Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

A genetically supported drug repurposing pipeline for diabetes treatment using electronic health records

Megan M. Shuey, Kyung Min Lee, Jacob Keaton, Nikhil K. Khankari, Joseph H. Breeyear, View ORCID ProfileVenexia M. Walker, Donald R. Miller, Kent R. Heberer, Peter D. Reaven, View ORCID ProfileShoa L. Clarke, Jennifer Lee, View ORCID ProfileJulie A. Lynch, View ORCID ProfileMarijana Vujkovic, Todd L. Edwards
doi: https://doi.org/10.1101/2022.12.14.22283414
Megan M. Shuey
1Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
2Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kyung Min Lee
3VA Informatics and Computer Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jacob Keaton
4Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
5Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Nikhil K. Khankari
1Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
2Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joseph H. Breeyear
2Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
6Nashville VA Medical Center, Nashville, TN, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Venexia M. Walker
7Medical Research Council, Integrative Epidemiology Unit, University of Bristol, Bristol, UK
8Bristol Medical School: Population Health Sciences, University of Bristol, Bristol, UK
9Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Venexia M. Walker
Donald R. Miller
10Center for Healthcare Organization and Implementation Research, Bedford VA Healthcare System, Bedford, MA, USA
11Center for Population Health, Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kent R. Heberer
12VA Palo Alto Health Care System, Palo Alto, CA, USA
13Departments of Medicine and Endocrinology, Stanford University School of Medicine, Stanford, CA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Peter D. Reaven
14Phoenix VA Health Care System, Phoenix, AZ, USA
15College of Medicine, University of Arizona, Phoenix, AZ, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shoa L. Clarke
16Departments of Medicine and Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Shoa L. Clarke
Jennifer Lee
12VA Palo Alto Health Care System, Palo Alto, CA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Julie A. Lynch
3VA Informatics and Computer Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
17School of Medicine, University of Utah, Salt Lake City, UT
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Julie A. Lynch
Marijana Vujkovic
18Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
19Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Marijana Vujkovic
  • For correspondence: todd.l.edwards{at}vumc.org vujkovic{at}pennmedicine.upenn.edu
Todd L. Edwards
2Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
5Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
6Nashville VA Medical Center, Nashville, TN, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: todd.l.edwards{at}vumc.org vujkovic{at}pennmedicine.upenn.edu
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Supplementary material
  • Data/Code
  • Preview PDF
Loading

Abstract

Objectives The identification of novel uses for existing drug therapies has the potential to provide a rapid, low-cost approach to drug (re)discovery. In the current study we developed and tested a genetically-informed drug-repurposing pipeline for diabetes management.

Design We developed and tested a genetically-informed drug-repurposing pipeline for diabetes management. This approach mapped genetically predicted gene expression signals from the largest genome-wide association study for type 2 diabetes mellitus to drug targets using publicly available databases to identify drug-gene pairs. These drug-gene pairs were then validated using a two-step approach: 1) a self-controlled case-series (SCCS) using electronic health records from a discovery and replication population, and 2) Mendelian randomization (MR).

Setting The SCCS experiments were completed using two EHRs: the Million Veterans Program (USA) as the discovery and the Vanderbilt University Medical Center (Tennessee, USA) as the replication.

Results After filtering on sample size, 20 candidate drug-gene pairs were validated and various medications demonstrated evidence of glycemic regulation including two anti-hypertensive classes: angiotensin-converting enzyme inhibitors as well as calcium channel blockers (CCBs). The CCBs demonstrated the strongest evidence of glycemic reduction in both validation approaches (SCCS HbA1c and glucose reduction: -0.11%, p=0.01 and -0.85 mg/dL, p=0.02, respectively; MR: OR=0.84, 95% CI=0.81, 0.87, p=5.0×10-25).

Conclusions Our results support CCBs as a strong candidate medication for blood glucose reduction in addition to cardiovascular disease reduction. Further, these results support the adaptation of this approach for use in future drug-repurposing efforts for other conditions.

Section 1: What is already known on this topic Medications with genetic support are significantly more likely to make it through clinical trials.

Section 2: What this study adds Our results identified two anti-hypertensive medication classes, calcium channel blockers and angiotensin-converting enzyme inhibitors, as genetically supported drug-repurposing targets that demonstrated glycemic measurement reduction in real-world clinical populations. These results suggest patients with diabetes or pre-diabetes could benefit from preferential use of these medication classes when they present with comorbid hypertension or other cardiovascular conditions. Finally, this study demonstrates a successful implementation of a novel genetically-supported drug-repurposing pipeline for diabetes treatment that can be readily adapted and applied to other diseases and as such it has the potential to identify/prioritize drug repurposing targets for these other conditions.

Introduction

An estimated 463 million individuals globally have a diabetes diagnosis1 and by 2045 this number is expected to reach 700 million. These rising case numbers are primarily due to type 2 diabetes (T2D) cases, the most prevalent form of the disease1. Major comorbidities of T2D include heart disease, peripheral artery disease, stroke, eye disease, kidney disease, and neuropathy2-4.

Early stages of glycemic dysregulation can be effectively managed with behavioral modifications and metformin monotherapy. However, as T2D progresses, the concurrent use of multiple medications acting on distinct pathophysiologic pathways is often needed to control blood glucose levels and reduce development of complications as T2D progresses5-8. Regardless, a high proportion of patients with T2D demonstrate poor or inadequate glycemic control9.

The reason for poor glycemic control is multifactorial. These may include poor medication adherence due to undesired side-effects and costs10,11, reduced treatment efficacy12,13, and ineffective management of secondary metabolic and glucoregulatory dysfunction14,15. Efforts to improve glycemic control include outreach to improve understanding of disease and development of new medications that target glucoregulatory dysfunctions to slow disease progression16-18. T2D and comorbidities are also often treated separately, which may lead to increased morbidity and mortality due to polypharmacy19.

Drug repurposing provides an efficient, cost-effective means to increase therapeutic options by identifying medications that are currently approved for other indications for treatment of a disease20. An example of this is acetylsalicylic acid, marketed as the analgesic Aspirin in 1899, which was subsequently discovered to both inhibit platelet aggregation21 and lower glucose22. The advance of high-throughput technologies, such as genomics and transcriptomics, supports the development of new computational approaches for drug repurposing and identification of possible adverse drug events23. These techniques leverage the highly druggable nature of human disease genes24 which may be implicated in diseases other than those indicated for derived medications25-27.

Serendipitous discoveries, usually a consequence of clinical observations of patients being treated for other conditions28, have historically driven drug repurposing. Findings from real-world observations, however, are subject to biases, such as confounding by indication, reverse causality, and selective data missingness29. Mendelian randomization (MR) may overcome some of the limitations of observational epidemiology29. Here we propose a computational drug repurposing approach that identifies potential therapeutic candidates for T2D by 1) applying computational methods to genome-wide association study (GWAS) results, 2) evaluating these candidates through a observational self-controlled case series conducted in the electronic health records (EHR) using serendipitous clinical observations in the Department of Veterans Affairs (VA) Corporate Data Warehouse and Vanderbilt University Medical Center’s synthetic derivative (VUMC SD), and 3) proxying the candidate’s drug exposure as predicted expression of the therapeutic gene target using S-PrediXcan in a MR analysis.

Methods

Gene-based medication discovery

As described previously30, we used summary statistics from the largest multi-ethnic GWAS of T2D, which included over 1.4 million participants, to estimate genetically predicted gene expression (GPGE) of individual genes and performed a transcriptome-wide association study (TWAS) using s-PrediXcan. The imputation of GPGE was based on version 7 predictors for 52 tissues including 48 from GTEx, two kidney tissues (glomerulus and tubule)31, and two tissues from an alpha and beta islet cell reference32 and signals were refined using colocalization analyses.30 This analysis identified 695 significant genes that we mapped to multiple drug targets using publicly available databases: ChEMBL33, Drug Gene Interaction Database (DGIdb)34, and National Cancer Institute Drug Dictionary35. The mapped genes were further refined by pairing the direction of GPGE and T2D risk with drug effects. For example, genes with increased GPGE that were associated with a decrease in T2D risk would need to map to a drug that acts as an activator or agonist of the associated protein or pathway.

Conversely, drugs that act as an antagonist, inhibitor or blocker would correspond with a gene for which increasing GPGE associated with an increase in T2D risk. This process is visualized in Figure 1, steps 1-4. Drug-gene pairs with correlated functions that were not identified previously for use in diabetes management were then included as experimental medications for the subsequent analyses to evaluate their potential for repurposing including a self-controlled case series and MR studies.

Figure 1.
  • Download figure
  • Open in new tab
Figure 1. Study design.

Steps 1-4 describe the process of identifying novel drug targets based on genetic evidence using a previously published genome-wide association study for Type 2 Diabetes Mellitus. Following identification of potential medications based on drug-gene pairing the results are validated using two approaches, self-controlled case series and mendelian randomization (MR). The general design of these two approaches are described briefly.

Self-controlled case series data sources

Two separate EHR systems were identified as data sources for the self-controlled case series. The discovery population included clinical, prescription, and laboratory data from the Corporate Data Warehouse (CDW) of the Veterans Health Administration (VHA), a national US data repository that provides access to the EHRs of all individuals who received care in the VHA. All study variables were extracted from the CDW in April 2020 by an experienced programmer on Microsoft SQL Server via the VA Informatics and Computing Infrastructure (VINCI) computing environment36.

The replication site data were collected from the VUMC SD, a de-identified copy of the electronic medical record with Health Insurance Portability and Accountability Act of 1996 (HIPAA) identifiers removed37. The SD contains clinical data on approximately 3.2 million individuals and includes basic demographics; text from clinical care notes; laboratory values; inpatient and outpatient medication data; International Classification of Disease (ICD) and Current Procedural Terminology (CPT) codes; and other diagnostic reports. Drug exposures were identified using previously described electronic-prescribing tools and MedEx38. The utility of these methods for medication extraction from the EHR have been shown previously39,40. All valid medication exposures required one of the following indications to be documented: route, frequency, dose, or duration. All study variables were extracted from the SD by an experienced programmer in January 2020.

Self-controlled case series study design

We utilized a self-controlled case series study design within the discovery and replication populations to evaluate the effects of novel medication use on hemoglobin A1c (HbA1c) and glucose. The design varied slightly between the two sites to accommodate variations in data availability and population. These variances are discussed below. The general design of this study is displayed in Figure 2.

Figure 2.
  • Download figure
  • Open in new tab
Figure 2. Examples of the medication exposure time period for the self-controlled case series for determination of patient inclusion or exclusion.

Patient A demonstrates a patient that would be included in the study based on study design. Specifically, this patient was prescribed a medication belonging to the experimental group and had a subsequent mention of the medication in their records in the following 6 months. They had no documented exposure to a medication belonging to one of the other medication groups in the period preceding t0. They also had glucose and hemoglobin A1c (HbA1c) measures collected in the six months before medication exposure and during the response periods. Conversely, patients B and C were excluded from this study. Patient B represents patients that were excluded from the study because they had a medication exposure from another medication group prior to t0. Patient C is an example of a patient that would be excluded due to insufficient data. In this example, Patient C had no glucose and HbA1c measurements in the six-month exposure period. Likewise, patients without a glucose or HbA1c in the six months before t0 would also be excluded from the study due to insufficient laboratory exposure.

Medications evaluated in this series were grouped in to three sets: 1) experimental, the gene-based medication set described previously; 2) diabetes/control (−), medications that are prescribed for the treatment of T2DM; and 3) control (+), medications belonging to classes that had previously described increasing effects on glucose or HbA1c or were implicated in a previous MR study41. The complete list of medications and their corresponding set are available in Supplemental Tables 1a and b. To ensure the effects on glucose or HbA1c were due to a specific medication, each individual medication series included patients that were prescribed the specific medication when the drug was first prescribed, t0, but excluded patients prescribed a medication in one of the other groups ever before, simultaneously at t0, or in the six-month follow-up period.

Further, in the discovery set the VA patients were only included in a medication series if they received at least one subsequent refill of the given medication within 180 days of t0, and had at least 90 days of cumulative exposure. Because records of a patient’s prescription fill is less readily available in the VUMC SD, e.g. many patients will fill their prescriptions at a non-VUMC pharmacy so records of prescription pick-ups are absent in their record, this study required included subject EHRs in VUMC SD to have at least two mentions of the medication within 180 days of the initial drug mention, t0, to proxy prescription refill.

Finally, all patients included in the medication series were required to have both a baseline and follow-up HbA1c and glucose measured to evaluate response. HbA1c and glucose measures were restricted to outpatient measures and non-physiologic measures were excluded, e.g. HbA1c less than 3% or greater than 18% and glucose less than 5 mg/dL or greater than 2,750 mg/dL. In the replication population we also excluded all laboratory measures that were collected within 9 months of a pregnancy ICD code or laboratory test (Supplemental Table 2). Because the discovery cohort is >90% male this exclusion was not applied.

The change in laboratory measure was defined as absolute change. The absolute change was calculated by taking the difference between the follow-up measure and the baseline measure. Baseline HbA1c was defined as the most recent measure in the six months prior to drug initiation. Follow-up HbA1c measure was defined as the first measure after 30 days and within the six months following drug initiation (Figure 2). For glucose, we used the mean of all measures in the six months prior to drug initiation as the baseline value and the mean of all measures in the six months after drug initiation as the follow-up value.

Statistical Analysis

We assessed patient characteristics at drug initiation by comparing demographics, smoking status, body mass index, glycemic status, and Charlson Comorbidity Index categories across the three drug groups for both study populations. Using a self-controlled case series design, we performed pairwise t-test to determine statistical significance of the change in laboratory measures. Drugs with a sample size less than 30 in the discovery population were not analyzed to increase precision. We used SAS 9.2 (Cary, NC) for all data preparation and analysis in the discovery population and Rv3.2 for the replication population. We also performed a random-effects meta-analysis to estimate between-study and within medication class effect sizes for glucose and HbA1c change using STATA.

Mendelian randomization

The MR analysis was structured around a subset of 12 genes identified as drug-gene pairs in Figure 1, step 4. Two-sample MR was conducted by leveraging summary statistics from existing GWAS on ten different drug indications (angina42, atrial fibrillation43, bipolar disorder44, coronary artery disease45, congestive heart failure46, epilepsy47, glaucoma48, pain49, rheumatoid arthritis50, systolic blood pressure51) in tandem with summary statistics from the largest T2D GWAS to date30. For the 12 genes, the MR analysis utilized only tissues with statistically significant GPGE (P<0.05) as the instrument and was conducted via the “TwoSampleMR” R package52.

We replicated a previously published approach using S-PrediXcan summary statistics as the instrumental variable in an MR analysis to proxy therapeutic targets41.Tissue-specific GPGE summary statistics for each trait and T2D were combined to calculate the IVW MR association as follows: Embedded Image where βtraitrepresents GPGE per trait; and βT2D and Embedded Image represent T2D GPGE and standard error, for t number of statistically significant GPGE tissues, respectively.

Corresponding odds ratios (ORs) and 95% confidence intervals (95% CI) were calculated using Embedded Image and se Embedded Image.

The estimates (and 95% CIs) obtained from the MR analysis represent T2D risk per standard deviation change in gene expression. Furthermore, MR Egger regression was used to evaluate directional pleiotropy53. Multivariable MR (MVMR) was conducted to estimate adjusted MR effects for indications that were correlated and shared therapeutic gene targets (e.g., ACE inhibitors are used to treat hypertension and congestive heart failure)54.

Patient and Public Involvement

For the VA discovery population, patient clinical data were analyzed as part of the Leveraging Electronic Health Information to Advance Precision Medicine research protocol, which has been approved by institutional review boards and research committees at 3 VA Medical Centers (Salt Lake City, Palo Alto, and West Haven) with approved waivers of informed consent and HIPAA authorization. For the replication population, VUMC SD, the associated project received non-human subjects determination and approval from the VUMC Institutional Review Board.

Results

Computational drug repurposing approach

We used a multi-step computational approach to drug repurposing that leveraged large scale GWAS and EHR data to identify and test potential non-diabetes medications for use in hemoglobin (HbA1c) and glucose control (Figure 1). Steps 1-4 describe the process of identifying drug-gene pairs for evaluation and validation. The evaluation stage takes a two-pronged approach to evaluate these drug-gene pairs: a) a self-controlled case series to evaluate the response of HbA1c and glucose to novel medication exposure and b) a Mendelian randomization study to proxy therapeutic effects of a subset of the identified gene-drug pairs.

Gene-based medication discovery (Figure 1, steps 1-4)

Briefly, genes were identified in a previous large-scale GWAS and TWAS of diabetes risk30. Publicly available databases of drug gene targets, indications, and interactions were consulted to identify drug-gene pairs. Drugs were selected such that the drug targeted a gene that was associated with diabetes risk via GPGE, the drug was not used in diabetes management, and where the action of the drug would be predicted to mitigate increases in diabetes risk.

Summary statistics for 19.8 million single nucleotide polymorphisms were utilized in S-PrediXcan models for GPGE estimation across 52 tissues. We identified 695 unique genes that were associated with diabetes risk at a p-value threshold of 1.92×10−7, including: 568 genes for which a relationship was not previously reported and 127 with a known relationship to diabetes. We further refined the list of target drug-gene pairs by comparing medication and gene effects. For example, if a drug was an activator or agonist of the associated pathway or enzyme activity, we expect that increasing GPGE of the corresponding gene would be associated with decreased diabetes risk, and vice-versa for inhibitors. We identified 283 drugs with repurposing potential for diabetes that targeted 54 genes, 7.7% of the unique genes reported from the TWAS.

Self-controlled case series

To test for the impact of novel medication start on HbA1c and glucose using EHRs, we designed a self-controlled case series to evaluate the change in these laboratory measures in the six months following medication initiation (Figure 2). We restricted medication starts to a single experimental medication, defined by a gene-drug pair identified previously, with no preceding prescription of control medications (established medications known to raise or lower HbA1c and glucose). These medications were classified as glucose-reducing or glucose-increasing control medications based on prior knowledge of their effects on HbA1c or glucose. Overall, 68 medications were identified as possible control medications (Supplemental Table 1a and b). Further, we repeated this self-controlled case series for the glucose-reducing or glucose-increasing medications using identical restrictions on medication exposure and novel start to evaluate design performance (Figure 2). We performed the self-controlled case series independently in two large EHR systems, the Veteran’s Administration (VA) and Vanderbilt University Medical Center Synthetic Derivative (VUMC SD), separately for HbA1c and glucose.

Hemoglobin A1c

The VA discovery population for the HbA1c self-controlled case series included 124,357 patients: 40,780 (32.8%) in the T2D/glucose-reducing group, 25,170 (20.2%) in the glucose-increasing group, and 58,407 (47.0%) in the experimental group. The baseline characteristics of these patients are summarized in Supplemental Table 3a. The VUMC SD replication population was one-tenth the size of the VA population, including 15,365 patients: 4,439 (28.9%) in the T2D/glucose-reducing group, 3,912 (25.5%) in the glucose-increasing group, and 7,014 (45.6%) in the experimental group. The baseline characteristics of these patients are summarized in Supplemental Table 3b.

The paired t-test results for HbA1c change in both the discovery and replication populations are summarized in Table 1. Analyzed medications were required to have at least 30 prescriptions in both sites. All T2D/glucose-reducing medications demonstrated substantial reductions in HbA1c in the discovery population with similar results in the replication set. The greatest reduction in HbA1c in the VA population was glyburide and glipizide, second-generation oral sulfonylureas, with mean changes of - 1.3% and -1.1%, respectively (p<0.001). The control (+) medications had more inconsistent effects on HbA1c across the two sites. Of the 16 glucose-increasing medications, only amitriptyline had evidence to support an increase in HbA1c in both populations. Seven additional medications were associated with increases in the discovery population but not in the replication. We observed consistent direction of efforts with these 7 medications in the replication population but the substantially lower sample sizes potentially reduced the power to detect effects.

View this table:
  • View inline
  • View popup
Table 1. HbA1c paired t-test results from VA LEAPS with replication at Vanderbilt (N>=30 at both sites)

Of the initial 283 experimental medications identified by drug-gene pairs, only 20 (7.1%) had sufficient data for inclusion in the HbA1c self-controlled case series. This set included four angiotensin-converting enzyme (ACE) inhibitors. Two demonstrated substantial reductions in HbA1c in the discovery and replication populations, and one, enalapril, had evidence for a reduction in the discovery but not the replication set.

Ramipril did not consistently reduce HbA1c in either set. Lisinopril demonstrated the largest reduction (p<0.001, mean change VA= -0.03% and VUMC=-0.24%) in both populations. The calcium channel blocker verapamil also demonstrated reductions in HbA1c in both sets. Most statins had inconsistent effects. Pravastatin demonstrated a decrease in HbA1c in both populations.

Glucose

The VA discovery population for the glucose self-controlled case series included 678,501 patients: 51,645 (7.6%) in the T2D/glucose-reducing group, 235,493 (34.7%) in the glucose-increasing group, and 391,363 (57.7%) in the experimental group. The baseline characteristics of these patients are summarized in Supplemental Table 4a. The VUMC SD replication population was about one-tenth of the size of the VA population, including 67,155 patients: 11,889 (17.7%) in the T2D/glucose-reducing group, 9,371 (14.0%) in the glucose-increasing group, and 45,895 (68.3%) in the experimental group. The baseline characteristics of these patients are summarized in Supplemental Table 4b.

The paired t-test results for glucose change in both the discovery and replication populations are summarized in Table 2. Consistent with the HbA1c results, glyburide and glipizide had the most substantial decrease in glucose (p<0.001) in the discovery set. For the most part, all T2D/glucose-reducing medications demonstrated decreases in glucose. Some medications, such as sitagliptin, decreased glucose in the replication but not the discovery set (VA, p=0.44 and VUMC, p=0.002). These results suggest high variability in random glucose measurements may decrease statistical precision. The glucose-increasing medications demonstrated similar variability in results with dexamethasone exhibiting the only consistent increase in glucose across both populations (p<0.001).

View this table:
  • View inline
  • View popup
Table 2. Glucose paired t-test results from VA LEAPS with replication at Vanderbilt (N>=30 at both sites)

Consistency between glucose and HbA1c findings

Compared with HbA1c, 35 (12.4%) of the 283 medications identified by drug-gene pairs that were included in the glucose analysis. Only one of these medications, oxcarbazepine, an anti-epileptic, had evidence for an effect in both sets. However, the direction of glucose change was inconsistent with glucose increasing in the VA discovery population and decreasing in the VUMC replication. None of the ACE inhibitors demonstrated a consistent impact on glucose in either population. In line with the HbA1c results, verapamil was associated with a consistent decrease in glucose in the VA (−0.82 mg/dL, p=0.03). In the VUMC population, the point estimate indicated that verapamil decreased glucose but the wide confidence interval included the null, potentially due to power limitations of this smaller sample set. There were also inconsistent results for statin medications. In the discovery set, rosuvastatin increased glucose but the remaining statins including simvastatin, atorvastatin, pravastatin, lovastatin, and Fluvastatin, decreased glucose.

Meta-analysis

We next performed a cross-site meta-analysis for both HbA1c and glucose in a self-controlled case series to estimate the combined effect for the given medications. The results of these analyses are available in Figures 3 and 4. There was evidence to support all T2D/glucose-reducing medications reducing HbA1c with effect sizes ranging from -0.33% for sitagliptin to -1.06% for glyburide (p<0.001 for all). For glucose, however, only half the T2D/control (−) medications were associated with a mean decrease in glucose. Four (26.7%) of the 15 glucose-increasing medications were associated with an increase in HbA1c as expected, with the most substantial increases being for propranolol and hydrochlorothiazide, respectively (0.12%, p=1.0e-5; and 0.08%, p=2.8×10−14). Both propranolol and hydrochlorothiazide also demonstrated increases in glucose. Interestingly, there was also some evidence that citalopram and fluoxetine decreased HbA1c (−0.02%, p=0.04; and -0.03%, p=0.01). However, evidence for their impact on glucose was limited. Corticosteroids had the most substantial impact on glucose. Dexamethasone and prednisone consistently increased glucose (7.87 mg/dL, p=9.4×10−3; 1.32 mg/dL, p<1.3×10−16). In the experimental group, only verapamil demonstrated a significant effect on HbA1c (−0.11%, p=0.01). This effect was also observed in the glucose analysis (−0.85 mg/dL, p=0.024). Three statins, simvastatin, rosuvastatin, and fluvastatin, had evidence for changes in glucose; however, the directions of effect varied among the medications.

Figure 3.
  • Download figure
  • Open in new tab
Figure 3. Medication group meta-analysis results from the self-controlled case series for hemoglobin A1c.

All medications included in the analysis series were grouped by medication class and meta-analyzed. A forest plot of these results for hemoglobin A1c from the self-controlled case series in the discovery and replication datasets is presented. Whether they belonged to the control, glucose-decreasing or glucose-increasing, or experimental group is noted on the left-hand side of the figure.

Figure 4.
  • Download figure
  • Open in new tab
Figure 4. Medication group meta-analysis results from the self-controlled case series for glucose.

All medications included in the analysis series were grouped by medication class and meta-analyzed. A forest plot of these results for glucose from the self-controlled case series in the discovery and replication datasets is presented. Whether they belonged to the control, glucose-decreasing or glucose-increasing, or experimental group is noted on the left-hand side of the figure.

To characterize the effect of medication group on HbA1c and glucose, we also performed a drug-class grouped meta-analysis (Figures 3 and 4, respectively). As expected, all six of the T2D/glucose-reducing medication groups lowered HbA1c with the most substantial decrease belonging to biguanides (−0.88%, p<1.6×10−13). We saw similar magnitudes of reductions in glucose for DPP4 inhibitors and insulin (p=0.10 and p=0.15, respectively). Three of the glucose-increasing medication groups, diuretics, tricyclic antidepressants, and beta blockers, demonstrated increases in HbA1c and glucose; while corticosteroids only demonstrated an increase in glucose. Only one experimental medication class, calcium channel blockers, substantially reduced HbA1c and glucose (−0.11%, p=0.01; and -0.85 mg/dL, p=0.02, respectively). The statin and anti-convulsant medication classes were associated with increases in glucose (0.72 mg/dL, p=2.1×10−7; and 0.71 mg/dL, p=5.9e-4, respectively).

Mendelian randomization

In Table 3, we report our T2D two-sample MR results using S-PrediXcan summary statistics for various indications with putative drug gene targets. In our MR analysis, the strongest evidence for T2D prevention were observed for drugs targeting genes involved in systolic blood pressure, angina, and atrial fibrillation. ACE inhibitors, as proxied by reduced ACE gene expression, were predicted to reduce systolic blood pressure by approximately 0.25 mmHg (p=0.28) per standard deviation decrease in ACE expression. Whereas for drugs targeting KCNJ11 expression (i.e., minoxidil and verapamil), the predicted change in systolic blood pressure was minimal per standard deviation change in KCNJ11 gene expression. A 16-18% T2D risk reduction was observed for ACE inhibitors (T2D OR=0.82, 95% CI=0.78, 0.86, p=3.3×10−17) and minoxidil/verapamil (T2D OR=0.84, 95% CI=0.81, 0.87, p=5.0×10−25) via changes in predicted systolic blood pressure. Reduced atrial fibrillation risk via decreased expression of HCN3 and SCN3A (via HCN channel blockers and sodium channel blockers, respectively) were associated with reduced T2D risk in the MR analysis; however, only sodium channel blockers were observed to reduce T2D risk (T2D OR=0.25, 95% CI=0.17, 0.39, p=4.7×10−11). Similarly, evidence for T2D risk reduction was also observed for angina risk reduction via verapamil as proxied by reduced CACNA1A expression (T2D OR=0.17, 95% CI=0.10, 0.29, p=2.1×10−10). However, after accounting for potential pleiotropy via changes in correlated indications, only ACE inhibitors demonstrated consistent evidence for a reduction in T2D risk (T2D MVMR OR=0.86, 95% CI=0.84, 0.89, P=4.8×10−06).

View this table:
  • View inline
  • View popup
Table 3.

Summary of estimated Mendelian randomization (MR) effects of drug-targeted genetically-predicted gene expression (GPGE) on indications (either continuous or dichotomous) with type 2 diabetes risk

Discussion

Our study has demonstrated an approach for genetics-informed drug repurposing for diabetes by leveraging the power of genomics data, drug annotation databases, EHRs, and contemporary statistical methods for causal inference. A strength of this approach is the rapid and cost-efficient ability to prioritize candidate drugs for clinical trials on the basis of pre-clinical experiments using patient studies and MR.

We demonstrated that calcium-channel blockers are a good target for repurposing for T2D, supported by evidence across all arms of the investigation. This medication demonstrated substantially lower reductions in glycemic indices than the traditional diabetes medications. Our results may be of particular importance to patients with pre-diabetes and hypertension where preferential use of CCBs for blood pressure control may have an additive impact on glycemic control compared with other antihypertensive therapies. We also observed reductions in glycemic indices with another anti-hypertensive medication group, ACE inhibitors. About half of the experimental ACE inhibitors demonstrated significant decreases in both glucose and HbA1c, however, the relative effect sizes varied across sites and depended on the medication type suggesting ACE inhibitor effectiveness to lower glycemic indices depends on type, dose, and possibly the population in which it was used. These findings are similar to clinical trials of ACE inhibitors that have also demonstrated inconsistent results.

The results of this study demonstrate the feasibility and strength of our approach for computational drug repurposing discovery. While many of the potential repurposing medications for glucose reduction have been explored in other trials, we believe these results demonstrate the feasibility of this approach for other disease conditions. Further, because these results derive from two real world clinical populations, they are likely to be robust. These results suggest medications that could be used preferentially to treat concomitant disease while providing potential additional benefit to patients at risk for diabetes or with inadequately controlled diabetes.

This study also demonstrates the importance of careful consideration of study design when performing secondary studies in EHR data sources across sites. The limitations related to use of these resources included ascertainment and utilization of pharmacy data, a continuous measurement that can be compared over time, and the relatively short timeframe under observation. Despite these limitations, the ability to rapidly evaluate impacts on glycemic indices provide a real-world presentation in two distinct cohorts of patients of the effects of the genetically identified medications. The additional support for a causal effect of the identified drugs provided by the MR analyses provides both epidemiological rigor and evidence of a biological mechanism at work in the phenomenon of the glucose-lowering effect of CCBs.

In conclusion, we present a computational approach to drug repurposing that has the potential to be used for other conditions and with other data resources. We demonstrated this approach with a SCCS design in two large EHR systems and with MR using extant results from large-scale GWAS. We identified CCBs as a candidate treatment for high glucose and observed some evidence for ACE inhibitors as well. This strategy can be implemented for other outcomes with access to sufficient quantities of EHR data and informatics expertise.

Data Availability

Summary level statistical results will be made available upon reasonable request to corresponding authors. Individual level data can be made available following institutional agreements.

Acknowledgements

The authors wish to acknowledge the efforts of Eric Tortenson and Max Breyer for their assistance with the initial data extraction from Vanderbilt University Medical Center Synthetic Derivative. Figures 1 and 2 were created with BioRender.com.

Footnotes

  • Copyright The Corresponding Author has the right to grant on behalf of all authors and does grant on behalf of all authors, an exclusive license (or non-exclusive for government employees) on a worldwide basis to the BMJ Publishing Group Ltd to permit this article (if accepted) to be published in BMJ editions and any other BMJPGL products and sublicences such use and exploit all subsidiary rights, as set out in our license.

  • Ethics Approval The studies obtained ethical approval from the affiliated organizations, Department of Veterans Affairs and Vanderbilt University Medical Center. For the Vanderbilt University Medical Center Synthetic derivative patient data is de-identified and do not meet the 45 CFR 46 definition for human subjects research. As such, the study received non-human subjects determination from the associated institutional review board.

  • Transparency Statement The corresponding authors affirm that the manuscript is an honest, accurate and transparent account of the reported study. No important aspects of the study have been omitted and all discrepancies from the study have been explained.

  • Conflicts of Interest No conflicts of interest

  • Funding Sources The dataset used for the replication analyses was obtained from the Vanderbilt University Medical Center Synthetic Derivative, which is supported by institutional funding, the 1S10RR025141-01 instrumentation award, and by the CTSA grant UL1TR000445 from National Center for Advancing Translational Sciences/National Institutes of Health. M.M.S. was supported by AHA 17SFRN33520017 and VA Merit I01 BX005399-01A1. N.K.K. is supported by NIH R00 CA215360. V.W. is supported by the Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK [MC_UU_00011/4] and the COVID-19 Longitudinal Health and Wellbeing National Core Study, which is funded by the UK Medical Research Council (MC_PC_20059). M.V. and P.R. are supported by 2I01BX003362-03A1. This work was supported using resources and facilities of the US Department of Veterans Affairs (VA), Veterans Health Administration, Cooperative Studies Program, grant number 825-MS-DI-33848, and used resources and facilities at the VA Informatics and Computing Infrastructure (VINCI), VA HSR RES 13-457.

  • Data Sharing Summary level statistical results will be made available upon reasonable request to corresponding authors. Individual level data can be made available following institutional agreements.

References

  1. 1.↵
    Saeedi, P. et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes Res Clin Pract 157, 107843 (2019).
    OpenUrlCrossRefPubMed
  2. 2.↵
    American Diabetes, A. Standards of Medical Care in Diabetes-2020 Abridged for Primary Care Providers. Clin Diabetes 38, 10–38 (2020).
    OpenUrlFREE Full Text
  3. 3.
    Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 414, 813–20 (2001).
    OpenUrlCrossRefPubMedWeb of Science
  4. 4.↵
    Danaei, G. et al. National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2.7 million participants. Lancet 378, 31–40 (2011).
    OpenUrlCrossRefPubMedWeb of Science
  5. 5.↵
    Buse, J.B. et al. 2019 update to: Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 63, 221–228 (2020).
    OpenUrlPubMed
  6. 6.
    Davies, M.J. et al. Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 61, 2461–2498 (2018).
    OpenUrlPubMed
  7. 7.
    Inzucchi, S.E. et al. Management of hyperglycaemia in type 2 diabetes, 2015: a patient-centred approach. Update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetologia 58, 429–42 (2015).
    OpenUrlCrossRefPubMed
  8. 8.↵
    Nathan, D.M. et al. Medical management of hyperglycaemia in type 2 diabetes mellitus: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetologia 52, 17–30 (2009).
    OpenUrlCrossRefPubMedWeb of Science
  9. 9.↵
    Karter, A.J. et al. Achieving good glycemic control: initiation of new antihyperglycemic therapies in patients with type 2 diabetes from the Kaiser Permanente Northern California Diabetes Registry. Am J Manag Care 11, 262–70 (2005).
    OpenUrlPubMedWeb of Science
  10. 10.↵
    de Vries, S.T. et al. Medication beliefs, treatment complexity, and non-adherence to different drug classes in patients with type 2 diabetes. J Psychosom Res 76, 134–8 (2014).
    OpenUrlCrossRefPubMed
  11. 11.↵
    Polonsky, W.H. & Henry, R.R. Poor medication adherence in type 2 diabetes: recognizing the scope of the problem and its key contributors. Patient Prefer Adherence 10, 1299–307 (2016).
    OpenUrlCrossRefPubMed
  12. 12.↵
    Kautzky-Willer, A., Kosi, L., Lin, J. & Mihaljevic, R. Gender-based differences in glycaemic control and hypoglycaemia prevalence in patients with type 2 diabetes: results from patient-level pooled data of six randomized controlled trials. Diabetes Obes Metab 17, 533–540 (2015).
    OpenUrlPubMed
  13. 13.↵
    DeSouza, C. et al. Efficacy and Safety of Semaglutide for Type 2 Diabetes by Race and Ethnicity: A Post Hoc Analysis of the SUSTAIN Trials. J Clin Endocrinol Metab 105(2020).
  14. 14.↵
    Hage, M., Zantout, M.S. & Azar, S.T. Thyroid disorders and diabetes mellitus. J Thyroid Res 2011, 439463 (2011).
    OpenUrlPubMed
  15. 15.↵
    Cryer, P.E. et al. Evaluation and management of adult hypoglycemic disorders: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 94, 709–28 (2009).
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    Knudsen, L.B. & Lau, J. The Discovery and Development of Liraglutide and Semaglutide. Front Endocrinol (Lausanne) 10, 155 (2019).
    OpenUrl
  17. 17.
    Neal, B., Perkovic, V. & Matthews, D.R. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med 377, 2099 (2017).
    OpenUrlPubMed
  18. 18.↵
    Zinman, B. et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med 373, 2117–28 (2015).
    OpenUrlCrossRefPubMed
  19. 19.↵
    Grundy, S.M. Drug therapy of the metabolic syndrome: minimizing the emerging crisis in polypharmacy. Nat Rev Drug Discov 5, 295–309 (2006).
    OpenUrlCrossRefPubMed
  20. 20.↵
    Ashburn, T.T. & Thor, K.B. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov 3, 673–83 (2004).
    OpenUrlCrossRefPubMedWeb of Science
  21. 21.↵
    Montinari, M.R., Minelli, S. & De Caterina, R. The first 3500years of aspirin history from its roots - A concise summary. Vascul Pharmacol 113, 1–8 (2019).
    OpenUrlPubMed
  22. 22.↵
    Goldfine, A.B. et al. The effects of salsalate on glycemic control in patients with type 2 diabetes: a randomized trial. Ann Intern Med 152, 346–57 (2010).
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.↵
    Pushpakom, S. et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov 18, 41–58 (2019).
    OpenUrlCrossRefPubMed
  24. 24.↵
    Finan, C. et al. The druggable genome and support for target identification and validation in drug development. Sci Transl Med 9(2017).
  25. 25.↵
    Brinkman, R.R., Dube, M.P., Rouleau, G.A., Orr, A.C. & Samuels, M.E. Human monogenic disorders - a source of novel drug targets. Nat Rev Genet 7, 249–60 (2006).
    OpenUrlCrossRefPubMedWeb of Science
  26. 26.
    Sanseau, P. et al. Use of genome-wide association studies for drug repositioning. Nat Biotechnol 30, 317–20 (2012).
    OpenUrlCrossRefPubMed
  27. 27.↵
    Wang, Z.Y., Fu, L.Y. & Zhang, H.Y. Can medical genetics and evolutionary biology inspire drug target identification? Trends Mol Med 18, 69–71 (2012).
    OpenUrlCrossRefPubMed
  28. 28.↵
    Robinson, E. Psychopharmacology: From serendipitous discoveries to rationale design, but what next? Brain Neurosci Adv 2, 2398212818812629 (2018).
    OpenUrl
  29. 29.↵
    Walker, V.M., Davey Smith, G., Davies, N.M. & Martin, R.M. Mendelian randomization: a novel approach for the prediction of adverse drug events and drug repurposing opportunities. Int J Epidemiol 46, 2078–2089 (2017).
    OpenUrlCrossRefPubMed
  30. 30.↵
    Vujkovic, M. et al. Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat Genet 52, 680–691 (2020).
    OpenUrlCrossRefPubMed
  31. 31.↵
    Ko, Y.A. et al. Genetic-Variation-Driven Gene-Expression Changes Highlight Genes with Important Functions for Kidney Disease. Am J Hum Genet 100, 940–953 (2017).
    OpenUrlCrossRefPubMed
  32. 32.↵
    Ackermann, A.M., Wang, Z., Schug, J., Naji, A. & Kaestner, K.H. Integration of ATAC-seq and RNA-seq identifies human alpha cell and beta cell signature genes. Mol Metab 5, 233–244 (2016).
    OpenUrl
  33. 33.↵
    Gaulton, A. et al. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res 40, D1100–7 (2012).
    OpenUrlCrossRefPubMedWeb of Science
  34. 34.↵
    Griffith, M. et al. DGIdb: mining the druggable genome. Nat Methods 10, 1209–10 (2013).
    OpenUrlCrossRefPubMedWeb of Science
  35. 35.↵
    National Cancer Institute Drug Dictionary.
  36. 36.↵
    Affairs, U.D.o.V. Health services research and development:VA Informatics and Computing Infrastructure. (2020).
  37. 37.↵
    Roden, D.M. et al. Development of a large-scale de-identified DNA biobank to enable personalized medicine. Clin Pharmacol Ther 84, 362–9 (2008).
    OpenUrlCrossRefPubMedWeb of Science
  38. 38.↵
    Xu, H. et al. MedEx: a medication information extraction system for clinical narratives. J Am Med Inform Assoc 17, 19–24 (2010).
    OpenUrlCrossRefPubMed
  39. 39.↵
    Shuey, M. et al. Retrospective cohort study to characterise the blood pressure response to spironolactone in patients with apparent therapy-resistant hypertension using electronic medical record data. BMJ Open 10, e033100 (2020).
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    Xu, H. et al. Facilitating pharmacogenetic studies using electronic health records and natural-language processing: a case study of warfarin. J Am Med Inform Assoc 18, 387–91 (2011).
    OpenUrlCrossRefPubMed
  41. 41.↵
    Khankari, N.K. et al. Using Mendelian randomisation to identify opportunities for type 2 diabetes prevention by repurposing medications used for lipid management. EBioMedicine 80, 104038 (2022).
    OpenUrl
  42. 42.↵
    Nelson, C.P. et al. Association analyses based on false discovery rate implicate new loci for coronary artery disease. Nat Genet 49, 1385–1391 (2017).
    OpenUrlCrossRefPubMed
  43. 43.↵
    Nielsen, J.B. et al. Biobank-driven genomic discovery yields new insight into atrial fibrillation biology. Nat Genet 50, 1234–1239 (2018).
    OpenUrlCrossRefPubMed
  44. 44.↵
    Hou, L. et al. Genome-wide association study of 40,000 individuals identifies two novel loci associated with bipolar disorder. Hum Mol Genet 25, 3383–3394 (2016).
    OpenUrlCrossRefPubMed
  45. 45.↵
    Nikpay, M. et al. A comprehensive 1,000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nat Genet 47, 1121–1130 (2015).
    OpenUrlCrossRefPubMed
  46. 46.↵
    Aragam, K.G. et al. Phenotypic Refinement of Heart Failure in a National Biobank Facilitates Genetic Discovery. Circulation (2018).
  47. 47.↵
    International League Against Epilepsy Consortium on Complex, E. Genome-wide mega-analysis identifies 16 loci and highlights diverse biological mechanisms in the common epilepsies. Nat Commun 9, 5269 (2018).
    OpenUrlCrossRefPubMed
  48. 48.↵
    Zhou, W. et al. Efficiently controlling for case-control imbalance and sample relatedness in large-scale genetic association studies. Nat Genet 50, 1335–1341 (2018).
    OpenUrlCrossRefPubMed
  49. 49.↵
    Johnston, K.J.A. et al. Genome-wide association study of multisite chronic pain in UK Biobank. PLoS Genet 15, e1008164 (2019).
    OpenUrlCrossRefPubMed
  50. 50.↵
    Okada, Y. et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506, 376–81 (2014).
    OpenUrlCrossRefPubMedWeb of Science
  51. 51.↵
    Giri, A. et al. Trans-ethnic association study of blood pressure determinants in over 750,000 individuals. Nat Genet 51, 51–62 (2019).
    OpenUrlCrossRefPubMed
  52. 52.↵
    Hemani, G. et al. The MR-Base platform supports systematic causal inference across the human phenome. Elife 7(2018).
  53. 53.↵
    Bowden, J., Davey Smith, G. & Burgess, S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol 44, 512–25 (2015).
    OpenUrlCrossRefPubMed
  54. 54.↵
    Burgess, S. & Thompson, S.G. Multivariable Mendelian randomization: the use of pleiotropic genetic variants to estimate causal effects. Am J Epidemiol 181, 251–60 (2015).
    OpenUrlCrossRefPubMed
Back to top
PreviousNext
Posted December 16, 2022.
Download PDF

Supplementary Material

Data/Code
Email

Thank you for your interest in spreading the word about medRxiv.

NOTE: Your email address is requested solely to identify you as the sender of this article.

Enter multiple addresses on separate lines or separate them with commas.
A genetically supported drug repurposing pipeline for diabetes treatment using electronic health records
(Your Name) has forwarded a page to you from medRxiv
(Your Name) thought you would like to see this page from the medRxiv website.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
A genetically supported drug repurposing pipeline for diabetes treatment using electronic health records
Megan M. Shuey, Kyung Min Lee, Jacob Keaton, Nikhil K. Khankari, Joseph H. Breeyear, Venexia M. Walker, Donald R. Miller, Kent R. Heberer, Peter D. Reaven, Shoa L. Clarke, Jennifer Lee, Julie A. Lynch, Marijana Vujkovic, Todd L. Edwards
medRxiv 2022.12.14.22283414; doi: https://doi.org/10.1101/2022.12.14.22283414
Twitter logo Facebook logo LinkedIn logo Mendeley logo
Citation Tools
A genetically supported drug repurposing pipeline for diabetes treatment using electronic health records
Megan M. Shuey, Kyung Min Lee, Jacob Keaton, Nikhil K. Khankari, Joseph H. Breeyear, Venexia M. Walker, Donald R. Miller, Kent R. Heberer, Peter D. Reaven, Shoa L. Clarke, Jennifer Lee, Julie A. Lynch, Marijana Vujkovic, Todd L. Edwards
medRxiv 2022.12.14.22283414; doi: https://doi.org/10.1101/2022.12.14.22283414

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Subject Area

  • Genetic and Genomic Medicine
Subject Areas
All Articles
  • Addiction Medicine (349)
  • Allergy and Immunology (668)
  • Allergy and Immunology (668)
  • Anesthesia (181)
  • Cardiovascular Medicine (2648)
  • Dentistry and Oral Medicine (316)
  • Dermatology (223)
  • Emergency Medicine (399)
  • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (942)
  • Epidemiology (12228)
  • Forensic Medicine (10)
  • Gastroenterology (759)
  • Genetic and Genomic Medicine (4103)
  • Geriatric Medicine (387)
  • Health Economics (680)
  • Health Informatics (2657)
  • Health Policy (1005)
  • Health Systems and Quality Improvement (985)
  • Hematology (363)
  • HIV/AIDS (851)
  • Infectious Diseases (except HIV/AIDS) (13695)
  • Intensive Care and Critical Care Medicine (797)
  • Medical Education (399)
  • Medical Ethics (109)
  • Nephrology (436)
  • Neurology (3882)
  • Nursing (209)
  • Nutrition (577)
  • Obstetrics and Gynecology (739)
  • Occupational and Environmental Health (695)
  • Oncology (2030)
  • Ophthalmology (585)
  • Orthopedics (240)
  • Otolaryngology (306)
  • Pain Medicine (250)
  • Palliative Medicine (75)
  • Pathology (473)
  • Pediatrics (1115)
  • Pharmacology and Therapeutics (466)
  • Primary Care Research (452)
  • Psychiatry and Clinical Psychology (3432)
  • Public and Global Health (6527)
  • Radiology and Imaging (1403)
  • Rehabilitation Medicine and Physical Therapy (814)
  • Respiratory Medicine (871)
  • Rheumatology (409)
  • Sexual and Reproductive Health (410)
  • Sports Medicine (342)
  • Surgery (448)
  • Toxicology (53)
  • Transplantation (185)
  • Urology (165)