Precision Medicine in Diabetes: A Consensus Report From the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD)

Wendy K Chung, Karel Erion, Jose C Florez, Andrew T Hattersley, Marie-France Hivert, Christine G Lee, Mark I McCarthy, John J Nolan, Jill M Norris, Ewan R Pearson, Louis Philipson, Allison T McElvaine, William T Cefalu, Stephen S Rich, Paul W Franks, Wendy K Chung, Karel Erion, Jose C Florez, Andrew T Hattersley, Marie-France Hivert, Christine G Lee, Mark I McCarthy, John J Nolan, Jill M Norris, Ewan R Pearson, Louis Philipson, Allison T McElvaine, William T Cefalu, Stephen S Rich, Paul W Franks

Abstract

The convergence of advances in medical science, human biology, data science, and technology has enabled the generation of new insights into the phenotype known as "diabetes." Increased knowledge of this condition has emerged from populations around the world, illuminating the differences in how diabetes presents, its variable prevalence, and how best practice in treatment varies between populations. In parallel, focus has been placed on the development of tools for the application of precision medicine to numerous conditions. This Consensus Report presents the American Diabetes Association (ADA) Precision Medicine in Diabetes Initiative in partnership with the European Association for the Study of Diabetes (EASD), including its mission, the current state of the field, and prospects for the future. Expert opinions are presented on areas of precision diagnostics and precision therapeutics (including prevention and treatment), and key barriers to and opportunities for implementation of precision diabetes medicine, with better care and outcomes around the globe, are highlighted. Cases where precision diagnosis is already feasible and effective (i.e., monogenic forms of diabetes) are presented, while the major hurdles to the global implementation of precision diagnosis of complex forms of diabetes are discussed. The situation is similar for precision therapeutics, in which the appropriate therapy will often change over time owing to the manner in which diabetes evolves within individual patients. This Consensus Report describes a foundation for precision diabetes medicine, while highlighting what remains to be done to realize its potential. This, combined with a subsequent, detailed evidence-based review (due 2022), will provide a roadmap for precision medicine in diabetes that helps improve the quality of life for all those with diabetes.

© 2020 by the American Diabetes Association and the European Association for the Study of Diabetes.

Figures

Figure 1
Figure 1
PMDI activities. PM, precision medicine; RFA, research funding announcement.
Figure 2
Figure 2
Precision diagnostics
Figure 3
Figure 3
Precision therapeutics
Figure 4
Figure 4
Precision prognostics
Figure 5
Figure 5
The path to precision diabetes medicine. HEA, health economic assessment. Adapted from Fitipaldi et al. (136).

References

    1. Hattersley AT, Patel KA. Precision diabetes: learning from monogenic diabetes. Diabetologia 2017;60:769–777
    1. De Franco E, Flanagan SE, Houghton JA, et al. . The effect of early, comprehensive genomic testing on clinical care in neonatal diabetes: an international cohort study. Lancet 2015;386:957–963
    1. Oram RA, Patel K, Hill A, et al. . A type 1 diabetes genetic risk score can aid discrimination between type 1 and type 2 diabetes in young adults. Diabetes Care 2016;39:337–344
    1. Grubb AL, McDonald TJ, Rutters F, et al. . A type 1 diabetes genetic risk score can identify patients with GAD65 autoantibody-positive type 2 diabetes who rapidly progress to insulin therapy. Diabetes Care 2019;42:208–214
    1. Sharp SA, Rich SS, Wood AR, et al. . Development and standardization of an improved type 1 diabetes genetic risk score for use in newborn screening and incident diagnosis. Diabetes Care 2019;42:200–207
    1. Udler MS, McCarthy MI, Florez JC, Mahajan A. Genetic risk scores for diabetes diagnosis and precision medicine. Endocr Rev 2019;40:1500–1520
    1. Dennis JM, Shields BM, Henley WE, Jones AG, Hattersley AT. Disease progression and treatment response in data-driven subgroups of type 2 diabetes compared with models based on simple clinical features: an analysis using clinical trial data. Lancet Diabetes Endocrinol 2019;7:442–451
    1. Fleming GA, Petrie JR, Bergenstal RM, Holl RW, Peters AL, Heinemann L. Diabetes digital app technology: benefits, challenges, and recommendations. A consensus report by the European Association for the Study of Diabetes (EASD) and the American Diabetes Association (ADA) Diabetes Technology Working Group. Diabetes Care 2020;43:250–260
    1. Fleming GA, Petrie JR, Bergenstal RM, Holl RW, Peters AL, Heinemann L. Diabetes digital app technology: benefits, challenges, and recommendations. A consensus report by the European Association for the Study of Diabetes (EASD) and the American Diabetes Association (ADA) Diabetes Technology Working Group. Diabetologia 2020;63:229–241
    1. American Diabetes Association 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes—2020. Diabetes Care 2020;43(Suppl. 1):S14–S31
    1. Thomas NJ, Jones SE, Weedon MN, Shields BM, Oram RA, Hattersley AT. Frequency and phenotype of type 1 diabetes in the first six decades of life: a cross-sectional, genetically stratified survival analysis from UK Biobank. Lancet Diabetes Endocrinol 2018;6:122–129
    1. Thomas NJ, Lynam AL, Hill AV, et al. . Type 1 diabetes defined by severe insulin deficiency occurs after 30 years of age and is commonly treated as type 2 diabetes. Diabetologia 2019;62:1167–1172
    1. Gloyn AL, Pearson ER, Antcliff JF, et al. . Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med 2004;350:1838–1849
    1. Pearson ER, Flechtner I, Njølstad PR, et al. .; Neonatal Diabetes International Collaborative Group . Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 2006;355:467–477
    1. Babenko AP, Polak M, Cavé H, et al. . Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N Engl J Med 2006;355:456–466
    1. Sagen JV, Raeder H, Hathout E, et al. . Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes 2004;53:2713–2718
    1. Bowman P, Sulen Å, Barbetti F, et al. .; Neonatal Diabetes International Collaborative Group . Effectiveness and safety of long-term treatment with sulfonylureas in patients with neonatal diabetes due to KCNJ11 mutations: an international cohort study. Lancet Diabetes Endocrinol 2018;6:637–646
    1. Steele AM, Shields BM, Wensley KJ, Colclough K, Ellard S, Hattersley AT. Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA 2014;311:279–286
    1. Stride A, Shields B, Gill-Carey O, et al. . Cross-sectional and longitudinal studies suggest pharmacological treatment used in patients with glucokinase mutations does not alter glycaemia. Diabetologia 2014;57:54–56
    1. Pearson ER, Starkey BJ, Powell RJ, Gribble FM, Clark PM, Hattersley AT. Genetic cause of hyperglycaemia and response to treatment in diabetes. Lancet 2003;362:1275–1281
    1. Pearson ER, Pruhova S, Tack CJ, et al. . Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4alpha mutations in a large European collection. Diabetologia 2005;48:878–885
    1. Bowman P, Flanagan SE, Edghill EL, et al. . Heterozygous ABCC8 mutations are a cause of MODY. Diabetologia 2012;55:123–127
    1. Shields BM, McDonald TJ, Ellard S, Campbell MJ, Hyde C, Hattersley AT. The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. Diabetologia 2012;55:1265–1272
    1. Carlsson A, Shepherd M, Ellard S, et al. . Absence of islet autoantibodies and modestly raised glucose values at diabetes diagnosis should lead to testing for MODY: lessons from a 5-year pediatric Swedish national cohort study. Diabetes Care 2020;43:82–89
    1. Ellard S, Colclough K, Patel KA, Hattersley AT. Prediction algorithms: pitfalls in interpreting genetic variants of autosomal dominant monogenic diabetes. J Clin Invest 2020;130:14–16
    1. Clissold RL, Hamilton AJ, Hattersley AT, Ellard S, Bingham C. HNF1B-associated renal and extra-renal disease-an expanding clinical spectrum. Nat Rev Nephrol 2015;11:102–112
    1. Tranebjaerg L, Barrett T, Rendtorff ND. WFS1 Wolfram syndrome spectrum disorder. In GeneReviews®. Adam MP, Ardinger HH, Pagon RA, et al., Eds. Seattle, WA, University of Washington, Seattle, 1993-2020 [Internet]. Available from . Accessed 13 May 2020
    1. Murphy R, Turnbull DM, Walker M, Hattersley AT. Clinical features, diagnosis and management of maternally inherited diabetes and deafness (MIDD) associated with the 3243A>G mitochondrial point mutation. Diabet Med 2008;25:383–399
    1. Tuomi T, Groop LC, Zimmet PZ, Rowley MJ, Knowles W, Mackay IR. Antibodies to glutamic acid decarboxylase reveal latent autoimmune diabetes mellitus in adults with a non-insulin-dependent onset of disease. Diabetes 1993;42:359–362
    1. Brophy S, Yderstraede K, Mauricio D, et al. .; Action LADA Group . Time to insulin initiation cannot be used in defining latent autoimmune diabetes in adults. Diabetes Care 2008;31:439–441
    1. Hawa MI, Kolb H, Schloot N, et al. .; Action LADA consortium . Adult-onset autoimmune diabetes in Europe is prevalent with a broad clinical phenotype: Action LADA 7. Diabetes Care 2013;36:908–913
    1. Battaglia M, Ahmed S, Anderson MS, et al. . Introducing the endotype concept to address the challenge of disease heterogeneity in type 1 diabetes. Diabetes Care 2020;43:5–12
    1. Onengut-Gumuscu S, Chen WM, Robertson CC, et al. .; SEARCH for Diabetes in Youth; Type 1 Diabetes Genetics Consortium . Type 1 diabetes risk in African-Ancestry participants and utility of an ancestry-specific genetic risk score. Diabetes Care 2019;42:406–415
    1. Rich SS. Genetics and its potential to improve type 1 diabetes care. Curr Opin Endocrinol Diabetes Obes 2017;24:279–284
    1. Onengut-Gumuscu S, Chen WM, Burren O, et al. .; Type 1 Diabetes Genetics Consortium . Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat Genet 2015;47:381–386
    1. Rickels MR, Evans-Molina C, Bahnson HT, et al. .; T1D Exchange β-Cell Function Study Group . High residual C-peptide likely contributes to glycemic control in type 1 diabetes. J Clin Invest 2020;130:1850–1862
    1. Hao W, Gitelman S, DiMeglio LA, Boulware D, Greenbaum CJ; Type 1 Diabetes TrialNet Study Group . Fall in C-peptide during first 4 years from diagnosis of type 1 diabetes: variable relation to age, HbA1c, and insulin dose. Diabetes Care 2016;39:1664–1670
    1. Shields BM, McDonald TJ, Oram R, et al. .; TIGI Consortium . C-peptide decline in type 1 diabetes has two phases: an initial exponential fall and a subsequent stable phase. Diabetes Care 2018;41:1486–1492
    1. Ahlqvist E, Storm P, Käräjämäki A, et al. . Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol 2018;6:361–369
    1. Zaharia OP, Strassburger K, Strom A, et al. .; German Diabetes Study Group . Risk of diabetes-associated diseases in subgroups of patients with recent-onset diabetes: a 5-year follow-up study. Lancet Diabetes Endocrinol 2019;7:684–694
    1. Little RR, Rohlfing CL, Tennill AL, et al. . Standardization of C-peptide measurements. Clin Chem 2008;54:1023–1026
    1. Udler MS, Kim J, von Grotthuss M, et al. .; Christopher D. Anderson on behalf of METASTROKE and the ISGC . Type 2 diabetes genetic loci informed by multi-trait associations point to disease mechanisms and subtypes: A soft clustering analysis. PLoS Med 2018;15:e1002654.
    1. Mahajan A, Wessel J, Willems SM, et al. .; ExomeBP Consortium; MAGIC Consortium; GIANT Consortium . Refining the accuracy of validated target identification through coding variant fine-mapping in type 2 diabetes. Nat Genet 2018;50:559–571
    1. Insel RA, Dunne JL, Atkinson MA, et al. . Staging presymptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association. Diabetes Care 2015;38:1964–1974
    1. Krischer JP; Type 1 Diabetes TrialNet Study Group . The use of intermediate endpoints in the design of type 1 diabetes prevention trials. Diabetologia 2013;56:1919–1924
    1. Ziegler AG, Rewers M, Simell O, et al. . Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. JAMA 2013;309:2473–2479
    1. Rewers M, Stene LC, Norris JM. Chapter 11: Risk factors for type 1 diabetes. In Diabetes in America, 3rd ed Cowie CC, Casagrande SS, Menke A, et al., Eds. Bethesda, MD, National Institutes of Health, 2018
    1. Skyler JS, Kirchner JP, Becker D, Rewers M. Chapter 37: Prevention of type 1 diabetes In Diabetes in America, 3rd ed Cowie CC, Casagrande SS, et al., Eds. Bethesda, MD, National Institutes of Health, 2018
    1. Knip M, Åkerblom HK, Becker D, et al. .; TRIGR Study Group . Hydrolyzed infant formula and early β-cell autoimmunity: a randomized clinical trial. JAMA 2014;311:2279–2287
    1. Vaarala O, Ilonen J, Ruohtula T, et al. . Removal of bovine insulin from cow’s milk formula and early initiation of beta-cell autoimmunity in the FINDIA pilot study. Arch Pediatr Adolesc Med 2012;166:608–614
    1. Hummel S, Pflüger M, Hummel M, Bonifacio E, Ziegler AG. Primary dietary intervention study to reduce the risk of islet autoimmunity in children at increased risk for type 1 diabetes: the BABYDIET study. Diabetes Care 2011;34:1301–1305
    1. Knip M, Åkerblom HK, Al Taji E, et al. .; Writing Group for the TRIGR Study Group . Effect of hydrolyzed infant formula vs conventional formula on risk of type 1 diabetes: the TRIGR randomized clinical trial. JAMA 2018;319:38–48
    1. Näntö-Salonen K, Kupila A, Simell S, et al. . Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a double-blind, randomised controlled trial. Lancet 2008;372:1746–1755
    1. Krischer JP, Schatz DA, Bundy B, Skyler JS, Greenbaum CJ; Writing Committee for the Type 1 Diabetes TrialNet Oral Insulin Study Group . Effect of oral insulin on prevention of diabetes in relatives of patients with type 1 diabetes: a randomized clinical trial. JAMA 2017;318:1891–1902
    1. Elding Larsson H, Lundgren M, Jonsdottir B, Cuthbertson D, Krischer J; DiAPREV-IT Study Group . Safety and efficacy of autoantigen-specific therapy with 2 doses of alum-formulated glutamate decarboxylase in children with multiple islet autoantibodies and risk for type 1 diabetes: a randomized clinical trial. Pediatr Diabetes 2018;19:410–419
    1. Lampeter EF, Klinghammer A, Scherbaum WA, et al. .; DENIS Group . The Deutsche Nicotinamide Intervention Study: an attempt to prevent type 1 diabetes. Diabetes 1998;47:980–984
    1. Gale EA, Bingley PJ, Emmett CL, Collier T; European Nicotinamide Diabetes Intervention Trial (ENDIT) Group . European Nicotinamide Diabetes Intervention Trial (ENDIT): a randomised controlled trial of intervention before the onset of type 1 diabetes. Lancet 2004;363:925–931
    1. Diabetes Prevention Trial–Type 1 Diabetes Study Group Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med 2002;346:1685–1691
    1. Vehik K, Cuthbertson D, Ruhlig H, Schatz DA, Peakman M, Krischer JP; DPT-1 and TrialNet Study Groups . Long-term outcome of individuals treated with oral insulin: Diabetes Prevention Trial–Type 1 (DPT-1) oral insulin trial. Diabetes Care 2011;34:1585–1590
    1. Vandemeulebroucke E, Gorus FK, Decochez K, et al. .; Belgian Diabetes Registry . Insulin treatment in IA-2A-positive relatives of type 1 diabetic patients. Diabetes Metab 2009;35:319–327
    1. Herold KC, Bundy BN, Long SA, et al. .; Type 1 Diabetes TrialNet Study Group . An anti-CD3 antibody, teplizumab, in relatives at risk for type 1 diabetes. N Engl J Med 2019;381:603–613
    1. Hober D, Alidjinou EK. Enteroviral pathogenesis of type 1 diabetes: queries and answers. Curr Opin Infect Dis 2013;26:263–269
    1. Hakola L, Takkinen HM, Niinistö S, et al. . Infant feeding in relation to the risk of advanced islet autoimmunity and type 1 diabetes in children with increased genetic susceptibility: a cohort study. Am J Epidemiol 2018;187:34–44
    1. Tapia G, Mårild K, Dahl SR, et al. . Maternal and newborn vitamin D-binding protein, vitamin D levels, vitamin D receptor genotype, and childhood type 1 diabetes. Diabetes Care 2019;42:553–559
    1. Norris JM, Kroehl M, Fingerlin TE, et al. . Erythrocyte membrane docosapentaenoic acid levels are associated with islet autoimmunity: the Diabetes Autoimmunity Study in the Young. Diabetologia 2014;57:295–304
    1. DECODE Study Group; the European Diabetes Epidemiology Group. Glucose tolerance and cardiovascular mortality: comparison of fasting and 2-hour diagnostic criteria. Arch Intern Med 2001;161:397–405
    1. Diabetes Prevention Program Research Group Within-trial cost-effectiveness of lifestyle intervention or metformin for the primary prevention of type 2 diabetes. Diabetes Care 2003;26:2518–2523
    1. American Diabetes Association 12. Older adults: Standards of Medical Care in Diabetes—2020. Diabetes Care 2020;43(Suppl. 1):S152–S162
    1. Haw JS, Galaviz KI, Straus AN, et al. . Long-term sustainability of diabetes prevention approaches: a systematic review and meta-analysis of randomized clinical trials. JAMA Intern Med 2017;177:1808–1817
    1. Knowler WC, Barrett-Connor E, Fowler SE, et al. .; Diabetes Prevention Program Research Group . Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med 2002;346:393–403
    1. Crandall J, Schade D, Ma Y, et al. .; Diabetes Prevention Program Research Group . The influence of age on the effects of lifestyle modification and metformin in prevention of diabetes. J Gerontol A Biol Sci Med Sci 2006;61:1075–1081
    1. Knowler WC, Fowler SE, Hamman RF, et al. .; Diabetes Prevention Program Research Group . 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet 2009;374:1677–1686
    1. Delahanty LM, Pan Q, Jablonski KA, et al. .; Diabetes Prevention Program Research Group . Effects of weight loss, weight cycling, and weight loss maintenance on diabetes incidence and change in cardiometabolic traits in the Diabetes Prevention Program. Diabetes Care 2014;37:2738–2745
    1. Papandonatos GD, Pan Q, Pajewski NM, et al. .; GIANT Consortium; Diabetes Prevention Program and the Look AHEAD Research Groups . Genetic predisposition to weight loss and regain with lifestyle intervention: analyses from the Diabetes Prevention Program and the Look AHEAD randomized controlled trials. Diabetes 2015;64:4312–4321
    1. Langenberg C, Sharp SJ, Franks PW, et al. . Gene-lifestyle interaction and type 2 diabetes: the EPIC interact case-cohort study. PLoS Med 2014;11:e1001647.
    1. Hivert MF, Christophi CA, Franks PW, et al. .; Diabetes Prevention Program Research Group . Lifestyle and metformin ameliorate insulin sensitivity independently of the genetic burden of established insulin resistance variants in Diabetes Prevention Program participants. Diabetes 2016;65:520–526
    1. Hivert MF, Jablonski KA, Perreault L, et al. .; DIAGRAM Consortium; Diabetes Prevention Program Research Group . Updated genetic score based on 34 confirmed type 2 diabetes loci is associated with diabetes incidence and regression to normoglycemia in the diabetes prevention program. Diabetes 2011;60:1340–1348
    1. Kilpeläinen TO, Qi L, Brage S, et al. . Physical activity attenuates the influence of FTO variants on obesity risk: a meta-analysis of 218,166 adults and 19,268 children. PLoS Med 2011;8:e1001116.
    1. Shungin D, Deng WQ, Varga TV, et al. .; GIANT Consortium . Ranking and characterization of established BMI and lipid associated loci as candidates for gene-environment interactions. PLoS Genet 2017;13:e1006812.
    1. Graff M, Scott RA, Justice AE, et al. .; CHARGE Consortium; EPIC-InterAct Consortium; PAGE Consortium . Genome-wide physical activity interactions in adiposity—a meta-analysis of 200,452 adults. PLoS Genet 2017;13:e1006528.
    1. Tyrrell J, Wood AR, Ames RM, et al. . Gene-obesogenic environment interactions in the UK Biobank study. Int J Epidemiol 2017;46:559–575
    1. Livingstone KM, Celis-Morales C, Papandonatos GD, et al. . FTO genotype and weight loss: systematic review and meta-analysis of 9563 individual participant data from eight randomised controlled trials. BMJ 2016;354:i4707.
    1. Godino JG, van Sluijs EM, Marteau TM, Sutton S, Sharp SJ, Griffin SJ. Lifestyle advice combined with personalized estimates of genetic or phenotypic risk of type 2 diabetes, and objectively measured physical activity: a randomized controlled trial. PLoS Med 2016;13:e1002185.
    1. Zeevi D, Korem T, Zmora N, et al. . personalized nutrition by prediction of glycemic responses. Cell 2015;163:1079–1094
    1. Jablonski KA, McAteer JB, de Bakker PI, et al. .; Diabetes Prevention Program Research Group . Common variants in 40 genes assessed for diabetes incidence and response to metformin and lifestyle intervention in the diabetes prevention program. Diabetes 2010;59:2672–2681
    1. Apolzan JW, Venditti EM, Edelstein SL, et al. .; Diabetes Prevention Program Research Group . Long-term weight loss with metformin or lifestyle intervention in the Diabetes Prevention Program Outcomes Study. Ann Intern Med 2019;170:682–690
    1. Lean ME, Leslie WS, Barnes AC, et al. . Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial. Lancet 2018;391:541–551
    1. Dennis JM, Shields BM, Jones AG, Pearson ER, Hattersley AT, Henley WE; MASTERMIND consortium . Evaluating associations between the benefits and risks of drug therapy in type 2 diabetes: a joint modeling approach. Clin Epidemiol 2018;10:1869–1877
    1. Dennis JM, Shields BM, Hill AV, et al. .; MASTERMIND Consortium . Precision medicine in type 2 diabetes: clinical markers of insulin resistance are associated with altered short- and long-term glycemic response to DPP-4 inhibitor therapy. Diabetes Care 2018;41:705–712
    1. Feng Y, Mao G, Ren X, et al. . Ser1369Ala variant in sulfonylurea receptor gene ABCC8 is associated with antidiabetic efficacy of gliclazide in Chinese type 2 diabetic patients. Diabetes Care 2008;31:1939–1944
    1. Zhang H, Liu X, Kuang H, Yi R, Xing H. Association of sulfonylurea receptor 1 genotype with therapeutic response to gliclazide in type 2 diabetes. Diabetes Res Clin Pract 2007;77:58–61
    1. Javorsky M, Klimcakova L, Schroner Z, et al. . KCNJ11 gene E23K variant and therapeutic response to sulfonylureas. Eur J Intern Med 2012;23:245–249
    1. Pearson ER, Donnelly LA, Kimber C, et al. . Variation in TCF7L2 influences therapeutic response to sulfonylureas: a GoDARTs study. Diabetes 2007;56:2178–2182
    1. Schroner Z, Javorsky M, Tkacova R, et al. . Effect of sulphonylurea treatment on glycaemic control is related to TCF7L2 genotype in patients with type 2 diabetes. Diabetes Obes Metab 2011;13:89–91
    1. Javorský M, Babjaková E, Klimčáková L, et al. . Association between TCF7L2 genotype and glycemic control in diabetic patients treated with gliclazide. Int J Endocrinol 2013;2013:374858.
    1. Kang ES, Park SY, Kim HJ, et al. . Effects of Pro12Ala polymorphism of peroxisome proliferator-activated receptor gamma2 gene on rosiglitazone response in type 2 diabetes. Clin Pharmacol Ther 2005;78:202–208
    1. Hsieh MC, Lin KD, Tien KJ, et al. . Common polymorphisms of the peroxisome proliferator-activated receptor-gamma (Pro12Ala) and peroxisome proliferator-activated receptor-gamma coactivator-1 (Gly482Ser) and the response to pioglitazone in Chinese patients with type 2 diabetes mellitus. Metabolism 2010;59:1139–1144
    1. Pei Q, Huang Q, Yang GP, et al. . PPAR-γ2 and PTPRD gene polymorphisms influence type 2 diabetes patients’ response to pioglitazone in China. Acta Pharmacol Sin 2013;34:255–261
    1. Zhou K, Bellenguez C, Spencer CC, et al. .; GoDARTS and UKPDS Diabetes Pharmacogenetics Study Group; Wellcome Trust Case Control Consortium 2; MAGIC investigators . Common variants near ATM are associated with glycemic response to metformin in type 2 diabetes. Nat Genet 2011;43:117–120
    1. Zhou K, Yee SW, Seiser EL, et al. .; MetGen Investigators; DPP Investigators; ACCORD Investigators . Variation in the glucose transporter gene SLC2A2 is associated with glycemic response to metformin. Nat Genet 2016;48:1055–1059
    1. Mahajan A, Taliun D, Thurner M, et al. . Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat Genet 2018;50:1505–1513
    1. Zhou K, Donnelly L, Burch L, et al. . Loss-of-function CYP2C9 variants improve therapeutic response to sulfonylureas in type 2 diabetes: a Go-DARTS study. Clin Pharmacol Ther 2010;87:52–56
    1. Dawed AY, Donnelly L, Tavendale R, et al. . CYP2C8 and SLCO1B1 variants and therapeutic response to thiazolidinediones in patients with type 2 diabetes. Diabetes Care 2016;39:1902–1908
    1. Shu Y, Sheardown SA, Brown C, et al. . Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J Clin Invest 2007;117:1422–1431
    1. Sundelin E, Gormsen LC, Jensen JB, et al. . Genetic polymorphisms in organic cation transporter 1 attenuates hepatic metformin exposure in humans. Clin Pharmacol Ther 2017;102:841–848
    1. Zhou K, Donnelly LA, Kimber CH, et al. . Reduced-function SLC22A1 polymorphisms encoding organic cation transporter 1 and glycemic response to metformin: a GoDARTS study. Diabetes 2009;58:1434–1439
    1. Dujic T, Zhou K, Yee SW, et al. . Variants in pharmacokinetic transporters and glycemic response to metformin: a MetGen meta-Analysis. Clin Pharmacol Ther 2017;101:763–772
    1. Kim YG, Hahn S, Oh TJ, Kwak SH, Park KS, Cho YM. Differences in the glucose-lowering efficacy of dipeptidyl peptidase-4 inhibitors between Asians and non-Asians: a systematic review and meta-analysis. Diabetologia 2013;56:696–708
    1. Davis TME, Mulder H, Lokhnygina Y, et al. .; TECOS Study Group . Effect of race on the glycaemic response to sitagliptin: insights from the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS). Diabetes Obes Metab 2018;20:1427–1434
    1. Williams LK, Padhukasahasram B, Ahmedani BK, et al. . Differing effects of metformin on glycemic control by race-ethnicity. J Clin Endocrinol Metab 2014;99:3160–3168
    1. Kim C, Newton KM, Knopp RH. Gestational diabetes and the incidence of type 2 diabetes: a systematic review. Diabetes Care 2002;25:1862–1868
    1. Das Gupta R, Gupta S, Das A, Biswas T, Haider MR, Sarker M. Ethnic predisposition of diabetes mellitus in the patients with previous history of gestational diabetes mellitus: a review. Expert Rev Endocrinol Metab 2018;13:149–158
    1. Lowe WL Jr, Scholtens DM, Sandler V, Hayes MG. Genetics of gestational diabetes mellitus and maternal metabolism. Curr Diab Rep 2016;16:15.
    1. Hayes MG, Urbanek M, Hivert MF, et al. .; HAPO Study Cooperative Research Group . Identification of HKDC1 and BACE2 as genes influencing glycemic traits during pregnancy through genome-wide association studies. Diabetes 2013;62:3282–3291
    1. Powe CE, Allard C, Battista MC, et al. . Heterogeneous contribution of insulin sensitivity and secretion defects to gestational diabetes mellitus. Diabetes Care 2016;39:1052–1055
    1. Benhalima K, Van Crombrugge P, Moyson C, et al. . Characteristics and pregnancy outcomes across gestational diabetes mellitus subtypes based on insulin resistance. Diabetologia 2019;62:2118–2128
    1. Cooray SD, Boyle JA, Soldatos G, Wijeyaratne LA, Teede HJ. Prognostic prediction models for pregnancy complications in women with gestational diabetes: a protocol for systematic review, critical appraisal and meta-analysis. Syst Rev 2019;8:270.
    1. Tobias DK. Prediction and prevention of type 2 diabetes in women with a history of GDM. Curr Diab Rep 2018;18:78.
    1. Aroda VR, Christophi CA, Edelstein SL, et al. .; Diabetes Prevention Program Research Group . The effect of lifestyle intervention and metformin on preventing or delaying diabetes among women with and without gestational diabetes: the Diabetes Prevention Program outcomes study 10-year follow-up. J Clin Endocrinol Metab 2015;100:1646–1653
    1. American Diabetes Association 14. Management of diabetes in pregnancy: Standards of Medical Care in Diabetes—2020. Diabetes Care 2020;43(Suppl. 1):S183–S192
    1. Spyer G, Macleod KM, Shepherd M, Ellard S, Hattersley AT. Pregnancy outcome in patients with raised blood glucose due to a heterozygous glucokinase gene mutation. Diabet Med 2009;26:14–18
    1. Sanyoura M, Letourneau L, Knight Johnson AE, et al. . GCK-MODY in the US monogenic diabetes registry: description of 27 unpublished variants. Diabetes Res Clin Pract 2019;151:231–236
    1. De Franco E, Caswell R, Houghton JA, Iotova V, Hattersley AT, Ellard S. Analysis of cell-free fetal DNA for non-invasive prenatal diagnosis in a family with neonatal diabetes. Diabet Med 2017;34:582–585
    1. Petrak F, Baumeister H, Skinner TC, Brown A, Holt RIG. Depression and diabetes: treatment and health-care delivery. Lancet Diabetes Endocrinol 2015;3:472–485
    1. Snoek FJ, Bremmer MA, Hermanns N. Constructs of depression and distress in diabetes: time for an appraisal. Lancet Diabetes Endocrinol 2015;3:450–460
    1. Moulton CD, Pickup JC, Ismail K. The link between depression and diabetes: the search for shared mechanisms. Lancet Diabetes Endocrinol 2015;3:461–471
    1. American Diabetes Association 5. Facilitating behavior change and well-being to improve health outcomes: Standards of Medical Care in Diabetes—2020. Diabetes Care 2020;43(Suppl. 1):S48–S65
    1. Fisher L, Polonsky WH, Hessler D. Addressing diabetes distress in clinical care: a practical guide. Diabet Med 2019;36:803–812
    1. Shields BM, Hicks S, Shepherd MH, Colclough K, Hattersley AT, Ellard S. Maturity-onset diabetes of the young (MODY): how many cases are we missing? Diabetologia 2010;53:2504–2508
    1. Shepherd M, Colclough K, Ellard S, Hattersley AT. Ten years of the national genetic diabetes nurse network: a model for the translation of genetic information into clinical care. Clin Med (Lond) 2014;14:117–121
    1. Owen KR. Monogenic diabetes in adults: what are the new developments? Curr Opin Genet Dev 2018;50:103–110
    1. Poudel A, Zhou JY, Story D, Li L. Diabetes and associated cardiovascular complications in American Indians/Alaskan Natives: a review of risks and prevention strategies. J Diabetes Res 2018;2018:2742565.
    1. Al Busaidi N, Shanmugam P, Manoharan D. Diabetes in the Middle East: government health care policies and strategies that address the growing diabetes prevalence in the Middle East. Curr Diab Rep 2019;19:8.
    1. Meyer RJ. Precision medicine, diabetes, and the U.S. Food and Drug Administration. Diabetes Care 2016;39:1874–1878
    1. Naylor R. Economics of genetic testing for diabetes. Curr Diab Rep 2019;19:23.
    1. Fitipaldi H, McCarthy MI, Florez JC, Franks PW. A global overview of precision medicine in type 2 diabetes. Diabetes 2018;67:1911–1922
    1. O’Brien RL, Brinster RL, Storb U. Somatic hypermutation of an immunoglobulin transgene in kappa transgenic mice. Nature 1987;326:405–409

Source: PubMed

3
Subscribe