You are on page 1of 31

Endocrine Reviews, 2022, XX, 1–31

https://doi.org/10.1210/endrev/bnac003
Advance access publication 18 January 2022
Review

A Clinical Update on Gestational Diabetes Mellitus


Arianne Sweeting,1,2, Jencia Wong,1,2 Helen R. Murphy,3,4,5 and Glynis P. Ross,1,2,
1
Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


2
Faculty of Medicine and Health, University of Sydney, Sydney, Australia
3
Diabetes in Pregnancy Team, Cambridge University Hospitals, Cambridge, UK
4
Norwich Medical School, Bob Champion Research and Education Building, University of East Anglia, Norwich, UK
5
Division of Women’s Health, Kings College London, London, UK
Correspondence: Arianne Sweeting, MBBS Hons, BSc, GradDip HL, FRACP, PhD, Department of Endocrinology, Royal Prince Alfred Hospital, Sydney,
Australia; Faculty of Medicine and Health, Level 2 Charles Perkins Centre D17, University of Sydney, Sydney, NSW 2006, Australia. Email: arianne.sweeting@
sydney.edu.au.

Abstract
Gestational diabetes mellitus (GDM) traditionally refers to abnormal glucose tolerance with onset or first recognition during pregnancy. GDM has
long been associated with obstetric and neonatal complications primarily relating to higher infant birthweight and is increasingly recognized as a
risk factor for future maternal and offspring cardiometabolic disease. The prevalence of GDM continues to rise internationally due to epidemiological
factors including the increase in background rates of obesity in women of reproductive age and rising maternal age and the implementation of
the revised International Association of the Diabetes and Pregnancy Study Groups’ criteria and diagnostic procedures for GDM. The current lack of
international consensus for the diagnosis of GDM reflects its complex historical evolution and pragmatic antenatal resource considerations given
GDM is now 1 of the most common complications of pregnancy. Regardless, the contemporary clinical approach to GDM should be informed not
only by its short-term complications but also by its longer term prognosis. Recent data demonstrate the effect of early in utero exposure to ma-
ternal hyperglycemia, with evidence for fetal overgrowth present prior to the traditional diagnosis of GDM from 24 weeks’ gestation, as well as the
durable adverse impact of maternal hyperglycemia on child and adolescent metabolism. The major contribution of GDM to the global epidemic of
intergenerational cardiometabolic disease highlights the importance of identifying GDM as an early risk factor for type 2 diabetes and cardiovascular
disease, broadening the prevailing clinical approach to address longer term maternal and offspring complications following a diagnosis of GDM.

Graphical Abstract

Key Words:  gestational diabetes mellitus, diagnosis, pathophysiology, genetics, outcomes, management, precision medicine, biomarkers, diabetes prevention,
COVID

Received: 7 July 2021. Editorial Decision: 4 January 2022. Corrected and Typeset: 17 February 2022
© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs licence (https://
creativecommons.org/licenses/by-nc-nd/4.0/), which permits non-commercial reproduction and distribution of the work, in any medium, provided the
original work is not altered or transformed in any way, and that the work is properly cited. For commercial re-use, please contact journals.permissions@
oup.com
2 Endocrine Reviews, 2022, Vol. XX, No. XX

Pregnancy Study Groups (IADPSG) diagnostic criteria in


Essential Points 2013 (11).
1. Gestational diabetes mellitus (GDM) is 1 of the most Since 1973, the screening approach to GDM frequently
common medical complications of pregnancy and is adopted a 2-step procedure with the 50-g 1-hour glucose
increasing in prevalence globally. challenge test (GCT) followed by the 100-g 3-hour OGTT
2. GDM is associated with obstetric and neonatal com- if the GCT was positive. This was based on data from
plications primarily due to increased birthweight O’Sullivan et  al, which showed that a 2-step diagnostic ap-
and is a major risk factor for future type 2 diabetes, proach to GDM using the GCT as the initial screening test
obesity, and cardiovascular disease in mother and and a glucose threshold of 7.9 mmol/L (143 mg/dL) was 79%
child. sensitive and 87% specific for diagnosing GDM on the 100-g

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


3. Detecting GDM is important because perinatal com- 3-h OGTT in a cohort of 752 women (12). The rationale for
plications and stillbirth risk are greatly reduced by this approach was the efficient identification of women most
treatment. at risk of GDM.
4. A precision medicine approach to GDM which In 1979, the US National Diabetes Data Group (NDDG)
recognizes severity and onset of maternal hypergly- published conversions of the original O’Sullivan and Mahan
cemia as well as genetic and physiologic subtypes 100-g 3-hour OGTT diagnostic criteria for GDM, reflecting
of GDM may address the current diagnostic con- the transition from venous whole blood glucose to plasma
troversy via accurate risk stratification and indi- blood glucose analysis (13). These revised criteria were sub-
vidualized treatment strategies, leading to improved sequently adopted by the American Diabetes Association
clinical care models and outcomes. (ADA) and internationally (9,14,15). In 1982, Carpenter and
5. The traditional focus on normalization of obstetric Coustan recommended lowering of the NDDG diagnostic
and neonatal outcomes achieved via short-term ante- criteria, reflecting newer preanalytical enzymatic methods
natal maternal glucose management should now shift that were more specific for plasma glucose (7,16). They also
to early postnatal prevention strategies to decrease the advised lowering the GCT glucose threshold to 7.5 mmol/L
progression from GDM to type 2 diabetes and ad- (135 mg/dL) based on their study of 381 women who under-
dress longer term maternal and offspring metabolic went the 100-g 3-h OGTT after screening positive on the
risk given the global epidemic of diabetes, obesity, and GCT, whereby a GCT glucose threshold ≤ 7.5  mmol/L
cardiovascular disease. (135  mg/dL) strongly correlated with a normal OGTT
(17). However, in the absence of clear evidence supporting
a specific glucose threshold for the GCT, the ADA and
Diabetes in pregnancy was first described in 1824 by the American College of Obstetricians and Gynecologists
Bennewitz in Germany (1), with subsequent case series in the (ACOG) continued to recommend a screen positive GCT
United Kingdom and United States reporting high perinatal glucose threshold from 7.2 to 7.8 mmol/L (130-140 mg/dL)
mortality rates in women with diabetes in pregnancy (2-4). for GDM (18,19).
In 1909, Williams reported arguably the first diagnostic cri- The ADA did however recommend the modified Carpenter
teria for diabetes in pregnancy in the United States, proposing and Coustan diagnostic glucose thresholds for GDM from
physiological and pathophysiological thresholds for “tran- 2000 (20), supported by the findings of the Toronto Tri-
sient glycosuria in pregnancy” (5). Hospital Gestational Diabetes Project (21,22). These data
In 1964, O’Sullivan and Mahan defined specific diag- demonstrated a positive correlation between increasing ma-
nostic criteria for gestational diabetes mellitus (GDM) in ternal hyperglycemia even below the NDDG diagnostic
the United States derived from the 100-g 3-hour oral glu- criteria for GDM and risk of obstetric and neonatal com-
cose tolerance test (OGTT) undertaken in the second and plications including preeclampsia, cesarean section, and
third trimester of pregnancy in 752 women (6). GDM was macrosomia (neonatal birthweight > 4000 g) (21,22). In add-
defined as ≥2 venous whole blood glucose values greater ition, several large cohort studies showed that women diag-
than 2 SD above the mean glucose values for pregnancy nosed (but not treated) with GDM based on the Carpenter
in their initial cohort. These glucose thresholds were pri- and Coustan criteria were at increased risk of perinatal com-
marily chosen because the resulting GDM prevalence of plications including hypertensive disorders of pregnancy, in-
2% corresponded to the background population preva- creased birthweight, macrosomia, neonatal hypoglycemia,
lence of diabetes, while the requirement of ≥2 elevated glu- hyperbilirubinemia, and shoulder dystocia, compared to
cose values sought to minimize the risk of preanalytical women diagnosed and treated as GDM by NDDG diagnostic
error (7). These thresholds were validated by their identifi- criteria (16,23-25). From 2003 the ADA additionally endorsed
cation of subsequent diabetes up to 8 years postpartum in the 1-step 75-g 2-hour OGTT for the diagnosis of GDM de-
an additional cohort of 1013 women. Increased perinatal rived from the modified Carpenter and Coustan fasting, 1-
mortality was also observed in women with ≥2 glucose and 2-hour glucose thresholds for the 100-g 3-hour OGTT,
values exceeding the proposed diagnostic criteria (6). In particularly for women at high-risk (26). This approach was
1965, the World Health Organization (WHO) concurrently deemed more cost-effective, albeit less validated, than the
recommended that GDM be diagnosed by either a 50- or 100-g 3-hour OGTT. The use of the modified Carpenter and
100-g OGTT using the 2-hour postload glucose value, but Coustan thresholds was associated with an almost 50% in-
the threshold used was the same as for diagnosing diabetes crease in prevalence of GDM (16,23).
in the nonpregnant population (8). The WHO continued The evolution of diagnostic criteria for GDM illustrates the
to diagnose GDM based on glucose thresholds for diabetes historic lack of consensus for the diagnosis of GDM, with
in the nonpregnant population (9,10) until its endorse- the presence or absence of disease varying dependent on ex-
ment of the International Association of the Diabetes and pert consensus. The underlying rationale for the diagnosis of
Endocrine Reviews, 2022, Vol. XX, No. XX 3

GDM also shifted over time toward identifying perinatal risk ACOG continue to recommend a 2-step testing approach,
rather than future maternal diabetes risk. with the initial screening GCT for all women and those who
screen positive proceeding to the diagnostic 100-g 3-hour
OGTT (19,37). This approach is also endorsed by ADA (18).
Current GDM Diagnostic Criteria However, the ACOG’s 2018 guidelines now acknowledge that
The seminal Hyperglycemia and Adverse Pregnancy individual practices and institutions may instead choose to use
Outcomes (HAPO) study sought to provide an evidence the IADPSG’s 1-step testing approach and diagnostic criteria
base to guide risk in GDM, and its results were published if appropriate for their population (19). The UK National
in 2008 (27). This large, international, prospective, observa- Institute for Health and Care Excellence (NICE) guidelines
tional study evaluated the relationship between glucose levels advise a selective screening approach, whereby women with

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


on the 75-g 2-hour OGTT performed at 24 to 32 weeks’ risk factors for GDM are recommended to undergo a diag-
gestation (mean 27.8 weeks’ gestation) in over 25 000 preg- nostic 75-g 2-hour OGTT at 26 to 28 weeks’ gestation, with
nant women with the following primary perinatal outcomes: diagnostic (fasting or 2-hour) glucose thresholds higher than
birthweight > 90th percentile for gestational age, primary the IADPSG diagnostic criteria for GDM (38). Several other
cesarean section delivery, neonatal hypoglycemia, and cord European bodies also currently recommend selective risk
blood serum C-peptide > 90th centile. Secondary outcomes factor-based screening, with only women fulfilling specific
were preeclampsia, preterm delivery (defined as delivery be- high-risk criteria proceeding to a diagnostic OGTT, even if
fore 37 weeks’ gestation), shoulder dystocia or birth injury, the IADPSG diagnostic criteria for GDM are applied (39,40).
hyperbilirubinemia, and neonatal intensive care admission. The revised IADPSG diagnostic criteria and testing approach
The results showed a continuous positive linear relation- to GDM in comparison to other international organizations
ship between maternal fasting; 1- and 2-hour plasma glucose are summarized in Table 1.
levels obtained on the OGTT, below those that were diag- It is important to consider the increase in GDM preva-
nostic of diabetes outside pregnancy; and risk of primary out- lence associated with the IADPSG diagnostic criteria in the
comes (27). Notably, there were no specific glucose thresholds context of the rising background rates of impaired glucose
at which obstetric and neonatal complications significantly tolerance, type 2 diabetes, and obesity among young adults
increased. and women of reproductive age (46,47). For example, al-
Based on these findings and supported by trials [the most 18% of HAPO study participants would have met the
Australian Carbohydrate Intolerance Study in Pregnant IADPSG diagnostic thresholds for GDM. By comparison, the
Women (ACHOIS) and the Maternal-Fetal Medicine Units rate of prediabetes in US adults aged between 20 and 44 years
Network (MFMU) trial] showing benefit of treatment is >29% (48,49).
of more severe and “mild” degrees of maternal hyper- Studies in Indian, Israeli, and US cohorts have suggested
glycemia, respectively (28,29), the IADPSG revised its that the IADPSG testing approach and intervention for
diagnostic criteria for GDM. Despite the lack of a clear GDM is cost-effective based on a combination of delaying
diagnostic glucose threshold in HAPO, the consensus of future type 2 diabetes and preventing perinatal complications
the IADPSG was to define diagnostic thresholds for the (50-53). For example, a US study found that the IADPSG
fasting, 1- and 2-hour glucose values for the 75-g 2-hour diagnostic criteria would be cost-effective if associated inter-
OGTT based on the average glucose values at which the vention decreased the absolute incidence of preeclampsia by
odds of the primary outcomes were 1.75 times the odds of >0.55% and cesarean delivery by >2.7% (53). In contrast,
these outcomes occurring at the mean glucose levels for the UK health economic data show that routinely identifying
HAPO cohort (30). The IADPSG consensus was also that GDM is not cost-effective based on perinatal outcomes (54)
only 1 elevated glucose level for the OGTT was required and that the universal WHO (IADPSG) testing approach is
for GDM diagnosis, as each glucose threshold represented less cost-effective than the NICE selective screening approach
broadly comparable level of risk. Thus, the main purpose (55).
of the diagnostic criteria for GDM post-HAPO was to de-
fine the level of risk associated with increased perinatal
complications. Contemporary Clinical Evidence Following the
Post-HAPO, there exist several different screening and Revised IADPSG GDM Diagnostic Criteria
testing approaches for the diagnosis of GDM internationally. The lack of randomized controlled trials (RCTs) evaluating
The IADPSG and WHO recommend universal testing of all outcomes in women diagnosed with GDM based on the
pregnant women between 24 to 28 weeks’ gestation with the IADPSG criteria and the clinical relevance of treating the re-
75-g 2-hour OGTT (11,30). These revised recommendations sulting milder degrees of hyperglycemia remain controversial
were largely endorsed by several organizations including the (56). Several retrospective studies have shown that women
ADA (18), Endocrine Society (31), International Federation diagnosed with GDM by the IADPSG criteria but who were
of Gynecology and Obstetrics (32), Australasian Diabetes previously classified as having normal glucose tolerance were
in Pregnancy Association (33), Japan Diabetes Society (34), still at increased risk for obstetric and neonatal complications,
Ministry of Health of China (35), and the European Board of including gestational hypertension, preeclampsia, cesarean de-
Gynecology and Obstetrics (36). livery, macrosomia, large-for-gestational-age (LGA), shoulder
The National Institutes of Health did not endorse the dystocia, and neonatal intensive care admission, compared
IADPSG recommendations, citing the expected increase in to women with normal glucose tolerance (57-59). For ex-
prevalence of GDM, cost, and intervention in the context ample, a 2015 retrospective study in Taiwan comparing preg-
of a lack of evidence for an associated improvement in peri- nancy outcomes in women diagnosed and treated for GDM
natal outcomes (37). The National Institutes of Health and using the 2-step (GCT followed by the 100-g 3-hour OGTT)
4 Endocrine Reviews, 2022, Vol. XX, No. XX

approach compared to the IADPSG 1-step approach found systematic review and meta-analysis of 25 studies (n = 4466
that the latter was associated with a reduction in gestational women) showed that even 1 abnormal value on the diagnostic
weight gain (GWG), birthweight, macrosomia, and LGA (60). 3-hour 100-g OGTT is associated with an increased risk of
Another retrospective study in the United Kingdom reported perinatal complications compared to women with a normal
that women who were diagnosed with GDM based on modi- GCT, and this risk was similar to that of women actually
fied IADPSG diagnostic glucose thresholds but who screened diagnosed with GDM (70).
negative for GDM on 2015 NICE diagnostic criteria had a The degree of benefit of treating women with GDM defined
higher risk of LGA, cesarean delivery, and polyhydramnios by the IADPSG diagnostic criteria is yet to be determined. The
(61). Other retrospective studies have also demonstrated potential benefit is inferred from the treatment of maternal
higher birthweight, birthweight z-score, ponderal index, and hyperglycemia described in the ACHOIS and MFMU inter-

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


increased rates of LGA and cesarean delivery in untreated vention trials (28,29), whereby maternal glucose levels over-
women diagnosed with GDM based on the IADPSG criteria, lapped with the thresholds recommended by the IADPSG. It
compared to women with normal glucose tolerance (62,63). is worth noting that there are differences in these 2 trials with
The recent randomized ScreenR2GDM trial compared regards to the diagnostic criteria used to define GDM and
1-step screening (75-g 2-hour OGTT) with 2-step screening (2 cohort characteristics (eg, women were excluded from the
GCT thresholds ≥7.2 mmol/L and ≥7.8 mmol/L used, followed MFMU trial if they had an abnormal glucose screening test
by the 100-g 3-hour OGTT) in 23 792 pregnant women in the prior to 24 weeks’ gestation or previous GDM), and thus the
United States (64). Despite doubling the diagnosis of GDM generalizability of these findings in women diagnosed with
with the 1-step approach (16.5% vs 8.5%), there were no GDM based on the IADPSG criteria remains contentious.
differences in pregnancy complications including LGA [rela-
tive risk (RR) 0.95; 97.5% CI 0.87-1.05], perinatal composite
outcome (RR 1.04; 97.5% CI 0.88-1.23), gestational hyper- Current Classification of Hyperglycemia in
tension or preeclampsia (RR 1.00; 97.5% CI 0.93-1.08), and Pregnancy and GDM
primary cesarean section (RR 0.98; 97.5% CI 0.93-1.02) be- The WHO first defined GDM in 1965 as “hyperglycemia of
tween the different screening approaches. These findings have diabetic levels occurring during pregnancy” (8). Thus, histor-
not resolved the diagnostic debate for GDM, with some ar- ically, the term “GDM” encompassed the entire spectrum of
guing that the 1-step approach therefore demonstrates insuffi- maternal hyperglycemia in pregnancy, from pregestational
cient perinatal benefit for the associated increased healthcare diabetes to hyperglycemia first detected in pregnancy. In
costs (65), while others have identified potential limitations 1979, the NDDG defined GDM as “glucose intolerance that
in study methodology (7,47,65,66). Despite randomization to has its onset or recognition during pregnancy” (13). This was
either testing strategy, the pragmatic trial design allowed clin- subsequently modified in 1985 at the Second International
icians to select a preferred strategy. Consequently, one third Workshop-Conference on Gestational Diabetes as “carbohy-
of women randomized to the 1-step approach did not adhere drate intolerance resulting in hyperglycemia of variable se-
to the assigned screening and were tested via the 2-step ap- verity with onset or first recognition during pregnancy” and
proach, compared to only 8% of women randomized to the remained the most widely used definition of GDM until re-
2-step approach. Although the study attempted to adjust for cently (71).
this difference using inverse probability weighting, residual Contemporary nomenclature and diagnostic criteria now
provider bias cannot be excluded (47). Given this was a popu- more clearly differentiate between women with pregestational
lation level analysis of GDM screening, GDM (treatment) diabetes and those with hyperglycemia first detected in preg-
status differed only for the 8% of women not diagnosed with nancy (30) (Fig. 1). Pregestational diabetes includes type 1
GDM based on the 2-step approach who may have otherwise diabetes, type 2 diabetes, and other types of diabetes such
been diagnosed with GDM based on the 1-step approach. as cystic fibrosis-related diabetes, steroid/medication-induced
Whether these women had potentially worse outcomes that diabetes, and monogenic diabetes.
may have been mitigated by treatment cannot be determined Hyperglycemia in pregnancy is now subclassified by the
by this study. However, given the rates of pharmacotherapy IADPSG into 2 separate categories, namely “overt diabetes
were similar between the 1- and 2-step cohorts at 43% and mellitus during pregnancy” (overt diabetes) and GDM (30).
46%, respectively (64), this strategy detected women with Similarly, the WHO has a binary definition of hyperglycemia
essentially an equivalent risk of hyperglycemia warranting in pregnancy but has replaced the term “overt diabetes”
pharmacotherapy (47). This observation is consistent with with “diabetes mellitus in pregnancy” (DIP) (11). The ra-
other studies in UK cohorts comparing the IADPSG testing tionale for the IADPSG recommendation for early testing in
approach to the less sensitive NICE and Canadian criteria, high-risk women is to diagnose DIP early in pregnancy. This
whereby women demonstrated insulin resistance and re- is because DIP, diagnosed based on nonpregnant diabetes
quired pharmacotherapy for control of hyperglycemia even glucose thresholds, recognizes the increasing prevalence of
at the most sensitive thresholds of the IADPSG diagnostic cri- undiagnosed preexisting diabetes in women of childbearing
teria (67). age as well as the greater risk associated with this degree
More generally, the GCT fails to detect approximately 20% of hyperglycemia (72-74). For example, a recent study in
to 25% of women with GDM, particularly those diagnosed almost 5000 women in France found that DIP was associ-
with GDM based on an elevated fasting glucose (68). The ated with a 3.5-fold greater risk of hypertensive disorders
frequency of GDM diagnosed by the OGTT fasting glucose in pregnancy compared to women with normal glucose tol-
threshold in the HAPO study ranged from 24% to 26% in erance, while early‐diagnosed DIP was associated with an
Thailand and Hong Kong to >70% in the United States (69). increased risk of congenital malformation (7.7% vs 1.0%
This highlights the variability and thus limitations of post- for women with normal glucose tolerance), suggesting that
glucose load screening based on ethnicity. Moreover, a recent early hyperglycemia in pregnancy may sometimes be present
Endocrine Reviews, 2022, Vol. XX, No. XX 5

Table 1.  Current international testing approach to gestational diabetes mellitus

Organization/ Selective vs Method of Screen positive Diagnostic Diagnostic (plasma glucose) threshold for GDM
country universal screening threshold test (mmol/L)
testing (mmol/L)

IADPSG (30) Universal One-step: 75-g 2-h 75-g Fasting ≥ 5.1


WHO (11) OGTT 2-hour 1-h ≥ 10.0
ADIPS (33) OGTT 2-h ≥ 8.5
FIGO (32) One abnormal value needed for diagnosis
JDS (34)
EBCOG (36)

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


Endocrine
Society (31)
China (35)
ADA (41) Universal One-step: 75-g 2-h ≥7.2 to 7.8a 75-g Fasting ≥ 5.1
OGTT 2-hour 1-h ≥ 10.0
Two-step: 50-g OGTT 2-h ≥ 8.5
GCT 100-g One abnormal value needed for diagnosis
3-hour Carpenter and Coustanb (17) or NDDG (13)
OGTT Fasting ≥ 5.3 Fasting ≥ 5.8
1-hour ≥ 10.0 1-hour ≥ 10.6
2-hour ≥ 8.6 2-hour ≥ 9.2
3-hour ≥ 7.8 3-hour ≥ 8.0
Two abnormal values needed for diagnosis
ACOGc (19) Universal Two-step: 50-g ≥7.2 to 7.8* 100-g Carpenter and Coustanb (17) or NDDG (13)
GCT OGTT Fasting ≥ 5.3 Fasting ≥ 5.8
1-hour ≥ 10.0 1-hour ≥ 10.6
2-hour ≥ 8.6 2-hour ≥ 9.2
3-hour ≥ 7.8 3-hour ≥ 8.0
Two abnormal values needed for diagnosisd
CDA (42) Universal Two-step: 50-g ≥7.8 50-g GCT ≥11.1 mmol/Le
GCT (preferred) 75-g Fasting ≥ 5.3
One-step: 75-g 2-hour 1-hour ≥ 10.6
2-h OGTT OGTT 2-hour ≥ 9.0
(alternative) One abnormal value needed for diagnosis
NICE (38) Selective Risk factorsf 75-g Fasting ≥ 7.0
2-hour 2-hour ≥ 7.8
OGTT One abnormal value needed for diagnosis
CNGOF (39) Selectiveg First ≥5.1
trimester Fasting ≥ 5.1
fasting 1-hour ≥ 10.0
glucose 2-hour ≥ 8.5
75-g One abnormal value needed for diagnosis
OGTTh
DDG/DGGG Universal Two-step: 50-g ≥7.5 50-g GCT ≥11.1 mmol/Le
(43) GCT 75-g Fasting ≥ 5.1
One-step: OGTT 1-hour ≥ 10.0
75-g OGTT 2-hour ≥ 8.5
(preferred) One abnormal value needed for diagnosis
DIPSI (44) Universal One-step: 75-g 75-g 2-hour ≥ 7.8i
OGTT OGTT

Abbreviations: ACOG, American College of Obstetricians and Gynecologists; ADA, American Diabetes Association; ADIPS, Australasian Diabetes in
Pregnancy Association; CDA, Canadian Diabetes Association; CNGOF, Organisme professionnel des médecins exerçant la gynécologie et l'obstétrique
en France; DDG, German Diabetes Association; DGGG, European Board of Gynecology and Obstetrics; DIPSI, Diabetes in Pregnancy Study Group of
India; FIGO, International Federation of Gynecology and Obstetrics; GCT, glucose challenge test; IADPSG, International Association of the Diabetes
and Pregnancy Study Groups; JDS, Japan Diabetes Society; NDDG, US National Diabetes Data Group; NICE, National Institute for Health and Care
Excellence; OGTT, oral glucose tolerance test; WHO, World Health Organization.
a
The ADA states that the choice of a specific positive GCT screening threshold is based upon the trade-off between sensitivity and specificity (41). ACOG
advises that in the absence of clear evidence that supports a specific GCT threshold value between 7.2 and 7.8 mmol/L, obstetricians and obstetric care
providers may select a single consistent GCT threshold for their practice based on factors such as community prevalence rates of GDM (19).
b
Plasma or serum glucose.
c
ACOG 2018 Clinical Practice Bulletin on GDM continues to recommend 2-step testing for GDM but states that individual practices and institutions may
choose to use the IADPSG’s 1-step testing approach and diagnostic criteria if appropriate for their population (19).
d
ACOG 2018 Clinical Practice Bulletin on GDM acknowledges that women who have even 1 abnormal value on the 100-g 3-hour OGTT have a
significantly increased risk of adverse perinatal outcomes compared to women without GDM but state that further research is needed to clarify the risk of
adverse outcomes and benefits of treatment in these women (19).
e
A glucose level ≥ 11.1 mmol/L following the initial screening GCT is classified as GDM, and there is no need for a subsequent 2-hour 75-g OGTT.
f
BMI > 30 kg/m2, previous macrosomia (≥4500 g), previous GDM, family history of diabetes, and family origin with a high prevalence of diabetes (South
Asian, Black Caribbean, Middle Eastern) (38).
g
Maternal age ≥ 35 years, body mass index ≥ 25 kg/m2, family history of diabetes, previous GDM, previous macrosomia (39).
h
If first trimester fasting glucose normal (ie, < 5.1 mmol/L).
i
Adapted from the WHO 1999 diagnostic criteria for GDM (45), using a nonfasting 75-g 2-hour OGTT (44).
6 Endocrine Reviews, 2022, Vol. XX, No. XX

at conception (75). However, DIP is not synonymous with macrosomia, and cesarean section (85). Similar to the HAPO
preexisting diabetes. In Australian, women with DIP who study, a clear glucose threshold was lacking, with pregnancy
performed an OGTT at 6 to 8 weeks postpartum, 21% had complications evident at fasting glucose levels <5.1 mmol/L
diabetes, 38% had impaired fasting glucose or impaired glu- (92 mg/dL). Third, maternal fasting glucose decreases in the
cose tolerance, and 41% returned to normal glucose toler- first trimester, most pronounced between 6 to 10 weeks’ gesta-
ance (76). tion [median decrease in glucose 0.11 mmol/L (1.98 mg/dL)]
Regardless of the specific nomenclature used, DIP is distinct (86), while studies have consistently shown that early fasting
from GDM, which is defined by lower glucose thresholds glucose is poorly predictive of GDM at 24 to 28 weeks’ ges-
on the OGTT and was historically considered to be a con- tation (86-88), leading to potential overdiagnosis of GDM.
dition of mid to late pregnancy. The ADA has not accepted In China, an early fasting glucose between 6.1  mmol/L to

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


this nomenclature and defines GDM based on timing of diag- 6.9  mmol/L (110-124  mg/dL) best corresponded to later
nosis: women diagnosed with diabetes in the first trimester GDM diagnosis (88), but this requires further validation.
are classified as having (preexisting) type 2 diabetes, while The WHO recommends the same diagnostic OGTT glu-
GDM is defined as diabetes diagnosed in later pregnancy cose thresholds for GDM in early pregnancy as those derived
and not meeting the diagnostic criteria for type 2 diabetes from HAPO by the IADPSG (11). However, the prognostic
(18). A  summary of the current international nomenclature value of these glucose levels in early pregnancy is yet to be es-
and diagnostic criteria for hyperglycemia in pregnancy is pre- tablished. Others have proposed an hemoglobin A1c (HbA1c)
sented in Table 2. risk threshold (89), based primarily on evidence that an early
HbA1c ≥ 5.9% (41 mmol/mol) detected all cases of DIP and pre-
dicted adverse pregnancy outcomes in a New Zealand cohort
Early GDM (90). However, studies in other cohorts have found that while
Most international guidelines now recommend early ante- an elevated HbA1c in early pregnancy is highly specific, it lacks
natal testing for women at high risk to identify women with sensitivity for identifying hyperglycemia and certain perinatal
DIP (11,18,30,38,39,42-44). This has resulted in increased complications (91,92), with no clear benefit of treating women
detection of milder degrees of hyperglycemia below the with HbA1c 5.7% to 6.4% (39-46  mmol/mol) in early preg-
threshold of DIP, referred to as GDM diagnosed prior to 24 nancy (93,94). A summary of the various international criteria
weeks’ gestation or early GDM. Studies in women with GDM for testing of GDM in early pregnancy is presented in Table 3.
have reported that between 27% and 66% of GDM can be Despite the lack of diagnostic clarity for early GDM,
detected in early pregnancy depending on the population as increasing evidence suggests that women with early GDM
well as the screening and diagnostic criteria used (77-81). represent a high-risk cohort (81). Early studies also reported
Recent studies evaluating the relationship between maternal worse pregnancy outcomes and increased insulin resistance in
glycemia and fetal growth trajectories confirm the early impact early GDM (78,95-97) but were confounded by the inclusion
of maternal glycemia on excess fetal growth and adiposity prior of women with pregestational diabetes. The first large retro-
to the diagnosis of standard GDM from 24 weeks’ gestation. spective cohort study excluding women with DIP showed that
A US multiethnic prospective cohort study of 2458 women en- women diagnosed and treated for early GDM, especially those
rolled between 8 and 13 weeks’ gestation included 107 (4.4%) diagnosed in the first trimester, were more insulin resistant
women with GDM (82). GDM was associated with an increase and at significantly greater risk for obstetric and neonatal
in estimated fetal weight from 20 weeks’ gestation, which be- complications compared to women diagnosed and treated for
came significant at 28 weeks’ gestation. Similarly, Sovio et al GDM from 24 weeks’ gestation (81). Other studies have since
showed that excessive fetal growth occurred between 20 to confirmed these findings (98,99). Concerningly, an increased
28 weeks’ gestation, prior to the diagnosis of GDM, especially risk of perinatal mortality and congenital abnormalities has
among women with higher body mass index [BMI (kg/m2)] also been reported in the offspring of women with early
(83). An Indian study also showed that excess subcutaneous GDM (75,78,95,96), with some data demonstrating that 5%
abdominal adiposity was first detected at 20 weeks’ gestation, of women with early GDM have abnormal fetal echocar-
at least 4 weeks prior to the diagnosis of GDM (84). Early ex- diograms (97). A  recent meta-analysis of 13 cohort studies
cess adiposity persisted despite adjustments for maternal age, showed greater perinatal mortality among women with early
BMI, GWG, fetal sex, and gestational age and remained higher GDM (RR 3.58; 95% CI 1.91-6.71) compared to women
at 32 weeks’ gestation (84). with a later diagnosis of GDM despite treatment (100).
Currently, there is no consensus for the preferred testing A recent study assessing the pathophysiological characteris-
approach or diagnostic glycemic thresholds for early GDM. tics of women diagnosed with GDM at a median of 16 weeks’
The IADPSG recommends diagnosing early GDM based on a gestation compared to those diagnosed from 24 weeks’ ges-
fasting glucose of 5.1 mmol/L to 6.9 mmol/L (92-124 mg/dL) tation using IADPSG diagnostic criteria reported that women
(30), consistent with the diagnostic fasting glucose threshold with early GDM had lower insulin sensitivity (defined by
for standard GDM. The utility of a single fasting glucose insulin-mediated glucose clearance during an OGTT), even
measurement for early GDM diagnosis warrants consider- after accounting for maternal BMI (101). Consistent with
ation. First, preanalytical glucose handling variation, particu- the pathophysiology of GDM, women with both early and
larly in the setting of a single glucose measurement, is a major standard GDM demonstrated impairment in pancreatic β-cell
issue for GDM diagnostic accuracy (discussed in the fol- function (102). These data underscore GDM phenotypic dif-
lowing text). Second, an Israeli cohort study of 6129 women ferences, specifically based on timing of diagnosis and degree
who underwent a fasting glucose test at a median of 9.5 of hyperglycemia (103).
weeks’ gestation demonstrated a positive association between A key issue is the current lack of high-quality evidence that
first trimester fasting glucose up to 5.8 mmol/L (104.5 mg/dL) diagnosing and treating early GDM improves pregnancy out-
and increased risk for subsequent diagnosis of GDM, LGA, comes. A  recent major RCT in the United States evaluating
Endocrine Reviews, 2022, Vol. XX, No. XX 7

early testing for GDM in 962 women with obesity included a fluoride tubes, with immediate placement in an ice slurry and
subgroup analysis of women diagnosed and treated for GDM centrifugation within 30 minutes (109). Citrate tubes are re-
[early n = 69 (15.0%) vs standard n = 56 (12.1%)] based on commended as an alternative where early centrifugation is
the 2-step testing approach (104). The average gestational not possible. These standards are important because a major
age at GDM diagnosis was similar at 24.3 ± 5.2 weeks for source of preanalytical glucose measurement error in sodium
the early screen group compared to 27.1 ± 1.7 weeks in the fluoride tubes is glycolysis by erythrocytes and leukocytes,
routine screen group. There was no difference in pregnancy which at room temperature lowers glucose levels prior to cen-
outcomes, although the primary composite perinatal out- trifugation at a rate of 5% to 7% per hour [~0.6  mmol/L
come (macrosomia, primary cesarean delivery, gestational (10  mg/dL)] (109,110). By 1 hour, this degree of glucose
hypertension, preeclampsia, hyperbilirubinemia, shoulder lowering is higher than the total analytical error threshold for

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


dystocia, and neonatal hypoglycemia) was nonsignificantly glucose based on biological variation (107).
higher in the early-screen group (56.9% vs 50.8%; P = 0.06). Recent studies have shown that OGTT preanalytical glu-
Requirement for insulin therapy was almost 4-fold higher, cose processing variability greatly impacts the prevalence of
while gestational age at delivery was lower (36.7 vs 38.7 GDM (67,111). Implementation of the AACC/ADA recom-
weeks’ gestation; P = 0.001) in women with early GDM. In mendations in a UK cohort resulted in higher mean glucose
a post hoc analysis of the Lifestyle in Pregnancy study (105), concentrations and 2.7-fold increased detection of GDM
no difference in pregnancy outcomes was shown between based on IADPSG criteria compared with the standard prac-
women randomized to either lifestyle intervention (n = 36) or tice of storing sodium fluoride tubes at room temperature and
standard treatment (n = 54) in early pregnancy. Whether dif- delaying centrifugation until collection of all 3 OGTT sam-
ferent glycemic targets are required reflecting physiological ples (112). This increase in GDM diagnosis was entirely at-
differences in early maternal glucose or whether additional tributable to control of glycolysis (107). Similarly, in a large
risk factors contributing to a more insulin resistant pheno- Australian multiethnic cohort (n = 12317), the rate of GDM
type such as maternal adiposity might also have a role re- diagnosis based on IADPSG criteria increased from 11.6%
main unanswered (81). The ongoing Treatment of Booking to 20.6% with early (within 10 minutes) vs delayed centri-
Gestational Diabetes Mellitus study, evaluating the impact of fugation (111). Mean glucose concentrations for the fasting,
immediate vs delayed care for gestational diabetes diagnosed 1-hour, and 2-hour OGTT samples were 0.24 mmol/L (5.4%),
at booking, will seek to determine whether or not there is 0.34 mmol/L (4.9%), and 0.16 mmol/L (2.3%) higher with
benefit from treating early GDM (106). early centrifugation, with the increase in GDM diagnosis pri-
marily due to the resulting increase in fasting glucose levels
(111). Importantly, the HAPO study, upon which the IADPSG
The Impact of Preanalytical Glucose diagnostic criteria for GDM was based, followed these AACC/
Processing Standards on the Diagnosis ADA preanalytical glucose processing standards (111).
of GDM
Although the contemporary testing approach to GDM re-
mains contentious, it is important to recognize that the diag- Incidence and Prevalence of GDM
nosis of GDM is based on the laboratory measurement of GDM is 1 of the most common medical complications of preg-
maternal glucose rather than a clinical diagnosis. Arguably nancy (73). In 2019, the International Diabetes Federation
then, a major issue in the contemporary diagnosis of GDM (IDF) estimated that 1 in 6 live births worldwide were com-
is optimizing preanalytical processing and measurement plicated by GDM (113). More than 90% of cases of hypergly-
of maternal plasma glucose to ensure diagnostic accuracy cemia in pregnancy occur in low- and middle-income countries
(107,108). This includes optimization of sample handling and (114), where the prevalence and severity of maternal and neo-
minimization of any analytic error. Unfortunately, stringent natal complications associated with GDM (47,113) contrast
preanalytical processing standards are not currently routinely with the near-normal pregnancy outcomes of modern manage-
applied. The American Association for Clinical Chemistry ment of GDM in developed countries (115).
(AACC) and ADA recommendations on laboratory testing The prevalence of GDM varies widely, depending on the
in diabetes advise collection of plasma glucose in sodium population, the specific screening and the diagnostic criteria

Figure 1.  Flowchart summarizing the contemporary nomenclature for hyperglycemia in pregnancy.
8 Endocrine Reviews, 2022, Vol. XX, No. XX

utilized. A  2012 systematic review of the diagnostic criteria recurrence rates of up to 84% (128). The risk of recurrence
used to define GDM reported a worldwide prevalence of GDM varies greatly depending on ethnicity (128). Ethnicities at in-
of 2% to 24.5% for the WHO criteria, 3.6% to 38% for the creased risk for development of type 2 diabetes, such as South
Carpenter and Coustan criteria, 1.4 to 50% for the NDDG cri- and East Asians, Hispanic, Black and Native Americans,
teria, and 2% to 19% for the IADPSG criteria (116). Aboriginal and Torres Strait Islanders, and Middle Easterners
Regardless of the specific diagnostic criteria or population, are also associated with an increased risk of GDM (129,130).
the prevalence of GDM continues to rise internationally, cor- A US study of over 123 000 women reported the prevalence of
responding to epidemiological factors including the back- GDM using the 2000 ADA diagnostic criteria to be the highest
ground rates of type 2 diabetes and increased incidence of among Filipinas (10.9%) and Asians (10.2%), followed by
obesity in women of childbearing age and rising maternal age Hispanics (6.8%), non-Hispanic Whites (4.5%) and Black

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


(117-124). Implementation of the revised IADPSG diagnostic Americans (4.4%) (131). Women who have had GDM are at
criteria have further increased the proportion of women being increased risk for subsequent type 2 diabetes, while family his-
diagnosed with GDM (69,125,126). The incidence of GDM tory of type 2 diabetes in a first-degree relative or sibling with
in the original HAPO study cohort applying the IADPSG GDM is a major risk factor for GDM (129,132-134).
diagnostic criteria ranged from 9.3% to 25.5% depending Increasing maternal age is also a risk factor for GDM
on study site (69). Recent international prevalence data also (129,133-135). The prospective First and Second Trimester
demonstrate marked variability in the rate of GDM, ranging Evaluation of Risk trial (n = 36 056)  demonstrated a con-
from 6.6% in Japan and Nepal to 45.3% of pregnancies in tinuous positive relationship between increasing maternal age
the United Arab Emirates (127). and risk for adverse pregnancy outcomes, including GDM
(135). Maternal age 35 to 39 years and ≥40 years was associ-
ated with an adjusted odds ratio (OR) for GDM of 1.8 (95%
Risk Factors for GDM CI 1.5-2.1) and 2.4 (95% CI 1.9-3.1), respectively (135).
Several modifiable and nonmodifiable risk factors for GDM Other studies in high-risk cohorts have reported a lesser risk
have been identified (Table 4). A history of GDM in a previous between increasing maternal age and GDM after adjustment
pregnancy is the strongest risk factor for GDM, with reported for other risk factors (136).

Table 2.  Classification and diagnostic criteria for hyperglycemia in pregnancy

Organization Results

IADPSG/EBCOG (30,36)
 GDM 75-g 2-hour OGTT
  Fasting glucose 5.1-6.9 mmol/L
  1-hour glucose ≥ 10.0 mmol/L
  2-hour glucose 8.5-11.0 mmol/L
  Overt diabetes during pregnancy Fasting glucose ≥ 7.0 mmol/L
Random glucose ≥ 11.1 mmol/La
HbA1c ≥ 6.5%
WHO/FIGO/ADIPS (11,32,33)
 GDM 75-g 2-hour OGTT
  Fasting glucose 5.1-6.9 mmol/L
  1-hour glucose ≥ 10.0 mmol/L
  2-hour glucose 8.5-11.0 mmol/L
  Diabetes mellitus in pregnancy Fasting glucose ≥ 7.0 mmol/L
2-hour glucose ≥ 11.1 mmol/L post 75-g OGTT
Random glucose ≥ 11.1 mmol/L in the presence of diabetes symptoms
ADA (41)
 GDM 1-step strategy: 2-step strategy: NDDG (13)
75-g 2-h OGTT 50-g 1-hour GCT ≥ 7.8 mmol/L   Fasting glucose ≥ 5.8 mmol/L
  Fasting glucose ≥ 5.1 mmol/L 100 g 3-hour OGTT   1-h glucose ≥ 10.6 mmol/L
  1-hour glucose ≥ 10.0 mmol/L   Carpenter and Coustan (17) or   2-h glucose ≥ 9.2 mmol/L
  2-hour glucose ≥ 8.5 mmol/L   Fasting glucose ≥ 5.3 mmol/L   3-h glucose ≥ 8.0 mmol/L
  1-hour glucose ≥ 10.0 mmol/L
  2-hour glucose ≥ 8.6 mmol/L
  3-hour glucose ≥ 7.8 mmol/L
  Type 2 diabetes mellitus Fasting glucose ≥ 7.0 mmol/L
2-hour glucose ≥ 11.1 mmol/L post 75 g 2-hour OGTT
Random glucose ≥ 11.1 mmol/L in the presence of diabetes symptoms
HbA1c ≥ 6.5%

75-g 2-hour OGTT: only 1 plasma glucose level needs to be elevated for the diagnosis of GDM. 100 g 3-hour OGTT: at least 2 plasma glucose levels need
to be elevated for the diagnosis of GDM.
Abbreviations: ADA, American Diabetes Association; ADIPS, Australasian Diabetes in Pregnancy Association; EBCOG, European Board & College of
Obstetrics and Gynaecology; FIGO, International Federation of Gynecology and Obstetrics; GCT, glucose challenge test; HbA1c, hemoglobulin A1c;
IADPSG/; International Association of the Diabetes and Pregnancy Study Groups; GDM, gestational diabetes mellitus; OGTT, oral glucose tolerance test;
WHO, World Health Organization.
a
The IADPSG recommends confirmation by fasting plasma glucose or HbA1c for the diagnosis of overt diabetes during pregnancy (30).
Endocrine Reviews, 2022, Vol. XX, No. XX 9

Maternal prepregnancy overweight (BMI 25-29.99 kg/m2) are maintained at lower levels than in healthy nonpregnant
or obesity (BMI ≥ 30  kg/m2) are common risk factors for women (175,176), and euglycemia is maintained by a corres-
GDM (129,130,133,134,136,137). The risk of GDM is in- ponding 200% to 250% increase in insulin secretion, most
creased almost 3-fold (95% CI 2.1-3.4) in women with notable in early pregnancy (161,167,177). Human placental
class  I  obesity (BMI 30-34.99  kg/m2) and 4-fold (95% CI lactogen, in addition to prolactin and growth hormone, pri-
3.1-5.2) in women with class  II obesity (BMI 35-39.99  kg/ marily regulate increased maternal β-cell insulin secretion and
m2), compared to women with a BMI < 30 kg/m2 (138). High proliferation during pregnancy (178-180). Rodent studies
GWG, particularly in the first trimester, is also associated with have demonstrated a 3- to 4-fold increase in β-cell mass
an increased risk for GDM (131,139,140). Further, women during pregnancy, mediated via hypertrophy, hyperplasia,
with obesity and high GWG are 3- to 4-fold more likely to de- neogenesis, and/or reduced apoptosis (181,182).

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


velop abnormal glucose tolerance compared to women who GDM is characterized by a relative insulin secretory
remained within the 1990 Institute of Medicine (IOM) re- deficit (177), in which maternal β-cell insulin secretion is
commendations for GWG (131,141). Interpregnancy weight unable to compensate for the progressive rise in insulin re-
gain is also a risk factor for GDM and perinatal complica- sistance during pregnancy (183). This leads to decreased
tions in a subsequent pregnancy (142) and may be a potential glucose uptake, increased hepatic gluconeogenesis, and ma-
confounder when considering the risk of GDM recurrence. ternal hyperglycemia (167). It is hypothesized that this re-
Studies have demonstrated an association between polycystic sults from the failure of β-cell mass expansion (182,184).
ovary syndrome and GDM, although this is significantly at- Hyperlipidemia, characterized predominantly by higher
tenuated after adjustment for maternal BMI (143,144). Other serum triglycerides, may also cause lipotoxic β-cell injury,
risk factors for GDM include multiparity (133,134), twin further impairing insulin secretion (185,186). The pathogen-
pregnancy (145,146), previous macrosomia (123), a history of esis of GDM therefore parallels that of type 2 diabetes, char-
perinatal complications (134), maternal small-for-gestational- acterized by both increased insulin resistance and relative
age (SGA) or LGA (134), physical inactivity (129,147,148), insulin deficiency arising from a reduction in β-cell function
low-fiber high-glycemic load diets (149), greater dietary fat and mass (187,188).
and lower carbohydrate intake (137), and medications such as Serial studies of the insulin secretory response in women
glucocorticoids and anti-psychotic agents (150,151). Maternal who develop GDM suggest that the abnormal insulin secre-
pre- and early pregnancy hypertension is also associated with tory response is present from prepregnancy and increases in
an increased risk of developing GDM (152,153). early pregnancy, prior to and independent of changes in in-
Overall, noting the variation in performance and utility of sulin sensitivity (170,189-191). These data suggest that many
clinical risk factors based on local population factors, pre- women with GDM may have chronic or preexisting β-cell
vious GDM and family history of diabetes appear to be the dysfunction, potentially mediated by circulating hormones
strongest clinical risk factors for GDM (154-157). Ethnicity, including leptin (191).
higher maternal age, and BMI are also strong predictors for
GDM (154-158).
Genetics of GDM
The genetics of GDM and glucose metabolism in pregnancy
Pathophysiology of GDM remain poorly defined. Data on epigenetic mechanisms in
Normal pregnancy is associated with marked changes in GDM are especially lacking and primarily limited to the po-
glycemic physiology (159,160). There is a progressive in- tential role of DNA methylation in mediating the intrauterine
crease in insulin resistance, predominantly due to increased effects of GDM on offspring outcomes (192,193).
circulating placental hormones including growth hormone, Most genetic studies have focused on variants associated
corticotrophin-releasing hormone, human placental lactogen, with type 2 diabetes and have demonstrated a similar associ-
prolactin, estrogen, and progesterone (161-166). Increased ation with GDM (194,195). A meta-analysis of 28 case-control
maternal adiposity particularly in early pregnancy also pro- studies (n = 23425) (196) identified 6 genetic polymorphisms
motes insulin resistance, contributing to facilitated lipolysis at loci involved in insulin secretion [insulin-like growth factor
by late pregnancy (167,168). The resultant increase in ma- 2 messenger RNA-binding protein 2 (IGF2BP2), melatonin
ternal free fatty acid (FFA) levels exacerbates maternal insulin receptor 1B (MTNR1B) and transcription factor 7-like 2
resistance by inhibiting maternal glucose uptake and stimu- (TCF7L2)] (197-199), insulin resistance [insulin receptor sub-
lating hepatic gluconeogenesis (168,169). By late pregnancy, strate 1 (IRS1) and peroxisome proliferator-activated receptor
studies have reported decreases in maternal glucose sensitivity gamma (PPARG)] (200,201), and inflammation [tumor ne-
between 40% and 80% in women with normal or increased crosis factor alpha (TNF-α)] (202) in type 2 diabetes. Overall,
BMI (170-172). Increased maternal insulin resistance results only MTNR1B, TCF7L2, and IRS1 were also significantly
in higher maternal postprandial glucose levels and FFAs for associated with GDM, supporting the role of both impaired
maternal growth (164,167,173) and increased facilitated dif- insulin secretion and insulin resistance in the pathogenesis
fusion across the placenta, leading to greater availability of of GDM as well as type 2 diabetes (196). Subgroup analysis
glucose for fetal growth (161,174). This progressive rise in showed the risk alleles of TCF7L2 and PPARG were signifi-
maternal insulin resistance underpins the delayed testing ap- cant only in Asian populations, while the association between
proach to GDM, aiming to maximize detection of GDM when IRS1 and TCF7L2 and GDM risk varied depending on diag-
insulin resistance is at its greatest in mid- to late gestation. nostic criteria and genotype methodology (196), highlighting
In addition to increased insulin resistance and elevated post- the need for further large confirmatory studies.
prandial glucose, adaptations in normal pregnancy include Two genome-wide association studies (GWAS) have
enhanced insulin secretion (160,165). Maternal glucose levels evaluated the genetic associations for GDM and glucose
10 Endocrine Reviews, 2022, Vol. XX, No. XX

metabolism (194,203). The first, a 2-stage GWAS in Korean associated with glucose or C-peptide levels in pregnancy,
women, compared 468 women with GDM and 1242 normo- although strength of association varied across cohorts
glycemic women using 2.19 million genotyped markers be- (194). Specifically, loci in glucokinase regulator (GCKR),
fore further genotyping 11 loci in 1714 women, identifying glucose-6-phosphatase 2 (G6PC2), proprotein convertase
2 loci significantly associated with GDM (203). A  variant subtilisin/kexin type 1 (PCSK1), protein phosphatase 1,
in cyclin-dependent kinase 5 regulatory subunit-associated regulatory subunit 3B (PPP1R3B), and MTNR1B were
protein 1-like 1 (CDKAL1) had the strongest association associated with fasting glucose. In addition, GCKR and
with GDM, followed by a variant near MTNR1B expressed PPP1R3B were associated with fasting C-peptide levels,
in pancreatic β-cells (204). The IGF2BP2 variant did not while MTNR1B was associated with 1-hour postload glu-
reach genome-wide significance with GDM in this study. cose. These loci have also previously been associated with

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


CDKAL1 was significantly associated with decreased fasting lipid metabolism (GCKR and PPP1R3B), glycogen me-
insulin concentration and homeostasis model assessment of tabolism (PPP1R3B), and obesity-related traits (PCSK1)
β-cell function in women with GDM, consistent with im- (209-214).
paired β-cell compensation. MTNR1B was associated with Two additional novel loci identified near hexokinase do-
decreased fasting insulin concentrations in women with main containing 1 (HKDC1) associated with 2-hour postload
GDM and increased fasting glucose concentrations in both glucose, and β-site amyloid polypeptide cleaving enzyme 2
women with and without GDM (203). Variants in CDKAL1 (BACE2) associated with fasting C-peptide, demonstrated
and MTNR1B have previously been associated with type 2 limited association with glycemic traits outside of compared
diabetes risk (205,206). to in pregnancy (215). In general, however, studies evaluating
A subsequent GWAS performed in a subset of the associations between genetic risk scores, glycemic traits in
HAPO cohort (n = 4528) comprising European, Thai, pregnancy, and GDM have also confirmed that genetic de-
Afro-Caribbean, and Hispanic women evaluated ma- terminants of fasting glucose and insulin, insulin secretion,
ternal metabolic traits in pregnancy (194). This study re- and insulin sensitivity reported outside of pregnancy influence
ported 5 variants associated with quantitative glycemic GDM risk (216). A  summary of the genes associated with
traits in the general population (207,208) that were also GDM is provided in Table 5.

Table 3.  International criteria for testing of gestational diabetes mellitus in early pregnancy

Organization Early pregnancy Method of testing Diagnostic test Criteria for diagnosing early GDM (mmol/L)
testing

IADPSG (30) Yes Selective—women at risk Fasting glucoseb ≥5.1


of overt diabetes during
pregnancya
WHO (11) Not specifiedc 75-g 2-hour OGTT Fasting 5.1-6.9 or
1-hour ≥ 10.0 or
2-hour 8.5-11.0
ADIPS (33) Yes Selective—women at risk 75-g 2-hour OGTT Fasting 5.1-6.9 or
of hyperglycemia in 1-hour ≥ 10.0 or
pregnancyd 2-hour 8.5-11.0
ADA (41) Yes Selective—women One-step: 75-g 2-hour Fasting 5.1-6.9 or
with risk factors for OGTT 1-hour ≥ 10.0 or
undiagnosed type 2 Two-step: 50-g GCT 2-hour 8.5-11.0
diabetese 100-g 3-hour OGTT ≥7.2 to 7.8
Carpenter and Coustan (17) NDDG (13)
Fasting ≥ 5.3  ≥ 5.8
1-hour ≥ 10.0  ≥ 10.6
2-hour ≥ 8.6  ≥ 9.2
3-hour ≥ 7.8  ≥ 8.0
ACOG (19) Yes Selective—women 75-g 2-h OGTT or Fasting ≥ 7.0 or
with risk factors for 50-g GCT 2-hour ≥ 11.1
undiagnosed type 2 Confirmatory ≥7.2 to 7.8
diabetes or GDMf 100-g 3-hour OGTT Carpenter and Coustan (17) NDDG (13)
Fasting ≥ 5.3  ≥ 5.8
1-hour ≥ 10.0  ≥ 10.6
2-hour ≥ 8.6  ≥ 9.2
3-hour ≥ 7.8  ≥ 8.0
EBCOG (36) Yes Selective—women at risk 75-g 2-hour OGTT Fasting 5.1-6.9 or
of overt diabetes during 1-hour ≥ 10.0 or
pregnancyg 2-hour 8.5-11.0
DDG/DGGG Yes Selective—women Random glucose or 7.8-11.05 mmol/L followed by a second blood
(43) with risk factors for Fasting glucose or glucose measurement or an OGTT
“manifest diabetes”h 75-g 2-hour OGTT 5.1-6.9
Fasting 5.1-6.9 or
1-hour ≥ 10.0 or
2-hour 8.5-11.0
Endocrine Reviews, 2022, Vol. XX, No. XX 11

Table 3.  Continued

Organization Early pregnancy Method of testing Diagnostic test Criteria for diagnosing early GDM (mmol/L)
testing

CNGOF (39) Yes Selectivei Fasting glucose ≥5.1


NICE (38) Yes Selectivej 75-g 2-hour OGTT Fasting ≥ 5.6
2-hour ≥ 7.8
DIPSI (44) Yes Universal 75-g 2-hour OGTTk 2-hour ≥ 7.8

75-g 2-h OGTT: Only 1 abnormal glucose level needs to be elevated for the diagnosis of GDM. 100-g 3-h OGTT: 2 abnormal glucose levels need to be
elevated for the diagnosis of GDM.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


Abbreviations: ADA, American Diabetes Association; ACOG, American College of Obstetricians and Gynecologists; ADIPS, Australasian Diabetes in Pregnancy
Association; CNGOF, Organisme professionnel des médecins exerçant la gynécologie et l'obstétrique en France; DDG, German Diabetes Association; DGGG,
European Board of Gynecology and Obstetrics; DIPSI, Diabetes in Pregnancy Study Group of India; EBCOG, European Board & College of Obstetrics and
Gynaecology; GCT, glucose challenge test; GDM, gestational diabetes mellitus; IADPSG, International Association of the Diabetes and Pregnancy Study Groups;
NICE, National Institute for Health and Care Excellence; OGTT, oral glucose tolerance test; WHO, World Health Organization.
a
High-risk criteria not explicitly defined.
b
IADPSG does not recommend routinely performing the 75-g 2-h OGTT prior to 24 weeks’ gestation but advises that a fasting glucose ≥ 5.1 mmol/L in
early pregnancy be classified as GDM (30).
c
GDM diagnosed at any time in pregnancy based on an abnormal 75-g 2-h OGTT (11).
d
High-risk criteria defined as previous hyperglycemia in pregnancy; previously elevated blood glucose level; maternal age ≥ 40 years; ethnicity: Asian, Indian
subcontinent, Aboriginal, Torres Strait Islander, Pacific Islander, Maori, Middle Eastern, non-White African; family history of diabetes (first-degree relative
with diabetes or sister with hyperglycemia in pregnancy); prepregnancy body mass index > 30 kg/m2; previous macrosomia (birth weight > 4500 g or > 90th
percentile); polycystic ovary syndrome; and medications: corticosteroids, antipsychotics (33).
e
High-risk criteria defined as body mass index ≥ 25 kg/m2 (≥ 23 kg/m2 in Asian Americans) plus 1 of the following: physical inactivity; previous GDM;
previous macrosomia (≥ 4000 g); previous stillbirth; hypertension; high density lipoprotein cholesterol ≤ 0.90 mmol/L; fasting triglycerides ≥ 2.82 mmol/L;
polycystic ovary syndrome; acanthosis nigricans; nonalcoholic steatohepatitis; morbid obesity and other conditions associated with insulin resistance;
hemoglobulin A1c ≥ 5.7%; impaired glucose tolerance or impaired fasting glucose; cardiovascular disease; family history of diabetes (first-degree relative);
and ethnicity: African American, American Indian, Asian American, Hispanic, Latina, or Pacific Islander ethnicity. Note that the ADA recommends testing
for GDM at 24 to 28 weeks’ gestation and have no specific definition for early GDM (41).
f
ACOG states that the best test for early GDM screening is not clear but suggest the testing approach and diagnostic criteria used to diagnose type 2
diabetes in the nonpregnant population and thus have no specific definition for early GDM (19).
g
High-risk criteria defined as previous GDM; overweight/obesity; family history of diabetes (first-degree relative with diabetes); previous macrosomia (>4000g or
>90th percentile); polycystic ovary syndrome; ethnicity: Mediterranean, South Asian, black African, North African, Caribbean, Middle Eastern, or Hispanic (36).
h
High-risk criteria defined as age ≥ 45 years; prepregnancy body mass index ≥ 30 kg/m2; physical inactivity; family history of diabetes; high-risk ethnicity
(eg. Asians, Latin Americans); previous macrosomia ≥ 4500 g; previous GDM; hypertension; prepregnancy dyslipidemia (high-density lipoprotein
cholesterol ≤ 0.90 mmol/L, fasting triglycerides ≥ 2.82 mmol/L); polycystic ovary syndrome; prediabetes in an earlier test; other clinical conditions
associated with insulin resistance (eg, acanthosis nigricans); history of coronary artery disease/peripheral artery disease/cerebral vascular disease;
medications associated with hyperglycemia (eg. glucocorticoids). Note that the DDG/DGGG recommends that a 75-g 2-h OGTT be the initial early test in
high-risk women (defined as women with ≥2 risk factors for GDM) (43).
i
High-risk criteria are defined as previous GDM, previous impaired glucose tolerance, and/or obesity (39).
j
High-risk criteria defined as body mass index> 30 kg/m2; previous macrosomia (≥4500 g); previous GDM; family history of diabetes (first-degree relative
with diabetes); minority ethnic family origin with a high prevalence of diabetes. The updated 2015 NICE guidelines state that women with previous GDM
should undergo early self-monitoring of blood glucose or a 75-g 2-hour OGTT as soon as possible after booking (first or second trimester), and a repeat
75-g 2-hour OGTT at 24 to 28 weeks’ gestation if the initial OGTT was negative (38).
k
2-hour postload glucose measured on nonfasting 75-g OGTT (44).

Table 4.  Key risk factors for gestational diabetes mellitus glucokinase (GCK)-MODY subtype (MODY2) (32%) were
most frequent in probands confirmed with MODY, followed
Previous GDM by HNF-4α (MODY1) and HNF-1β (MODY5) (224).
An ethnicity with a high prevalence of diabetes Women with GCK-MODY often first present following
Maternal age > 35 years antenatal screening for GDM, with an estimated prevalence
of 1% of all GDM “cases” actually GCK-MODY (220,222).
Family history of diabetes (first-degree relative with diabetes)
GCK-MODY is caused by mutations in the glucokinase gene,
Obesity (BMI > 30 kg/m2)
leading to a greater set point for glucose stimulated insulin
Previous macrosomia (birthweight > 4500 g) release (219). Clinically, GCK-MODY is defined by mild,
Polycystic ovary syndrome stable fasting hyperglycemia [fasting glucose 98-150  mg/
Iatrogenic: glucocorticoids and antipsychotic medication dL (5.4-8.3  mmol/L)] and low rates of microvascular and
macrovascular complications (220). It should be suspected
Abbreviations: BMI, body mass index; GDM, gestational diabetes mellitus. following a positive OGTT in pregnancy if the fasting glu-
cose is ≥5.5 mmol/L, the glucose increment from the fasting
Maturity-onset Diabetes of the Young to 2-hour (75-g) OGTT is small (<4.6 mmol/L), and there is
Maturity-onset diabetes of the young (MODY) is the most a positive family history of mild hyperglycemia or diabetes.
common form of monogenic diabetes; inherited forms of dia- In addition, a combination of fasting glucose ≥ 100  mg/dL
betes characterized by defects in single genes regulating β-cell (5.6 mmol/L) and BMI < 25 kg/m2 has been shown to have a
development and function (217,218). MODY consists of sev- sensitivity of 68% and a specificity of 99% for differentiating
eral autosomal dominant forms of diabetes accounting for up GCK-MODY from GDM (220). Importantly, management
to 2% of all diabetes diagnoses (219). A diagnosis of MODY differs from that of GDM because the need for intensive ma-
requires confirmatory molecular genetic testing, and thus ternal glycemic control largely depends on whether the GCK-
MODY is frequently misdiagnosed as preexisting diabetes or MODY mutation is also present in the fetus (220,225,226).
GDM, accounting for up to 5% of GDM “cases” (220-223). Maternal insulin therapy is therefore only recommended in
A  UK study reported that HNF-1α (MODY3) (52%) and the presence of increased fetal abdominal growth (>75th
12 Endocrine Reviews, 2022, Vol. XX, No. XX

centile) measured on serial ultrasounds from 26 weeks’ ges- than women treated with insulin therapy (249). In contrast,
tation, as this indicates that the fetus does not have the GCK the HAPO study did not demonstrate excess perinatal mor-
mutation (220). tality in their untreated cohort (27).
Modern management of GDM and associated maternal
risk factors is associated with near-normal birthweight in
Consequences of GDM developed countries (115,257). This is important because
GDM is associated with excess neonatal and maternal short- birthweight is the major risk factor for shoulder dystocia,
and long-term morbidity, summarized in Table 6. brachial plexus injury, neonatal hypoglycemia, and neonatal
respiratory distress syndrome in the offspring of women
with and without GDM (242). A retrospective cohort study
Neonatal Complications

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


of 36 241 pregnancies in the United States reported that the
risk of shoulder dystocia among infants of women without
The Pedersen hypothesis describes the pathophysiology con-
GDM compared to women with GDM was 0.9% vs 1.6%
tributing to perinatal complications in GDM (229). Maternal
if birthweight was <4000  g and 6.0% vs 10.5% if birth-
hyperglycemia results in fetal hyperglycemia via facilitated
weight was ≥4000  g (macrosomia) (242). The risk of neo-
diffusion of glucose by the glucose transporter 1 (GLUT1)
natal hypoglycemia in infants with birthweight < 4000 g was
(230). Fetal hyperglycemia results in fetal hyperinsulinemia,
1.2% vs 2.6% and 2.4% vs 5.3% for birthweight ≥ 4000 g,
promoting fetal anabolism, excessive fetal adiposity, and
in women without GDM compared to women with GDM,
accelerated growth, leading to LGA and macrosomia (231-
respectively. Similar findings were seen for brachial plexus in-
239). Maternal hyperlipidemia also contributes to excess
jury and neonatal respiratory distress syndrome. Thus, GDM
fetal growth (233,240). Macrosomia and LGA increase the
confers increased risk of perinatal complications independent
risk of cesarean section, birth trauma, and perinatal compli-
of birthweight.
cations including shoulder dystocia, brachial plexus injury
The risk of stillbirth is also greater in women with GDM.
and fracture, and perinatal asphyxia (27,132,237,238,241-
A large US retrospective analysis examined stillbirth rates at
243). Increased risk of perinatal asphyxia is associated with
various stages of gestation in over 4 million women, including
fetal death in utero, polycythemia, and hyperbilirubinemia
193 028 women with GDM. The overall risk of stillbirth from
(27,244-246). Fetal hyperinsulinemia can also increase the
36 to 42 weeks’ gestation was higher in women with GDM
risk of metabolic abnormalities including neonatal hypogly-
compared to women without GDM (17.1 vs 12.7 per 10 000
cemia, hyperbilirubinemia, and respiratory distress syndrome
deliveries; RR 1.34; 95% CI 1.2-1.5) (253). This increased
postpartum (27,244). The risk appears to be greater among
risk of stillbirth was also observed at each gestational week:
offspring of women with more severe hyperglycemia (247).
3.3 to 8.6 per 10 000 ongoing pregnancies in women with
Figure 2 summarizes the perinatal consequences of GDM.
GDM compared to 2.1 to 6.4 per 10 000 ongoing pregnan-
cies in women without GDM from 36 to 41 weeks’ gestation
Short-term Risk (253). For women with GDM, the relative risk of stillbirth
was highest in week 37 (RR 1.84, 95% CI 1.5-2.3). Notably,
In the HAPO study, higher maternal glucose levels were as-
the risk of stillbirth is highest in women with undiagnosed
sociated with an increased risk of LGA, shoulder dystocia or
GDM. In a UK prospective case-control study (n = 1024),
birth injury, and neonatal hypoglycemia (27). A  recent sys-
women with undiagnosed GDM based on a fasting glucose
tematic review (n = 207 172) confirmed similar positive linear
level ≥ 5.6mmol/L (≥100 mg/dL) had a 4-fold greater risk of
associations for maternal glycemia based on maternal glucose
late stillbirth (defined as occurring ≥28 weeks’ gestation) com-
thresholds for the GCT, 75-g 2-hour OGTT, or 100-g 3-hour
pared to women with fasting glucose < 5.6mmol/L (<100 mg/
OGTT and risk of cesarean section, induction of labor (IOL),
dL) (74). In contrast, women at risk of GDM based on NICE
LGA, macrosomia, and shoulder dystocia (248). GDM has
risk factors who were diagnosed with GDM on the OGTT
also been associated with an increased risk of preterm birth,
had a similar risk of stillbirth to women who were not at risk
birth trauma, neonatal respiratory distress syndrome, and
of GDM. This suggests that diagnosing and managing GDM
hypertrophic cardiomyopathy (27,244,249). An increased
reduces the risk of stillbirth to near-normal levels (74).
risk of congenital malformations in the offspring has been
reported, although whether this persists after adjustment for
maternal age, BMI, ethnicity, and other contributing factors
is unknown (250). A  French cohort study (n = 796 346) re- Long-term Risk in the Offspring
ported a 30% higher risk of cardiac malformations in the Recent epidemiological studies suggest an increased risk of
offspring of women with GDM compared to women with later adverse cardiometabolic sequelae in the offspring of
normal glucose tolerance, after excluding women with likely women with GDM (227,258). A  large Danish population-
undiagnosed pregestational diabetes (249). However, this in- based cohort study (n = 2 432 000) demonstrated an asso-
creased risk only reached statistical significance in women ciation between maternal diabetes and an increased rate of
treated with insulin therapy. Maternal BMI, which was not early onset cardiovascular disease (CVD; ≤40  years of age)
evaluated in these studies, may account for these findings among offspring (259). GDM specifically was associated with
(251,252). Similarly, a reported increase in perinatal mor- a 19% increased risk of early onset CVD (95% CI 1.07-1.32).
tality after 35 weeks’ gestation in the offspring of women A  longitudinal UK study provides potential mechanistic in-
with GDM may also be confounded by obesity (253-256). An sight, finding that GDM was associated with alterations in
increased risk of perinatal mortality after 37 weeks’ gestation fetal cardiac function and structure, with reduced systolic and
was demonstrated in French women with GDM on dietary diastolic ventricular function persisting in infancy (260). This
intervention, possibly because these women delivered later is consistent with the association between in utero exposure to
Endocrine Reviews, 2022, Vol. XX, No. XX 13

maternal hyperglycemia and fetal programming first reported As demonstrated in HAPO and other studies, women with
in the Native American Pima population, characterized by a GDM also have an increased risk of gestational hypertension
high prevalence of obesity, type 2 diabetes, and GDM (261). and preeclampsia (267-269). Consistent with the association
The recent HAPO Follow Up Study (HAPO-FUS), which between diabetes and microvascular disease, abnormalities
was not confounded by treatment of maternal glycemia, in- in glucose metabolism affect trophoblast invasion, leading
cluded 4832 children 10 to 14  years of age whose mothers to impaired placentation and greater risk for preeclampsia
were participants of HAPO (227). The HAPO-FUS demon- (270). The mechanism likely relates to insulin resistance and
strated a durable impact of maternal glycemia with long-term inflammatory pathway activation (271,272), with in vitro
offspring glucose metabolism, including at glucose levels studies showing that elevated glucose concentrations inhibit
lower than those diagnostic for GDM (227). A  generally trophoblast invasiveness by preventing uterine plasminogen

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


linear relationship between maternal antenatal glucose and activator activity (272).
offspring glucose levels and related outcomes was observed.
Increasing maternal glucose categories were associated with Long-term Maternal Risk Following GDM
a higher risk of impaired fasting glucose and impaired glu- Women diagnosed with GDM based on pre-IADPSG diag-
cose tolerance and higher timed glucose measures and HbA1c nostic criteria are at increased risk of GDM in future preg-
levels and were inversely associated with insulin sensitivity nancies, with reported recurrence rates of 30% to 84%
and disposition index by 14  years of age, independent of (128). A  diagnosis of GDM is also associated with up to a
maternal and childhood BMI and family history of diabetes 20-fold greater lifetime risk of type 2 diabetes (273,274).
(227). A  positive association was observed between GDM A  recent large meta-analysis and systematic review (20
defined by any criteria and glucose levels and impaired glu- studies, n = 1 332 373 including 67 956 women with GDM)
cose tolerance in the offspring at ages 10 to 14  years and showed that women with a history of GDM have a 10-fold
an inverse association with offspring insulin sensitivity (262). increased risk of developing type 2 diabetes, mostly within
Higher frequencies of childhood obesity and measures of adi- the first 5 years post-GDM (273). HAPO-FUS demonstrated
posity across increasing categories of maternal OGTT glucose that over 50% of women whose OGTT thresholds met (un-
levels were also noted (262). Recent evidence for increased treated) IADPSG diagnostic criteria for GDM had devel-
glucose-linked hypothalamic activation in offspring aged 7 to oped impaired glucose tolerance after 14 years of follow-up
11 years previously exposed to maternal obesity and GDM in (275). These data highlight the importance of a management
utero, which predicted higher subsequent BMI, represents 1 approach to GDM that focuses on early prevention of type
possible mechanism for this increased childhood obesity risk 2 diabetes. For example, the updated NICE guidelines now
(263). recommend diabetes prevention for all women with previous
GDM (276,277).
Previous GDM is also associated with cardiovascular
Maternal Complications
risk factors such as obesity, hypertension, and dyslipidemia
Short-term Risk (274,278-280). The lifetime risk of cardiovascular disease fol-
Women with GDM are at an increased risk of obstetric inter- lowing GDM is almost 3-fold higher in women who develop
vention including IOL, cesarean section (27-29,264,265), and type 2 diabetes and 1.5 fold higher even in women without
complications associated with delivery including perineal la- type 2 diabetes (280). Studies also report a 26% greater risk
cerations and uterine rupture, predominantly relating to fetal of hypertension and a 43% greater risk of myocardial infarc-
macrosomia and polyhydramnios (266). tion or stroke in women with previous GDM compared to

Table 5.  Genes linked to gestational diabetes mellitus

Gene Gene name Function


symbol

MTNR1B Melatonin receptor 1B Receptor mediating the action of melatonin, including its inhibitory effect on insulin
secretion
TCF7L2 Transcription factor 7-like 2 Blood glucose homeostasis
IRS1 Insulin receptor substrate 1 Receptor mediating the control of various cellular processes by insulin
CDKAL1 Cyclin-dependent kinase 5 regulatory Proinsulin to insulin conversion
subunit-associated protein 1-like 1
GCKR Glucokinase regulator Inhibits glucokinase in liver and pancreatic islet cells
G6PC2 Glucose-6-phosphatase 2 Glucose metabolism
PCSK1 Proprotein convertase subtilisin/ Endoprotease involved in proteolytic activation of polypeptide hormones and neuropeptides
kexin type 1 precursors including proinsulin, proglucagon-like peptide 1, and pro-opiomelanocortin
PPP1R3B Protein phosphatase 1, regulatory Regulates glycogen metabolism
subunit 3B
HKDC1 Hexokinase domain containing 1 Involved in glucose homeostasis and hepatic lipid accumulation
BACE2 Beta-site amyloid polypeptide Proteolytic processing of CLTRNa in pancreatic β-cells
cleaving enzyme 2

Genes were identified and selected from the genome-wide association studies (194,203). The name and function of each gene was determined from
GeneCards (https://www.genecards.org).
a
Collectrin, amino acid transport regulator is a stimulator of β-cell replication.
14 Endocrine Reviews, 2022, Vol. XX, No. XX

women without GDM (281,282). The significance of GDM (29). Following a positive GCT between 24 and 30 + 6 weeks’
as a risk factor for type 2 diabetes and cardiovascular disease gestation, “mild” GDM was defined on a positive 100-g
has been recently recognized by international organizations 3-hour OGTT by a fasting glucose < 5.3 mmol/L (95 mg/dL),
including the American Heart Association (283). and at least 2 postload glucose thresholds that exceeded the
2000 ADA diagnostic thresholds [1-, 2-, or 3-hour thresholds
10.0  mmol/L (180  mg/dL), 8.6  mmol/L (155  mg/dL), and
Management of GDM
7.8 mmol/L (140 mg/dL), respectively]. Women with previous
Benefits of Intervention on Perinatal Outcomes GDM were excluded from the study. Dietary intervention,
Contemporary changes to the detection and management of SMBG, and insulin therapy, if required, to achieve a fasting
GDM have been associated with almost comparable neonatal glucose target < 5.3  mmol/L (95  mg/dL) and 2-hour post-

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


birthweight and adiposity outcomes to the background ma- prandial glucose target < 6.7 mmol/L (121 mg/dL) was asso-
ternity population in developed countries (115). ciated with reduced rates of macrosomia (5.9% vs 14.3%;
The ACHOIS trial (n = 1000) was the first large RCT P < 0.001), LGA (7.1% vs 14.5%; P < 0.001), shoulder dys-
to evaluate whether treatment of women with GDM re- tocia (1.5% vs 4.0%; P = 0.02), cesarean section (26.9% vs
duced the risk of perinatal complications (28). GDM 33.8%; P = 0.02), and preeclampsia and gestational hyper-
was diagnosed based on a combination of fasting glu- tension (8.6% vs 13.6%; P = 0.01) compared to routine care.
cose < 7.8  mmol/L (140  mg/dL) and 2-hour postload glu- However, the intervention did not lead to a significant dif-
cose 7.8 to 11.0  mmol/L (140-199  mg/dL), respectively, ference in the primary composite outcome of stillbirth, peri-
using the 75-g 2-hour OGTT between 24 and 34 weeks’ natal death, and neonatal complications (hyperbilirubinemia,
gestation, following screening with either positive clinical hypoglycemia, hyperinsulinemia, and birth trauma) (29).
risk factors or the GCT (28). ACHOIS demonstrated that a Treatment targets in the MFMU trial were lower than that
combination of dietary advice, self-monitoring of maternal of the ACHOIS trial, and whether this may account for the
glucose levels (SMBG), and insulin therapy, if required, to reduction in cesarean section not shown in the ACHOIS trial
achieve SMBG targets [fasting glucose 3.5-5.5 mmol/L (63- is unclear. These key findings, supported by other studies
99  mg/dL), preprandial glucose ≤ 5.5  mmol/L (99  mg/dL), (22,284), were highlighted by the IADPSG to support the
and 2-hour postprandial glucose ≤ 7.0  mmol/L (126  mg/ lowering of the GDM diagnostic criteria and treating mild
dL)], reduced the rate of serious perinatal complications hyperglycemia (30).
(a composite of death, shoulder dystocia, nerve palsy, and A recent Cochrane review (8 RCTs; n = 1418) reported that
fracture) compared to routine care (1% vs 4%; P = 0.01). GDM treatment, including dietary intervention and insulin
In addition, such interventions were associated with a re- therapy, reduced a composite outcome of perinatal morbidity
duced incidence of macrosomia (10% vs 21%; P < 0.001), (death, shoulder dystocia, bone fracture, and nerve palsy) by
preeclampsia (12% vs 18%; P = 0.02), and improved ma- 68% compared to routine antenatal care (285). Treatment
ternal health-related quality of life (28). was also associated with reductions in macrosomia, LGA,
In 2009, the MFMU trial (n = 958) reported that treatment and preeclampsia but an increase in IOL and neonatal inten-
of “mild” GDM was also associated with improved outcomes sive care admission.

Table 6.  Maternal and neonatal complications of gestational diabetes mellitus

Complications Maternal Neonatal

Short term Preeclampsia Stillbirth


Gestational hypertension Neonatal death
Hydramnios Preterm birth
Urinary tract/vaginal infections Congenital malformations
Instrumental delivery Macrosomia
Cesarean delivery Cardiomyopathy
Traumatic labor/perineal tears Birth trauma:
Postpartum hemorrhage   Shoulder dystocia
Difficulty initiating and/or maintaining breastfeeding   Bone fracture
  Brachial plexus injury
Hypoglycemia
Hyperbilirubinemia
Respiratory distress syndrome
Long term Recurrence of GDM Metabolic syndrome
Type 2 diabetes mellitus Hyperinsulinemia
Hypertension Childhood obesity
Ischemic heart disease Excess abdominal adiposity
Nonalcoholic fatty liver disease Higher blood pressure
Dyslipidemia Possible earlier onset cardiovascular disease
Chronic kidney disease Possible attention-deficit hyperactivity
disorder
Autism spectrum disorder

Sources: Scholtens et al (227) and Saravanan (228).


Abbreviation: GDM, gestational diabetes mellitus.
Endocrine Reviews, 2022, Vol. XX, No. XX 15

Lifestyle Intervention Gestational Weight Gain


The main objective of GDM management is to attain ma- The IOM has published recommendations for weight gain
ternal normoglycemia because evidence suggests that ex- during pregnancy based on prepregnancy BMI (289), but
cessive fetal growth can be attenuated by maintaining near no specific recommendations for weight gain in GDM exist
normal glucose levels (286,287). The foundation of this ap- (286). In women with overweight or obesity, studies have sug-
proach is medical nutrition therapy. Given carbohydrates gested that weight reduction or gain ≤ 5 kg increased the risk
are the primary determinant of maternal postprandial glu- of SGA (308). A recent systematic review based on data from
cose levels, current dietary practice aims to modify carbohy- almost 740 000 women demonstrated that GWG of 5 kg to
drate quality (glycemic index) and distribution (32,288,289). 9 kg in women with class I obesity (BMI 30-34.99 kg/m2), 1
The original nutritional approach for GDM decreased total to <5 kg for class II obesity (35-39.99 kg/m2), and no GWG

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


carbohydrate intake to 33% to 40% of total energy intake for women with class III obesity (BMI ≥ 40kg/m2), minimized
(EI) and was associated with reduced postprandial glycemia the combined risk of LGA, SGA, and cesarean section (309).
and fetal overgrowth (290). More recent evidence suggests A meta-analysis (n = 88 599)  evaluating the relationship
that higher carbohydrate intake and quality (lower glycemic between GWG and pregnancy outcomes in GDM specifically
index) between 60% and 70% EI can also limit maternal showed that GWG greater than the IOM recommendations was
hyperglycemia (291-293). Nevertheless, there remain limited associated with an increased risk of pharmacotherapy, as well as
data to support a specific dietary intervention for GDM (294). of hypertensive disorders of pregnancy, cesarean section, LGA,
A  recent meta-analysis (18 RCTs; n = 1151) showed that and macrosomia (310). GWG below the IOM recommenda-
enhancing nutritional quality (modified dietary intervention, tions was protective for LGA (RR 0.71; 95% CI 0.56-0.90) and
defined as a dietary intervention different from the usual one macrosomia (RR 0.57; 95% CI 0.40-0.83) and did not increase
used in the control group) after GDM diagnosis, irrespective the risk of SGA (RR 1.40; 95% CI 0.86-2.27) (289). This sug-
of the specific dietary approach, improved maternal fasting gests that GWG targets in GDM may need to be lower than
and postprandial glycemia, and reduced pharmacotherapy re- the current recommendations for normal pregnancy. However,
quirements, birthweight, and macrosomia (295). from a practical perspective, only 30% of women gained less
Guidelines therefore currently recommend a range of than the recommended IOM GWG targets (310).
carbohydrate intake between 33% and 55% EI (32,288,289).
Studies have reported improved pregnancy outcomes in GDM
with both lower carbohydrate (42%E) and high‐carbohydrate Maternal Glucose Targets
(55%E) diets (296), reflected in the most recent Academy of Fasting and postprandial glucose testing with either the 1- or
Nutrition and Dietetics guidelines, which state that beneficial 2-hour postprandial glucose value is recommended in women
effects on pregnancy outcomes in GDM are seen with a range with GDM. The 1-hour postprandial glucose approximates to
of carbohydrate intakes (288). The IOM guidelines recom- the peak glucose excursion in pregnancy in women without
mend a carbohydrate intake of at least 175 g/day and a total diabetes and those with type 1 diabetes (175). Studies have
daily caloric intake of 2000 to 2500 kilocalories during preg- shown that the 1-hour postprandial peak glucose level cor-
nancy (289). The ACOG recommends a lower carbohydrate relates with amniotic fluid insulin levels, reflecting fetal
diet (33-40%E) (297). However, the ADA has raised concerns hyperinsulism (311) and with fetal abdominal circumference
over the corresponding higher maternal fat intake, fetal lipid in women with type 1 diabetes (286). An RCT that compared
exposure, and overgrowth resulting from lowering carbohy- pre- to postprandial maternal SMBG values showed that ti-
drate intake (298) and withdrew specific dietary guidelines trating insulin therapy based on the 1-hour postprandial
for GDM in 2005 (299). values was associated with improved maternal glycemic con-
Given maternal glucose primarily supports fetal growth trol and may better attenuate the risk of neonatal complica-
and brain development (300), theoretically if the maternal tions attributed to fetal hyperinsulinemia (312).
diet is too low in carbohydrate, the maternal-fetal glucose Treatment targets based on maternal SMBG levels vary
gradient may be compromised. Restriction of total maternal internationally (Table 7). There is some suggestion that
EI is associated with reduced fetal growth (301). A  recent lower glucose targets may improve pregnancy outcomes in
systematic review similarly showed that lower carbohydrate GDM (176,313,314), but this is yet to be evaluated in ad-
intake correlated with lower birthweight and greater inci- equately powered RCTs. Conversely, lower glycemic targets
dence of SGA (302), with a lower carbohydrate threshold of may be associated with an increased risk of SGA (315-317)
47% EI associated with appropriate fetal growth (302,303). and maternal and fetal hypoglycemia (318,319). A  small
Importantly, the lower carbohydrate threshold independent study evaluating stringent glycemic targets in 180 women
of energy restriction in GDM is yet to be established. with GDM failed to demonstrate additional benefits, with
Related safety concerns with lower carbohydrate diets in- no differences in the rates of cesarean section, birthweight,
clude the potential risk of higher fetal exposure to maternal macrosomia, or SGA in the offspring of women randomized
ketones (304) and micronutrient deficiency (305,306). In to intensive [preprandial glucose ≤ 5.0  mmol/L (90  mg/dL)
vitro studies have shown that ketones suppress trophoblast and 1-hour postprandial glucose ≤ 6.7 mmol/L (121 mg/dL)]
uptake of glucose, jeopardizing glucose transfer across the compared to standard treatment targets [preprandial glu-
placenta (307). Clinically, a prospective US cohort study cose ≤ 5.8  mmol/L (104.5  mg/dL) and 1-hour postprandial
of women with preexisting diabetes, GDM, or normal glu- glucose ≤ 7.8 mmol/L (140 mg/dL)] (320).
cose tolerance demonstrated an inverse correlation between
higher maternal third trimester beta-hydroxybutyrate and
FFAs and lower offspring intellectual development scores at Insulin Therapy
2 to 5 years of age, although total carbohydrate, EI, and ma- Insulin has traditionally been the preferred treatment for
ternal BMI were not reported (304). GDM if maternal glucose levels remain elevated on medical
16 Endocrine Reviews, 2022, Vol. XX, No. XX

Maternal insulin resistance Maternal circulating glucose and free fatty acids

Placental transfer of glucose and free fatty acids to fetus

Fetal circulating insulin and IGF-1 levels

Lung surfactant synthesis & function Fetal substrate uptake Neonatal hypoglycemia

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


Macrosomia

Iatrogenic preterm delivery Shoulder dystocia Hypoxia Cardiomyopathy

Respiratory distress syndrome Brachial plexus injury


Stillbirth Polycythemia

Hyperbilirubinemia

Figure 2.  Perinatal consequences of gestational diabetes mellitus.

nutrition therapy (267). Depending on targets, approxi- thus insulin is generally preferred as first-line pharmaco-
mately 50% of women with GDM are prescribed insulin therapy following lifestyle intervention.
therapy to maintain normoglycemia (321,322), with a com- Glyburide is commonly prescribed as first-line therapy for
bination of evening intermediate-acting insulin if fasting GDM in the United States (328). An early study evaluating
glucose levels are elevated and mealtime rapid-acting insulin the efficacy of glyburide vs insulin therapy in 404 women
when indicated. Additional daytime intermediate-acting in- with GDM reported no differences in maternal glucose levels
sulin may also be needed to control prelunch or predinner or neonatal outcomes between the treatment groups (329).
hyperglycemia. However, subsequent studies show that approximately 20%
Decreasing insulin doses in the third trimester may simply of women treated with glyburide required additional in-
reflect the physiological increase in maternal insulin sensi- sulin therapy to achieve adequate maternal glycemia (330).
tivity observed at this stage of pregnancy (176,323). However, Moreover, a large retrospective US study of almost 111 000
substantial insulin dose reduction, recurrent maternal hypo- women with GDM, in which 4982 women were treated with
glycemia, and/or slowing of fetal growth or preeclampsia may glyburide and 4191 women were treated with insulin, re-
indicate underlying pathophysiological placental insufficiency ported that glyburide was associated with an increased risk of
(324), impacting the timing of delivery and intensity of ob- neonatal complications including neonatal intensive care ad-
stetric monitoring. mission, respiratory distress syndrome, hypoglycemia, birth
Risk factors for insulin therapy include earlier diagnosis of injury, and LGA compared to insulin therapy (331). Although
GDM (81), the pattern and degree of elevation of the 75-g transplacental transfer of glyburide to the fetus is highly vari-
2-hour OGTT diagnostic glucose thresholds (325), and eth- able, it can reach 50% to 70% of maternal plasma concen-
nicity (325). Other risk factors including gestational age tration (332), potentially causing direct stimulation of fetal
and HbA1c level at the time of GDM diagnosis, BMI, and insulin production (333).
family history of diabetes account for only 9% of the at- The use of metformin in pregnancy continues to rise (334).
tributable risk for insulin therapy (321). A recent Australian However, its use remains controversial, due to the poten-
study found that maternal age > 30  years, family history of tial concerns regarding long-term metabolic programming
diabetes, prepregnancy obesity, previous GDM, early diag- effects of placental transfer of metformin to the fetus, with
nosis of GDM, fasting glucose ≥ 5.3 mmol/L (96 mg/dL) and some studies suggesting similar plasma concentrations of
HbA1c ≥ 5.5% (37  mmol/mol) at diagnosis were all inde- metformin in the maternal and fetal circulation (335). A recent
pendent predictors for insulin therapy (326). Insulin usage systematic review and meta-analysis of 28 studies (n = 3976)
could also be estimated according to the number of predictors evaluating growth in offspring of women with GDM exposed
present, with up to 93% of women with 6 to 7 predictors to metformin compared to insulin therapy found that neo-
using insulin therapy compared with less than 15% of women nates exposed to metformin had lower birthweights (mean
with 0 to 1 predictors (326). difference −107.7 g; 95% CI −182.3 to −32.7), decreased risk
of LGA (OR 0.78; 95% CI 0.62-0.99), and macrosomia (OR
0.59; 95% CI 0.46-0.77) and lower ponderal indices than
Oral Pharmacotherapy neonates whose mothers were treated with insulin (336). No
Oral pharmacotherapy options include glyburide and difference in the risk of SGA was found, in contrast to out-
metformin. Oral pharmacotherapy is associated with im- comes in women with type 2 diabetes, with the Metformin
proved cost effectiveness, compliance, and acceptability com- in Women with Type 2 Diabetes RCT observing more than
pared to insulin therapy (327). However, there are issues double the rate of SGA (95% CI 1.16-3.71) in the metformin
regarding efficacy and safety, particularly longer term, and treated cohort, in association with lower insulin doses, HbA1c,
Endocrine Reviews, 2022, Vol. XX, No. XX 17

and GWG (337). Offspring of women with GDM exposed to fetal programming via their effects on cellular metabolism,
metformin also demonstrate accelerated postnatal growth at hepatic gluconeogenesis, and insulin sensitivity (metformin)
18 to 24 months of age (2 studies; n = 411; mean difference (347) and fetal hyperinsulinemia (glyburide) is unknown
in weight 440 g; 95% CI 50-830), resulting in higher BMI at (348).
5 to 9 years of age (3 studies; n = 520; BMI mean difference
0.78 kg/m2, 95% CI 0.23-1.33) (336).
The Metformin in Gestational Diabetes trial randomized Obstetric Management
751 women to receive either metformin or insulin therapy, A recent Cochrane review consisting of only 3 small RCTs
finding no significant difference in the composite neo- (n = 524) reported insufficient (very low certainty) evidence
natal outcome of neonatal hypoglycemia, respiratory dis- to evaluate the use of fetal biometry in guiding the med-

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


tress syndrome, hyperbilirubinemia, low Apgar scores, birth ical management of GDM (349). Nevertheless, serial fetal
trauma, and preterm birth (322). There was a trend toward growth ultrasounds, particularly assessing fetal abdominal
increased preterm birth and decreased maternal GWG in circumference, are potentially useful in guiding the intensity
women treated with metformin, while severe neonatal hypo- of maternal glucose targets and insulin therapy (350-352).
glycemia was highest in those treated with insulin. Almost Studies have demonstrated that neonates with an estimated
50% of women treated with metformin required the addition fetal weight ≥ 75th percentile on early third trimester ultra-
of insulin therapy (322). Other studies have reported that be- sound were 10-fold more likely to be LGA compared to neo-
tween 14.0% and 55.8% of women treated with metformin nates with an estimated fetal weight < 75th percentile (353).
also require insulin therapy to achieve optimal glycemic con- Measured fetal abdominal circumference < 90th percentile on
trol (338,339). The Metformin in Gestational Diabetes: The 2 ultrasounds at 3- to 4-week intervals has also been shown
Offspring Follow-Up 2-year follow-up study found that chil- to provide high reliability in excluding the risk of LGA (351).
dren exposed to metformin had increased subcutaneous fat Moreover, a recent retrospective study (n = 275) found that
localized to the arm compared with children whose mothers estimated fetal weight or abdominal circumference up to the
were treated with insulin alone (340). By 7 and 9  years of 30th percentile on third trimester ultrasound was associated
age the children exposed to metformin had similar offspring with a greater risk of adverse neonatal outcomes, comparable
total and abdominal body fat percentage and metabolic bio- to that observed with abdominal circumference or estimated
chemistry including fasting glucose, insulin, and lipids but fetal weight > 95th percentile in women with hyperglycemia
were larger overall based on measures including weight, arm in pregnancy (including GDM) (354). These findings suggest
and waist circumference, waist-to-height ratio, and dual- the potential utility of fetal biometry at thresholds other than
energy X-ray absorptiometry fat mass and lean mass (341). defining SGA or LGA in identifying higher risk pregnancies
These findings are consistent with a recent follow-up study of in GDM.
metformin therapy in pregnant women with polycystic ovary The optimal timing of delivery in GDM is complex, guided
syndrome, which showed that children exposed to metformin by maternal glycemic control in addition to maternal and fetal
in utero had higher BMI and rates of overweight and obesity factors, and has not been definitively established. Current
at 4 years of age (342). guidelines recommend delivery by 40 + 6 weeks’ gestation
A recent Cochrane review (8 RCTs; n = 1487) evaluating in low-risk women with GDM managed with diet  alone
the use of metformin, glyburide, and acarbose in women and from 39 + 0 to 39 + 6 weeks’ gestation for women with
with GDM found that the benefits and potential harms of GDM well controlled with therapy (38,277,355). A  recent
these therapies in comparison to each other are unclear Canadian population-based cohort study examining the
(343). Other meta-analyses comparing glyburide, metformin, week-specific risks of severe pregnancy complications in
and insulin have shown that metformin was associated with women with diabetes included 138 917 women with GDM
lower GWG, gestational hypertension, and postprandial ma- and 2 553 243 women without diabetes over a 10-year
ternal glucose levels compared to either glyburide or insulin period (356). There was no significant difference in gesta-
(344,345), but metformin was associated with an increased tional age-specific maternal mortality or morbidity (defined
risk of preterm birth compared to insulin (345). Compared as ≥1 of the following in the immediate perinatal period: ob-
to metformin, glyburide was associated with a higher risk of stetric embolism, obstetric shock, postpartum hemorrhage
increased birthweight, LGA, macrosomia, neonatal hypogly- with hysterectomy or other procedures to control bleeding,
cemia, and increased GWG (344). More recently, a small RCT sepsis, thromboembolism, or uterine rupture) between iat-
(n = 104) suggested that glyburide and metformin were com- rogenic delivery and expectant management in women with
parable in terms of maternal glycemia and perinatal outcomes GDM. However, iatrogenic delivery was associated with an
(346). Treatment success after second-line (oral) therapy was increased risk of neonatal mortality and morbidity (birth or
higher in the (first-line) metformin vs glyburide cohort (87% fetal asphyxia, grade 3 or 4 intraventricular hemorrhage,
vs 50%; P = 0.03), suggesting that metformin may be the neonatal convulsions, other disturbances of cerebral status
preferred first-line therapy. Overall, most women required of newborn, respiratory distress syndrome, birth injury,
either a combination of metformin and glyburide to achieve shoulder dystocia, stillbirth or neonatal death) at 36 to 37
glycemic control and/or replacement of first-line oral therapy weeks’ gestation (76.7 and 27.8 excess cases per 1000 de-
due to hypoglycemia and gastrointestinal side effects, sug- liveries, respectively) but a lower risk of neonatal morbidity
gesting neither agent alone is likely to be successful in most and mortality at 38 to 40 weeks’ gestation (7.9, 27.3, and
women with GDM. Combined oral pharmacotherapy had 15.9 fewer cases per 1000 deliveries, respectively) compared
an efficacy rate of 89%, with only 11% of women required with expectant management, suggesting that delivery at 38,
third-line therapy with insulin (346). However, the effects of 39, or 40 weeks’ gestation may provide the best neonatal
dual oral therapy crossing the placenta on long-term potential outcomes in women with GDM (356).
18 Endocrine Reviews, 2022, Vol. XX, No. XX

Longer Term Management of Women Precision Medicine in GDM: Physiological


Following GDM Heterogeneity, Subtype Classification, Risk
Up to one third of women with GDM diagnosed by pre- Prediction, and Biomarker Utility
IADPSG criteria will have glucose levels consistent with dia- Precision medicine seeks to improve diagnostics, prognostics,
betes or prediabetes on postpartum testing at 6 to 12 weeks prediction, and therapeutics in diabetes, including GDM, by
(357). Thus, a repeat OGTT or fasting glucose as early as evaluating and translating various biological axes including
6 to 12 weeks’ postpartum is recommended to confirm ma- metabolomics, genomics, lipidomics, proteomics, technology,
ternal glucose status (41,277). Only around 25% of women clinical risk factors and biomarkers, and mathematical and
are tested at this time point with compliance with postpartum computer modeling into clinical practice (384). The Precision
testing ranging between 23% and 58% (357,358). In women Medicine in Diabetes Initiative was launched in 2018 by the

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


with GDM with overweight or obesity, a reduction in ADA, in partnership with the European Association for the
interpregnancy BMI of ≥2.0  kg/m2 reduces the risk of sub- Study of Diabetes, with their first consensus report published
sequent GDM by 74% (359). Longer term, women should in 2020 (384).
perform regular cardiometabolic health assessment and op- In GDM, precision medicine represents the increasing
timization of lifestyle measures to reduce their greater risk understanding of heterogeneity within its genotype and pheno-
of type 2 diabetes and cardiovascular disease (282,360,361). type (170,385-388) to identify and translate subclassification
Up to 74% of women with obesity and previous GDM de- of GDM into more personalized clinical care (388). For ex-
velop type 2 diabetes compared with <25% of women who ample, physiologic subtypes of GDM based on the underlying
achieve a normal BMI postpartum following GDM (362). It mechanisms leading to maternal hyperglycemia have been
is unclear how relevant these studies in older women are for recently characterized (386). Among 809 women from the
current clinical care given recent data that 50% of women Genetics of Glucose Regulation in Gestation and Growth
develop type 2 diabetes within 5 to 10 years post-GDM diag- pregnancy cohort, heterogeneity in the contribution of insulin
nosis (273). The Diabetes Prevention Program demonstrated resistance and deficiency to GDM were characterized based
that lifestyle intervention and metformin therapy improved on validated indices of insulin sensitivity and secretory re-
insulin sensitivity and preserved β-cell function in women sponse measured during the 75-g OGTT performed between
with a history of previous GDM (363). Early type 2 diabetes 24 and 30 weeks’ gestation (388). Compared to women
prevention following GDM is therefore an essential compo- with normal glucose tolerance, women with insulin resistant
nent of the contemporary GDM detection and management GDM (51% of GDM) had higher BMI and fasting glucose,
paradigm (276). hypertriglyceridemia, and hyperinsulinemia, larger infants,
and almost double the risk of GDM-associated pregnancy
complications. In contrast, women with predominantly in-
Treatment of GDM and Long-term Offspring sulin secretion defects had comparable BMI, fasting glucose,
Outcomes infant birthweight, and risk of adverse outcomes to those
Importantly, despite a reduction in the risk of macrosomia with normal glucose tolerance (388).
at birth, the ACHOIS and MFMU follow-up studies did not Other studies have also suggested that greater insulin
demonstrate a beneficial impact on childhood obesity and resistance in GDM carries a higher risk of perinatal com-
glucose tolerance at 5 to 10 years of age in the offspring of plications (389). A recent multicenter prospective study of
women who received treatment for maternal hyperglycemia 1813 women evaluating subtypes of GDM based on insulin
(364,365). Other prospective cohort studies similarly sug- resistance (389) found that women with GDM and high
gest that the offspring of women with treated GDM still insulin resistance [n = 189 (82.9%)] had a higher BMI,
have a greater risk of obesity, type 2 diabetes, the metabolic systolic blood pressure, fasting glucose, and lipid levels in
syndrome, and cardiovascular disease from early childhood early pregnancy compared to women with normal glucose
and adolescence (258,366-380). For example, a 2017 Danish tolerance or those diagnoses with insulin-sensitive GDM.
National Birth Cohort study (n = 561) reported increased adi- Insulin-sensitive women with GDM [n = 39 (17.1%)] had
posity, an adverse cardiometabolic profile, and earlier onset a significantly lower BMI than women with normal glu-
puberty among adolescent females of women with GDM cose tolerance but similar blood pressure, early pregnancy
(381). A  prospective offspring cohort study of women with fasting glucose and lipid levels, and pregnancy outcomes.
GDM who achieved good antenatal glycemic control dem- Despite no differences in insulin treatment and early
onstrated that offspring adiposity (adipose tissue quantity postpartum glucose intolerance among the GDM sub-
measured using magnetic resonance imaging) was similar types, women with GDM and high insulin resistance had
in the GDM and normal glucose tolerance groups within 2 a greater than 2-fold risk of preterm birth and an almost
weeks postpartum but was 16.0% greater (95% CI 6.0-27.1; 5-fold increased risk of neonatal hypoglycemia compared
P = 0.002) by 2 months of age (382). The mechanism for this with women with normal glucose tolerance. This suggests
greater adiposity and rapid weight gain in early infancy is the high insulin resistance GDM subtype has a greater risk
uncertain given both groups were predominantly breastfed. of pregnancy complications potentially arising from the re-
Consistent with the ACHOIS and MFMU follow-up studies sultant fetal hyperinsulinemia (389).
(364,365), these data suggest that the current approach to The contemporary precision medicine approach to GDM
glycemic control in GDM may not mitigate its impact on also includes the increasing exploration of early pregnancy
longer term infant health. Further, this pathway may be po- risk prediction and risk management models (390). The trad-
tentially mediated by excess infant adiposity, which correlates itional binary clinical risk factor approach to identifying
with childhood adiposity (383). Table 8 presents practical tips women at high risk in early pregnancy is limited by poor sen-
for managing women with GDM. sitivity and specificity, with studies showing that clinical risk
Endocrine Reviews, 2022, Vol. XX, No. XX 19

factor-based screening fails to identify 10% to over 30% of alternative to the OGTT, including measurement of fasting
women with GDM (391-396). The Pregnancy Outcome for plasma glucose, random plasma glucose, and HbA1c (417-
Women with Pre-gestational Diabetes Along the Irish Atlantic 419). A secondary analysis of 5974 women from the HAPO
Seaboard study found that the prevalence of women with study (420), reported that the UK, Canadian, and Australian
GDM who had no risk factors was low, ranging from 2.7% COVID-19–modified diagnostic approaches reduced the
to 5.4% (397). However, despite the absence of risk factors, frequency of GDM by 81%, 82%, and 25%, respectively.
these women with GDM had more pregnancy complications Short-term pregnancy complications in the subgroup of
than those with normal glucose tolerance (397). Other studies women now with undiagnosed GDM (“missed GDM”)
have also reported that women without risk factors diagnosed were comparable to women diagnosed with GDM based
with GDM have comparable pregnancy outcomes to women on the Canadian-modified diagnostic criteria, slightly lower

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


with GDM identified as high risk (393). Thus, clinical risk for the UK-modified criteria, but significantly lower for the
factors alone are not predictive of GDM risk for all women. Australasian Diabetes in Pregnancy Association–modified
Although some improvement in the predictive accuracy for criteria. While all approaches recommend universal testing,
GDM is seen in clinical risk scoring approaches (158,398), the Australian approach adopts a lower fasting glucose
greater improvement via multivariate risk prediction and threshold of 4.7 mmol/L to identify women who require an
mathematical or computer models combining clinical risk OGTT and does not include HbA1c measurement (420).
factors and biomarkers have been reported in the GDM re- A retrospective UK study of over 18 000 women sought to
search setting (154-156,399-403). define evidence-based recommendations for pragmatic GDM
Biomarkers are defined as a biological observation that sub- testing during the COVID-19 pandemic (421), reporting
stitutes and ideally predicts the clinically relevant endpoint that ~5% of women would be identified as GDM based on
(ie, GDM) (404). Biomarker discovery and application in the a random glucose threshold ≥ 8.5  mmol/L (153  mg/dL) at
early detection of GDM has become a major research area. 12 weeks’ gestation and fasting glucose ≥ 5.2 to 5.4 mmol/L
However, few biomarkers are specific enough for clinical ap- (94-97 mg/dL) or HbA1c ≥ 5.7% (39 mmol/mol) measured
plication (405). Most novel biomarkers with potential utility at 28 weeks’ gestation. Each test predicted some, but not all,
for the prediction of GDM are involved in pathophysiological obstetric and perinatal complications, lacking the sensitivity
pathways related to insulin resistance, dyslipidemia, and type of the OGTT for the diagnosis of GDM but overall may
2 diabetes (402,406) but are frequently mediated by maternal provide adequate risk stratification where the OGTT is not
obesity (240,407). Early pregnancy risk prediction models feasible (421).
for GDM combining clinical risk factors and biomarkers
have included various measures of maternal glucose, lipids,
adipokines, inflammatory markers, and pragmatic aneuploidy Conclusion
and preeclampsia screening markers, with model performance GDM is one of the most common complications of pregnancy
(area under the curve) up to 0.91 (153,154,399,402,403,408- and is increasing in global prevalence. Diagnosing GDM is im-
416). Limitations to the clinical application of novel bio- portant because perinatal complications and stillbirth risk are
markers and model performance include heterogeneity in the reduced by treatment. Despite the benefit of identifying and
testing approach to GDM and cohort characteristics, poten- treating GDM, much of the current (short-term) diagnostic
tial overestimation of model performance due to overfitting and management approach to GDM remains contentious.
of the data to the index study population, the lack of external These differences confound interpretation and application
clinical validation studies, and limited regulatory guidance of trial data, preventing a single standard international ap-
for validating biomarker assays (405). proach to GDM.
Recent data indicates near normal birthweight and ma-
ternity population outcomes in women with GDM based
The COVID-19 Pandemic and GDM on modern IADPSG criteria in developed countries,
The COVID-19 pandemic has led to dynamic changes in demonstrating that even treatment of “milder” maternal
the testing approach and model of care for women with hyperglycemia improves pregnancy outcomes. However,
GDM to minimize the risk of virus transmission and be- most cases of GDM occur in low- and middle-income coun-
cause of decreased clinical capacity. Several temporary tries where perinatal risks are far greater and universal 1-step
pragmatic diagnostic strategies have been suggested as an testing may be more practical. There are limited RCT data to

Table 7.  Recommended glycemic treatment targets in GDM

Fasting plasma Preprandial plasma 1-hour post-prandial 2-h post-prandial plasma


glucose (mmol/L) glucose (mmol/L) plasma glucose (mmol/L) glucose (mmol/L)

ADIPS (33) ≤5.0 ≤7.4 ≤6.7


ADA (41) ≤5.3 ≤7.8 ≤6.7
CDA (42)
NICE (38) <5.3 <7.8 <6.4
ACHOIS (37) 3.5-5.5 ≤5.5 ≤7.0
MFMU (38) <5.3 <6.7

Abbreviations: ACHOIS, Australian Carbohydrate Intolerance Study in Pregnant Women Study; ADA, American Diabetes Association; ADIPS, Australasian
Diabetes in Pregnancy Society; CDA, Canadian Diabetes Association; NICE, UK National Institute for Health and Care Excellence; MFMU, National
Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network.
20 Endocrine Reviews, 2022, Vol. XX, No. XX

Table 8.  Practical tips for managing women with GDM

Period Tips

Preconception All women should be encouraged to plan for pregnancy.


Optimize modifiable risk factors prior to pregnancy (eg. BMI, diet, physical activity).
Glucose assessment in high-risk women to detect undiagnosed preexisting glucose intolerance or diabetes.
During All pregnant women should be encouraged to have a nutritionally dense diet and undertake regular exercise during pregnancy
pregnancy unless there are obstetric contraindications.
All pregnant women should be given personalized gestational weight gain targets and have their weight monitored at clinical
reviews.
High-risk women who have not undergone prepregnancy glucose assessment should be tested early for diabetes in pregnancy.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


Test all pregnant women without known diabetes/early GDM for GDM at 24 to 28 weeks’ gestation according to
recommended screening and diagnostic criteria.
GDM management ideally involves a multidisciplinary team with regular diabetes and obstetric assessment and includes patient
education, lifestyle modification and support.
Women should monitor their blood glucose levels. Pharmacotherapy, usually insulin, should be commenced if glucose levels are
elevated despite lifestyle optimization. Metformin can be considered unless there are concerns with inadequate fetal growth.
Timing of delivery is an individualized decision based on maternal and fetal well-being in addition to maternal glycemic control.
Postpartum Early postpartum OGTT to assess glucose status.
Regular long-term follow-up focused on diabetes and vascular risk factor modification and assessment to reduce subsequent
risk of GDM, diabetes, and cardiovascular disease.
Family lifestyle support, which includes optimizing diet, physical activity, and weight in the offspring.

Abbreviations: GDM, gestational diabetes mellitus; OGTT, oral glucose tolerate test.

guide diagnosis and management in this setting, and further Other important areas for research include the evaluation
evidence is urgently needed. In developed countries including of dietary interventions establishing the optimal carbohydrate
the United Kingdom, the main issue arguably does not per- threshold in GDM, further clarity on the potential long-term
tain to women diagnosed with GDM but rather high-risk impact of intrauterine metformin on the offspring, as well as
women who remain unscreened (associated with factors such the efficacy of preconception and early pregnancy preventive
as lower socioeconomic status and higher BMI) who are at strategies targeting risk factors other than glycemia, such as
highest risk of stillbirth (74). maternal obesity and GWG. Improved obstetric assessment
The background to the various GDM diagnostic criteria is of placental function, especially in late pregnancy, to inform
informative in demonstrating that no approach clearly sep- timing of delivery and identify women at highest risk of still-
arates risk groups. It is also now evident that a continuum birth in GDM is also needed.
of risk for GDM exists based on both the timing and degree The complications of GDM may indeed be greater based
of maternal hyperglycemia. This underscores the difficulty of on the severity of maternal glycemia and associated vascular
defining absolute glucose thresholds at a single timepoint in risk factors. Nevertheless, the traditional focus on diagnostic
pregnancy for the diagnosis of GDM and is confounded fur- criteria and short-term antenatal maternal glucose manage-
ther by variation in glucose measurement due to preanalytical ment fails to address the importance of identifying “milder”
glucose processing and reproducibility issues. Thus, current (IADPSG-defined) GDM as a risk factor for future maternal
diagnostic glucose thresholds for GDM must inevitably re- and offspring diabetes and CVD risk. It should also be ap-
flect compromise and consensus. parent that the increasing prevalence of GDM largely reflects
A precision medicine approach that recognizes GDM sub- the worsening metabolic health burden including prediabetes
type and heterogeneity, enhanced by further research into the and obesity in women of childbearing age. The clinical focus
genetics of GDM and validation of novel biomarkers and new for GDM must therefore urgently shift to early postnatal pre-
technologies such as continuous glucose monitoring may im- vention strategies to decrease the progression from GDM to
prove risk stratification, optimize clinical models of care, and type 2 diabetes and address longer term maternal and off-
facilitate more individualized and consumer-friendly detec- spring cardiometabolic risk post-GDM via a life course man-
tion and treatment strategies. agement approach.
The recent HAPO-FUS data confirming the long-term im-
pact of maternal hyperglycemia on maternal and offspring
metabolic health (227,262) highlight an important paradigm Financial Support
shift. The approach to GDM should reflect an evidence base A.S.  was supported by an NHMRC Fellowship Grant
that evaluates diagnostic glucose thresholds and measure- (GNT1148952).
ment within a framework that includes timing of detection
and treatment trials with long-term clinical and health eco-
nomic outcomes. For example, if the ongoing Treatment of Disclosure Summary
Booking Gestational Diabetes Mellitus trial demonstrates a A.S., J.W., H.M., and G.P.R. have nothing to declare.
benefit for early GDM detection and treatment, there are im-
plications for the prevailing diagnostic GDM glucose thresh-
olds in later pregnancy. This is because these thresholds were References
derived from the risk of perinatal complications in a heteroge- 1. Bennewitz H. De Diabete Mellito, Gravidatatis Symptomate. MD
neous GDM cohort, which included women who would fulfill thesis, University of Berlin; 1824.
early GDM criteria. 2. Barker DJP. Mothers Babies and Diseases in Later Life. BMJ; 1994.
Endocrine Reviews, 2022, Vol. XX, No. XX 21

 3. Duncan  J. On puerperal diabetes. Trans Obstet Soc Lond. 26. American Diabetes Association. Gestational diabetes mellitus.
1882;24:256-285. Diabetes Care. 2003;26(suppl 1):S103-S105.
  4. Miller HC. The effect of diabetic and prediabetic pregnancies on 27. Metzger BE, Lowe LP, Dyer AR, et al; HAPO Study Cooperative
the fetus and newborn infant. J Pediatr. 1946;29(4):455-461. Group. Hyperglycemia and adverse pregnancy outcomes. N Engl J
 5. Williams  J. The clinical significance of glycosuria in pregnant Med. 2008;358(19):1991-2002.
women. Am J Med Sci. 1909;137:1-26. 28. Crowther CA, Hiller JE, Moss JR, et al. Effect of treatment of ges-
  6. O’Sullivan JB, Mahan CM. Criteria for the oral glucose tolerance tational diabetes mellitus on pregnancy outcomes. N Engl J Med.
test in pregnancy. Diabetes. 1964;13:278-285. 2005;352(24):2477-2486.
 7. Coustan  DR. Gestational diabetes. In: Harris  MI, Cowie  CC, 29. Landon MB, Spong CY, Thom E, et al. A multicenter, randomized
Stern  MP, Boyko  EJ, Reiber  GE, Bennett  PH, eds. Diabetes in trial of treatment for mild gestational diabetes. N Engl J Med.
America. National Institute of Health; 1995:703-717. 2009;361(14):1339-1348.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


 8. World Health Organization. Technical Report Series. No 310. 30. Metzger BE, Gabbe SG, Persson B, et al; International Association
Diabetes Mellitus. Report of a WHO Expert Committee. 1965. of Diabetes and Pregnancy Study Groups Consensus Panel.
 9. World Health Organization. Technical Report Series. No 646. International Association of Diabetes and Pregnancy Study Groups
Second Report on Diabetes Mellitus. Report of a WHO Expert recommendations on the diagnosis and classification of hypergly-
Committee. 1980. cemia in pregnancy. Diabetes Care. 2010;33(3):676-682.
10. World Health Organization. Technical Report Series. No 727. 31. Blumer  I, Hadar  E, Hadden  DR, et  al. Diabetes and pregnancy:
Second Report on Diabetes Mellitus. Report of a WHO Expert an Endocrine Society clinical practice guideline. J Clin Endocrinol
Committee. 1985. Metab. 2013;98(11):4227-4249.
11. World Health Organisation. Diagnostic criteria and classifi- 32. Hod M, Kapur A, Sacks DA, et al. The International Federation of
cation of hyperglycaemia first detected in pregnancy: a World Gynecology and Obstetrics (FIGO) Initiative on gestational dia-
Health Organization guideline. Diabetes Res Clin Pract. betes mellitus: a pragmatic guide for diagnosis, management, and
2014;103(3):341-363. care. Int J Gynaecol Obstet. 2015;131(suppl 3):S173-S211.
12. O’Sullivan  JB, Mahan  CM, Charles  D, Dandrow  RV. Screening 33. Nankervis  A, McIntyre  HD, Moses  R, et  al. ADIPS Consensus
criteria for high-risk gestational diabetic patients. Am J Obstet Guidelines for the Testing and Diagnosis of Hyperglycaemia in
Gynecol. 1973;116(7):895-900. Pregnancy in Australia and New Zealand. Modified November 2014.
13. National Diabetes Data Group. Classification and diagnosis of dia- http://adips.org/downloads/2014ADIPSGDMGuidelinesV18.11.
betes mellitus and other categories of glucose intolerance. Diabetes. 2014_000.pdf. Accessed June 1, 2021.
1979;28(12):1039-1057. 34. Japan Diabetes Society. Evidence-based practice guideline for the
14. American College of Obstetricians and Gynecologists. Management treatment for diabetes in Japan 2013. Last updated November 26,
of Diabetes Mellitus During Pregnancy. Technical Bulletin No. 92. 2020. http://www.jds.or.jp/modules/en/index.php?content_id=44.
1986. Accessed June 1, 2021.
15. American Diabetes Association. Gestational diabetes mellitus. 35. Yang  HX. Diagnostic criteria for gestational diabetes mellitus.
Diabetes Care. 1986;9(4):430-431. Chin Med J. 2012;125(7):1212-1213.
16. Ferrara A, Hedderson MM, Quesenberry CP, Selby JV. Prevalence 36. Benhalima K, Mathieu C, Damm P, et al. A proposal for the use of
of gestational diabetes mellitus detected by the National Diabetes uniform diagnostic criteria for gestational diabetes in Europe: an
Data Group or the Carpenter and Coustan plasma glucose opinion paper by the European Board & College of Obstetrics and
thresholds. Diabetes Care. 2002;25(9):1625-1630. Gynaecology (EBCOG). Diabetologia. 2015;58(7):1422-1429.
17. Carpenter MW, Coustan DR. Criteria for screening tests for gesta- 37. Vandorsten  JP, Dodson  WC, Espeland  MA, et  al. NIH consensus
tional diabetes. Am J Obstet Gynecol. 1982;144(7):768-773. development conference: diagnosing gestational diabetes mellitus.
18. American Diabetes Association. 2. Classification and diagnosis NIH Consens State Sci Statements. 2013;29(1):1-31.
of diabetes: standards of medical care in diabetes-2018. Diabetes 38. National Institute for Health and Care Excellence. Diabetes in Pregnancy:
Care. 2018;41(suppl 1):S13-S27. Management of Diabetes and its Complications from Pre-conception to
19. American College of Obstetricians and Gynecologists. Practice the Postnatal Period. NICE Clinical Guideline NG3. 2015.
Bulletin No. 190: gestational diabetes mellitus. Obstet Gynecol. 39. Vambergue  A. Expert consensus on gestational diabetes mellitus.
2018;131(2):e49-e64. Diabetes Metab. 2010;36(6 Pt 2):511.
20. American Diabetes Association. Gestational diabetes mellitus. 40. Associazione Medici Diabetologi and Società Italiana di
Diabetes Care. 2000;23(suppl 1):S77-S79. Diabetologia. Italian National Health System Guidelines for
21. Sermer  M, Naylor  CD, Farine  D, et  al. The Toronto tri-hospital the screening of gestational diabetes mellitus. May 28, 2014.
gestational diabetes project: a preliminary review. Diabetes Care. http://www.standarditaliani.it/skin/www.standarditaliani.it/pdf/
1998;21(Suppl 2):B33-B42. STANDARD_2014_May28.pdf. Accessed June 1, 2021.
22. Sermer  M, Naylor  CD, Gare  DJ, et  al. Impact of increasing 41. American Diabetes Association. 14. Management of diabetes in
carbohydrate intolerance on maternal-fetal outcomes in pregnancy: standards of medical care in diabetes-2020. Diabetes
3637 women without gestational diabetes: the Toronto Tri- Care. 2020;43(suppl 1):S183-SS92.
Hospital Gestational Diabetes Project. Am J Obstet Gynecol. 42. Canadian Diabetes Association Clinical Practice Guidelines
1995;173(1):146-156. Expert Committee. Clinical practice guidelines for the prevention
23. Berggren EK, Boggess KA, Stuebe AM, Jonsson Funk M. National and management of diabetes in Canada. Canadian J Diabetes.
Diabetes Data Group vs Carpenter-Coustan criteria to diagnose 2013;37(suppl 1):S1-S3.
gestational diabetes. Am J Obstet Gynecol. 2011;205(3):253 e1- 43. Kleinwechter H, Schafer-Graf U, Buhrer C, et al. Gestational dia-
253.e7. betes mellitus (GDM) diagnosis, therapy and follow-up care: prac-
24. Cheng  YW, Block-Kurbisch  I, Caughey  AB. Carpenter-Coustan tice Guideline of the German Diabetes Association (DDG) and the
criteria compared with the National Diabetes Data Group German Association for Gynaecology and Obstetrics (DGGG).
thresholds for gestational diabetes mellitus. Obstet Gynecol. Exp Clin Endocrinol Diabetes. 2014;122(7):395-405.
2009;114(2 Pt 1):326-332. 44. Seshiah V, Das AK, Balaji V, et al. Gestational diabetes mellitus—
25. Chou  CY, Lin  CL, Yang  CK, et  al. Pregnancy outcomes of guidelines. J Assoc Physicians India. 2006;54:622-628.
Taiwanese women with gestational diabetes mellitus: a comparison 45. World Health Organization. Definition, Diagnosis and
of Carpenter-Coustan and National Diabetes Data Group criteria. Classification of Diabetes Mellitus and its Complications. Report
J Womens Health (Larchmt). 2010;19(5):935-939. of WHO Consultation. 1999.
22 Endocrine Reviews, 2022, Vol. XX, No. XX

46. Coustan  DR, Lowe  LP, Metzger  BE, et  al; on behalf of the 64. Hillier  TA, Pedula  KL, Ogasawara  KK, et  al. A pragmatic,
International Association of Diabetes in Pregnancy Study Groups. randomized clinical trial of gestational diabetes screening. N Engl J
The Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Med. 2021;384(10):895-904.
study: paving the way for new diagnostic criteria for gestational 65. Casey  B. Gestational diabetes—on broadening the diagnosis. N
diabetes mellitus. Am J Obstet Gynecol. 2010;202(6):654 e1-654 Engl J Med. 2021;384(10):965-966.
e6. 66. Dunne F, Lindsay R, Loeken M. This is the decade to find the so-
47. McIntyre  HD, Oats  JJ, Kihara  AB, et  al. Update on diagnosis of lution for gestational diabetes mellitus screening and treatments.
hyperglycemia in pregnancy and gestational diabetes mellitus from Diabet Med. 2021;38(8):e14602.
FIGO’s Pregnancy & Non-Communicable Diseases Committee. Int 67. O’Malley EG, Reynolds CME, O’Kelly R, et al. The diagnosis of
J Gynaecol Obstet. 2021;154(2):189-194. gestational diabetes mellitus (GDM) using a 75 g oral glucose tol-
48. Menke  A, Casagrande  S, Geiss  L, Cowie  CC. Prevalence of and erance test: a prospective observational study. Diabetes Res Clin

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


trends in diabetes among adults in the United States, 1988-2012. Pract. 2020;163:108144.
JAMA. 2015;314(10):1021-1029. 68. van Leeuwen M, Louwerse MD, Opmeer BC, et al. Glucose chal-
49. Centers for Disease Control and Prevention. National Diabetes lenge test for detecting gestational diabetes mellitus: a systematic
Statistics Report, 2020. US Department of Health and Human review. BJOG. 2012;119(4):393-401.
Services; 2020. 69. Sacks  DA, Hadden  DR, Maresh  M, et  al. Frequency of gesta-
50. Lieberman  N, Kalter-Leibovici  O, Hod  M. Global adaptation of tional diabetes mellitus at collaborating centers based on IADPSG
IADPSG recommendations: a national approach. Int J Gynaecol consensus panel-recommended criteria: the Hyperglycemia and
Obstet. 2011;115(suppl 1):S45-S47. Adverse Pregnancy Outcome (HAPO) study. Diabetes Care.
51. Marseille E, Lohse N, Jiwani A, et al. The cost-effectiveness of ges- 2012;35(3):526-528.
tational diabetes screening including prevention of type 2 diabetes: 70. Roeckner  JT, Sanchez-Ramos  L, Jijon-Knupp  R, et  al. Single ab-
application of a new model in India and Israel. J Matern Fetal normal value on 3-hour oral glucose tolerance test during preg-
Neonatal Med. 2013;26(8):802-810. nancy is associated with adverse maternal and neonatal outcomes:
52. Werner  EF, Pettker  CM, Zuckerwise  L, et  al. Screening for ges- a systematic review and metaanalysis. Am J Obstet Gynecol.
tational diabetes mellitus: are the criteria proposed by the 2016;215(3):287-297.
International Association of the Diabetes and Pregnancy Study 71. Freinkel N, Metzger BE, Phelps RL, et al. Gestational diabetes mel-
Groups cost-effective? Diabetes Care. 2012;35(3):529-535. litus: heterogeneity of maternal age, weight, insulin secretion, HLA
53. Mission JF, Ohno MS, Cheng YW, Caughey AB. Gestational dia- antigens, and islet cell antibodies and the impact of maternal me-
betes screening with the new IADPSG guidelines: a cost-effective- tabolism on pancreatic B-cell and somatic development in the off-
ness analysis. Am J Obstet Gynecol. 2012;207(4):326 e1-326 e9. spring. Diabetes. 1985;34(suppl 2):1-7.
54. Farrar  D, Simmonds  M, Griffin  S, et  al. The identification and 72. Schaefer UM, Songster G, Xiang A, et al. Congenital malformations
treatment of women with hyperglycaemia in pregnancy: an anal- in offspring of women with hyperglycemia first detected during
ysis of individual participant data, systematic reviews, meta- pregnancy. Am J Obstet Gynecol. 1997;177(5):1165-1171.
analyses and an economic evaluation. Health Technol Assess. 73. Omori  Y, Jovanovic  L. Proposal for the reconsideration
2016;20(86):1-348. of the definition of gestational diabetes. Diabetes Care.
55. Jacklin  PB, Maresh  MJ, Patterson  CC, et  al. A cost-effectiveness 2005;28(10):2592-2593.
comparison of the NICE 2015 and WHO 2013 diagnostic criteria 74. Stacey T, Tennant P, McCowan L, et al. Gestational diabetes and
for women with gestational diabetes with and without risk factors. the risk of late stillbirth: a case-control study from England, UK.
BMJ Open. 2017;7(8):e016621. BJOG. 2019;126(8):973-982.
56. Cundy  T, Ackermann  E, Ryan  EA. Gestational diabetes: new 75. Cosson E, Bentounes SA, Nachtergaele C, et al. Prognosis associ-
criteria may triple the prevalence but effect on outcomes is unclear. ated with sub-types of hyperglycaemia in pregnancy. J Clin Med.
BMJ. 2014;348:g1567. 2021;10(17):3904.
57. O’Sullivan  EP, Avalos  G, O’Reilly  M, et  al. Atlantic Diabetes in 7 6. Wong  T, Ross  GP, Jalaludin  BB, Flack  JR. The clinical sig-
Pregnancy (DIP): the prevalence and outcomes of gestational nificance of overt diabetes in pregnancy. Diabet Med.
diabetes mellitus using new diagnostic criteria. Diabetologia. 2013;30(4):468-474.
2011;54(7):1670-1675. 77. Bartha JL, Martinez-Del-Fresno P, Comino-Delgado R. Early diag-
58. Lapolla  A, Dalfra  MG, Ragazzi  E, et  al. New International nosis of gestational diabetes mellitus and prevention of diabetes-
Association of the Diabetes and Pregnancy Study Groups (IADPSG) related complications. Eur J Obstet Gynecol Reprod Biol.
recommendations for diagnosing gestational diabetes compared 2003;109(1):41-44.
with former criteria: a retrospective study on pregnancy outcome. 78. Bartha JL, Martinez-Del-Fresno P, Comino-Delgado R. Gestational
Diabet Med. 2011;28(9):1074-1077. diabetes mellitus diagnosed during early pregnancy. Am J Obstet
59. Benhalima K, Hanssens M, Devlieger R, et al. Analysis of pregnancy Gynecol. 2000;182(2):346-350.
outcomes using the new IADPSG recommendation compared with 79. Berkowitz GS, Roman SH, Lapinski RH, et al. Maternal character-
the Carpenter and Coustan criteria in an area with a low preva- istics, neonatal outcome, and the time of diagnosis of gestational
lence of gestational diabetes. Int J Endocrinol. 2013;2013:248121. diabetes. Am J Obstet Gynecol. 1992;167(4 Pt 1):976-8282.
60. Hung TH, Hsieh TT. The effects of implementing the International 80. Meyer  WJ, Carbone  J, Gauthier  DW, et  al. Early gestational
Association of Diabetes and Pregnancy Study Groups criteria glucose screening and gestational diabetes. J Reprod Med.
for diagnosing gestational diabetes on maternal and neonatal 1996;41(9):675-679.
outcomes. PLoS One. 2015;10(3):e0122261. 81. Sweeting AN, Ross GP, Hyett J, et al. Gestational diabetes mellitus
61. Meek  CL, Lewis  HB, Patient  C, et  al. Diagnosis of gesta- in early pregnancy: evidence for poor pregnancy outcomes despite
tional diabetes mellitus: falling through the net. Diabetologia. treatment. Diabetes Care. 2016;39(1):75-81.
2015;58(9):2003-2012. 82. Li  M, Hinkle  SN, Grantz  KL, et  al. Glycaemic status during
62. Djelmis J, Pavic M, Mulliqi Kotori V, et al. Prevalence of gestational pregnancy and longitudinal measures of fetal growth in a multi-
diabetes mellitus according to IADPSG and NICE criteria. Int J racial US population: a prospective cohort study. Lancet Diabetes
Gynaecol Obstet. 2016;135(3):250-254. Endocrinol. 2020;8(4):292-300.
63. Ethridge JK Jr, Catalano PM, Waters TP. Perinatal outcomes asso- 83. Sovio  U, Murphy  HR, Smith  GC. Accelerated fetal growth
ciated with the diagnosis of gestational diabetes made by the inter- prior to diagnosis of gestational diabetes mellitus: a pro-
national association of the diabetes and pregnancy study groups spective cohort study of nulliparous women. Diabetes Care.
criteria. Obstet Gynecol. 2014;124(3):571-578. 2016;39(6):982-987.
Endocrine Reviews, 2022, Vol. XX, No. XX 23

  84. Venkataraman H, Ram U, Craik S, et al. Increased fetal adiposity 104. Harper  LM, Jauk  V, Longo  S, et  al. Early gestational diabetes
prior to diagnosis of gestational diabetes in South Asians: more ev- screening in obese women: a randomized controlled trial. Am J
idence for the “thin-fat” baby. Diabetologia. 2017;60(3):399-405. Obstet Gynecol. 2020;222(5):495 e1-495 e8.
  85. Riskin-Mashiah  S, Damti  A, Younes  G, et  al. Normal fasting 105. Vinter CA, Tanvig MH, Christensen MH, et al. Lifestyle interven-
plasma glucose levels during pregnancy: a hospital-based study. J tion in Danish obese pregnant women with early gestational dia-
Perinat Med. 2011;39(2):209-211. betes mellitus according to WHO 2013 criteria does not change
 86. Mills  JL, Jovanovic  L, Knopp  R, et  al. Physiological reduction pregnancy outcomes: results from the LiP (Lifestyle in Pregnancy)
in fasting plasma glucose concentration in the first trimester study. Diabetes Care. 2018;41(10):2079-2085.
of normal pregnancy: the diabetes in early pregnancy study. 106. Simmons  D, Hague  WM, Teede  HJ, et  al. Hyperglycaemia in
Metabolism. 1998;47(9):1140-1144. early pregnancy: the Treatment of Booking Gestational Diabetes
 87. Corrado  F, D’Anna  R, Cannata  ML, et  al. Correspondence be- Mellitus (TOBOGM) study: a randomised controlled trial. Med J

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


tween first-trimester fasting glycaemia, and oral glucose tol- Aust. 2018;209(9):405-406.
erance test in gestational diabetes diagnosis. Diabetes Metab. 107. Bruns  DE, Metzger  BE, Sacks  DB. Diagnosis of gestational dia-
2012;38(5):458-61. betes mellitus will be flawed until we can measure glucose. Clin
 88. Zhu  WW, Yang  HX, Wei  YM, et  al. Evaluation of the value Chem. 2020;66(2):265-267.
of fasting plasma glucose in the first prenatal visit to diag- 108. Bogdanet D, O’Shea P, Lyons C, et al. The oral glucose tolerance
nose gestational diabetes mellitus in china. Diabetes Care. test-is it time for a change?-a literature review with an emphasis
2013;36(3):586-590. on pregnancy. J Clin Med. 2020;9(11):3451.
  89. McIntyre  HD, Sacks  DA, Barbour  LA, et  al. Issues with the di- 109. Sacks  DB, Arnold  M, Bakris  GL, et  al. Position statement exec-
agnosis and classification of hyperglycemia in early pregnancy. utive summary: guidelines and recommendations for laboratory
Diabetes Care. 2016;39(1):53-54. analysis in the diagnosis and management of diabetes mellitus.
 90. Hughes  RC, Moore  MP, Gullam  JE, et  al. An early pregnancy Diabetes Care. 2011;34(6):1419-1423.
HbA1c ≤5.9% (41  mmol/mol) is optimal for detecting diabetes 110. Chan  AY, Swaminathan  R, Cockram  CS. Effectiveness of so-
and identifies women at increased risk of adverse pregnancy dium fluoride as a preservative of glucose in blood. Clin Chem.
outcomes. Diabetes Care. 2014;37(11):2953-2959. 1989;35(2):315-317.
  91. Sweeting AN, Ross GP, Hyett J, et al. Baseline HbA1c to identify 111. Potter  JM, Hickman  PE, Oakman  C, et  al. Strict preanalytical
high-risk gestational diabetes: utility in early vs standard gesta- oral glucose tolerance test blood sample handling is essen-
tional diabetes. J Clin Endocrinol Metab. 2017;102(1):150-156. tial for diagnosing gestational diabetes mellitus. Diabetes Care.
  92. Immanuel  J, Simmons  D, Desoye  G, et  al. Performance of early 2020;43(7):1438-1441.
pregnancy HbA1c for predicting gestational diabetes mellitus and 112. Daly  N, Flynn  I, Carroll  C, et  al. Impact of implementing
adverse pregnancy outcomes in obese European women. Diabetes preanalytical laboratory standards on the diagnosis of gestational
Res Clin Pract. 2020;168:108378. diabetes mellitus: a prospective observational study. Clin Chem.
  93. Osmundson SS, Norton ME, El-Sayed YY, et al. Early screening 2016;62(2):387-391.
and treatment of women with prediabetes: a randomized 113. International Diabetes Federation. IDF Diabetes Atlas. 9th ed.
controlled trial. Am J Perinatol. 2016;33(2):172-179. 2019.
  94. Roeder HA, Moore TR, Wolfson T, Ramos GA. Treating hyper- 114. Guariguata L, Linnenkamp U, Beagley J, et al. Global estimates of
glycemia in the first trimester: a randomized controlled trial. Am the prevalence of hyperglycaemia in pregnancy. Diabetes Res Clin
J Obstet Gynecol MFM. 2017;1(1):33-41. Pract. 2014;103(2):176-185.
  95. Hawkins JS, Lo JY, Casey BM, et al. Diet-treated gestational di- 115. Prentice  PM, Olga  L, Petry  CJ, et  al. Reduced size at birth and
abetes mellitus: comparison of early vs routine diagnosis. Am J persisting reductions in adiposity in recent, compared with earlier,
Obstet Gynecol. 2008;198(3):287 e1-287 e6. cohorts of infants born to mothers with gestational diabetes mel-
  96. Most  OL, Kim  JH, Arslan  AA, et  al. Maternal and neonatal litus. Diabetologia. 2019;62(11):1977-1987.
outcomes in early glucose tolerance testing in an obstetric popula- 116. Hartling  L, Dryden  DM. Screening and diagnosing gesta-
tion in New York city. J Perinat Med. 2009;37(2):114-117. tional diabetes mellitus. Evid Rep Technol Assess (Full Rep).
  97. Gupta S, Dolin C, Jadhav A, et al. Obstetrical outcomes in patients 2012;210:1-327.
with early onset gestational diabetes. J Matern Fetal Neonatal 117. Bottalico  JN. Recurrent gestational diabetes: risk factors, di-
Med. 2016;29(1):27-31. agnosis, management, and implications. Semin Perinatol.
 98. Harreiter  J, Simmons  D, Desoye  G, et  al. IADPSG and WHO 2007;31(3):176-184.
2013 gestational diabetes mellitus criteria identify obese women 118. Anna V, van der Ploeg H, P, Cheung NW, et al. Sociodemographic
with marked insulin resistance in early pregnancy. Diabetes Care. correlates of the increasing trend in prevalence of gestational dia-
2016;39(7):e90-e92. betes mellitus in a large population of women between 1995 and
  99. Egan  AM, Vellinga  A, Harreiter  J, et  al. Epidemiology of ges- 2005. Diabetes Care. 2008;31(12):2288-2293.
tational diabetes mellitus according to IADPSG/WHO 2013 119. Chu  SY, Callaghan  WM, Kim  SY, et  al. Maternal obe-
criteria among obese pregnant women in Europe. Diabetologia. sity and risk of gestational diabetes mellitus. Diabetes Care.
2017;60(10):1913-1921. 2007;30(8):2070-2076.
100. Immanuel J, Simmons D. Screening and treatment for early-onset 120. Torloni MR, Betran AP, Horta BL, et al. Prepregnancy BMI and
gestational diabetes mellitus: a systematic review and meta- the risk of gestational diabetes: a systematic review of the litera-
analysis. Curr Diab Rep. 2017;17(11):115. ture with meta-analysis. Obes Rev. 2009;10(2):194-203.
101. Bozkurt L, Gobl CS, Pfligl L, et al. Pathophysiological character- 121. Mokdad AH, Ford ES, Bowman BA, et al. Prevalence of obesity,
istics and effects of obesity in women with early and late mani- diabetes, and obesity-related health risk factors, 2001. JAMA.
festation of gestational diabetes diagnosed by the International 2003;289(1):76-79.
Association of Diabetes and Pregnancy Study Groups criteria. J 122. Mulla  WR, Henry  TQ, Homko  CJ. Gestational diabetes
Clin Endocrinol Metab. 2015;100(3):1113-1120. screening after HAPO: has anything changed? Curr Diab Rep.
102. Bozkurt  L, Gobl  CS, Hormayer  AT, et  al. The impact of 2010;10(3):224-228.
preconceptional obesity on trajectories of maternal lipids during 123. Petry CJ. Gestational diabetes: risk factors and recent advances in
gestation. Sci Rep. 2016;6:29971. its genetics and treatment. Br J Nutr. 2010;104(6):775-787.
103. Sweeting  AN, Ross  GP, Hyett  J, et  al. Gestational diabetes in 124. Hunt  KJ, Schuller  KL. The increasing prevalence of diabetes in
the first trimester: is early testing justified? Lancet Diabetes pregnancy. Obstet Gynecol Clin North Am. 2007;34(2):173-199,
Endocrinol. 2017;5(8):571-573. vii.
24 Endocrine Reviews, 2022, Vol. XX, No. XX

125. Agarwal  MM, Dhatt  GS, Shah  SM. Gestational diabetes mel- 147. Chasan-Taber  L, Schmidt  MD, Pekow  P, et  al. Physical activity
litus: simplifying the international association of diabetes and and gestational diabetes mellitus among Hispanic women. J
pregnancy diagnostic algorithm using fasting plasma glucose. Womens Health (Larchmt). 2008;17(6):999-1008.
Diabetes Care. 2010;33(9):2018-2020. 148. Mottola  MF. The role of exercise in the prevention and
126. Moses RG, Morris GJ, Petocz P, et al. The impact of potential new treatment of gestational diabetes mellitus. Curr Diab Rep.
diagnostic criteria on the prevalence of gestational diabetes mel- 2008;8(4):299-304.
litus in Australia. Med J Aust. 2011;194(7):338-340. 149. Zhang  C, Liu  S, Solomon  CG, et  al. Dietary fiber intake, die-
127. Brown  FM, Wyckoff  J. Application of one-step IADPSG versus tary glycemic load, and the risk for gestational diabetes mellitus.
two-step diagnostic criteria for gestational diabetes in the real Diabetes Care. 2006;29(10):2223-2230.
world: impact on health services, clinical care, and outcomes. 150. Kucukgoncu S, Guloksuz S, Celik K, et al. Antipsychotic exposure
Curr Diab Rep. 2017;17(10):85. in pregnancy and the risk of gestational diabetes: a systematic re-

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


128. Kim  C, Berger  DK, Chamany  S. Recurrence of gesta- view and meta-analysis. Schizophr Bull. 2020;46(2):311-318.
tional diabetes mellitus: a systematic review. Diabetes Care. 151. Galbally M, Frayne J, Watson SJ, et al. The association between
2007;30(5):1314-1319. gestational diabetes mellitus, antipsychotics and severe mental
129. Solomon CG, Willett WC, Carey VJ, et al. A prospective study of illness in pregnancy: a multicentre study. Aust N Z J Obstet
pregravid determinants of gestational diabetes mellitus. JAMA. Gynaecol. 2020;60(1):63-69.
1997;278(13):1078-1083. 152. Hedderson MM, Ferrara A. High blood pressure before and during
130. Chamberlain  C, McNamara  B, Williams  ED, et  al. Diabetes in early pregnancy is associated with an increased risk of gestational
pregnancy among indigenous women in Australia, Canada, diabetes mellitus. Diabetes Care. 2008;31(12):2362-2367.
New Zealand and the United States. Diabetes Metab Res Rev. 153. Lao  TT, Ho  LF. First-trimester blood pressure and gestational
2013;29(4):241-256. diabetes in high-risk Chinese women. J Soc Gynecol Investig.
131. Hedderson  MM, Gunderson  EP, Ferrara  A. Gestational weight 2003;10(2):94-98.
gain and risk of gestational diabetes mellitus. Obstet Gynecol. 154. Sweeting  AN, Appelblom  H, Ross  GP, et  al. First trimester pre-
2010;115(3):597-604. diction of gestational diabetes mellitus: a clinical model based
132. Kjos  SL, Buchanan  TA. Gestational diabetes mellitus. N Engl J on maternal demographic parameters. Diabetes Res Clin Pract.
Med. 1999;341(23):1749-1756. 2017;127:44-50.
133. Di Cianni G, Volpe L, Lencioni C, et al. Prevalence and risk factors 155. Syngelaki  A, Visser  GH, Krithinakis  K, et  al. First trimester
for gestational diabetes assessed by universal screening. Diabetes screening for gestational diabetes mellitus by maternal factors and
Res Clin Pract. 2003;62(2):131-137. markers of inflammation. Metabolism. 2016;65(3):131-137.
134. Cypryk  K, Szymczak  W, Czupryniak  L, et  al. Gestational di- 156. Nanda S, Savvidou M, Syngelaki A, et al. Prediction of gestational
abetes mellitus—an analysis of risk factors. Endokrynol Pol. diabetes mellitus by maternal factors and biomarkers at 11 to 13
2008;59(5):393-397. weeks. Prenat Diagn. 2011;31(2):135-141.
135. Cleary-Goldman  J, Malone  FD, Vidaver  J, et  al. Impact of ma- 157. van  Leeuwen  M, Opmeer  BC, Zweers  EJ, et  al. Estimating the
ternal age on obstetric outcome. Obstet Gynecol. 2005;105(5 Pt risk of gestational diabetes mellitus: a clinical prediction model
1):983-990. based on patient characteristics and medical history. BJOG.
136. Yang H, Wei Y, Gao X, et al. Risk factors for gestational diabetes 2010;117(1):69-75.
mellitus in Chinese women: a prospective study of 16,286 preg- 158. Naylor CD, Sermer M, Chen E, et al; Toronto Trihospital Gestational
nant women in China. Diabet Med. 2009;26(11):1099-1104. Diabetes Project Investigators. Selective screening for gestational
137. Morisset AS, St-Yves A, Veillette J, et al. Prevention of gestational diabetes mellitus. N Engl J Med. 1997;337(22):1591-1596.
diabetes mellitus: a review of studies on weight management. 159. Catalano PM. Trying to understand gestational diabetes. Diabet
Diabetes Metab Res Rev. 2010;26(1):17-25. Med. 2014;31(3):273-281.
138. Weiss  JL, Malone  FD, Emig  D, et  al. Obesity, obstetric 160. Catalano PM. Carbohydrate metabolism and gestational diabetes.
complications and cesarean delivery rate—a population-based Clin Obstet Gynecol. 1994;37(1):25-38.
screening study. Am J Obstet Gynecol. 2004;190(4):1091-1097. 161. Lain  KY, Catalano  PM. Metabolic changes in pregnancy. Clin
139. Gibson  KS, Waters  TP, Catalano  PM. Maternal weight gain Obstet Gynecol. 2007;50(4):938-948.
in women who develop gestational diabetes mellitus. Obstet 162. Costrini  NV, Kalkhoff  RK. Relative effects of pregnancy, estra-
Gynecol. 2012;119(3):560-565. diol, and progesterone on plasma insulin and pancreatic islet in-
140. Morisset AS, Tchernof A, Dube MC, et al. Weight gain measures sulin secretion. J Clin Invest. 1971;50(5):992-999.
in women with gestational diabetes mellitus. J Womens Health 163. Kalkhoff RK, Kissebah AH, Kim HJ. Carbohydrate and lipid me-
(Larchmt). 2011;20(3):375-380. tabolism during normal pregnancy: relationship to gestational
141. Tovar  A, Must  A, Bermudez  OI, et  al. The impact of gesta- hormone action. Semin Perinatol. 1978;2(4):291-307.
tional weight gain and diet on abnormal glucose tolerance 164. Leturque A, Hauguel S, Sutter Dub MT, et al. Effects of placental
during pregnancy in Hispanic women. Matern Child Health J. lactogen and progesterone on insulin stimulated glucose metabo-
2009;13(4):520-530. lism in rat muscles in vitro. Diabete Metab. 1989;15(4):176-181.
142. Teulings N, Masconi KL, Ozanne SE, et al. Effect of interpregnancy 165. Ryan EA, Enns L. Role of gestational hormones in the induction of
weight change on perinatal outcomes: systematic review and insulin resistance. J Clin Endocrinol Metab. 1988;67(2):341-347.
meta-analysis. BMC Pregnancy Childbirth. 2019;19(1):386. 166. Alvarez JJ, Montelongo A, Iglesias A, Lasuncion MA, Herrera E.
143. Lo JC, Feigenbaum SL, Escobar GJ, et al. Increased prevalence of Longitudinal study on lipoprotein profile, high density lipoprotein
gestational diabetes mellitus among women with diagnosed poly- subclass, and postheparin lipases during gestation in women. J
cystic ovary syndrome: a population-based study. Diabetes Care. Lipid Res. 1996;37(2):299-308.
2006;29(8):1915-1917. 167. Barbour  LA, McCurdy  CE, Hernandez  TL, Kirwan  JP,
144. Mikola M, Hiilesmaa V, Halttunen M, et al. Obstetric outcome Catalano PM, Friedman JE. Cellular mechanisms for insulin re-
in women with polycystic ovarian syndrome. Hum Reprod. sistance in normal pregnancy and gestational diabetes. Diabetes
2001;16(2):226-229. Care. 2007;30(suppl 2):S112-S119.
145. Dinham GK, Henry A, Lowe SA, et al. Twin pregnancies compli- 168. Bomba-Opon D, Wielgos M, Szymanska M, et al. Effects of free
cated by gestational diabetes mellitus: a single centre cohort study. fatty acids on the course of gestational diabetes mellitus. Neuro
Diabet Med. 2016;33(12):1659-1667. Endocrinol Lett. 2006;27(1-2):277-280.
146. Rauh-Hain JA, Rana S, Tamez H, et al. Risk for developing ges- 169. Boden  G, Chen  X, Ruiz  J, et  al. Mechanisms of fatty
tational diabetes in women with twin pregnancies. J Matern Fetal acid-induced inhibition of glucose uptake. J Clin Invest.
Neonatal Med. 2009;22(4):293-299. 1994;93(6):2438-2446.
Endocrine Reviews, 2022, Vol. XX, No. XX 25

170. Catalano  PM, Huston  L, Amini  SB, et  al. Longitudinal changes 192. Dluski DF, Wolińska E, Skrzypczak M. Epigenetic changes in ges-
in glucose metabolism during pregnancy in obese women with tational diabetes mellitus. Int J Mol Sci. 2021;22(14):7649.
normal glucose tolerance and gestational diabetes mellitus. Am J 193. Elliott  HR, Sharp  GC, Relton  CL, et  al. Epigenetics and gesta-
Obstet Gynecol. 1999;180(4):903-916. tional diabetes: a review of epigenetic epidemiology studies and
171. Buchanan  TA, Metzger  BE, Freinkel  N, et  al. Insulin sensitivity their use to explore epigenetic mediation and improve prediction.
and B-cell responsiveness to glucose during late pregnancy in Diabetologia. 2019;62(12):2171-2178.
lean and moderately obese women with normal glucose tol- 194. Hayes  MG, Urbanek  M, Hivert  MF, et  al. Identification of
erance or mild gestational diabetes. Am J Obstet Gynecol. HKDC1 and BACE2 as genes influencing glycemic traits during
1990;162(4):1008-1014. pregnancy through genome-wide association studies. Diabetes.
172. Sivan E, Chen X, Homko CJ, et al. Longitudinal study of carbo- 2013;62(9):3282-3291.
hydrate metabolism in healthy obese pregnant women. Diabetes 195. Liu  S, Liu  Y, Liao  S. Heterogeneous impact of type 2 dia-

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


Care. 1997;20(9):1470-1475. betes mellitus-related genetic variants on gestational glycemic
173. Caro  JF, Dohm  LG, Pories  WJ, et  al. Cellular alterations in traits: review and future research needs. Mol Genet Genomics.
liver, skeletal muscle, and adipose tissue responsible for insulin 2019;294(4):811-847.
resistance in obesity and type II diabetes. Diabetes Metab Rev. 196. Wu L, Cui L, Tam WH, et al. Genetic variants associated with ges-
1989;5(8):665-689. tational diabetes mellitus: a meta-analysis and subgroup analysis.
174. Freinkel N, Metzger BE, Nitzan M, et al. Faciltated anabolism in late Sci Rep. 2016;6:30539.
pregnancy: some novel maternal compensations for accelerated star- 197. Chon  SJ, Kim  SY, Cho  NR, et  al. Association of variants in
vation. In: Malaise  WJ, Pirart  J, Vallence-Own  J, eds. International PPARgamma(2), IGF2BP2, and KCNQ1 with a susceptibility to
Congress Series 312. Excerpta Medica; 1974:474-488. gestational diabetes mellitus in a Korean population. Yonsei Med
175. Yogev Y, Ben-Haroush A, Chen R, et al. Diurnal glycemic profile J. 2013;54(2):352-357.
in obese and normal weight nondiabetic pregnant women. Am J 198. Kim  JY, Cheong  HS, Park  BL, et  al. Melatonin receptor 1 B
Obstet Gynecol. 2004;191(3):949-953. polymorphisms associated with the risk of gestational diabetes
176. Hernandez TL, Friedman JE, Van Pelt RE, et al. Patterns of gly- mellitus. BMC Med Genet. 2011;12:82.
cemia in normal pregnancy: should the current therapeutic targets 199. Klein  K, Haslinger  P, Bancher-Todesca  D, et  al. Transcription
be challenged? Diabetes Care. 2011;34(7):1660-1668. factor 7-like 2 gene polymorphisms and gestational diabetes mel-
177. Kuhl  C. Etiology and pathogenesis of gestational diabetes. litus. J Matern Fetal Neonatal Med. 2012;25(9):1783-1786.
Diabetes Care. 1998;21(suppl 2):B19-B26. 200. Alharbi  KK, Khan  IA, Abotalib  Z, et  al. Insulin receptor
178. Parsons  JA, Brelje  TC, Sorenson  RL. Adaptation of islets of substrate-1 (IRS-1) Gly927Arg: correlation with gesta-
Langerhans to pregnancy: increased islet cell proliferation and tional diabetes mellitus in Saudi women. Biomed Res Int.
insulin secretion correlates with the onset of placental lactogen 2014;2014:146495.
secretion. Endocrinology. 1992;130(3):1459-1466. 201. Tok EC, Ertunc D, Bilgin O, et al. PPAR-gamma2 Pro12Ala pol-
179. Sorenson RL, Brelje TC. Adaptation of islets of Langerhans to preg- ymorphism is associated with weight gain in women with ges-
nancy: beta-cell growth, enhanced insulin secretion and the role of tational diabetes mellitus. Eur J Obstet Gynecol Reprod Biol.
lactogenic hormones. Horm Metab Res. 1997;29(6):301-307. 2006;129(1):25-30.
180. Nielsen  JH, Galsgaard  ED, Moldrup  A, et  al. Regulation of 202. Montazeri S, Nalliah S, Radhakrishnan AK. Is there a genetic var-
beta-cell mass by hormones and growth factors. Diabetes. iation association in the IL-10 and TNF alpha promoter gene with
2001;50(suppl 1):S25-S29. gestational diabetes mellitus? Hereditas. 2010;147(2):94-102.
181. Parsons  JA, Bartke  A, Sorenson  RL. Number and size of islets 203. Kwak  SH, Kim  SH, Cho  YM, et  al. A genome-wide association
of Langerhans in pregnant, human growth hormone-expressing study of gestational diabetes mellitus in Korean women. Diabetes.
transgenic, and pituitary dwarf mice: effect of lactogenic 2012;61(2):531-541.
hormones. Endocrinology. 1995;136(5):2013-2021. 204. Lyssenko V, Nagorny CL, Erdos MR, et al. Common variant in
182. Rieck  S, White  P, Schug  J, et  al. The transcriptional re- MTNR1B associated with increased risk of type 2 diabetes and
sponse of the islet to pregnancy in mice. Mol Endocrinol. impaired early insulin secretion. Nat Genet. 2009;41(1):82-88.
2009;23(10):1702-1712. 205. Wellcome Trust Case Control Consortium. Genome-wide associ-
183. Buchanan  TA, Kitzmiller  JL. Metabolic interactions of diabetes ation study of 14,000 cases of seven common diseases and 3,000
and pregnancy. Annu Rev Med. 1994;45:245-260. shared controls. Nature. 2007;447(7145):661-678.
184. Rieck S, Kaestner KH. Expansion of beta-cell mass in response to 206. Bouatia-Naji  N, Bonnefond  A, Cavalcanti-Proenca  C, et  al.
pregnancy. Trends Endocrinol Metab. 2010;21(3):151-158. A variant near MTNR1B is associated with increased fasting
185. Koukkou  E, Watts  GF, Lowy  C. Serum lipid, lipoprotein and plasma glucose levels and type 2 diabetes risk. Nat Genet.
apolipoprotein changes in gestational diabetes mellitus: a cross-sec- 2009;41(1):89-94.
tional and prospective study. J Clin Pathol. 1996;49(8):634-637. 207. Saxena R, Elbers CC, Guo Y, et al. Large-scale gene-centric meta-
186. Nolan  CJ. Controversies in gestational diabetes. Best Pract Res analysis across 39 studies identifies type 2 diabetes loci. Am J
Clin Obstet Gynaecol. 2011;25(1):37-49. Hum Genet. 2012;90(3):410-425.
187. Talchai C, Xuan S, Lin HV, Sussel L, Accili D. Pancreatic beta cell 208. Manning  AK, Hivert  MF, Scott  RA, et  al. A genome-wide ap-
dedifferentiation as a mechanism of diabetic beta cell failure. Cell. proach accounting for body mass index identifies genetic variants
2012;150(6):1223-1234. influencing fasting glycemic traits and insulin resistance. Nat
188. Halban PA, Polonsky KS, Bowden DW, et al. beta-cell failure in Genet. 2012;44(6):659-669.
type 2 diabetes: postulated mechanisms and prospects for preven- 209. Sparso T, Andersen G, Nielsen T, et al. The GCKR rs780094 poly-
tion and treatment. Diabetes Care. 2014;37(6):1751-1758. morphism is associated with elevated fasting serum triacylglycerol,
189. Catalano PM, Tyzbir ED, Wolfe RR, et al. Carbohydrate metabo- reduced fasting and OGTT-related insulinaemia, and reduced risk
lism during pregnancy in control subjects and women with gesta- of type 2 diabetes. Diabetologia. 2008;51(1):70-75.
tional diabetes. Am J Physiol. 1993;264(1 Pt 1):E60-E67. 210. Vaxillaire  M, Cavalcanti-Proenca  C, Dechaume  A, et  al. The
190. Homko  C, Sivan  E, Chen  X, et  al. Insulin secretion during and common P446L polymorphism in GCKR inversely modulates
after pregnancy in patients with gestational diabetes mellitus. J fasting glucose and triglyceride levels and reduces type 2 dia-
Clin Endocrinol Metab. 2001;86(2):568-573. betes risk in the DESIR prospective general French population.
191. Powe  CE, Huston  Presley  LP, Locascio  JJ, et  al. Augmented in- Diabetes. 2008;57(8):2253-2257.
sulin secretory response in early pregnancy. Diabetologia. 211. Agius L. Glucokinase and molecular aspects of liver glycogen me-
2019;62(8):1445-1452. tabolism. Biochem J. 2008;414(1):1-18.
26 Endocrine Reviews, 2022, Vol. XX, No. XX

212. Chasman  DI, Pare  G, Mora  S, et  al. Forty-three loci associated 235. Spellacy  WN, Miller  S, Winegar  A, et  al. Macrosomia—ma-
with plasma lipoprotein size, concentration, and cholesterol con- ternal characteristics and infant complications. Obstet Gynecol.
tent in genome-wide analysis. PLoS Genet. 2009;5(11):e1000730. 1985;66(2):158-161.
213. Seidah NG. The proprotein convertases, 20 years later. Methods 236. Jang  HC, Cho  NH, Min  YK, et  al. Increased macrosomia and
Mol Biol. 2011;768:23-57. perinatal morbidity independent of maternal obesity and ad-
214. Benzinou  M, Creemers  JW, Choquet  H, et  al. Common vanced age in Korean women with GDM. Diabetes Care.
nonsynonymous variants in PCSK1 confer risk of obesity. Nat 1997;20(10):1582-1588.
Genet. 2008;40(8):943-945. 237. Langer  O, Yogev  Y, Most  O, et  al. Gestational diabetes:
215. Hayes  A, Chevalier  A, D’Souza  M, et  al. Early childhood obe- the consequences of not treating. Am J Obstet Gynecol.
sity: association with healthcare expenditure in Australia. Obesity 2005;192(4):989-997.
(Silver Spring). 2016;24(8):1752-8. 238. Weiss PA, Haeusler M, Tamussino K, et al. Can glucose tolerance test

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


216. Powe  CE, Nodzenski  M, Talbot  O, et  al. Genetic determinants predict fetal hyperinsulinism? BJOG. 2000;107(12):1480-1485.
of glycemic traits and the risk of gestational diabetes mellitus. 239. Boulet SL, Alexander GR, Salihu HM, et al. Macrosomic births in
Diabetes. 2018;67(12):2703-2709. the united states: determinants, outcomes, and proposed grades of
217. Fajans SS, Bell GI, Polonsky KS. Molecular mechanisms and clin- risk. Am J Obstet Gynecol. 2003;188(5):1372-1378.
ical pathophysiology of maturity-onset diabetes of the young. N 240. Ryckman KK, Spracklen CN, Smith CJ, et al. Maternal lipid levels
Engl J Med. 2001;345(13):971-980. during pregnancy and gestational diabetes: a systematic review
218. Hattersley AT, Patel KA. Precision diabetes: learning from mono- and meta-analysis. BJOG. 2015;122(5):643-651.
genic diabetes. Diabetologia. 2017;60(5):769-777. 241. Jovanovic  L, Pettitt  DJ. Gestational diabetes mellitus. JAMA.
219. Dickens  LT, Naylor  RN. Clinical management of women 2001;286(20):2516-2518.
with monogenic diabetes during pregnancy. Curr Diab Rep. 242. Esakoff TF, Cheng YW, Sparks TN, et al. The association between
2018;18(3):12. birthweight 4000 g or greater and perinatal outcomes in patients
220. Chakera AJ, Steele AM, Gloyn AL, et al. Recognition and manage- with and without gestational diabetes mellitus. Am J Obstet
ment of individuals with hyperglycemia because of a heterozygous Gynecol. 2009;200(6):672 e1-672 e4.
glucokinase mutation. Diabetes Care. 2015;38(7):1383-1392. 243. Henriksen T. The macrosomic fetus: a challenge in current obstet-
221. Urbanova J, Brunerova L, Nunes M, et al. Identification of MODY rics. Acta Obstet Gynecol Scand. 2008;87(2):134-145.
among patients screened for gestational diabetes: a clinician’s 244. Reece EA, Leguizamon G, Wiznitzer A. Gestational diabetes: the
guide. Arch Gynecol Obstet. 2020;302(2):305-314. need for a common ground. Lancet. 2009;373(9677):1789-1797.
222. Wang Z, Ping F, Zhang Q, et al. Preliminary screening of mutations 245. Cetin  H, Yalaz  M, Akisu  M, et  al. Polycythaemia in infants of
in the glucokinase gene of Chinese patients with gestational dia- diabetic mothers: beta-hydroxybutyrate stimulates erythropoietic
betes. J Diabetes Investig. 2018;9(1):199-203. activity. J Int Med Res. 2011;39(3):815-821.
223. Gjesing AP, Rui G, Lauenborg J, et al. High prevalence of diabetes- 246. Farrar  D, Fairley  L, Santorelli  G, et  al. Association between
predisposing variants in MODY genes among Danish women hyperglycaemia and adverse perinatal outcomes in south Asian and
with gestational diabetes mellitus. J Endocr Soc. 2017;1(6): white British women: analysis of data from the Born in Bradford
681-690. cohort. Lancet Diabetes Endocrinol. 2015;3(10):795-804.
224. Shields  BM, Hicks  S, Shepherd  MH, et  al. Maturity-onset dia- 247. Balsells M, Corcoy R, Adelantado JM, et al. Gestational diabetes
betes of the young (MODY): how many cases are we missing? mellitus: metabolic control during labour. Diabetes Nutr Metab.
Diabetologia. 2010;53(12):2504-2508. 2000;13(5):257-262.
225. Spyer G, Hattersley AT, Sykes JE, et al. Influence of maternal and 248. Farrar  D, Simmonds  M, Bryant  M, et  al. Hyperglycaemia and
fetal glucokinase mutations in gestational diabetes. Am J Obstet risk of adverse perinatal outcomes: systematic review and meta-
Gynecol. 2001;185(1):240-241. analysis. BMJ. 2016;354:i4694.
226. Hattersley  AT, Beards  F, Ballantyne  E, et  al. Mutations in the 249. Billionnet  C, Mitanchez  D, Weill  A, et  al. Gestational diabetes
glucokinase gene of the fetus result in reduced birth weight. Nat and adverse perinatal outcomes from 716,152 births in France in
Genet. 1998;19(3):268-270. 2012. Diabetologia. 2017;60(4):636-644.
227. Scholtens DM, Kuang A, Lowe LP, et al. Hyperglycemia and ad- 250. Allen VM, Armson BA. SOGC Clinical Practice Guideline: terat-
verse pregnancy outcome follow-up study (HAPO FUS): maternal ogenicity associated with pre-existing and gestational diabetes. J
glycemia and childhood glucose metabolism. Diabetes Care. Obstet Gynaecol Can. 2007;29(11):927-934.
2019;42(3):381-392. 251. Owens  LA, O’Sullivan  EP, Kirwan  B, et  al. ATLANTIC DIP:
228. Saravanan P; Diabetes in Pregnancy Working Group. Gestational the impact of obesity on pregnancy outcome in glucose-tolerant
diabetes: opportunities for improving maternal and child health. women. Diabetes Care. 2010;33(3):577-579.
Lancet Diabetes Endocrinol. 2020;8(9):793-800. 252. Dennedy MC, Avalos G, O’Reilly MW, et al. ATLANTIC-DIP: raised
229. Pedersen  J. Hyperglycaemia-hyperinsulinism theory and birth- maternal body mass index (BMI) adversely affects maternal and
weight. In: The Pregnant Diabetic and Her Newborn: Problems fetal outcomes in glucose-tolerant women according to International
and Management. Williams and Wilkins; 1977:211-220. Association of Diabetes and Pregnancy Study Groups (IADPSG)
230. Illsley NP. Glucose transporters in the human placenta. Placenta. criteria. J Clin Endocrinol Metab. 2012;97(4):E608-E612.
2000;21(1):14-22. 253. Rosenstein MG, Cheng YW, Snowden JM, et al. The risk of stillbirth
231. Pedersen  J. Weight and length at birth of infants of diabetic and infant death stratified by gestational age in women with gesta-
mothers. Acta Endocrinol (Copenh). 1954;16(4):330-342. tional diabetes. Am J Obstet Gynecol. 2012;206(4):309 e1-309 e7.
232. Whitelaw  A. Subcutaneous fat in newborn infants of diabetic 254. Mitanchez D. Foetal and neonatal complications in gestational dia-
mothers: an indication of quality of diabetic control. Lancet. betes: perinatal mortality, congenital malformations, macrosomia,
1977;1(8001):15-18. shoulder dystocia, birth injuries, neonatal complications. Diabetes
233. Vrijkotte  TG, Krukziener  N, Hutten  BA, et  al. Maternal lipid Metab. 2010;36(6 Pt 2):617-627.
profile during early pregnancy and pregnancy complications 255. Cundy T, Gamble G, Townend K, et al. Perinatal mortality in type
and outcomes: the ABCD study. J Clin Endocrinol Metab. 2 diabetes mellitus. Diabet Med. 2000;17(1):33-39.
2012;97(11):3917-3925. 256. Hutcheon JA, Kuret V, Joseph KS, et al. Immortal time bias in the
234. Yang  X, Hsu-Hage  B, Zhang  H, et  al. Women with im- study of stillbirth risk factors: the example of gestational diabetes.
paired glucose tolerance during pregnancy have signifi- Epidemiology. 2013;24(6):787-790.
cantly poor pregnancy outcomes. Diabetes Care. 2002;25(9): 257. Poston L, Bell R, Croker H, et al. Effect of a behavioural interven-
1619-1624. tion in obese pregnant women (the UPBEAT study): a multicentre,
Endocrine Reviews, 2022, Vol. XX, No. XX 27

randomised controlled trial. Lancet Diabetes Endocrinol. preconception to the postnatal period. NICE Guidelines NG3.
2015;3(10):767-777. 2015. Last updated December 16, 2020. https://www.nice.org.uk/
258. Tam  WH, Ma  RCW, Ozaki  R, et  al. In utero exposure to ma- guidance/ng3
ternal hyperglycemia increases childhood cardiometabolic risk in 278. Carr  DB, Utzschneider  KM, Hull  RL, et  al. Gestational dia-
Offspring. Diabetes Care. 2017;40(5):679-686. betes mellitus increases the risk of cardiovascular disease in
259. Yu  Y, Arah  OA, Liew  Z, et  al. Maternal diabetes during preg- women with a family history of type 2 diabetes. Diabetes Care.
nancy and early onset of cardiovascular disease in offspring: 2006;29(9):2078-2083.
population based cohort study with 40 years of follow-up. BMJ. 279. Gunderson EP, Chiang V, Pletcher MJ, et al. History of gestational
2019;367:l6398. diabetes mellitus and future risk of atherosclerosis in mid-life: the
260. Hod M. The fetal heart in gestational diabetes: long-term effects. Coronary Artery Risk Development in Young Adults study. J Am
BJOG. 2021;128(2):280. Heart Assoc. 2014;3(2):e000490.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


261. Dabelea D, Hanson RL, Lindsay RS, et al. Intrauterine exposure 280. Retnakaran R. Glucose tolerance status in pregnancy: a window
to diabetes conveys risks for type 2 diabetes and obesity: a study to the future risk of diabetes and cardiovascular disease in young
of discordant sibships. Diabetes. 2000;49(12):2208-2211. women. Curr Diabetes Rev. 2009;5(4):239-244.
262. Lowe WL Jr., Lowe LP, Kuang A, et al. Maternal glucose levels 281. Tobias DK, Hu FB, Forman JP, et al. Increased risk of hyperten-
during pregnancy and childhood adiposity in the Hyperglycemia sion after gestational diabetes mellitus: findings from a large pro-
and Adverse Pregnancy Outcome Follow-up Study. Diabetologia. spective cohort study. Diabetes Care. 2011;34(7):1582-1584.
2019;62(4):598-610. 282. Tobias  DK, Stuart  JJ, Li  S, et  al. Association of history of ges-
263. Page  KA, Luo  S, Wang  X, et  al. Children exposed to maternal tational diabetes with long-term cardiovascular disease risk in
obesity or gestational diabetes mellitus during early fetal devel- a large prospective cohort of US Women. JAMA Intern Med.
opment have hypothalamic alterations that predict future weight 2017;177(12):1735-1742.
gain. Diabetes Care. 2019;42(8):1473-1480. 283. Brown HL, Warner JJ, Gianos E, et al. Promoting risk identifica-
264. Anderberg E, Kallen K, Berntorp K. The impact of gestational di- tion and reduction of cardiovascular disease in women through
abetes mellitus on pregnancy outcome comparing different cut- collaboration with obstetricians and gynecologists: a presidential
off criteria for abnormal glucose tolerance. Acta Obstet Gynecol advisory from the American Heart Association and the American
Scand. 2010;89(12):1532-1537. College of Obstetricians and Gynecologists. Circulation.
265. Ju  H, Rumbold  AR, Willson  KJ, et  al. Borderline gestational 2018;137(24):e843-ee52.
diabetes mellitus and pregnancy outcomes. BMC Pregnancy 284. Knudsen LS, Christensen IJ, Lottenburger T, et al. Pre-analytical
Childbirth. 2008;8:31. and biological variability in circulating interleukin 6 in healthy
266. Jastrow N, Roberge S, Gauthier RJ, et al. Effect of birth weight on subjects and patients with rheumatoid arthritis. Biomarkers.
adverse obstetric outcomes in vaginal birth after cesarean delivery. 2008;13(1):59-78.
Obstet Gynecol. 2010;115(2 Pt 1):338-343. 285. Alwan N, Tuffnell DJ, West J. Treatments for gestational diabetes.
267. Metzger  BE, Coustan  DR. Summary and recommendations of Cochrane Database Syst Rev. 2009;3:CD003395.
the Fourth International Workshop-Conference on Gestational 286. Jovanovic-Peterson  L, Peterson  CM, Reed  GF, et  al. Maternal
Diabetes Mellitus. The Organizing Committee. Diabetes Care. postprandial glucose levels and infant birth weight: the Diabetes
1998;21(suppl 2):B161-B167. in Early Pregnancy Study. The National Institute of Child Health
268. Hiden U, Lassance L, Tabrizi NG, et al. Fetal insulin and IGF-II and Human Development—Diabetes in Early Pregnancy Study.
contribute to gestational diabetes mellitus (GDM)-associated Am J Obstet Gynecol. 1991;164(1 Pt 1):103-111.
up-regulation of membrane-type matrix metalloproteinase 1 287. Bain E, Crane M, Tieu J, et al. Diet and exercise interventions for
(MT1-MMP) in the human feto-placental endothelium. J Clin preventing gestational diabetes mellitus. Cochrane Database Syst
Endocrinol Metab. 2012;97(10):3613-3621. Rev. 0443;2015;4:CD01.
269. Dunne  FP, Avalos  G, Durkan  M, et  al. ATLANTIC DIP: preg- 288. Duarte-Gardea  MO, Gonzales-Pacheco  DM, Reader  DM, et  al.
nancy outcomes for women with type 1 and type 2 diabetes. Ir Academy of nutrition and dietetics gestational diabetes evi-
Med J. 2012;105(5 suppl):6-9. dence-based nutrition practice guideline. J Acad Nutr Diet.
270. Roberts JM, Redman CW. Pre-eclampsia: more than pregnancy- 2018;118(9):1719-1742.
induced hypertension. Lancet. 1993;341(8858):1447-1451. 289. Rasmussen  KM, Yaktine  AL, eds.; Institute of Medicine and
271. Desoye  G, Hauguel-de  Mouzon  S. The human placenta in ges- National Research Council Committee to Reexamine IOM
tational diabetes mellitus. The Insulin and Cytokine Network. Pregnancy Weight Guidelines. Weight Gain During Pregnancy:
Diabetes Care. 2007;30(suppl 2):S120-S126. Reexamining the Guidelines. National Academies Press; 2009.
272. Belkacemi  L, Lash  GE, Macdonald-Goodfellow  SK, et  al. 290. Jovanovic-Peterson  L, Peterson  CM. Dietary manipulation as a
Inhibition of human trophoblast invasiveness by high glu- primary treatment strategy for pregnancies complicated by dia-
cose concentrations. J Clin Endocrinol Metab. 2005;90(8): betes. J Am Coll Nutr. 1990;9(4):320-325.
4846-4851. 291. Hernandez TL, Van Pelt RE, Anderson MA, et al. A higher-complex
273. Vounzoulaki E, Khunti K, Abner SC, et al. Progression to type 2 carbohydrate diet in gestational diabetes mellitus achieves glucose
diabetes in women with a known history of gestational diabetes: targets and lowers postprandial lipids: a randomized crossover
systematic review and meta-analysis. BMJ. 2020;369:m1361. study. Diabetes Care. 2014;37(5):1254-1262.
274. Daly  B, Toulis  KA, Thomas  N, et  al. Increased risk of ischemic 292. Hernandez  TL, Van  Pelt  RE, Anderson  MA, et  al. Women with
heart disease, hypertension, and type 2 diabetes in women with gestational diabetes mellitus randomized to a higher-complex car-
previous gestational diabetes mellitus, a target group in general bohydrate/low-fat diet manifest lower adipose tissue insulin re-
practice for preventive interventions: a population-based cohort sistance, inflammation, glucose, and free fatty acids: a pilot study.
study.. PLoS Med. 2018;15(1):e1002488. Diabetes Care. 2016;39(1):39-42.
275. Lowe WL Jr, Scholtens DM, Kuang A, et al. Hyperglycemia and 293. Asemi  Z, Tabassi  Z, Samimi  M, et  al. Favourable effects of the
adverse pregnancy outcome follow-up study (HAPO FUS): ma- Dietary Approaches to Stop Hypertension diet on glucose toler-
ternal gestational diabetes mellitus and childhood glucose metab- ance and lipid profiles in gestational diabetes: a randomised clin-
olism. Diabetes Care. 2019;42(3):372-380. ical trial. Br J Nutr. 2013;109(11):2024-2030.
276. Murphy HR. 2020 NICE guideline update: good news for preg- 294. Han S, Middleton P, Shepherd E, et al. Different types of dietary
nant women with type 1 diabetes and past or current gestational advice for women with gestational diabetes mellitus. Cochrane
diabetes. Diabet Med. 2021;38(6):e14576. Database Syst Rev. 2017;2:CD009275.
277. National Institute for Health and Care Excellence. Diabetes in 295. Yamamoto  JM, Kellett  JE, Balsells  M, et  al. Gestational dia-
pregnancy: management of diabetes and its complications from betes mellitus and diet: a systematic review and meta-analysis of
28 Endocrine Reviews, 2022, Vol. XX, No. XX

randomized controlled trials examining the impact of modified 317. Langer  O, Rodriguez  DA, Xenakis  EM, et  al. Intensified versus
dietary interventions on maternal glucose control and neonatal conventional management of gestational diabetes. Am J Obstet
birth weight. Diabetes Care. 2018;41(7):1346-1361. Gynecol. 1994;170(4):1036-1046.
296. Major  CA, Henry  MJ, De  Veciana  M, et  al. The effects of car- 318. Pregnancy outcomes in the diabetes control and complications
bohydrate restriction in patients with diet-controlled gestational trial. Am J Obstet Gynecol. 1996;174(4):1343-1353.
diabetes. Obstet Gynecol. 1998;91(4):600-604. 319. Martis  R, Brown  J, Alsweiler  J, et  al. Different intensities of
297. American College of Obstetrics and Gynecology. Practice glycaemic control for women with gestational diabetes mellitus.
Bulletin No. 137: gestational diabetes mellitus. Obstet Gynecol. Cochrane Database Syst Rev. 2016;4:CD011624.
2013;122(2 Pt 1):406-416. 320. Snyder  JMI, Melzter  S, Nadeau  J. Gestational diabetes and gly-
298. Metzger  BE, Buchanan  TA, Coustan  DR, et  al. Summary and cemic control: a randomized clinical trial. Am J Obstet Gynecol.
recommendations of the Fifth International Workshop-Conference 1998;178(1 Pt 2):S55.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


on Gestational Diabetes Mellitus. Diabetes Care. 2007;30(suppl 321. Pertot  T, Molyneaux  L, Tan  K, et  al. Can common clinical
2):S251-S260. parameters be used to identify patients who will need in-
299. American Diabetes Association. Standards of medical care in dia- sulin treatment in gestational diabetes mellitus? Diabetes Care.
betes-2011. Diabetes Care. 2011;34(suppl 1):S11-S61. 2011;34(10):2214-2216.
300. Battaglia  FC, Meschia  G. An Introduction to Fetal Physiology. 322. Rowan  JA, Hague  WM, Gao  W, et  al. Metformin versus in-
Academic Press; 1986. sulin for the treatment of gestational diabetes. N Engl J Med.
301. Harding  JE, Johnston  BM. Nutrition and fetal growth. Reprod 2008;358(19):2003-2015.
Fertil Dev. 1995;7(3):539-547. 323. Murphy  HR, Rayman  G, Duffield  K, et  al. Changes in the gly-
302. Sweeting  A, Markovic  TP, Mijatovic  J, et  al. The carbohydrate cemic profiles of women with type 1 and type 2 diabetes during
threshold in pregnancy and gestational diabetes: how low can we pregnancy. Diabetes Care. 2007;30(11):2785-2791.
go? Nutrients. 2021;13(8):2599. 324. Padmanabhan  S, McLean  M, Cheung  NW. Falling in-
303. Morisaki  N, Nagata  C, Yasuo  S, et  al. Optimal protein intake sulin requirements are associated with adverse obstetric
during pregnancy for reducing the risk of fetal growth restric- outcomes in women with preexisting diabetes. Diabetes Care.
tion: the Japan Environment and Children’s Study. Br J Nutr. 2014;37(10):2685-2692.
2018;120(12):1432-1440. 325. Wong  VW. Gestational diabetes mellitus in five ethnic groups:
304. Rizzo T, Metzger BE, Burns WJ, et al. Correlations between ante- a comparison of their clinical characteristics. Diabet Med.
partum maternal metabolism and intelligence of offspring. N Engl 2012;29(3):366-371.
J Med. 1991;325(13):911-916. 326. Barnes  RA, Wong  T, Ross  GP, et  al. A novel validated model
305. Shaw  GM, Yang  W. Women’s periconceptional lowered car- for the prediction of insulin therapy initiation and adverse per-
bohydrate intake and NTD-affected pregnancy risk in the inatal outcomes in women with gestational diabetes mellitus.
era of prefortification with folic acid. Birth Defects Res. Diabetologia. 2016;59(11):2331-2338.
2019;111(5):248-253. 327. Ryu  RJ, Hays  KE, Hebert  MF. Gestational diabetes mellitus
306. Mijatovic  J, Louie  JCY, Buso  MEC, et  al. Effects of a modestly management with oral hypoglycemic agents. Semin Perinatol.
lower carbohydrate diet in gestational diabetes: a randomized 2014;38(8):508-515.
controlled trial. Am J Clin Nutr. 2020;112(2):284-292. 328. Camelo  Castillo  W, Boggess  K, Sturmer  T, et  al. Trends in
307. Shubert PJ, Gordon MC, Landon MB, et al. Ketoacids attenuate glyburide compared with insulin use for gestational diabetes
glucose uptake in human trophoblasts isolated from first-trimester treatment in the United States, 2000-2011. Obstet Gynecol.
chorionic villi. Am J Obstet Gynecol. 1996;175(1):56-62. 2014;123(6):1177-1184.
308. Catalano PM, Mele L, Landon MB, et al. Inadequate weight gain 329. Langer  O, Conway  DL, Berkus  MD, et  al. A comparison of
in overweight and obese pregnant women: what is the effect on glyburide and insulin in women with gestational diabetes mellitus.
fetal growth? Am J Obstet Gynecol. 2014;211(2):137 e1-137 e7. N Engl J Med. 2000;343(16):1134-1138.
309. Faucher MA, Barger MK. Gestational weight gain in obese women 330. Kremer CJ, Duff P. Glyburide for the treatment of gestational dia-
by class of obesity and select maternal/newborn outcomes: a sys- betes. Am J Obstet Gynecol. 2004;190(5):1438-1439.
tematic review. Women Birth. 2015;28(3):e70-e79. 331. Camelo  Castillo  W, Boggess  K, Sturmer  T, et  al. Association
310. Viecceli  C, Remonti  LR, Hirakata  VN, et  al. Weight gain ade- of adverse pregnancy outcomes with glyburide vs insulin in
quacy and pregnancy outcomes in gestational diabetes: a meta- women with gestational diabetes. JAMA Pediatr. 2015;169(5):
analysis. Obes Rev. 2017;18(5):567-580. 452-458.
311. Kainer F, Weiss PA, Huttner U, et al. Levels of amniotic fluid in- 332. Schwartz RA, Rosenn B, Aleksa K, et al. Glyburide transport across
sulin and profiles of maternal blood glucose in pregnant women the human placenta. Obstet Gynecol. 2015;125(3):583-588.
with diabetes type-I. Early Hum Dev. 1997;49(2):97-105. 333. Cheng YW, Chung JH, Block-Kurbisch I, et al. Treatment of ges-
312. de Veciana M, Major CA, Morgan MA, et al. Postprandial versus tational diabetes mellitus: glyburide compared to subcutaneous
preprandial blood glucose monitoring in women with gesta- insulin therapy and associated perinatal outcomes. J Matern Fetal
tional diabetes mellitus requiring insulin therapy. N Engl J Med. Neonatal Med. 2012;25(4):379-384.
1995;333(19):1237-1241. 334. Lindsay RS, Loeken MR. Metformin use in pregnancy: promises
313. Hernandez TL. Glycemic targets in pregnancies affected by dia- and uncertainties. Diabetologia. 2017;60(9):1612-1619.
betes: historical perspective and future directions. Curr Diab Rep. 335. Charles B, Norris R, Xiao X, et al. Population pharmacokinetics
2015;15(1):565. of metformin in late pregnancy. Ther Drug Monit. 2006;28(1):
314. Abell SK, Boyle JA, Earnest A, et al. Impact of different glycaemic 67-72.
treatment targets on pregnancy outcomes in gestational diabetes. 336. Tarry-Adkins  JL, Aiken  CE, Ozanne  SE. Neonatal, infant, and
Diabet Med. 2019;36(2):177-183. childhood growth following metformin versus insulin treatment
315. Garner P, Okun N, Keely E, et al. A randomized controlled trial for gestational diabetes: a systematic review and meta-analysis.
of strict glycemic control and tertiary level obstetric care versus PLoS Med. 2019;16(8):e1002848.
routine obstetric care in the management of gestational diabetes: 337. Feig  DS, Donovan  LE, Zinman  B, et  al. Metformin in women
a pilot study. Am J Obstet Gynecol. 1997;177(1):190-195. with type 2 diabetes in pregnancy (MiTy): a multicentre, inter-
316. Langer  O, Levy  J, Brustman  L, et  al. Glycemic control in gesta- national, randomised, placebo-controlled trial. Lancet Diabetes
tional diabetes mellitus--how tight is tight enough: small for gesta- Endocrinol. 2020;8(10):834-844.
tional age versus large for gestational age? Am J Obstet Gynecol. 338. Niromanesh S, Alavi A, Sharbaf FR, et al. Metformin compared
1989;161(3):646-653. with insulin in the management of gestational diabetes
Endocrine Reviews, 2022, Vol. XX, No. XX 29

mellitus: a randomized clinical trial. Diabetes Res Clin Pract. 358. Gabbe  SG, Landon  MB, Warren-Boulton  E, et  al. Promoting
2012;98(3):422-429. health after gestational diabetes: a National Diabetes Education
339. Khin MO, Gates S, Saravanan P. Predictors of metformin failure Program call to action. Obstet Gynecol. 2012;119(1):171-176.
in gestational diabetes mellitus (GDM). Diabetes Metab Syndr. 359. Ehrlich SF, Hedderson MM, Feng J, et al. Change in body mass
2018;12(3):405-410. index between pregnancies and the risk of gestational diabetes in
340. Rowan JA, Rush EC, Obolonkin V, et al. Metformin in gestational a second pregnancy. Obstet Gynecol. 2011;117(6):1323-1330.
diabetes: the offspring follow-up (MiG TOFU): body composition 360. Kim  C, Newton  KM, Knopp  RH. Gestational diabetes and the
at 2 years of age. Diabetes Care. 2011;34(10):2279-2284. incidence of type 2 diabetes: a systematic review. Diabetes Care.
341. Rowan  JA, Rush  EC, Plank  LD, et  al. Metformin in gestational 2002;25(10):1862-1868.
diabetes: the offspring follow-up (MiG TOFU): body composition 361. Bellamy L, Casas JP, Hingorani AD, et al. Type 2 diabetes mellitus
and metabolic outcomes at 7-9 years of age. BMJ Open Diabetes after gestational diabetes: a systematic review and meta-analysis.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


Res Care. 2018;6(1):e000456. Lancet. 2009;373(9677):1773-1779.
342. Hanem LGE, Stridsklev S, Juliusson PB, et al. Metformin use in 362. Dornhorst  A, Bailey  PC, Anyaoku  V, et  al. Abnormalities of
PCOS pregnancies increases the risk of offspring overweight at glucose tolerance following gestational diabetes. Q J Med.
4 years of age: follow-up of two RCTs. J Clin Endocrinol Metab. 1990;77(284):1219-1228.
2018;103(4):1612-1621. 363. Aroda VR, Christophi CA, Edelstein SL, et al. The effect of life-
343. Brown J, Martis R, Hughes B, et al. Oral anti-diabetic pharma- style intervention and metformin on preventing or delaying dia-
cological therapies for the treatment of women with gestational betes among women with and without gestational diabetes: the
diabetes. Cochrane Database Syst Rev. 2017;1:CD011967. Diabetes Prevention Program outcomes study 10-year follow-up.
344. Balsells  M, Garcia-Patterson  A, Sola  I, et  al. Glibenclamide, J Clin Endocrinol Metab. 2015;100(4):1646-1653.
metformin, and insulin for the treatment of gestational diabetes: a 364. Pirc LK, Owens JA, Crowther CA, et al. Mild gestational diabetes
systematic review and meta-analysis. BMJ. 2015;350:h102. in pregnancy and the adipoinsular axis in babies born to mothers
345. Poolsup  N, Suksomboon  N, Amin  M. Efficacy and safety of in the ACHOIS randomised controlled trial. BMC Pediatr.
oral antidiabetic drugs in comparison to insulin in treating 2007;7:18.
gestational diabetes mellitus: a meta-analysis. PLoS One. 365. Landon  MB, Rice  MM, Varner  MW, et  al. Mild gestational di-
2014;9(10):e109985. abetes mellitus and long-term child health. Diabetes Care.
346. Nachum Z, Zafran N, Salim R, et al. Glyburide versus metformin 2015;38(3):445-452.
and their combination for the treatment of gestational dia- 366. Dabelea D, Pettitt DJ. Intrauterine diabetic environment confers
betes mellitus: a randomized controlled study. Diabetes Care. risks for type 2 diabetes mellitus and obesity in the offspring, in
2017;40(3):332-337. addition to genetic susceptibility. J Pediatr Endocrinol Metab.
347. Sacco  F, Calderone  A, Castagnoli  L, et  al. The cell-autonomous 2001;14(8):1085-1091.
mechanisms underlying the activity of metformin as an anticancer 367. Pettitt  DJ, Baird  HR, Aleck  KA, et  al. Excessive obesity in off-
drug. Br J Cancer. 2016;115(12):1451-1456. spring of Pima Indian women with diabetes during pregnancy. N
348. Barbour  LA, Davies  JK. Comment on Nachum et  al. Glyburide Engl J Med. 1983;308(5):242-245.
versus metformin and their combination for the treatment of ges- 368. Gillman MW, Rifas-Shiman S, Berkey CS, Field AE, Colditz GA.
tational diabetes mellitus: a randomized controlled study. Diabetes Maternal gestational diabetes, birth weight, and adolescent obe-
Care. 2017;40:332-337. Diabetes Care. 2017;40(8):e115. sity. Pediatrics. 2003;111(3):e221-e226.
349. Rao U, de Vries B, Ross GP, et al. Fetal biometry for guiding the 369. Plagemann A, Harder T, Kohlhoff R, et al. Overweight and obe-
medical management of women with gestational diabetes mellitus sity in infants of mothers with long-term insulin-dependent di-
for improving maternal and perinatal health. Cochrane Database abetes or gestational diabetes. Int J Obes Relat Metab Disord.
Syst Rev. 2019;9:CD012544. 1997;21(6):451-456.
350. Schaefer-Graf  UM, Kjos  SL, Fauzan  OH, et  al. A randomized 370. Pribylova  H, Dvorakova  L. Long-term prognosis of infants of
trial evaluating a predominantly fetal growth-based strategy to diabetic mothers. Relationship between metabolic disorders in
guide management of gestational diabetes in Caucasian women. newborns and adult offspring. Acta Diabetol. 1996;33(1):30-34.
Diabetes Care. 2004;27(2):297-302. 371. Silverman BL, Metzger BE, Cho NH, et al. Impaired glucose tol-
351. Schaefer-Graf  UM, Wendt  L, Sacks  DA, et  al. How many erance in adolescent offspring of diabetic mothers: relationship to
sonograms are needed to reliably predict the absence of fetal fetal hyperinsulinism. Diabetes Care. 1995;18(5):611-617.
overgrowth in gestational diabetes mellitus pregnancies? Diabetes 372. Crume TL, Ogden L, West NA, et al. Association of exposure to di-
Care. 2011;34(1):39-43. abetes in utero with adiposity and fat distribution in a multiethnic
352. Kjos SL, Schaefer-Graf UM. Modified therapy for gestational dia- population of youth: the Exploring Perinatal Outcomes among
betes using high-risk and low-risk fetal abdominal circumference Children (EPOCH) Study. Diabetologia. 2011;54(1):87-92.
growth to select strict versus relaxed maternal glycemic targets. 373. West  NA, Crume  TL, Maligie  MA, et  al. Cardiovascular risk
Diabetes Care. 2007;30(suppl 2):S200-S205. factors in children exposed to maternal diabetes in utero.
353. Nelson L, Wharton B, Grobman WA. Prediction of large for gesta- Diabetologia. 2011;54(3):504-507.
tional age birth weights in diabetic mothers based on early third- 374. Hillier TA, Pedula KL, Schmidt MM, et al. Childhood obesity and
trimester sonography. J Ultrasound Med. 2011;30(12):1625-1628. metabolic imprinting: the ongoing effects of maternal hypergly-
354. McLean A, Katz M, Oats J, et al. Rethinking third trimester ul- cemia. Diabetes Care. 2007;30(9):2287-2292.
trasound measurements and risk of adverse neonatal outcomes 375. Whincup PH, Kaye SJ, Owen CG, et al. Birth weight and risk of type
in pregnancies complicated by hyperglycaemia: a retrospective 2 diabetes: a systematic review. JAMA. 2008;300(24):2886-2897.
study. Aust N Z J Obstet Gynaecol. 2021;61(3):366-372. 376. Guerrero-Romero  F, Aradillas-Garcia  C, Simental-Mendia  LE,
355. American College of Obstetricians and Gynecologists. ACOG et al. Birth weight, family history of diabetes, and metabolic syn-
Committee Opinion No. 560: medically indicated late-preterm drome in children and adolescents. J Pediatr. 2010;156(5):719-
and early-term deliveries. Obstet Gynecol. 2013;121(4):908-910. 723, 23 e1.
356. Metcalfe  A, Hutcheon  JA, Sabr  Y, et  al. Timing of delivery in 377. Harder  T, Roepke  K, Diller  N, et  al. Birth weight, early weight
women with diabetes: a population-based study. Acta Obstet gain, and subsequent risk of type 1 diabetes: systematic review
Gynecol Scand. 2020;99(3):341-349. and meta-analysis. Am J Epidemiol. 2009;169(12):1428-1436.
357. Hunt  KJ, Logan  SL, Conway  DL, et  al. Postpartum screening 378. Reece  EA. The fetal and maternal consequences of gestational
following GDM: how well are we doing? Curr Diab Rep. diabetes mellitus. J Matern Fetal Neonatal Med. 2010;23(3):
2010;10(3):235-241. 199-203.
30 Endocrine Reviews, 2022, Vol. XX, No. XX

379. Ornoy A. Prenatal origin of obesity and their complications: ges- 399. Syngelaki A, Pastides A, Kotecha R, et al. First-trimester screening
tational diabetes, maternal overweight and the paradoxical effects for gestational diabetes mellitus based on maternal characteristics
of fetal growth restriction and macrosomia. Reprod Toxicol. and history. Fetal Diagn Ther. 2015;38(1):14-21.
2011;32(2):205-212. 400. Savvidou M, Nelson SM, Makgoba M, et al. First-trimester pre-
380. Page  KA, Romero  A, Buchanan  TA, et  al. Gestational diabetes diction of gestational diabetes mellitus: examining the potential
mellitus, maternal obesity, and adiposity in offspring. J Pediatr. of combining maternal characteristics and laboratory measures.
2014;164(4):807-810. Diabetes. 2010;59(12):3017-3022.
381. Grunnet LG, Hansen S, Hjort L, et al. Adiposity, dysmetabolic traits, 401. Lamain-de Ruiter M, Kwee A, Naaktgeboren CA, et al. Prediction
and earlier onset of female puberty in adolescent offspring of women models for the risk of gestational diabetes: a systematic review.
with gestational diabetes mellitus: a clinical study within the Danish Diagn Progn Res. 2017;1:3.
National Birth Cohort. Diabetes Care. 2017;40(12):1746-1755. 402. Sweeting  AN, Wong  J, Appelblom  H, et  al. A novel early preg-

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022


382. Logan KM, Emsley RJ, Jeffries S, et al. Development of early ad- nancy risk prediction model for gestational diabetes mellitus.
iposity in infants of mothers with gestational diabetes mellitus. Fetal Diagn Ther. 2019;45(2):76-84.
Diabetes Care. 2016;39(6):1045-1051. 403. Sweeting AN, Wong J, Appelblom H, et al. A first trimester pre-
383. Catalano PM, Farrell K, Thomas A, et al. Perinatal risk factors for diction model for gestational diabetes utilizing aneuploidy and
childhood obesity and metabolic dysregulation. Am J Clin Nutr. pre-eclampsia screening markers. J Matern Fetal Neonatal Med.
2009;90(5):1303-1313. 2018;31(16):2122-2130.
384. Chung WK, Erion K, Florez JC, et al. Precision medicine in dia- 404. Aronson JK, Ferner RE. Biomarkers-a general review. Curr Protoc
betes: a consensus report from the American Diabetes Association Pharmacol. 2017;76:9 23 19 17.
(ADA) and the European Association for the Study of Diabetes 405. Allinson  JL. Clinical biomarker validation. Bioanalysis.
(EASD). Diabetes Care. 2020;43(7):1617-1635. 2018;10(12):957-968.
385. Cheney C, Shragg P, Hollingsworth D. Demonstration of hetero- 406. Sattar  N, Wannamethee  SG, Forouhi  NG. Novel biochemical
geneity in gestational diabetes by a 400-kcal breakfast meal toler- risk factors for type 2 diabetes: pathogenic insights or prediction
ance test. Obstet Gynecol. 1985;65(1):17-23. possibilities? Diabetologia. 2008;51(6):926-940.
386. Powe CE, Allard C, Battista MC, et al. Heterogeneous contribu- 407. O’Malley  EG, Reynolds  CME, Killalea  A, et  al. The use of
tion of insulin sensitivity and secretion defects to gestational dia- biomarkers at the end of the second trimester to predict
betes mellitus. Diabetes Care. 2016;39(6):1052-1055. Gestational Diabetes Mellitus. Eur J Obstet Gynecol Reprod Biol.
387. Catalano PM, Tyzbir ED, Roman NM, et al. Longitudinal changes 2020;250:101-106.
in insulin release and insulin resistance in nonobese pregnant 408. Richardson  AC, Carpenter  MW. Inflammatory mediators in
women. Am J Obstet Gynecol. 1991;165(6 Pt 1):1667-1672. gestational diabetes mellitus. Obstet Gynecol Clin North Am.
388. Powe  CE, Hivert  MF, Udler  MS. Defining heterogeneity 2007;34(2):213-224, viii.
among women with gestational diabetes mellitus. Diabetes. 409. Eleftheriades M, Papastefanou I, Lambrinoudaki I, et al. Elevated
2020;69(10):2064-2074. placental growth factor concentrations at 11-14 weeks of ges-
389. Benhalima  K, Van  Crombrugge  P, Moyson  C, et  al. tation to predict gestational diabetes mellitus. Metabolism.
Characteristics and pregnancy outcomes across gestational dia- 2014;63(11):1419-1425.
betes mellitus subtypes based on insulin resistance. Diabetologia. 410. Lovati  E, Beneventi  F, Simonetta  M, et  al. Gestational dia-
2019;62(11):2118-2128. betes mellitus: including serum pregnancy-associated plasma
390. Sweeting  A, Park  F, Hyett  J. The first trimester: prediction and protein-A testing in the clinical management of primipa-
prevention of the great obstetrical syndromes. Best Pract Res Clin rous women? A  case-control study. Diabetes Res Clin Pract.
Obstet Gynaecol. 2015;29(2):183-193. 2013;100(3):340-347.
391. Coustan DR, Nelson C, Carpenter MW, et al. Maternal age and 411. White  SL, Lawlor  DA, Briley  AL, et  al. Early antenatal pre-
screening for gestational diabetes: a population-based study. diction of gestational diabetes in obese women: develop-
Obstet Gynecol. 1989;73(4):557-561. ment of prediction tools for targeted intervention. PLoS One.
392. Lavin JP Jr. Screening of high-risk and general populations for ges- 2016;11(12):e0167846.
tational diabetes. Clinical application and cost analysis. Diabetes. 412. Rasanen JP, Snyder CK, Rao PV, et al. Glycosylated fibronectin as
1985;34(suppl 2):24-27. a first-trimester biomarker for prediction of gestational diabetes.
393. Weeks JW, Major CA, de Veciana M, et al. Gestational diabetes: Obstet Gynecol. 2013;122(3):586-594.
does the presence of risk factors influence perinatal outcome? Am 413. Watanabe  N, Morimoto  S, Fujiwara  T, et  al. Prediction
J Obstet Gynecol. 1994;171(4):1003-1007. of gestational diabetes mellitus by soluble (pro)renin re-
394. Cosson E, Benbara A, Pharisien I, et al. Diagnostic and prognostic ceptor during the first trimester. J Clin Endocrinol Metab.
performances over 9 years of a selective screening strategy for ges- 2013;98(6):2528-2535.
tational diabetes mellitus in a cohort of 18,775 subjects. Diabetes 414. Theriault  S, Giguere  Y, Masse  J, et  al. Early prediction of ges-
Care. 2013;36(3):598-603. tational diabetes: a practical model combining clinical and bio-
395. Chevalier N, Fenichel P, Giaume V, et al. Universal two-step screening chemical markers. Clin Chem Lab Med. 2016;54(3):509-518.
strategy for gestational diabetes has weak relevance in French 415. Syngelaki A, Kotecha R, Pastides A, et al. First-trimester biochem-
Mediterranean women: should we simplify the screening strategy for ical markers of placentation in screening for gestational diabetes
gestational diabetes in France? Diabetes Metab. 2011;37(5):419-425. mellitus. Metabolism. 2015;64(11):1485-1489.
396. Moses  RG, Moses  J, Davis  WS. Gestational diabetes: do lean 416. Gobl CS, Bozkurt L, Rivic P, et al. A two-step screening algorithm
young Caucasian women need to be tested? Diabetes Care. including fasting plasma glucose measurement and a risk estima-
1998;21(11):1803-1806. tion model is an accurate strategy for detecting gestational dia-
397. Avalos  GE, Owens  LA, Dunne  F, et  al. Applying current betes mellitus. Diabetologia. 2012;55(12):3173-3181.
screening tools for gestational diabetes mellitus to a European 417. Australasian Diabetes in Pregnancy Society. Diagnostic testing
population: is it time for change? Diabetes Care. 2013;36(10): for gestational diabetes mellitus (GDM) during the COVID 19
3040-3044. pandemic: antenatal and postnatal testing advice: a statement
398. Teede  HJ, Harrison  CL, Teh  WT, et  al. Gestational diabetes: from the Australasian Diabetes in Pregnancy Society (ADIPS).
development of an early risk prediction tool to facilitate Last updated April 7, 2020. Accessed June 12, 2021. https://www.
opportunities for prevention. Aust N Z J Obstet Gynaecol. adips.org/documents/COVID19-WITHQLDGUIDELINES07042
2011;51(6):499-504. 01150ADIPSADSADEADAupdated.pdf
Endocrine Reviews, 2022, Vol. XX, No. XX 31

418. Royal College of Obstetricians. Guidance for maternal medi- 4 20. McIntyre  HD, Gibbons  KS, Ma  RCW, et  al. Testing for
cine in the evolving coronavirus Covid-19 pandemic. Published gestational diabetes during the COVID-19 pandemic.
July 10, 2020. Accessed June 30, 2021. https://www.rcog.org. An evaluation of proposed protocols for the United
uk/globalassets/documents/guidelines/2020-07-10-guidance-for- Kingdom, Canada and Australia. Diabetes Res Clin Pract.
maternal-medicine.pdf 2020;167:108353.
419. Simmons D, Rudland VL, Wong V, et al. Options for screening for 421. Meek CL, Lindsay RS, Scott EM, et al. Approaches to screening
gestational diabetes mellitus during the SARS-CoV-2 pandemic. for hyperglycaemia in pregnant women during and after the
Aust N Z J Obstet Gynaecol. 2020;60(5):660-666. COVID-19 pandemic. Diabet Med. 2021;38(1):e14380.

Downloaded from https://academic.oup.com/edrv/advance-article/doi/10.1210/endrev/bnac003/6511028 by guest on 15 July 2022

You might also like