease health-enhancing physical activity. J Clin Epidemiol 2003;
Study 2013. Lancet 2014; 384: 766–81. 56: 1163–69.
2 The International Weight Management in Pregnancy (i-WIP) 19 Brown MA, Hague WM, Higgins J, et al. The detection,
Collaborative Group. Effect of diet and physical activity based investigation and management of hypertension in pregnancy: full
interventions in pregnancy on gestational weight gain and consensus statement. Aust N Z J Obstet Gynaecol 2000; 40: 139–55.
pregnancy outcomes: meta-analysis of individual participant data 20 Ware JE, Sherbourne CD. The MOS 36 item short form health
from randomised trials. BMJ 2017; 358: j3119. survey (SF36). 1. Conceptual framework and item selection.
3 Dodd JM, Grivell RM, Nguyen A-M, Chan A, Robinson JS. Maternal Med Care 1992; 30: 473–83.
and perinatal health outcomes by body mass index category. 21 Marteau TM, Bekker H. The development of a six-item short-form
Aust N Z J Obstet Gynaecol 2011; 51: 136–40. of the state scale of the Spielberger State-Trait Anxiety Inventory
4 Woo Baidal JA, Locks LM, Cheng ER, Blake-Lamb TL, Perkins ME, (STAI). Br J Clin Psychol 1992; 31: 301–06.
Taveras EM. Risk factors for childhood obesity in the first 1000 days: 22 Cox JL, Holden JM, Sagovsky R. Detection of postnatal depression.
a systematic review. Am J Prev Med 2016; 50: 761–79. Development of the 10-item Edinburgh Postnatal Depression Scale.
5 Peaceman AM, Clifton RG, Phelan S, et al. Lifestyle interventions Br J Psychiatry 1987; 154: 782–86.
limit gestational weight gain in women with overweight or obesity: 23 Dodd JM, Deussen AR, Mohamad I, et al. The effect of antenatal
LIFE-Moms prospective meta-analysis. Obesity (Silver Spring) 2018; lifestyle advice for women who are overweight or obese on
26: 1396–404. secondary measures of neonatal body composition: the LIMIT
6 Catalano PM, Hauguel-De Mouzon S. Is it time to revisit the randomised trial. BJOG 2016; 123: 244–53.
Pedersen hypothesis in the face of the obesity epidemic? 24 Australian Bureau of Statistics. Socio-economic Indexes for Areas
Am J Obstet Gynecol 2011; 204: 479–87. (SEIFA) 2011. Australian Bureau of Statistics, 2013. http://www.abs.
7 Wiernsperger NF, Bailey CJ. The antihyperglycaemic effect of gov.au/AUSSTATS/abs@.nsf/DetailsPage/2033.0.55.0012011
metformin: therapeutic and cellular mechanisms. Drugs 1999; (accessed Nov 2, 2017).
58 (suppl 1): 31–39. 25 White I, Royston P, Wood A. Multiple imputation using chained
8 Rowan JA, Hague WM, Gao W, Battin MR, Moore MP, for the MIG equations: issues and guidance for practice. Stat Med 2011;
Trial Investigators. Metformin versus insulin for the treatment of 30: 377–99.
gestational diabetes. N Engl J Med 2008; 358: 2003–15. 26 Dodd JM, Grivell RM, Deussen AR, Hague W. Metformin for
9 Chiswick C, Reynolds RM, Denison F, et al. Effect of metformin on women who are overweight or obese during pregnancy for
maternal and fetal outcomes in obese pregnant women improving maternal and infant outcomes.
(EMPOWaR): a randomised, double-blind, placebo-controlled trial. Cochrane Database Syst Rev 2018; 7: CD010564.
Lancet Diabetes Endocrinol 2015; 3: 778–86. 27 Dodd JM, Cramp CS, Sui Z, et al. The effects of antenatal dietary
10 Syngelaki A, Nicolaides KH, Balani J, et al. Metformin versus and lifestyle advice for women who are overweight or obese on
placebo in obese pregnant women without diabetes mellitus. maternal diet and physical activity: the LIMIT randomised trial.
N Engl J Med 2016; 374: 434–43. BMC Med 2014; 12: 161.
11 Dodd JM, Grivell RM, Deussen AR, Dekker G, Louise J, Hague W. 28 Rowan JA, Rush EC, Plank LD, et al. Metformin in gestational
Metformin and dietary advice to improve insulin sensitivity and diabetes: the offspring follow-up (MiG TOFU): body composition
promote gestational restriction of weight among pregnant women and metabolic outcomes at 7–9 years of age.
who are overweight or obese: the GRoW randomised trial. BMJ Open Diabetes Res Care 2018; 6: e000456.
BMC Pregnancy Childbirth 2016; 16: 359. 29 Abd El Fattah E. Can metformin limit weight gain in the obese with
12 Dodd JM, Turnbull DA, McPhee AJ, et al. Antenatal lifestyle advice pregnancy? Int J Reprod Contracept Obstet Gynecol 2016; 5: 818–25.
for women who are overweight or obese: the LIMIT randomised 30 Hanson MA, Barker M, Dodd JM, et al. Interventions to prevent
trial. BMJ 2014; 348: g1285. maternal obesity before conception, during pregnancy, and post
13 Australian guide to healthy eating. https://www.eatforhealth.gov.au/ partum. Lancet Diabetes Endocrinol 2017; 5: 65–76.
guidelines/australian-guide-healthy-eating (accessed Nov 22, 2018).

10 www.thelancet.com/diabetes-endocrinology Published online December 4, 2018 http://dx.doi.org/10.1016/S2213-8587(18)30310-3

Das könnte Ihnen auch gefallen