GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 28, No. 3 ( 2022-03), p. 599-599
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 1484517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 26, No. 8 ( 2020-08-01), p. 1247-1255
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 1484517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Diabetes, American Diabetes Association, Vol. 68, No. Supplement_1 ( 2019-06-01)
    Abstract: Background: There is an unmet need for accurate cost-effective estimation of future T1D risk. Methods: We derived a combined T1D prediction model using 7883 children followed closely from birth for a median of 9 yr, considering T1D genetic risk score (GRS), family T1D history (FH), standard islet autoantibodies (IA), race, birth circumstances, early growth and nutrient status. T1D developed in 326. Results: Machine learning and traditional methods (Cox models) performed equivalently as measured by time-dependent AUC ROC. Future T1D was accurately predicted by combining only 3 variables: GRS, FH and IA status. Accuracy of combined scores increased with age at scoring. By age 2, they were highly predictive for T1D in the next 5 years (AUC & gt;0.91, 95% CI 0.88-0.95) (see X on Figure). A 2-yr-old with 2 IA, positive FH and high GRS ( & gt;12) would have T1D risk over the next 1, 3 and 5 years of 14% (8-19%), 36% (25-45%) and 51% (39%-61%) respectively. A 2-yr-old with 1 IA, no FH and moderate GRS (10-11) has T1D risk of 0.8% (0.6-1.2%), 2.6% (1.9-3.2%) and 4.3% (3.4%-5.2%) respectively. After newborn genetic screening, only simple venous sampling in routine healthcare settings is required. Conclusion: This approach allows updated individual risk estimates by age, and in the future may enable release of low risk individuals from surveillance long after initial newborn screening for more cost-efficient population based pediatric T1D prediction. Disclosure L.A. Ferrat: None. K. Vehik: None. S.A. Sharp: None. Å. Lernmark: None. A. Ziegler: None. M. Rewers: None. J. She: None. J. Toppari: None. B. Akolkar: None. J. Krischer: None. M.N. Weedon: None. S.S. Rich: None. R.A. Oram: Other Relationship; Self; Randox Laboratories Ltd. W. Hagopian: Research Support; Self; Novo Nordisk A/S. Funding National Institute of Diabetes and Digestive and Kidney Diseases; National Institute of Allergy and Infectious Diseases; Eunice Kennedy Shriver National Institute of Child Health and Human Development; National Institute of Environmental Health Sciences; JDRF; Centers for Disease Control and Prevention
    Type of Medium: Online Resource
    ISSN: 0012-1797 , 1939-327X
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2019
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Diabetes, American Diabetes Association, Vol. 68, No. Supplement_1 ( 2019-06-01)
    Abstract: Accurate classification of diabetes type guides correct treatment. We recently showed a type 1 diabetes (T1D) genetic risk score (GRS-1), combined with clinical features and biomarkers discriminates between T1D and type 2 (T2D) and MODY in European adults. We aimed to test the ability of an improved 67 SNP T1D score (GRS-2) to discriminate T1D in an ethnically diverse U.S. pediatric population. We included 1818 SEARCH Study participants who had a C-peptide at a median (IQR) of 8 (6-9) years post diagnosis. T1D was defined based on severe insulin deficiency (fasting C-peptide & lt;0.25 ng/ml) at follow-up. We generated genetic risk scores from SNP data, and assessed discriminative power of the T1D GRS-2 using the area under the receiver operating curve (AUC) in the three largest ethnic groups in SEARCH: white (n=1101, 95% T1D), black (n=228, 59% T1D) and Hispanic (n=257, 77% T1D). We then assessed the discriminative power of GRSs combined with autoantibody and clinical data. The T1D GRS-2 was discriminative of T1D across all three ethnic groups AUC (95% CI) for GRS-1 vs. GRS-2 was 0.88 (0.84, 0.93) vs. 0.88 (0.83, 0.92) in white, 0.82 (0.77, 0.87) vs. 0.87 (0.83, 0.92) in blacks, and 0.87 (0.82, 0.92) vs. 0.9 (0.86, 0.95) in Hispanics). Combined risk scores were most accurate at classifying T1D. AUC for different scores were: 0.93 (0.91, 0.95) for type assigned by provider at diagnosis, 0.95 (0.93, 0.96) for age and BMI, 0.95 (0.94, 0.97) for GAD/IA-2/ZNT8 autoantibodies, and 0.99 (0.99, 1.00) for a combined approach using autoantibodies, GRS-2 and clinical features. These results were similar across all race/ethnic groups. A new T1D GRS-2 is discriminative of T1D in a racial/ethnically diverse pediatric population. Using GRS2 in a combined model of clinical features, autoantibodies and genetics offers near perfect classification of T1D and could be used to accurately classify diabetes type. Disclosure R.A. Oram: Other Relationship; Self; Randox Laboratories Ltd. S.A. Sharp: None. C. Pihoker: None. L.A. Ferrat: None. G. Imperatore: None. S. Saydah: None. A.H. Williams: None. L.E. Wagenknecht: None. J.M. Lawrence: None. M.N. Weedon: None. R. Dagostino: Consultant; Self; Acelity, Amgen Inc. W. Hagopian: Research Support; Self; Novo Nordisk A/S. J. Divers: None. D. Dabelea: None. Funding National Institutes of Health (UC4DK108173)
    Type of Medium: Online Resource
    ISSN: 0012-1797 , 1939-327X
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2019
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Diabetes, American Diabetes Association, Vol. 71, No. Supplement_1 ( 2022-06-01)
    Abstract: Introduction: Screening for type 1 diabetes (T1D) genetic risk in early life can prevent life threatening complications such as diabetic ketoacidosis and allow cost-effective recruitment into intervention and immunotherapy trials. Celiac disease (CD) frequently presents as a comorbidity in those with T1D and shares a strong genetic basis with T1D. Single nucleotide polymorphism (SNP) based genetic risk scores (GRS) for both T1D and CD have shown to be very predictive of future disease but are difficult to generate and typically require SNP array genotyping. We aimed to develop a combined GRS screening panel that could be genotyped from a single dried blood spot at birth. Methods: We developed assays for proxy SNPs of common HLA-DQ haplotypes reported in previous GRS and additional loci from the most recent available genome-wide association studies (GWAS) . We used backwards stepwise regression to identify a subset of variants able to be genotyped with DNA eluted from 800 6mm dried blood spots. We developed and validated neural network models to quantify genetic risk of both T1D and CD using T1DGC and UK Celiac case-control SNP array data, validated in UK Biobank. Assays were developed with LGC Genomics and validated on 675 Seattle area samples. Results: The complete panel consisted of 71 validated SNP assays, including 11 backup variants for key loci. We generated neural nework models which demonstrate equivalent or greater predictive power (AUC: T1D=0.914, CD=0.893) to previously published GRS yet require much less expertise to apply. We have developed an algorithm for preparation of raw genotyping data and subsequent generation of GRS requiring little expertise to apply. Conclusion: A 71 SNP blood-spot screening panel is highly effective at screening genetic risk associated with T1D and CD at birth. Using a neural network model the panel enables widely available, easy to generate and inexpensive population screening of genetic risk for T1D and CD. Disclosure S. A. Sharp: None. J. M. Locke: None. Y. Xu: None. D. P. Fraser: None. L. A. Ferrat: None. M. N. Weedon: None. M. Inouye: None. R. A. Oram: Consultant; Janssen Research & Development, LLC, Research Support; Randox R & D. W. Hagopian: Research Support; Janssen Research & Development, LLC. Funding Diabetes UK (16/0005529) JDRF (3-SRA-2019-827-S-B)
    Type of Medium: Online Resource
    ISSN: 0012-1797
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2022
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Diabetes, American Diabetes Association, Vol. 70, No. Supplement_1 ( 2021-06-01)
    Abstract: Background: Type 1 diabetes (T1D) genetic risk score (GRS) and islet autoantibody (AAb) specificity and titers influence T1D risk; however, these factors collected at baseline in cross sectional studies have not been effectively incorporated in the current models for prediction of progression along pre-clinical stages of T1D. Our aim was to develop an updated model of T1D prediction for research and clinical practice. Methods: AAb-positive relatives of individuals with T1D (n=1,233, 52% female, 88% European ancestry, age mean 15 yrs [SD 12.2]) from the TrialNet Pathway to Prevention were followed for a median of 3.1 years (IQR 1.1 -7.1). T1D developed in 498/1,233 (40.4%). T1D GRS, AAb number, AAb combination, AAb titer, age and sex were evaluated on the combined T1D prediction model, estimating its performance using 3-fold cross-validation (10x repeated). Results: Machine learning (survival random forest) and traditional methods (Cox models, extended Cox model) performed equivalently as measured by 5 yr horizon time-dependent AUC ROC (respectively 0.74, 0.74, 0.72), on calibration by integrated Brier score (respectively 0.15, 0.15, 0.16), and by calibration plots. AAb combinations with specific variables predicted T1D. In multivariate analysis, T1D GRS was a significant predictor of T1D in individuals positive for GADA, IAA or both. For any autoantibody combination including IA2A, the IA2A titer was predictive while the T1D GRS was not. Variable importance analyses showed that T1D was predicted by IA2A titer level, followed by T1D GRS and combinations of specific AAbs. Conclusion: We modeled individual estimates of T1D risk with T1D GRS, AAb characteristics, age, and sex. T1D prediction improved by adding T1D GRS and AAb characteristics to other variables, and was similar using machine learning or traditional models. T1D GRS was a significant predictor only in the absence of IA2A. In the presence of IA2A, IA2A titer was the most influential factor. Disclosure L. A. Ferrat: None. M. J. Redondo: Advisory Panel; Self; Provention Bio, Inc. A. Steck: None. H. M. Parikh: None. L. You: None. S. Onengut-gumuscu: None. P. Gottlieb: Advisory Panel; Self; Janssen Research & Development, LLC, Tolerion, Inc., Viacyte, Inc., Other Relationship; Self; ImmunoMolecular Therapeutics, Inc., Research Support; Self; Caladrius Biosciences, Inc., Immune Tolerance Network, Mercia Pharma Inc., National Institute of Diabetes and Digestive and Kidney Diseases, Precigen, Inc., Tolerion, Inc. S. S. Rich: None. J. Krischer: None. R. A. Oram: Consultant; Self; Janssen Research & Development, LLC. Funding National Institute of Diabetes and Digestive and Kidney Diseases (1R01DK121843-01); JDRF (3-SRA-2019-827-S-B)
    Type of Medium: Online Resource
    ISSN: 0012-1797 , 1939-327X
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2021
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Diabetes, American Diabetes Association, Vol. 71, No. Supplement_1 ( 2022-06-01)
    Abstract: T1D screening and monitoring studies suggest opportunities for scaling up to public health population-based programs. Genetic Risk Scores (GRS) for T1D are also demonstrating the ability to identify those at higher risk of progressing to T1D as well as an increased opportunity for implementation into public health settings potential as a result of practical considerations, including falling gene sequencing costs.Previous studies have shown that screening and monitoring children for T1D progression significantly decreases DKA events at diagnosis, which has been shown to have both short- and long-term benefits. Furthermore, identification of those at higher risk of T1D provides an opportunity to intervene and delay T1D onset. A non-stochastic, state transition cohort simulation projected 20US births through adolescence. Data from previously collected natural history studies, including TEDDY, DIPP & DAISY were used to model the patient journey. Children seroconverting to islet autoantibodies (IAB) each year and children with lower T1D risk were simulated as separate cohorts with distinct progressions. Interception strategies were layered over this natural history projection. Success of GRS screening followed by annual IAB and glucose monitoring to intercept T1D progression and reduce DKA events was explored. Therapeutic intervention to delay T1D onset was also simulated.Real-world benchmarks for newborn screening (85%) and patient follow-up (80%) as well as rapid T1D progression ( & lt;12 months) for some children limited maximum interception at dysglycemia to 52% of children progressing to T1D by age 15 using a GRS screening strategy. Short-term net incremental costs range reach $1.1B at current test costs or $225K per T1D case interception. Breakeven costs could be achieved with $40 IAB test cost and $5 GRS costs with a therapeutic to delay T1D onset by 3 years or possibly by including assessment potential long-term benefits of early intervention and improved glycemic control. Disclosure M.Trusheim: Other Relationship; CO BIO CONSULTING LLC. S.Kostense: Employee; Janssen Research & Development, LLC. L.A.Ferrat: None. R.Mcqueen: Consultant; Merck & Co., Inc., Monument Analytics, Research Support; Eli Lilly and Company. R.D.Neusner: Employee; Janssen Pharmaceuticals, Inc. R.A.Oram: Consultant; Janssen Research & Development, LLC, Research Support; Randox R & D. M.Rewers: Consultant; Janssen Research & Development, LLC, Medscape, Provention Bio, Inc., Research Support; Dexcom, Inc., JDRF, Roche Diagnostics USA. J.L.Dunne: Employee; Janssen Research & Development, LLC, JDRF.
    Type of Medium: Online Resource
    ISSN: 0012-1797
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2022
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Diagnostic and Prognostic Research, Springer Science and Business Media LLC, Vol. 4, No. 1 ( 2020-12)
    Abstract: There is much interest in the use of prognostic and diagnostic prediction models in all areas of clinical medicine. The use of machine learning to improve prognostic and diagnostic accuracy in this area has been increasing at the expense of classic statistical models. Previous studies have compared performance between these two approaches but their findings are inconsistent and many have limitations. We aimed to compare the discrimination and calibration of seven models built using logistic regression and optimised machine learning algorithms in a clinical setting, where the number of potential predictors is often limited, and externally validate the models. Methods We trained models using logistic regression and six commonly used machine learning algorithms to predict if a patient diagnosed with diabetes has type 1 diabetes (versus type 2 diabetes). We used seven predictor variables (age, BMI, GADA islet-autoantibodies, sex, total cholesterol, HDL cholesterol and triglyceride) using a UK cohort of adult participants (aged 18–50 years) with clinically diagnosed diabetes recruited from primary and secondary care ( n = 960, 14% with type 1 diabetes). Discrimination performance (ROC AUC), calibration and decision curve analysis of each approach was compared in a separate external validation dataset ( n = 504, 21% with type 1 diabetes). Results Average performance obtained in internal validation was similar in all models (ROC AUC ≥ 0.94). In external validation, there were very modest reductions in discrimination with AUC ROC remaining ≥ 0.93 for all methods. Logistic regression had the numerically highest value in external validation (ROC AUC 0.95). Logistic regression had good performance in terms of calibration and decision curve analysis. Neural network and gradient boosting machine had the best calibration performance. Both logistic regression and support vector machine had good decision curve analysis for clinical useful threshold probabilities. Conclusion Logistic regression performed as well as optimised machine algorithms to classify patients with type 1 and type 2 diabetes. This study highlights the utility of comparing traditional regression modelling to machine learning, particularly when using a small number of well understood, strong predictor variables.
    Type of Medium: Online Resource
    ISSN: 2397-7523
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2886634-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Diabetes, American Diabetes Association, Vol. 72, No. Supplement_1 ( 2023-06-20)
    Abstract: Autoimmune loss of beta-cell function (measured by C-peptide) is the hallmark of type 1 diabetes (T1D) targeted by interventions that aim to prevent T1D or its progression after onset. We sought to determine whether T1D genetic risk score-2 (T1D-GRS2) and single nucleotide polymorphisms (SNPs) that have been previously associated with C-peptide preservation after T1D diagnosis (e.g., SNPs in CLEC16A, G6CP2, INS, JAZF1, PTPN22, SLC30A8 and TCF7L2) influence C-peptide levels before diagnosis. We studied islet autoantibody (Ab)-positive participants in the TrialNet Pathway to Prevention Study who had T1DExomeChip data and assessed the influence of these 12 SNPs and the T1D-GRS2 on area under the curve (AUC) C-peptide levels during oral glucose tolerance tests conducted between 0-9 months prior to the diagnosis of T1D. Participants (n=702) had a mean age of 13.5±10.3 years, 47% were female, mean BMI was 20.7±6.0 kg/m2, and mean HbA1c 5.4±0.4%. The T1D high-risk HLA-DR3-DQ2 haplotype was present in 47% and the high-risk HLA-DR4-DQ8 haplotype was present in 67% of participants. We performed univariate and multivariate analyses adjusting for BMI, age, sex, number of positive Ab, and the first 3 principal components of ancestry. A higher T1D-GRS2 was associated with lower C-peptide AUC 0-9 months prior to T1D diagnosis in univariate (β=-0.06, P & lt;0.0001) and multivariate (β=-0.03, p=0.008) analyses. Participants with the JAZF1 rs864745 G allele had lower C-peptide AUC 0-9 months prior to T1D diagnosis in univariate (β=-0.10, p=0.003) and multivariate (β=-0.05, p=0.047) analysis. In conclusion, the JAZF1 rs864745 G allele (which has also been associated with type 2 diabetes risk) and higher T1D-GRS2 predict lower C-peptide AUC prior to the diagnosis of T1D. Studies on their effect on response to trials to prevent or delay T1D onset are warranted. Disclosure T. M. Triolo: None. S. S. Rich: None. A. Steck: None. M. J. Redondo: None. H. M. Parikh: None. M. Tosur: Advisory Panel; Provention Bio, Inc. L. A. Ferrat: Consultant; Johnson & Johnson. L. You: None. P. Gottlieb: Advisory Panel; ViaCyte, Inc., Board Member; ImmunoMolecular Therapeutics, Research Support; Imcyse, Hemsley Charitable Trust, Novartis, National Institute of Diabetes and Digestive and Kidney Diseases, Precigen, Inc., Dompé, Nova Pharmaceuticals, Provention Bio, Inc. R. A. Oram: Consultant; Janssen Research & Development, LLC, Research Support; Randox R & D. S. Onengut-gumuscu: None. J. Krischer: None. Funding National Institutes of Health (R01DK121843, R01DK124395)
    Type of Medium: Online Resource
    ISSN: 0012-1797
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2023
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Public Library of Science (PLoS) ; 2018
    In:  PLOS Computational Biology Vol. 14, No. 3 ( 2018-3-2), p. e1006009-
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...