Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Ahead-of print > Article
Original Article
Pharmacotherapy Novel Insights into the Causal Relationship between Antidiabetic Drugs and Adverse Perinatal Outcomes: A Mendelian Randomization Study
Chang Suorcid, Xueqing He, Xiaona Chang, Juan Tian, Guang Wangorcidcorresp_icon, Jia Liuorcidcorresp_icon

DOI: https://doi.org/10.4093/dmj.2024.0521
Published online: June 2, 2025
  • 716 Views
  • 56 Download

Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China

corresp_icon Corresponding authors: Guang Wang orcid Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang district, Beijing 100020, China E-mail: drwg6688@126.com
Jia Liu orcid Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang district, Beijing 100020, China E-mail: liujia0116@126.com
• Received: August 30, 2024   • Accepted: January 14, 2025

Copyright © 2025 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Background
    Hyperglycemia during pregnancy increases the risk of adverse perinatal outcomes and birth defects. Evidence regarding the long-term safety of antidiabetic drugs during pregnancy is still lacking.
  • Methods
    A two-sample Mendelian randomization (MR) study was performed to assess the causal association between six antidiabetic drug targets (ABCC8, DPP4, INSR, GLP1R, PPARG, and SLC5A2) and seven adverse perinatal outcomes and five congenital malformation outcomes. Inverse variance weighted (IVW) was adopted as the main MR method, and sensitivity analysis using traditional MR methods was performed to evaluate the robustness of the results.
  • Results
    We observed strong evidence that sodium-glucose cotransporter 2 (SGLT2) inhibitors (odds ratio [OR], 0.084; 95% confidence interval [CI], 0.009 to 0.834; P=0.034) reduces the risk of preterm birth; genetic variation in sulfonylurea drug targets (OR, 0.015; 95% CI, 2.50E-04 to 0.919; P=0.045) and genetic variation in thiazolidinedione drug targets (OR, 0.007; 95% CI, 4.16E-04 to 0.121; P=0.001) reduced the risk of eclampsia/preeclampsia; glucagon-like peptide 1 (GLP-1) analogues target (β=–0.549; 95% CI, –0.958 to –0.140; P=0.009) was inversely associated with fetal birth weight; thiazolidinedione target was inversely associated with gestational age (β=–0.952; 95% CI, –1.785 to –0.118; P=0.025); SGLT2 inhibitors reduced the risk of cardiocirculatory malformations (OR, 0.001; 95% CI, 8.75E-06 to 0.126; P=0.005).
  • Conclusion
    Most antidiabetic drugs are safe when used during the perinatal period. Of note, GLP-1 analogues may lead to a risk of low birth weight, while thiazolidinediones may lead to a reduction in fetal gestational age.
• This study assessed the safety of antidiabetic drugs during the perinatal period.
• Most antidiabetic drugs seem genetically safe, but perinatal adverse risks remain.
• Our findings guide medication use in pregnancy complicated by hyperglycemia.
Hyperglycemia has become one of the most common diseases in pregnancy [1]. According to the survey, 21.1 million (16.7%) women suffered from hyperglycemia during pregnancy in 2021 [2]. Hyperglycemia during pregnancy increases the risk of adverse perinatal outcomes such as low birth weight, preeclampsia, and preterm birth and birth defects [3-5]. Adequate glycemic control can reduce the risk of these adverse perinatal outcomes [6]. Although some patients can maintain expected glycemic control through lifestyle intervention, some patients still fail to reach proper blood glucose and need drug intervention. So far, insulin and metformin are the only two approved glucose-lowering drugs for pregnancy due to the lack of evidence on the long-term safety of oral glucose-lowering drugs during pregnancy [7,8]. Studies have shown that metformin can cross the blood-placental barrier, and no animal studies or clinical trials have found metformin to have teratogenic potential [9].
Mendelian randomization (MR) is a method for causal inference based on genetic variation. Because single nucleotide polymorphism (SNP) alleles segregate randomly following Mendelian laws, MR methods have the advantage of mitigating confounding factors compared to other research methods [10]. Drug target MR analysis has been used in clinical trial design. It has become an effective technique for evaluating the potential risk of drugs, agonists, inhibitors, and antagonists targeting protein-coding genes in related diseases [11]. Therefore, this study aims to comprehensively discuss the relationship between commonly used antidiabetic drug targets and perinatal outcomes (including premature birth, eclampsia/preeclampsia, birth weight, pregnancy duration, postpartum hemorrhage, premature rupture of membranes, spontaneous abortion, and various types of congenital malformations) to evaluate further the safety of the use of antidiabetic drugs in the perinatal period.
Study design
This study adopted a two-sample MR design (Fig. 1). Exposure and outcome data for this design were obtained from two independent, nonoverlapping populations. Proxies for antidiabetic drug use were composed by identifying genetic variants (cis-variants) within genes encoding protein targets for antidiabetic drugs in the genome-wide association study (GWAS) Glycated Hemoglobin Summary Dataset (https://www.ebi.ac.uk/gwas/studies/GCST90002244). To ensure the validity of potential causal effects, MR analyses needed to fulfill three core assumptions: (1) genetic variants are robustly associated with the exposure (relevance); (2) genetic variants are independent of confounders (exchangeability); and (3) genetic variants influence the outcome only through the exposure (exclusion restriction) [12]. This study is reported based on the guidelines outlined in the Strengthening the Reporting of Observational Studies in Epidemiology Using Mendelian Randomization (STROBE-MR) protocol [13].
Selection of genetic instruments
Seven antidiabetic drug targets were identified from Drugbank (https://go.drugbank.com/), including dipeptidyl peptidase 4 (DPP4, DPP4 inhibitors target), insulin receptor (INSR, insulin/insulin analogues target), solute carrier family 5 member 2 (SLC5A2) (sodium-glucose cotransporter 2 [SGLT2] inhibitors target), glucagon-like peptide 1 receptor (GLP1R, GLP-1 analogues target), ATP binding cassette subfamily C member 8 (ABCC8) and potassium inwardly rectifying channel subfamily J member 11 (KCNJ11) (sulfonylureas targets), and peroxisome proliferator-activated receptor gamma (PPARG, thiazolidinediones target). Metformin was excluded from further analysis because its target could not be identified. Since the chromosomal locations of ABCC8 and KCNJ11 are similar, we chose the midpoint between them as the dividing line to distinguish them (Table 1).
We identified cis-expression Quantitative Trait Loci (cis-eQTL) within each coding gene (±1 Mb base pairs of the gene position) and selected those that retained significant correlations with glycosylated hemoglobin levels. Because the retrieved SNPs significantly associated with cis-variants were fewer (less than 3) at the genome-wide significance level P<5×10−8, we gradually extended the P value to 5×10−6, or even 0.05, thereby identifying the SNPs used for analysis [14]. We set the linkage disequilibrium (LD) parameter to r2<0.2 within 100 kb, and SNPs with palindromic structures were deleted to ensure the reliability of the results [11].
The influence of weak instrument bias on the results was eliminated by calculating the F statistic of the selected SNPs and removing SNPs with F<10. If an SNP was not present in the resulting GWAS, it was replaced with an alternative SNP in high LD (r2>0.80) using SNiPA (https://snipa.helmholtzmuenchen.de/snipa3/index.php). If there is no suitable alternative SNP, it is discarded.
Outcome data
To determine the reliability of the selected instrumental variables (IVs), MR analysis was performed using type 2 diabetes mellitus (T2DM) as a positive control. GWAS data for T2DM were obtained from the GWAS Catalog database. Eclampsia/preeclampsia, preterm birth, premature rupture of membranes, postpartum hemorrhage, spontaneous abortion, birth weight, pregnancy duration, and congenital malformations were selected as the primary outcomes of our research. In our study, eclampsia/preeclampsia, premature rupture of membranes, preterm birth, postpartum hemorrhage, and spontaneous abortion GWAS data from the FinnGen database (released 10) [15]. GWAS data for birth weight and gestational age were obtained from the Early Growth Genetics (EGG) Consortium. Considering the widespread harm caused by drug-induced diseases, we included data on congenital malformations on a large scale. Data on various congenital malformations come from the UK Biobank (UKB), including skin malformations, heart and circulatory system malformations, digestive system malformations, genitourinary system malformations, and coagulation malformations. The detailed data information is shown in Supplementary Table 1.
Statistical analysis
We evaluated the effects of antidiabetic drugs on perinatal outcomes and congenital malformations based on a two-sample MR analysis. The inverse variance weighted (IVW) method was mainly used to evaluate the causal relationship between exposure factors and outcome factors [16], and the remaining four methods were used as supplementary analysis, including MR-Egger, weighted median, simple model, and weighted model. The choice of fixed-effects model or random-effects model was determined by heterogeneity test. Specifically, Cochran’s Q test was used to determine whether there was heterogeneity. When the heterogeneity index showed that P<0.05 of the Q test and I2>50%, the random-effects model should be selected because it can provide more precise estimates and confidence intervals (CI) than the fixed-effects IVW method. If no significant heterogeneity was found, the fixed-effects model was used. Furthermore, the MR-Egger intercept test and MR pleiotropy residuals and outliers (MR-PRESSO) were used to assess relative pleiotropy. Outlier SNPs will be detected by MR-PRESSO outlier test and P index level of 0.05.
Sensitivity analysis
We used the intercept term of MR-Egger regression to represent horizontal pleiotropy and performed Cochrane’s Q value to assess the heterogeneity of IVW estimators, with P<0.05 indicating evidence of heterogeneity. In addition, we used MR-PRESSO as a supplement to assess horizontal pleiotropy [17]. MR-PRESSO is designed to detect outlier genetic variants and correct for horizontal multivariate validity by removing outliers. MR-PRESSO can also determine whether the causal effect changes substantially before and after removing outliers in MR analysis.
Ethical statement
Our analysis used publicly available GWAS summary statistics. No new data were collected, and no new ethical approval was required.
Selection and validation of genetic instruments
By applying the thresholds we set in our method analysis, a total of 28 significant cis-eQTL SNPs were included as genetic instruments, including four SNPs representing ABCC8, four SNPs representing DPP4, eight SNPs representing GLP1R, five SNPs representing INSR, and three SNPs representing PPARG and four SNPs representing SLC5A2. Afterward, the influence of weak IVs on the results was eliminated by calculating the F statistic. In this process, the F statistics of the IVs of KCNJ11 were all <10 and therefore were excluded from further analysis. The F statistics of the remaining studied IVs range from 10.016 to 49.904, indicating that our MR analysis is hardly affected by weak instrument bias. The detailed information of all included SNPs are listed in Supplementary Table 2. Then, we performed a two-sample MR analysis using T2DM as a positive control, and the results are shown in Supplementary Table 3. The results showed that all antidiabetic drug target genes involved in this study were significantly associated with a reduced risk of T2DM. No significant multiple effects and heterogeneity were observed in the results.
Impact of genetic variation in antidiabetic drug targets on perinatal outcomes and risk of congenital malformations
In our preliminary analysis using the IVW approach, we observed strong evidence that SGLT2 inhibitors (odds ratio [OR], 0.084; 95% CI, 0.009 to 0.834; P=0.034) reduced the risk of preterm birth. Genetic variation in sulfonylurea drug targets (OR, 0.015; 95% CI, 2.50E-04 to 0.919; P=0.045) and genetic variation in thiazolidinedione drug targets (OR, 0.007; 95% CI, 4.16E-04 to 0.121; P=0.001) reduced the risk of eclampsia/preeclampsia. GLP-1 analogues target (β=–0.549; 95% CI, –0.958 to –0.140; P=0.009) was inversely associated with fetal birth weight, indicating that GLP-1 analogues may cause reduced fetal birth weight. Thiazolidinedione target was inversely associated with gestational age (β=–0.952; 95% CI, –1.785 to –0.118; P=0.025) (Table 2, Figs. 2 and 3).
In the MR analysis of congenital malformations, we found that SGLT2 inhibitors had a positive effect on cardiocirculatory malformations (OR, 0.001; 95% CI, 8.75E-06 to 0.126; P=0.005) (Table 2, Fig. 4). We did not observe significant heterogeneity or multiplicity of effects in our findings.
This study systematically evaluated the relationship between six antidiabetic drugs and multiple perinatal outcomes through drug-targeted MR analysis. We found that SGLT2 inhibitors were associated with a reduced risk of preterm birth and congenital malformations of the cardiocirculatory system. GLP-1 analogues were associated with reduced fetal birth weight. Both sulfonylureas and thiazolidinedione drug targets were associated with reduced risk of eclampsia/preeclampsia. We also found a negative correlation between thiazolidinedione drug targets and gestational age.
GLP-1 receptor agonists (GLP-1RA) is a new antidiabetic drug that maintains blood glucose homeostasis by stimulating insulin secretion from β-cells and inhibiting glucagon release from α-cells [18-20]. The current study found that GLP-1 analogues are generally safe and have no significant impact on perinatal outcomes and fetal congenital malformations. Consistently, a prospective multicenter observational study evaluated pregnancy outcomes after first-trimester exposure to GLP-1RA and found that these drugs did not increase the risk of adverse pregnancy outcomes and birth defects [21]. However, we found that GLP-1 analogues are associated with reduced fetal birth weight. An animal study suggests that maternal exposure to GLP-1RA during pregnancy or lactation is associated with fetal growth retardation and weight loss [22]. This may be because GLP-1 analogues influence appetite control and feelings of fullness by acting on the hypothalamus [23]. They reduce food intake and slow gastric emptying, leading to weight loss [24-26]. Therefore, the application of GLP-1RA should alert the risk of low-birth-weight infants. In addition, SGLT2 inhibitors, another common antidiabetic drug, can effectively reduce blood glucose levels in patients with T2DM by reducing renal tubular glucose reabsorption and have a certain weight loss effect [27]. However, we did not find that SGLT2 inhibitors are associated with low-birth-weight infants. Interestingly, we found that SGLT2 inhibitors were associated with a reduced risk of preterm birth and congenital malformations of the cardiocirculatory system. So far, a series of studies have proven that SGLT2 inhibitors have anti-inflammatory and anti-fibrotic effects [28-30]. Research shows that chronic inflammation throughout the body is a risk factor for premature birth in women [31]. Sulfonylureas are oral antidiabetic drugs whose main mechanism of action is to inhibit adenosine triphosphate-sensitive potassium channels (Katp channels) and increase insulin secretion from pancreatic β-cells [32-34]. We found that sulfonylureas reduced the risk of eclampsia. Previous studies have shown that hyperglycemia increases the production of various metabolic injuries, leading to insulin resistance in the placenta, stimulating the transcription and expression of proinflammatory cytokines, promoting placental cell apoptosis and dysfunction, and finally leading to eclampsia [35,36]. Therefore, effective blood glucose control will reduce the risk of eclampsia. Interestingly, we found that thiazolidinediones can also significantly reduce the risk of eclampsia, which is related to the effect of the drugs themselves on improving insulin resistance. Previous studies have demonstrated that PPAR is an important transcription factor involved in various placental pathways and plays a role in placental development and function [37]. Thiazolidinediones can have beneficial effects on stressed preeclamptic placentas by activating cell growth and anti-oxidative stress pathways, including the heme oxygenase 1 (Hmox1) gene, and restoring pathways disrupted by PPARG in preeclampsia [38]. However, our study also found that thiazolidinediones may lead to a reduction in fetal gestational age. A previous study found that women who carry homozygous variants of the PPARG gene have a 43% chance of being born small for gestational age due to perinatal brain injury [39].
There are few human clinical data on the effects of maternal use of antidiabetic drugs during pregnancy or lactation on the fetus, making it impossible to assess the safety of perinatal use of antidiabetic drugs reliably. GWAS is a hypothesis-free study design used to test the association between thousands or millions of genetic variants and phenotypes [40]. The principal aim of a GWAS is to identify variants associated with phenotypes, whose alleles have been randomly assigned to individuals before any exposure or outcome [40,41]. Compared with traditional regression analysis, MR analysis can avoid the influence of potential confounding factors. Due to the randomness of genetic variation, the distribution of exposure variables (such as a disease, biomarker, or drug effect) is usually independent of the pregnancy status or other similar subgroup divisions of the participants [42]. Therefore, even in different subgroups (such as pregnant and non-pregnant populations), MR methods can still provide robust causal inferences [43,44]. We present the first large-scale study of the association between multiple antidiabetic drugs and adverse perinatal outcomes and fetal congenital malformations. Considering the interference of the mother’s genetic background on the results, we chose data on fetal congenital malformations as the outcome, in which the effects of these SNPs are mainly manifested in fetal physiology and focused more precisely on the genetic background of the fetus [45,46]. We provide evidence that using GLP-1 analogues and thiazolidinediones may contribute to adverse perinatal outcomes. With the increased availability of GWAS data, drug-target MR can effectively identify targets with an efficiency of up to 70% [11]. The results of positive controls found that the genetically predicted drug effects were consistent with clinical evidence and mechanisms, indicating the robustness of the selected IVs [22,47,48]. This concept will give a new direction for choosing medication regimens for patients with hyperglycemia during pregnancy. However, our study also has some limitations. First, the data we selected for congenital malformations of the heart and large vessels include a small amount of non-European population data, which may have confounded the results. However, their proportion in the overall population is minimal, and their impact on the results is negligible. Second, during our investigation of the link between antidiabetic drugs and congenital malformations, we excluded ABCC8 and INSR from the analysis due to the limited number of SNPs and the low reliability of the findings. Finally, MR is limited to predicting adverse events related to the drug mechanism. In contrast, off-target effects that are not related to the drug mechanism of action cannot be predicted by MR [49]. However, drug-target MR was performed using cis-variants, reducing the possibility of horizontal pleiotropy, which are selected in and around drug-target encoding loci [46]. Moreover, we also performed sensitivity analyses using robust methods, including weighted median and MR Egger methods, which further supported the absence of horizontal pleiotropy in these MR analyses [11]. DPP4 inhibitors, insulin/insulin analogues, and thiazolidinediones were found to be possibly associated with an increased risk of spontaneous abortion, although their P were not significant (P>0.05). A more rigorous approach should be to expand the sample size and population groups and conduct an in-depth analysis. Meanwhile, more efforts should be made to combine both laboratory studies and clinical trials to clarify the effect of antidiabetic drugs on adverse perinatal outcomes.
In conclusion, our findings provide new evidence for the perinatal use of antidiabetic medications in pregnant women with hyperglycemia. Most antidiabetic drugs are safe to use during the perinatal period and reduce the risk of adverse perinatal outcomes. Of note, GLP-1 analogues may lea1d to a risk of low birth weight, while thiazolidinediones may lead to a reduction in fetal gestational age.
Supplementary materials related to this article can be found online at https://doi.org/10.4093/dmj.2024.0521.
Supplementary Table 1.
Genome-wide association study summary data and expression quantitative trait loci studies’ data information
dmj-2024-0521-Supplementary-Table-1.pdf
Supplementary Table 2.
Instrumental variables for antidiabetic drugs
dmj-2024-0521-Supplementary-Table-2.pdf
Supplementary Table 3.
MR analysis results of positive control
dmj-2024-0521-Supplementary-Table-3.pdf

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

AUTHOR CONTRIBUTIONS

Conception or design: C.S., G.W., J.L.

Acquisition, analysis, or interpretation of data: C.S., X.H., X.C., J. T.

Drafting the work or revising: C.S.

Final approval of the manuscript: all authors.

FUNDING

This work was supported by grants from the Chinese National Natural Science Foundation (No. 82072527), Beijing Natural Science Foundation (L248018) to Jia Liu and Beijing Hospitals Authority’s Ascent Plan (DFL20220302) to Guang Wang.

ACKNOWLEDGMENTS

The authors appreciate all the subjects involved in this study.

Fig. 1.
Overview of study design and analysis strategy. SNP, single nucleotide polymorphism; PPARG, peroxisome proliferator-activated receptor gamma; SLC5A2, solute carrier family 5 member 2; GLP1R, glucagon-like peptide 1 receptor; DPP4, dipeptidyl peptidase 4; INSR, insulin receptor; ABCC8, ATP binding cassette subfamily C member 8; LD, linkage disequilibrium; IV, instrumental variable; KCNJ11, potassium inwardly rectifying channel subfamily J member 11; HbA1c, glycosylated hemoglobin; MR, Mendelian randomization; T2DM, type 2 diabetes mellitus.
dmj-2024-0521f1.jpg
Fig. 2.
The forest plot of the effects of antidiabetic drugs on adverse perinatal outcomes. Odds ratio (OR), 95% confidence interval (CI), and P values were calculated for the respective method of Mendelian randomization (MR) analysis. P<0.05 was considered significant. nSNP, number of single nucleotide polymorphisms; IVW, inverse variance weighted; DPP4, dipeptidyl peptidase 4; GLP-1, glucagon-like peptide 1; SGLT2, sodium-glucose cotransporter 2.
dmj-2024-0521f2.jpg
Fig. 3.
The forest plot of the effects of antidiabetic drugs on birthweight and gestational age. Odds ratio, 95% confidence interval (CI), and P values were calculated for the respective method of Mendelian randomization (MR) analysis. P<0.05 was considered significant. nSNP, number of single nucleotide polymorphisms; IVW, inverse variance weighted; DPP4, dipeptidyl peptidase 4; GLP-1, glucagon-like peptide 1; SGLT2, sodium-glucose cotransporter 2.
dmj-2024-0521f3.jpg
Fig. 4.
The forest plot of the effects of antidiabetic drugs on congenital malformation. Odds ratio (OR), 95% confidence interval (CI), and P values were calculated for the respective method of Mendelian randomization (MR) analysis. P<0.05 was considered significant. nSNP, number of single nucleotide polymorphisms; SGLT2, sodium-glucose cotransporter 2; IVW, inverse variance weighted; GLP-1, glucagon-like peptide 1; DPP4, dipeptidyl peptidase 4.
dmj-2024-0521f4.jpg
dmj-2024-0521f5.jpg
Table 1.
Summary information of antidiabetic drugs classes, targets, and encoding genes
Drug class Drug target Encoding genes Gene region (in GRCh37 from Ensembl) Included in analysis
Dipeptidyl peptidase 4 (DPP4) inhibitors Dipeptidyl peptidase 4 DPP4 2: 162848755-162930725 Yes
Sodium-glucose cotransporter 2 (SGLT2) inhibitors Sodium/glucose cotransporter 2 SLC5A2 16: 31494444-31502090 Yes
Insulin/insulin analogues Insulin receptor INSR 19: 7112276-7294425 Yes
Glucagon-like peptide 1 (GLP-1) analogues Glucagon-like peptide 1 receptor GLP1R 6: 39016557-39059079 Yes
Sulfonylureas ATP-sensitive potassium channel KCNJ11 11: 17406795-17410893 No
ABCC8 11: 17414045-17498392 Yes
Thiazolidinediones Peroxisome proliferator-activated receptor gamma PPARG 3: 12328867-12475843 Yes
Table 2.
Results of two-sample MR analysis of antidiabetic drugs and adverse pregnancy outcomes
Drug class Proxy genes Outcome Method nSNP P value OR/β (95% CI) Cochran Q value Q-P value MR-Egger intercept P intercept
Sulfonylureas ABCC8 Eclampsia/preeclampsia IVW 3 0.045 0.015 (0.00025 to 0.919) 0.313 0.855 2.30E-02 0.695
GLP-1 analogues GLP1R Birthweight IVW 8 0.009 –0.549 (–0.958 to –0.140) 6.005 0.539 –3.40E-03 0.311
Thiazolidinediones PPARG Eclampsia/preeclampsia IVW 3 0.001 0.007 (0.000416 to 0.121) 1.789 0.409 7.43E-02 0.418
Thiazolidinediones PPARG Gestational age IVW 3 0.025 –0.952 (–1.785 to –0.118) 0.005 0.997 –6.06E-04 0.977
SGLT2 inhibitors SLC5A2 Preterm birth IVW 4 0.034 0.084 (0.009 to 0.834) 0.782 0.854 8.40E-02 0.705
SGLT2 inhibitors SLC5A2 Congenital anomalies of the heart and circulatory system IVW 4 0.005 0.001 (0.000009 to 0.126) 5.278 0.153 –2.05E-01 0.709

OR, 95% CI, and P values were calculated for the respective method of MR analysis.

MR, Mendelian randomization; nSNP, number of single nucleotide polymorphisms; OR, odds ratio; CI, confidence interval; IVW, inverse variance weighting; GLP-1, glucagon-like peptide 1; SGLT2, sodium-glucose cotransporter 2.

  • 1. Damm P, Houshmand-Oeregaard A, Kelstrup L, Lauenborg J, Mathiesen ER, Clausen TD. Gestational diabetes mellitus and long-term consequences for mother and offspring: a view from Denmark. Diabetologia 2016;59:1396-9.ArticlePubMedPDF
  • 2. Malaza N, Masete M, Adam S, Dias S, Nyawo T, Pheiffer C. A systematic review to compare adverse pregnancy outcomes in women with pregestational diabetes and gestational diabetes. Int J Environ Res Public Health 2022;19:10846.ArticlePubMedPMC
  • 3. Kelly RS, Giorgio RT, Chawes BL, Palacios NI, Gray KJ, Mirzakhani H, et al. Applications of metabolomics in the study and management of preeclampsia: a review of the literature. Metabolomics 2017;13:86.ArticlePubMedPMCPDF
  • 4. Gilbert L, Gross J, Lanzi S, Quansah DY, Puder J, Horsch A. How diet, physical activity and psychosocial well-being interact in women with gestational diabetes mellitus: an integrative review. BMC Pregnancy Childbirth 2019;19:60.ArticlePubMedPMCPDF
  • 5. Perez-Perez A, Vilarino-Garcia T, Guadix P, Duenas JL, Sanchez-Margalet V. Leptin and nutrition in gestational diabetes. Nutrients 2020;12:1970.ArticlePubMedPMC
  • 6. Landon MB, Spong CY, Thom E, Carpenter MW, Ramin SM, Casey B, et al. A multicenter, randomized trial of treatment for mild gestational diabetes. N Engl J Med 2009;361:1339-48.ArticlePubMedPMC
  • 7. American Diabetes Association Professional Practice Committee. 15. Management of diabetes in pregnancy: standards of care in diabetes-2024. Diabetes Care 2024;47(Suppl 1):S282-94.PubMed
  • 8. Fan X, Wang C, Chen L, Zhao X, Wu Y, Zeng G. Improved pregnancy outcome in gestational diabetes mellitus patients treated with insulin aspart and metformin: a comparative study. Am J Transl Res 2024;16:1177-87.ArticlePubMedPMC
  • 9. Sciacca L, Bianchi C, Burlina S, Formoso G, Manicardi E, Sculli MA, et al. Position paper of the Italian Association of Medical Diabetologists (AMD), Italian Society of Diabetology (SID), and the Italian Study Group of Diabetes in pregnancy: metformin use in pregnancy. Acta Diabetol 2023;60:1421-37.ArticlePubMedPMCPDF
  • 10. Davies NM, Holmes MV, Davey Smith G. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ 2018;362:k601.ArticlePubMedPMC
  • 11. Schmidt AF, Finan C, Gordillo-Maranon M, Asselbergs FW, Freitag DF, Patel RS, et al. Genetic drug target validation using Mendelian randomisation. Nat Commun 2020;11:3255.ArticlePubMedPMCPDF
  • 12. Emdin CA, Khera AV, Kathiresan S. Mendelian randomization. JAMA 2017;318:1925-6.ArticlePubMed
  • 13. Skrivankova VW, Richmond RC, Woolf BA, Yarmolinsky J, Davies NM, Swanson SA, et al. Strengthening the Reporting of Observational Studies in Epidemiology Using Mendelian Randomization: the STROBE-MR statement. JAMA 2021;326:1614-21.ArticlePubMed
  • 14. Zhu Y, Li M, Wang H, Yang F, Pang X, Du R, et al. Genetically proxied antidiabetic drugs targets and stroke risk. J Transl Med 2023;21:681.ArticlePubMedPMCPDF
  • 15. Kurki MI, Karjalainen J, Palta P, Sipila TP, Kristiansson K, Donner KM, et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature 2023;613:508-18.PubMedPMC
  • 16. Bowden J, Del Greco MF, Minelli C, Davey Smith G, Sheehan N, Thompson J. A framework for the investigation of pleiotropy in two-sample summary data Mendelian randomization. Stat Med 2017;36:1783-802.ArticlePubMedPMCPDF
  • 17. Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet 2018;50:693-8.ArticlePubMedPMCPDF
  • 18. Michalowska J, Miller-Kasprzak E, Bogdanski P. Incretin hormones in obesity and related cardiometabolic disorders: the clinical perspective. Nutrients 2021;13:351.ArticlePubMedPMC
  • 19. Holst JJ, Vilsboll T, Deacon CF. The incretin system and its role in type 2 diabetes mellitus. Mol Cell Endocrinol 2009;297:127-36.ArticlePubMed
  • 20. Kim W, Egan JM. The role of incretins in glucose homeostasis and diabetes treatment. Pharmacol Rev 2008;60:470-512.ArticlePubMed
  • 21. Dao K, Shechtman S, Weber-Schoendorfer C, Diav-Citrin O, Murad RH, Berlin M, et al. Use of GLP1 receptor agonists in early pregnancy and reproductive safety: a multicentre, observational, prospective cohort study based on the databases of six Teratology Information Services. BMJ Open 2024;14:e083550.ArticlePubMedPMC
  • 22. Muller DR, Stenvers DJ, Malekzadeh A, Holleman F, Painter RC, Siegelaar SE. Effects of GLP-1 agonists and SGLT2 inhibitors during pregnancy and lactation on offspring outcomes: a systematic review of the evidence. Front Endocrinol (Lausanne) 2023;14:1215356.ArticlePubMedPMC
  • 23. Abdullah Bin Ahmed I. A comprehensive review on weight gain following discontinuation of glucagon-like peptide-1 receptor agonists for obesity. J Obes 2024;2024:8056440.PubMedPMC
  • 24. Farr OM, Sofopoulos M, Tsoukas MA, Dincer F, Thakkar B, Sahin-Efe A, et al. GLP-1 receptors exist in the parietal cortex, hypothalamus and medulla of human brains and the GLP-1 analogue liraglutide alters brain activity related to highly desirable food cues in individuals with diabetes: a crossover, randomised, placebo-controlled trial. Diabetologia 2016;59:954-65.ArticlePubMedPMCPDF
  • 25. Tamayo-Trujillo R, Ruiz-Pozo VA, Cadena-Ullauri S, Guevara-Ramirez P, Paz-Cruz E, Zambrano-Villacres R, et al. Molecular mechanisms of semaglutide and liraglutide as a therapeutic option for obesity. Front Nutr 2024;11:1398059.ArticlePubMedPMC
  • 26. Rowlands J, Heng J, Newsholme P, Carlessi R. Pleiotropic effects of GLP-1 and analogs on cell signaling, metabolism, and function. Front Endocrinol (Lausanne) 2018;9:672.ArticlePubMedPMC
  • 27. Huttasch M, Roden M, Kahl S. Obesity and MASLD: is weight loss the (only) key to treat metabolic liver disease? Metabolism 2024;157:155937.ArticlePubMed
  • 28. Mancini SJ, Boyd D, Katwan OJ, Strembitska A, Almabrouk TA, Kennedy S, et al. Canagliflozin inhibits interleukin-1β-stimulated cytokine and chemokine secretion in vascular endothelial cells by AMP-activated protein kinase-dependent and -independent mechanisms. Sci Rep 2018;8:5276.ArticlePubMedPMCPDF
  • 29. Lu Q, Liu J, Li X, Sun X, Zhang J, Ren D, et al. Empagliflozin attenuates ischemia and reperfusion injury through LKB1/AMPK signaling pathway. Mol Cell Endocrinol 2020;501:110642.ArticlePubMed
  • 30. Ye Y, Bajaj M, Yang HC, Perez-Polo JR, Birnbaum Y. SGLT-2 inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with type 2 diabetes: further augmentation of the effects with saxagliptin, a DPP4 inhibitor. Cardiovasc Drugs Ther 2017;31:119-32.ArticlePubMedPDF
  • 31. Cheng H, Zhu Z, Chi P, Alifu X, Zhuang Y, Si S, et al. Association of systemic chronic inflammation during pregnancy in different periods and its trajectories with preterm birth. Am J Reprod Immunol 2024;91:e13848.ArticlePubMed
  • 32. Peterson SM, Juliana CA, Hu CF, Chai J, Holliday C, Chan KY, et al. Optimization of a glucagon-like peptide 1 receptor antagonist antibody for treatment of hyperinsulinism. Diabetes 2023;72:1320-9.ArticlePubMedPMCPDF
  • 33. Hou T, Chen L. Sulfonylurea receptor 2 (SUR2), intricate sensors for intracellular Mg-nucleotides. Bioessays 2024;46:e2300151.ArticlePubMed
  • 34. Rubaiy HN. The therapeutic agents that target ATP-sensitive potassium channels. Acta Pharm 2016;66:23-34.ArticlePubMed
  • 35. Wang H, Zhou W, She G, Yu B, Sun L. Downregulation of hsa_circ_0005243 induces trophoblast cell dysfunction and inflammation via the β-catenin and NF-κB pathways. Reprod Biol Endocrinol 2020;18:51.ArticlePubMedPMCPDF
  • 36. Ludidi A, Siboto A, Nkosi A, Xulu ND, Khathi A, Sibiya NH, et al. High-fat, high-carbohydrate diet-induced prediabetes preconception in Sprague-Dawley rats as a risk factor for the development of preeclampsia: assessing changes in placental metabolic insults. Front Nutr 2023;10:1241785.ArticlePubMedPMC
  • 37. Silva A, Motta A. Peroxisome proliferator-activated receptors and placental function in women with polycystic ovary syndrome. Reproduction 2024;167:e230325.ArticlePubMed
  • 38. Grimaldi B, Kohan-Ghadr HR, Halari CD, Nandi P, Kingdom JC, Drewlo S. Rosiglitazone-mediated activation of PPARγ restores HO1 expression in the human preeclamptic placenta. Hypertension 2023;80:2386-96.ArticlePubMed
  • 39. Adibi JJ, Zhao Y, Koistinen H, Mitchell RT, Barrett ES, Miller R, et al. Molecular pathways in placental-fetal development and disruption. Mol Cell Endocrinol 2024;581:112075.ArticlePubMed
  • 40. Larsson SC, Butterworth AS, Burgess S. Mendelian randomization for cardiovascular diseases: principles and applications. Eur Heart J 2023;44:4913-24.ArticlePubMedPMCPDF
  • 41. Sekula P, Del Greco MF, Pattaro C, Kottgen A. Mendelian randomization as an approach to assess causality using observational data. J Am Soc Nephrol 2016;27:3253-65.ArticlePubMedPMC
  • 42. Rogne T, Gill D, Liew Z, Shi X, Stensrud VH, Nilsen TI, et al. Mediating factors in the association of maternal educational level with pregnancy outcomes: a Mendelian randomization study. JAMA Netw Open 2024;7:e2351166.ArticlePubMedPMC
  • 43. Wang R, Zhao J, Li L, Huo Y. Associations between lipid-lowering drugs and pregnancy and perinatal outcomes: a Mendelian randomization study. J Hypertens 2024;42:727-34.ArticlePubMed
  • 44. Ma Y, Cai J, Liu LW, Wen T, Huang W, Hou W, et al. Causal relationships exist between polycystic ovary syndrome and adverse pregnancy and perinatal outcomes: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024;15:1327849.ArticlePubMedPMC
  • 45. Yang K, Qin X, Wang H, Zhou W. Exploring genetic causal relationship between antidepressants use and congenital malformations. Reprod Toxicol 2024;130:108736.ArticlePubMed
  • 46. Ardissino M, Slob EA, Reddy RK, Morley AP, Schuermans A, Hill P, et al. Genetically proxied low-density lipoprotein cholesterol lowering via PCSK9-inhibitor drug targets and risk of congenital malformations. Eur J Prev Cardiol 2024;31:955-65.ArticlePubMedPMCPDF
  • 47. Tang B, Wang Y, Jiang X, Thambisetty M, Ferrucci L, Johnell K, et al. Genetic variation in targets of antidiabetic drugs and Alzheimer disease risk: a Mendelian randomization study. Neurology 2022;99:e650-9.PubMedPMC
  • 48. Cesta CE, Rotem R, Bateman BT, Chodick G, Cohen JM, Furu K, et al. Safety of GLP-1 receptor agonists and other second-line antidiabetics in early pregnancy. JAMA Intern Med 2024;184:144-52.ArticlePubMed
  • 49. Ference BA, Ray KK, Catapano AL, Ference TB, Burgess S, Neff DR, et al. Mendelian randomization study of ACLY and cardiovascular disease. N Engl J Med 2019;380:1033-42.ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      • PubReader PubReader
      • ePub LinkePub Link
      • Cite this Article
        Cite this Article
        export Copy Download
        Close
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        Novel Insights into the Causal Relationship between Antidiabetic Drugs and Adverse Perinatal Outcomes: A Mendelian Randomization Study
        Close
      • XML DownloadXML Download
      Figure
      • 0
      • 1
      • 2
      • 3
      • 4
      Novel Insights into the Causal Relationship between Antidiabetic Drugs and Adverse Perinatal Outcomes: A Mendelian Randomization Study
      Image Image Image Image Image
      Fig. 1. Overview of study design and analysis strategy. SNP, single nucleotide polymorphism; PPARG, peroxisome proliferator-activated receptor gamma; SLC5A2, solute carrier family 5 member 2; GLP1R, glucagon-like peptide 1 receptor; DPP4, dipeptidyl peptidase 4; INSR, insulin receptor; ABCC8, ATP binding cassette subfamily C member 8; LD, linkage disequilibrium; IV, instrumental variable; KCNJ11, potassium inwardly rectifying channel subfamily J member 11; HbA1c, glycosylated hemoglobin; MR, Mendelian randomization; T2DM, type 2 diabetes mellitus.
      Fig. 2. The forest plot of the effects of antidiabetic drugs on adverse perinatal outcomes. Odds ratio (OR), 95% confidence interval (CI), and P values were calculated for the respective method of Mendelian randomization (MR) analysis. P<0.05 was considered significant. nSNP, number of single nucleotide polymorphisms; IVW, inverse variance weighted; DPP4, dipeptidyl peptidase 4; GLP-1, glucagon-like peptide 1; SGLT2, sodium-glucose cotransporter 2.
      Fig. 3. The forest plot of the effects of antidiabetic drugs on birthweight and gestational age. Odds ratio, 95% confidence interval (CI), and P values were calculated for the respective method of Mendelian randomization (MR) analysis. P<0.05 was considered significant. nSNP, number of single nucleotide polymorphisms; IVW, inverse variance weighted; DPP4, dipeptidyl peptidase 4; GLP-1, glucagon-like peptide 1; SGLT2, sodium-glucose cotransporter 2.
      Fig. 4. The forest plot of the effects of antidiabetic drugs on congenital malformation. Odds ratio (OR), 95% confidence interval (CI), and P values were calculated for the respective method of Mendelian randomization (MR) analysis. P<0.05 was considered significant. nSNP, number of single nucleotide polymorphisms; SGLT2, sodium-glucose cotransporter 2; IVW, inverse variance weighted; GLP-1, glucagon-like peptide 1; DPP4, dipeptidyl peptidase 4.
      Graphical abstract
      Novel Insights into the Causal Relationship between Antidiabetic Drugs and Adverse Perinatal Outcomes: A Mendelian Randomization Study
      Drug class Drug target Encoding genes Gene region (in GRCh37 from Ensembl) Included in analysis
      Dipeptidyl peptidase 4 (DPP4) inhibitors Dipeptidyl peptidase 4 DPP4 2: 162848755-162930725 Yes
      Sodium-glucose cotransporter 2 (SGLT2) inhibitors Sodium/glucose cotransporter 2 SLC5A2 16: 31494444-31502090 Yes
      Insulin/insulin analogues Insulin receptor INSR 19: 7112276-7294425 Yes
      Glucagon-like peptide 1 (GLP-1) analogues Glucagon-like peptide 1 receptor GLP1R 6: 39016557-39059079 Yes
      Sulfonylureas ATP-sensitive potassium channel KCNJ11 11: 17406795-17410893 No
      ABCC8 11: 17414045-17498392 Yes
      Thiazolidinediones Peroxisome proliferator-activated receptor gamma PPARG 3: 12328867-12475843 Yes
      Drug class Proxy genes Outcome Method nSNP P value OR/β (95% CI) Cochran Q value Q-P value MR-Egger intercept P intercept
      Sulfonylureas ABCC8 Eclampsia/preeclampsia IVW 3 0.045 0.015 (0.00025 to 0.919) 0.313 0.855 2.30E-02 0.695
      GLP-1 analogues GLP1R Birthweight IVW 8 0.009 –0.549 (–0.958 to –0.140) 6.005 0.539 –3.40E-03 0.311
      Thiazolidinediones PPARG Eclampsia/preeclampsia IVW 3 0.001 0.007 (0.000416 to 0.121) 1.789 0.409 7.43E-02 0.418
      Thiazolidinediones PPARG Gestational age IVW 3 0.025 –0.952 (–1.785 to –0.118) 0.005 0.997 –6.06E-04 0.977
      SGLT2 inhibitors SLC5A2 Preterm birth IVW 4 0.034 0.084 (0.009 to 0.834) 0.782 0.854 8.40E-02 0.705
      SGLT2 inhibitors SLC5A2 Congenital anomalies of the heart and circulatory system IVW 4 0.005 0.001 (0.000009 to 0.126) 5.278 0.153 –2.05E-01 0.709
      Table 1. Summary information of antidiabetic drugs classes, targets, and encoding genes

      Table 2. Results of two-sample MR analysis of antidiabetic drugs and adverse pregnancy outcomes

      OR, 95% CI, and P values were calculated for the respective method of MR analysis.

      MR, Mendelian randomization; nSNP, number of single nucleotide polymorphisms; OR, odds ratio; CI, confidence interval; IVW, inverse variance weighting; GLP-1, glucagon-like peptide 1; SGLT2, sodium-glucose cotransporter 2.

      Su C, He X, Chang X, Tian J, Wang G, Liu J. Novel Insights into the Causal Relationship between Antidiabetic Drugs and Adverse Perinatal Outcomes: A Mendelian Randomization Study. Diabetes Metab J. 2025 Jun 2. doi: 10.4093/dmj.2024.0521. Epub ahead of print.
      Received: Aug 30, 2024; Accepted: Jan 14, 2025
      DOI: https://doi.org/10.4093/dmj.2024.0521.

      Diabetes Metab J : Diabetes & Metabolism Journal
      Close layer
      TOP