Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

A combined risk score enhances prediction of type 1 diabetes among susceptible children

An Author Correction to this article was published on 14 February 2022

This article has been updated

Abstract

Type 1 diabetes (T1D)—an autoimmune disease that destroys the pancreatic islets, resulting in insulin deficiency—often begins early in life when islet autoantibody appearance signals high risk1. However, clinical diabetes can follow in weeks or only after decades, and is very difficult to predict. Ketoacidosis at onset remains common2,3 and is most severe in the very young4,5, in whom it can be life threatening and difficult to treat6,7,8,9. Autoantibody surveillance programs effectively prevent most ketoacidosis10,11,12 but require frequent evaluations whose expense limits public health adoption13. Prevention therapies applied before onset, when greater islet mass remains, have rarely been feasible14 because individuals at greatest risk of impending T1D are difficult to identify. To remedy this, we sought accurate, cost-effective estimation of future T1D risk by developing a combined risk score incorporating both fixed and variable factors (genetic, clinical and immunological) in 7,798 high-risk children followed closely from birth for 9.3 years. Compared with autoantibodies alone, the combined model dramatically improves T1D prediction at ≥2 years of age over horizons up to 8 years of age (area under the receiver operating characteristic curve ≥ 0.9), doubles the estimated efficiency of population-based newborn screening to prevent ketoacidosis, and enables individualized risk estimates for better prevention trial selection.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Average time-dependent ROC AUCs for the three-variable model by age at prediction scoring.
Fig. 2: ROC curves derived from models incorporating different numbers of variables.
Fig. 3: Performance of the three-variable model at a 5-year horizon.
Fig. 4: Three strategies for population-based newborn screening and surveillance follow-up.

Similar content being viewed by others

Data availability

Clinical metadata and GRS genotyping data analyzed for this study are available in the NIDDK Central Repository at https://www.niddkrepository.org/studies/teddy, in accordance with the NIDDK’s controlled-access authorization process.

Code availability

The R code used in these analyses is available in the NIDDK Central Repository at https://www.niddkrepository.org/studies/teddy, in accordance with the NIDDK’s controlled-access authorization process.

Change history

References

  1. Ziegler, A. G. et al. Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. J. Am. Med. Assoc.309, 2473–2479 (2013).

    CAS  Google Scholar 

  2. Dabelea, D. et al. Trends in the prevalence of ketoacidosis at diabetes diagnosis: the SEARCH for Diabetes in Youth Study. Pediatrics133, e938–e945 (2014).

    PubMed  PubMed Central  Google Scholar 

  3. Alonso, G. T. et al. Diabetic ketoacidosis at diagnosis of type 1 diabetes in Colorado children, 2010–2017. Diabetes Care43, 117–121 (2020).

    PubMed  Google Scholar 

  4. Jefferies, C. et al. 15-year incidence of diabetic ketoacidosis at onset of type 1 diabetes in children from a regional setting (Auckland, New Zealand). Sci. Rep.5, 10358 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Iovane, B. et al. Diabetic ketoacidosis at the onset of type 1 diabetes in young children: is it time to launch a tailored campaign for DKA prevention in children <5 years? Acta Biomed.89, 67–71 (2018).

    PubMed  PubMed Central  Google Scholar 

  6. Rewers, M. et al. Assessment and monitoring of glycemic control in children and adolescents with diabetes. Pediatr. Diabetes8, 408–418 (2007).

    PubMed  Google Scholar 

  7. Usher-Smith, J. A., Thompson, M. J., Sharp, S. J. & Walter, F. M. Factors associated with the presence of diabetic ketoacidosis at diagnosis of diabetes in children and young adults: a systematic review. Br. Med. J.343, d4092 (2011).

    Google Scholar 

  8. Desai, D., Mehta, D., Mathias, P., Menon, G. & Schubart, U. K. Health care utilization and burden of diabetic ketoacidosis in the U.S. over the past decade: a nationwide analysis. Diabetes Care41, 1631–1638 (2018).

    PubMed  Google Scholar 

  9. Glaser, N. et al. Risk factors for cerebral edema in children with diabetic ketoacidosis. N. Engl. J. Med.344, 264–269 (2001).

    CAS  PubMed  Google Scholar 

  10. Barker, J. M. et al. Clinical characteristics of children diagnosed with type 1 diabetes through intensive screening and follow-up. Diabetes Care27, 1399–1404 (2004).

    PubMed  Google Scholar 

  11. Winkler, C., Schober, E., Ziegler, A. G. & Holl, R. W. Markedly reduced rate of diabetic ketoacidosis at onset of type 1 diabetes in relatives screened for islet autoantibodies. Pediatr. Diabetes13, 301–306 (2012).

    Google Scholar 

  12. Elding Larsson, H. et al. Children followed in the TEDDY study are diagnosed with type 1 diabetes at an early stage of disease. Pediatr. Diabetes15, 118–126 (2014).

    PubMed  Google Scholar 

  13. Meehan, C., Fout, B., Ashcraft, J., Schatz, D. A. & Haller, M. J. Screening for T1D risk to reduce DKA is not economically viable. Pediatr. Diabetes16, 565–572 (2015).

    CAS  PubMed  Google Scholar 

  14. Bonifacio, E. et al. Effects of high-dose oral insulin on immune responses in children at high risk for type 1 diabetes. J. Am. Med. Assoc.313, 1541–1549 (2015).

    CAS  Google Scholar 

  15. Noble, J. A. & Valdes, A. M. Genetics of the HLA region in the prediction of type 1 diabetes. Curr. Diabetes Rep.11, 533–542 (2011).

    CAS  Google Scholar 

  16. Rewers, M. & Ludvigsson, J. Environmental risk factors for type 1 diabetes. Lancet387, 2340–2348 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Beyerlein, A., Wehweck, F., Ziegler, A.-G. & Pflueger, M. Respiratory infections in early life and the development of islet autoimmunity in children at increased type 1 diabetes risk. JAMA Pediatr.167, 800–807 (2013).

    PubMed  Google Scholar 

  18. Verge, C. et al. Prediction of type I diabetes in first-degree relatives using a combination of insulin, GAD, and ICA512bdc/IA-2 autoantibodies. Diabetes45, 926–933 (1996).

    CAS  PubMed  Google Scholar 

  19. Sosenko, J. M. et al. Glucose and C-peptide changes in the perionset period of type 1 diabetes in the diabetes prevention trial–type 1. Diabetes Care31, 2188–2192 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Redondo, M. J. et al. A type 1 diabetes genetic risk score predicts progression of islet autoimmunity and development of type 1 diabetes in individuals at risk. Diabetes Care41, 1887–1894 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Winkler, C. et al. Feature ranking of type 1 diabetes susceptibility genes improves prediction of type 1 diabetes. Diabetologia57, 2521–2529 (2014).

    CAS  PubMed  Google Scholar 

  22. Oram, R. A. et al. A type 1 diabetes genetic risk score can aid discrimination between type 1 and type 2 diabetes in young adults. Diabetes Care39, 337–344 (2015).

    PubMed  Google Scholar 

  23. Beyerlein, A. et al. Progression from islet autoimmunity to clinical type 1 diabetes is influenced by genetic factors: results from the prospective TEDDY study. J. Med. Genet.56, 602–605 (2019).

    CAS  PubMed  Google Scholar 

  24. Bonifacio, E. et al. Genetic scores to stratify risk of developing multiple islet autoantibodies and type 1 diabetes: a prospective study in children. PLoS Med.15, e1002548 (2018).

    PubMed  PubMed Central  Google Scholar 

  25. Hippich, M. et al. Genetic contribution to the divergence in type 1 diabetes risk between children from the general population and children from affected families. Diabetes68, 847–857 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Familial risk of type I diabetes in European children. The EURODIAB ACE Study Group & The EURODIAB ACE Substudy 2 Study Group. Diabetologia41, 1151–1156 (1998).

  27. LaGasse, J. M. et al. Successful prospective prediction of type 1 diabetes in schoolchildren through multiple defined autoantibodies: an 8-year follow-up of the Washington State Diabetes Prediction Study. Diabetes Care25, 505–511 (2002).

    PubMed  Google Scholar 

  28. Ziegler, A. G. et al. Primary prevention of beta-cell autoimmunity and type 1 diabetes—the Global Platform for the Prevention of Autoimmune Diabetes (GPPAD) perspectives. Mol. Metab.5, 255–262 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Hagopian, W. A. et al. TEDDY—The Environmental Determinants of Diabetes in the Young: an observational clinical trial. Ann. NY Acad. Sci.1079, 320–326 (2006).

    PubMed  Google Scholar 

  30. Sharp, S. A. et al. Development and standardization of an improved type 1 diabetes genetic risk score for use in newborn screening and incident diagnosis. Diabetes Care42, 200–207 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Rewers, M. et al. The Environmental Determinants of Diabetes in the Young (TEDDY) study: 2018 update. Curr. Diabetes Rep.18, 136 (2018).

    Google Scholar 

  32. Krischer, J. P. et al. Genetic and environmental interactions modify the risk of diabetes-related autoimmunity by 6 years of age: the TEDDY study. Diabetes Care40, 1194–1202 (2017).

    PubMed  PubMed Central  Google Scholar 

  33. Krischer, J. P. et al. Predicting islet cell autoimmunity and type 1 diabetes: an 8-year TEDDY study progress report. Diabetes Care42, 1051–1060 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Hippisley-Cox, J., Coupland, C. & Brindle, P. Development and validation of QRISK3 risk prediction algorithms to estimate future risk of cardiovascular disease: prospective cohort study. Br. Med. J.357, j2099 (2017).

    Google Scholar 

  35. National Institutes of Health Consensus Development Panel National Institutes of Health Consensus Development Conference Statement: phenylketonuria: screening and management, October 16–18, 2000. Pediatrics108, 972–982 (2001).

    Google Scholar 

  36. Edge, J. A., Ford-Adams, M. E. & Dunger, D. B. Causes of death in children with insulin dependent diabetes 1990–96. Arch. Dis. Child.81, 318–323 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Rose, G. Sick individuals and sick populations. Int. J. Epidemiol.14, 32–38 (1985).

    CAS  PubMed  Google Scholar 

  38. Hyöty, H., Leon, F. & Knip, M. Developing a vaccine for type 1 diabetes by targeting coxsackievirus B. Expert Rev. Vaccines17, 1071–1083 (2018).

    PubMed  Google Scholar 

  39. Redondo, M. J., Oram, R. A. & Steck, A. K. Genetic risk scores for type 1 diabetes prediction and diagnosis. Curr. Diabetes Rep.17, 129 (2017).

    Google Scholar 

  40. Cheng, B.-W. et al. Autoantibodies against islet cell antigens in children with type 1 diabetes mellitus. Oncotarget9, 16275–16283 (2018).

    PubMed  PubMed Central  Google Scholar 

  41. TEDDY Study Group The Environmental Determinants of Diabetes in the Young (TEDDY) study. Ann. NY Acad. Sci.1150, 1–13 (2008).

    Google Scholar 

  42. Hagopian, W. A. et al. The Environmental Determinants of Diabetes in the Young (TEDDY): genetic criteria and international diabetes risk screening of 421,000 infants. Pediatr. Diabetes12, 733–743 (2011).

    PubMed  PubMed Central  Google Scholar 

  43. Lönnrot, M. et al. A method for reporting and classifying acute infectious diseases in a prospective study of young children: TEDDY. BMC Pediatr.15, 24 (2015).

    PubMed  PubMed Central  Google Scholar 

  44. Bonifacio, E. et al. Harmonization of glutamic acid decarboxylase and islet antigen-2 autoantibody assays for National Institute of Diabetes and Digestive and Kidney Diseases consortia. J. Clin. Endocrinol. Metab.95, 3360–3367 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Cortes, A. & Brown, M. A. Promise and pitfalls of the immunochip. Arthritis Res. Ther.13, 101 (2011).

    PubMed  PubMed Central  Google Scholar 

  46. Krischer, J. P. et al. The 6 year incidence of diabetes-associated autoantibodies in genetically at-risk children: the TEDDY study. Diabetologia58, 980–987 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Dafni, U. Landmark analysis at the 25-year landmark point. Circ. Cardiovasc. Qual. Outcomes4, 363–371 (2011).

    PubMed  Google Scholar 

  48. Anderson, J. R., Cain, K. C. & Gelber, R. D. Analysis of survival by tumor response. J. Clin. Oncol.1, 710–719 (1983).

    CAS  PubMed  Google Scholar 

  49. Klein, J. P. & Moeschberger, M. L. Survival Analysis. Techniques for Censored and Truncated Data (Springer, 2003).

  50. Venables, W. N. & Ripley, B. Modern Applied Statistics with S (Springer, 2002).

  51. Blanche, P., Dartigues, J.-F. & Jacqmin-Gadda, H. Estimating and comparing time-dependent areas under receiver operating characteristic curves for censored event times with competing risks. Stat. Med.32, 5381–5397 (2013).

    PubMed  Google Scholar 

Download references

Acknowledgements

The TEDDY study is included in ClinicalTrials.gov under the identifier NCT00279318. The TEDDY Study Group is funded by U01 DK63829, U01 DK63861, U01 DK63821, U01 DK63865, U01 DK63863, U01 DK63836, U01 DK63790, UC4 DK63829, UC4 DK63861, UC4 DK63821, UC4 DK63865, UC4 DK63863, UC4 DK63836, UC4 DK95300, UC4 DK100238, UC4 DK106955, UC4 DK112243, UC4 DK117483 and UC4 DK100238, and by contract number HHSN267200700014C from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institute of Allergy and Infectious Diseases (NIAID), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institute of Environmental Health Sciences (NIEHS), Centers for Disease Control and Prevention (CDC) and JDRF. This work is supported in part by the National Institutes of Health/National Center for Advancing Translational Sciences Clinical and Translational Science Awards (UL1 TR000064 (University of Florida) and UL1 TR001082 (University of Colorado)) and Diabetes Research Center (5P30 DK017047; University of Washington). R.A.O. is supported by a Diabetes UK Harry Keen Fellowship (16/0005529). S.A.S. is supported by a Diabetes UK PhD studentship (17/0005757). M.N.W. is supported by the Wellcome Trust Institutional Strategic Support Fund (WT097835MF). R.A.O., L.A.F., W.A.H. and K.V. are supported by a JDRF strategic research agreement (3-SRA-2019-827-S-B). The authors thank the following former staff members at TEDDY Study Group centers: Michael Abbondondolo, Lori Ballard, David Hadley, Hye-Seung Lee, Wendy McLeod, Steven Meulemans, Henry Erlich, Steven J. Mack and Anna Lisa Fear.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

L.A.F., W.A.H., R.A.O., K.V. and S.A.S. designed the study, contributed to analysis and wrote the manuscript. Å.L., M.J.R., J.-X.S., A.-G.Z., J.T., B.A., J.P.K. and M.N.W. contributed to analysis and reviewed the manuscript. All authors contributed to discussions and editing of the manuscript.

Corresponding author

Correspondence to William A. Hagopian.

Ethics declarations

Competing interests

R.A.O. holds a UK Medical Research Council Institutional Confidence in Concept grant to develop a ten-SNP biochip T1D genetic test in collaboration with Randox. A.-G.Z. is a co-applicant on patent application WO/2019/002364 Al covering the use of a GRS to identify and treat individuals with high T1D genetic risk. Neither of these genetic risk tests is identical to the more extensive GRS2 used in this paper. The other authors declare no competing interests.

Peer review

Peer review information

Saheli Sadanand was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Variables previously shown or susceptible to be associated with T1D auto-immunity evaluated in univariate analysis.

Time ROC AUC and p-value (two side Wald test) are computed at landmark age 2 years and horizon of 8 years (n = 6,805). Abbreviations: Type 1 diabetes (T1D), Family history (FH), Islet Autoantibodies (AB), insulinoma Antigen-2 Autoantibody (IA2A), Glutamic Acid Decarboxylase Autoantibody (GADA), Insulin AutoAntibody (IAA), Genetic Risk score (GRS2). The referent sex is female. A concise list of references for this table is provided in the Supplementary Information file associated with this paper.

Extended Data Fig. 2 Time dependent ROC curves comparing the performance of various genetic risk scores in the TEDDY cohort.

Shown are curves for GRS1, GRS2 and the combined TEDDY GRS to predict T1D from a landmark age of birth and a horizon interval of 8 years (n= 7,798).

Extended Data Fig. 3 Family history adds predictive power to the T1D GRS2.

T1D GRS2 alone (a) is compared to T1D GRS2 + FH (b) at nine different landmark scoring ages and over four different horizon times. Although 95% confidence intervals always overlapped, among 34 total combinations, T1D GRS2 + FH gave a larger AUC ROC in 24 of these combinations. Results were similar in 9 combinations, and in only one instance was T1D GRS2 better. T1D GRS2 + FH superiority was greatest at landmarks ≤3 years of age. The number of children at each landmark age were 7798 (birth), 7563 (1 year), 7123 (1.5 years), 6805 (2 years), 6316 (3 years), 5973 (4 years), 5706 (5 years), 5517 (6 years) and 5323 (7 years).

Extended Data Fig. 4 T1D GRS2 and family history add predictive power to AB.

AB alone (a) is compared to the three-variable model of AB, GRS2 and FH. (b) at eight different landmark scoring ages and over four different horizon times. Although 95% confidence intervals overlapped, among 30 total combinations, the three-variable model yielded larger AUC ROC in 28 of these combinations and similar results in the remaining 2 combinations. The differences were often substantial, especially at landmarks ≤4 years of age. The number of children at each landmark age were 7798 (birth), 7563 (1 year), 7123 (1.5 years), 6805 (2 years), 6316 (3 years), 5973 (4 years), 5706 (5 years), 5517 (6 years) and 5323 (7 years).

Extended Data Fig. 5 Hazard ratio for each variable at different ages at prediction scoring landmarks.

Each point represents the hazard ratio at a landmark age (x abscises), the shaded region its respective 95% confidence interval. The number of children at each landmark age were 7798 (birth), 7563 (1 year), 7123 (1.5 years), 6805 (2 years), 6316 (3 years), 5973 (4 years), 5706 (5 years), 5517 (6 years) and 5323 (7 years).

Extended Data Fig. 6 Time dependent ROC of different models now considering only children positive for at least one AB (n = 252).

The landmark age is 2 years. At the 3 year time horizon the CRS (AB+GRS2+FH) performs similarly to AB only, but at the 8 year horizon the CRS is more predictive.

Extended Data Fig. 7 Individual estimated future T1D risk probability percentages (and 95% confidence intervals) for 24 different scenarios combining a GRS risk level and FH background with different AB status calcluated at age 2 years.

“++” represents a T1D genetic risk score at 80th percentile of the general (UK) population. “+++” represents a T1D genetic risk score at 90th percentile of the general (UK) population. “++++” represents a T1D genetic risk score at 99th percentile of the general (UK) population.

Extended Data Fig. 8 Comparison of newborn screening strategies aiming to predict ≥75% of the children who will develop T1D before age 10.

In the “Classic” design, the 9.3% of screened newborn population containing 75% of the T1D cases, are all followed for 10 years. In the “Simple Adaptive” design, 10.7% of the screened newborns containing 79.8% of the T1D cases, are followed for variable lengths determined by CRS-based risk, and 4.8% of T1D cases miss AB detection before onset, leaving 75% detected in advance. In the “Advanced Adaptive” design, 11.2% of the screened newborns containing 81.6 % of T1D cases are followed closely or less closely determined by CRS-based risk, 6.6% of cases miss AB detection before onset, again leaving 75% detected. Numbers are computed by using the performance of each strategy on TEDDY data. Tests per child are computed using TEDDY data and simulation to take into account right censoring in TEDDY data.

Extended Data Fig. 9 Visit number calculation for each design.

Table A. Visit number calculations for the “Classic” design. Infants initially selected for high. GRS2 genetic risk were all followed quarterly until age 3, and every 6 months until age 6, then annually thereafter. This simulation was made on the TEDDY dataset. Table B. Visit number calculations for the “Simple Adaptive” design. Infants selected for high genetic risk were initially followed as in the Classic strategy, but the T1D CRS was recalculated at annual landmarks, at which time any child whose T1D probability by age 10 had decreased to <0.8% was eliminated from further follow-up. Of new cases, 94% had high risk detected before onset. This simulation was made on the TEDDY dataset. Table C. Visit number calculations for the “Advanced Adaptive” design. Infants selected for high genetic risk were initially followed as in the Classic strategy, but at birth and annually thereafter, a T1D CRS calculation was used to reallocate children among the quarterly or annual surveillance groups based on T1D probability in 2 years of ≥0.6% or <0.6%, respectively. Of new cases, 92% had high risk detected before onset. Simulation made on the TEDDY dataset.

Extended Data Fig. 10 GRS2 violin plot in the Type 1 Diabetes Genetics Consortium (T1DGC) and TEDDY datasets.

T1DGC is more representative of the general background population. The genetic pre- selection in TEDDY based on the major T1D risk locus HLA-DR-DQ, renders the T1D GRS2 higher in TEDDY, even in T1D free subjects. Further, the separation between T1D and non-T1D subjects in TEDDY is much less. There are 7,798 observations in TEDDY including 305 with T1D. There are 15729 observations in T1DGC including 6483 with T1D. The lines in the violin plots respectively indicate the 25th, 50th and 75th percentiles, while the lowest and the highest point of each violin plot indicates the minimum and the maximum, respectively, for each group of individuals.

Supplementary information

Supplementary Information

References cited in Extended Data Fig. 1, and Supplementary Tables 1–4.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ferrat, L.A., Vehik, K., Sharp, S.A. et al. A combined risk score enhances prediction of type 1 diabetes among susceptible children. Nat Med 26, 1247–1255 (2020). https://doi.org/10.1038/s41591-020-0930-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-0930-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing