1
|
Johannesen J. Why study the T1D remission phase in the pediatric population? J Pediatr (Rio J) 2025; 101:117-118. [PMID: 39701549 PMCID: PMC11889656 DOI: 10.1016/j.jped.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Affiliation(s)
- Jesper Johannesen
- Copenhagen University Hospital, Herlev, Denmark; Steno Diabetes Center Copenhagen, Copenhagen, Denmark; University of Copenhagen, Department of Clinical Medicine, Copenhagen, Denmark.
| |
Collapse
|
2
|
Barmpagianni A, Karamanakos G, Anastasiou IA, Kountouri A, Lambadiari V, Liatis S. The relationship between residual insulin secretion and subclinical cardiovascular risk indices in young adults with type 1 diabetes. J Diabetes Complications 2025; 39:108946. [PMID: 39731973 DOI: 10.1016/j.jdiacomp.2024.108946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/25/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Patients with type 1 diabetes (DM1), even in the setting of adequate glycaemic control, have an excess risk for developing cardiovascular disease. Residual insulin secretion (RIS), measured by detectable C-peptide levels in patients with DM1, might protect against diabetes-related complications. This study aimed to examine the relationship between residual insulin secretion and prognostic markers of cardiovascular complications in patients with DM1. METHODS A total of 137 patients with DM1 were included in this analysis. They were of young age (<45 years), with an established diagnosis of over two years before the study entry and without a history of cardiovascular complications. All patients underwent complete clinical and laboratory evaluation. A c-peptide measurement of ≥0.05 ng/ml was used to identify the presence of RIS. Pulse wave velocity (PWV), cardiac autonomic function assessed both at rest, by total power of heart rate variability and dynamically, by the expiration to inspiration (e/i) index, albumin to creatinine ratio (ACR), and high sensitivity CRP (hs-CRP) were used as predictive biomarkers of cardiovascular complications. RESULTS Female participants represented 63.5% of the population [mean age: 29.7 (±8.1) years, mean HbA1c: 7.6% (±1.4), median diabetes duration:15 (10-21) years, median age at diabetes diagnosis: 13 (8-17) years]]. The median value of fasting c-peptide was 0.04 (0.03-0.05) ng/ml, and RIS was detected in 32 patients (23.4%). Patients with RIS had a shorter diabetes duration, an older age at diagnosis and a lower BMI, while no significant association was found between residual c-peptide and age or HbA1c. RIS was significantly associated with lower PWV values [8.1 m/s² (7-8.7) vs 9.2 m/s² (7.8-10.1), p <0,001], higher total power values [1124 Hz (600-3277) vs 577 Hz (207-2091), p <0,001], and higher E/I measurements [1.4 (1.2-1.5) vs. 1.3 (1.2-1.4), p=0.01]. No significant association was noted between RIS and either ACR or hs-CRP. In multivariable linear regression analysis, the association between RIS and lower PWV values remained significant (p= 0.007) regardless of age, sex, diabetes duration or age of diagnosis, blood pressure and BMI. Similarly, residual insulin secretion retained a significant independent association with total power (p= 0.032) and E/I (p=0.045). CONCLUSION In young patients with DM1, free of macrovascular complications, residual insulin secretion is independently associated with more favorable prognostic markers of subclinical atherosclerosis and cardiac autonomic function.
Collapse
Affiliation(s)
- Aikaterini Barmpagianni
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece.
| | - Georgios Karamanakos
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Ioanna A Anastasiou
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Aikaterini Kountouri
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Vaia Lambadiari
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| | - Stavros Liatis
- National and Kapodistrian University of Athens First Department of Propaedeutic and Internal Medicine, Laiko General Hospital Athens, Attiki, Greece
| |
Collapse
|
3
|
Roshandel D, Spiliopoulou A, McGurnaghan SJ, Iakovliev A, Lipschutz D, Hayward C, Bull SB, Klein BE, Lee KE, Kinney GL, Rewers M, Costacou T, Miller RG, McKeigue PM, Paterson AD, Colhoun HM. Genetics of C-Peptide and Age at Diagnosis in Type 1 Diabetes. Diabetes 2025; 74:223-233. [PMID: 39556808 PMCID: PMC11755686 DOI: 10.2337/db24-0340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
ARTICLE HIGHLIGHTS Identified genetic loci for C-peptide and type 1 diabetes (T1D) age at diagnosis (AAD) explain only a small proportion of their variation. We aimed to identify additional genetic loci associated with C-peptide and AAD. Some HLA allele/haplotypes associated with T1D also contributed to variability of C-peptide and AAD, whereas outside the HLA region, T1D loci were mostly not associated with C-peptide or AAD. Genetic variation within CTSH can affect AAD. There is still residual heritability of C-peptide and AAD outside of HLA that could benefit from larger meta-genome-wide association studies.
Collapse
Affiliation(s)
- Delnaz Roshandel
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Athina Spiliopoulou
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, U.K
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, U.K
| | | | - Andrii Iakovliev
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, U.K
| | - Debby Lipschutz
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, U.K
| | - Caroline Hayward
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, U.K
| | - Shelley B. Bull
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Barbara E.K. Klein
- School of Medicine and Public Health, Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI
| | - Kristine E. Lee
- School of Medicine and Public Health, Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI
| | - Gregory L. Kinney
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, CO
| | - Marian Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tina Costacou
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| | - Rachel G. Miller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| | - Paul M. McKeigue
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, U.K
| | - Andrew D. Paterson
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Helen M. Colhoun
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
4
|
Chen X, Yu J, Liu Y, Wang X, Ping F, Li W, Zhang H, Xu L, Li Y. Positive Islet Cell Cytoplasmic Antibody and Long-Term Use of Lipid-Lowering Agents Are Positively Correlated With Peripheral Atherosclerosis in Patients With Autoimmune Diabetes: A Cross-Sectional Study. J Diabetes Res 2025; 2025:1933825. [PMID: 39949401 PMCID: PMC11824714 DOI: 10.1155/jdr/1933825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 02/16/2025] Open
Abstract
Aims: This cross-sectional study is aimed at determining whether systemic inflammation, diabetic autoantibodies, and islet β cell dysfunction play a role in the progression of macrovascular complications in patients with autoimmune diabetes. Methods: 202 patients with autoimmune diabetes aged ≥ 35 years and hospitalized in Peking Union Medical College Hospital were enrolled in this study. The patients were divided into three groups based on the severity of peripheral atherosclerosis. Biomarkers of systemic inflammation, diabetes autoantibodies, islet β cell function, and other covariates validated to be associated with macrovascular complications were collected. Correlations between the severity of peripheral atherosclerosis and systemic inflammation, diabetic autoantibodies, and islet β cell function were examined using an ordinal logistic regression model. Results: Of the enrolled patients, 39.1% were male, with a median age of 53 (43, 60) years and a diabetes duration of 96 (36, 216) months. 58 patients had no lesions in the peripheral arteries, 72 had atherosclerosis in the carotid or lower extremity arteries, and the rest had lesions in both arteries. In the multifactor ordinal logistic regression test, positive islet cell cytoplasmic antibody (ICA) and long-term use of lipid-lowering agents were independently associated with peripheral atherosclerosis after adjusting for age and diabetes duration. Conclusions: The correlation between positive ICA and atherosclerosis suggests inflammation at an early stage plays a role in macrovascular complications in autoimmune diabetes. The association between long-term use of lipid-lowering agents and atherosclerosis suggests the need for early screening and intervention for dyslipidemia in patients with autoimmune diabetes.
Collapse
Affiliation(s)
- Xinyue Chen
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Yu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yiwen Liu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuechen Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fan Ping
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Li
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Huabing Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lingling Xu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuxiu Li
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Mallone R, Bismuth E, Thivolet C, Benhamou PY, Hoffmeister N, Collet F, Nicolino M, Reynaud R, Beltrand J. Screening and care for preclinical stage 1-2 type 1 diabetes in first-degree relatives: French expert position statement. DIABETES & METABOLISM 2025; 51:101603. [PMID: 39675522 DOI: 10.1016/j.diabet.2024.101603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
The natural history of type 1 diabetes (T1D) evolves from stage 1 (islet autoimmunity with normoglycemia; ICD-10 diagnostic code E10.A1) to stage 2 (autoimmunity with dysglycemia; E10.A2) and subsequent clinical stage 3 (overt hyperglycemia), which is commonly the first time of referral. Autoantibody testing can diagnose T1D at its preclinical stages 1-2 and lead to earlier initiation of care, particularly for first-degree relatives of people living with T1D, who are at higher genetic risk. Preclinical T1D screening and monitoring aims to avoid inaugural ketoacidosis and prolong preservation of endogenous insulin secretion, thereby improving glycemic control and reducing long-term morbidity. Moreover, early management can help coping with T1D and correct modifiable risk factors (obesity, sedentary lifestyle). New treatments currently under clinical deployment or trials also offer the possibility of delaying clinical progression. All these arguments lead to the proposition of a national screening and care pathway open to interested first-degree relatives. This pathway represents a new expertise to acquire for healthcare professionals. By adapting international consensus guidance to the French specificities, the proposed screening strategy involves testing for ≥ 2 autoantibodies (among IAA, anti-GAD, anti-IA-2) in relatives aged 2-45 years. Negative screening (∼95 % of cases) should be repeated every 4 years until the age of 12. A management workflow is proposed for relatives screening positive (∼5 % of cases), with immuno-metabolic monitoring by autoantibody testing, OGTT, glycemia and/or HbA1c of variable frequency, depending on T1D stage, age, patient preference and available resources, as well as the definition of expert centers for preclinical T1D.
Collapse
Affiliation(s)
- Roberto Mallone
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France; Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service de Diabétologie et Immunologie Clinique, Hôpital Cochin, Paris, France; Indiana Biosciences Research Institute, Indianapolis, IN, USA.
| | - Elise Bismuth
- Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service d'Endocrinologie et Diabétologie Pédiatrique, Hôpital Robert Debré, Paris, France
| | - Charles Thivolet
- Hospices Civils de Lyon, Université de Lyon, Centre du diabète DIAB-eCARE, Lyon, France
| | - Pierre-Yves Benhamou
- Université Grenoble Alpes, INSERM U1055, LBFA, Endocrinologie, CHU Grenoble Alpes, France
| | | | - François Collet
- CHU Lille, Psychiatrie de Liaison et psycho-oncologie, Lille, France
| | - Marc Nicolino
- Hospices Civils de Lyon, Université de Lyon, Service d'Endocrinologie et Diabétologie Pédiatrique, Lyon, France
| | - Rachel Reynaud
- Assistance Publique Hôpitaux de Marseille, Université Aix-Marseille, Service de Pédiatrie Multidisciplinaire, Hôpital de la Timone, Marseille, France
| | - Jacques Beltrand
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France; Assistance Publique Hôpitaux de Paris, Université Paris Cité, Service d'Endocrinologie, Gynécologie et Diabétologie Pédiatrique, Necker Hospital, Paris, France
| |
Collapse
|
6
|
Jones AG, Shields BM, Oram RA, Dabelea DM, Hagopian WA, Sharp SA, Lustigova E, Shah AS, Knupp J, Mottl AK, D’Agostino RB, Williams A, Marcovina SM, Pihoker C, Divers J, Redondo MJ. Clinical Prediction Models Combining Routine Clinical Measures Have High Accuracy in Identifying Youth-Onset Type 2 Diabetes Defined by Maintained Endogenous Insulin Secretion: The SEARCH for Diabetes in Youth Study. Diabetes Care 2024; 47:2110-2119. [PMID: 38252849 PMCID: PMC11655402 DOI: 10.2337/dc23-1815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/21/2023] [Indexed: 01/24/2024]
Abstract
OBJECTIVE With high prevalence of obesity and overlapping features between diabetes subtypes, accurately classifying youth-onset diabetes can be challenging. We aimed to develop prediction models that, using characteristics available at diabetes diagnosis, can identify youth who will retain endogenous insulin secretion at levels consistent with type 2 diabetes (T2D). RESEARCH DESIGN AND METHODS We studied 2,966 youth with diabetes in the prospective SEARCH for Diabetes in Youth study (diagnosis age ≤19 years) to develop prediction models to identify participants with fasting C-peptide ≥250 pmol/L (≥0.75 ng/mL) after >3 years' (median 74 months) diabetes duration. Models included clinical measures at the baseline visit, at a mean diabetes duration of 11 months (age, BMI, sex, waist circumference, HDL cholesterol), with and without islet autoantibodies (GADA, IA-2A) and a type 1 diabetes genetic risk score (T1DGRS). RESULTS Models using routine clinical measures with or without autoantibodies and T1DGRS were highly accurate in identifying participants with C-peptide ≥0.75 ng/mL (17% of participants; 2.3% and 53% of those with and without positive autoantibodies) (area under the receiver operating characteristic curve [AUCROC] 0.95-0.98). In internal validation, optimism was very low, with excellent calibration (slope 0.995-0.999). Models retained high performance for predicting retained C-peptide in older youth with obesity (AUCROC 0.88-0.96) and in subgroups defined by self-reported race and ethnicity (AUCROC 0.88-0.97), autoantibody status (AUCROC 0.87-0.96), and clinically diagnosed diabetes types (AUCROC 0.81-0.92). CONCLUSIONS Prediction models combining routine clinical measures at diabetes diagnosis, with or without islet autoantibodies or T1DGRS, can accurately identify youth with diabetes who maintain endogenous insulin secretion in the range associated with T2D.
Collapse
Affiliation(s)
| | | | | | | | | | - Seth A. Sharp
- Department of Genetics, Stanford University School of Medicine, Stanford, CA
| | - Eva Lustigova
- Kaiser Permanente Southern California, Los Angeles, CA
| | - Amy S. Shah
- University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | | | - Amy K. Mottl
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | | | | | - Maria J. Redondo
- Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
| |
Collapse
|
7
|
Hill TG, Gao R, Benrick A, Kothegala L, Rorsman N, Santos C, Acreman S, Briant LJ, Dou H, Gandasi NR, Guida C, Haythorne E, Wallace M, Knudsen JG, Miranda C, Tolö J, Clark A, Davison L, Størling J, Tarasov A, Ashcroft FM, Rorsman P, Zhang Q. Loss of electrical β-cell to δ-cell coupling underlies impaired hypoglycaemia-induced glucagon secretion in type-1 diabetes. Nat Metab 2024; 6:2070-2081. [PMID: 39313541 PMCID: PMC11599053 DOI: 10.1038/s42255-024-01139-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
Diabetes mellitus involves both insufficient insulin secretion and dysregulation of glucagon secretion1. In healthy people, a fall in plasma glucose stimulates glucagon release and thereby increases counter-regulatory hepatic glucose production. This response is absent in many patients with type-1 diabetes (T1D)2, which predisposes to severe hypoglycaemia that may be fatal and accounts for up to 10% of the mortality in patients with T1D3. In rats with chemically induced or autoimmune diabetes, counter-regulatory glucagon secretion can be restored by SSTR antagonists4-7 but both the underlying cellular mechanism and whether it can be extended to humans remain unestablished. Here, we show that glucagon secretion is not stimulated by low glucose in isolated human islets from donors with T1D, a defect recapitulated in non-obese diabetic mice with T1D. This occurs because of hypersecretion of somatostatin, leading to aberrant paracrine inhibition of glucagon secretion. Normally, KATP channel-dependent hyperpolarization of β-cells at low glucose extends into the δ-cells through gap junctions, culminating in suppression of action potential firing and inhibition of somatostatin secretion. This 'electric brake' is lost following autoimmune destruction of the β-cells, resulting in impaired counter-regulation. This scenario accounts for the clinical observation that residual β-cell function correlates with reduced hypoglycaemia risk8.
Collapse
Affiliation(s)
- Thomas G Hill
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Rui Gao
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Anna Benrick
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Lakshmi Kothegala
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
- Department of Developmental Biology and Genetics (DBG), Indian Institute of Science (IISc), Bengaluru, India
| | - Nils Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Cristiano Santos
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Samuel Acreman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Linford J Briant
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Haiqiang Dou
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Nikhil R Gandasi
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
- Department of Developmental Biology and Genetics (DBG), Indian Institute of Science (IISc), Bengaluru, India
| | - Claudia Guida
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Elizabeth Haythorne
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Marsha Wallace
- Nuffield Department of Clinical Medicine, University of Oxford, Roosevelt Drive, Oxford, UK
- The Royal Veterinary College, Hatfield, Hertfordshire, UK
| | - Jakob G Knudsen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Miranda
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Johan Tolö
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Lucy Davison
- Nuffield Department of Clinical Medicine, University of Oxford, Roosevelt Drive, Oxford, UK
- The Royal Veterinary College, Hatfield, Hertfordshire, UK
| | - Joachim Størling
- Steno Diabetes Center Copenhagen, Translational Type 1 Diabetes Research, Herlev, Denmark
| | - Andrei Tarasov
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Frances M Ashcroft
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
- Metabolic Research Unit, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden.
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
- Oxford National Institute for Health Research, Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
8
|
Alan MS, Tayebi A, Afshar EJ, Alan SS, Alan MS, Fazeli R, Sohbatzade T, Samimisedeh P, Rastad H. Association of detectable C-peptide levels with glycemic control and chronic complications in individuals with type 1 diabetes mellitus: A systematic review and meta-analysis. J Diabetes Complications 2024; 39:108867. [PMID: 39879848 DOI: 10.1016/j.jdiacomp.2024.108867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/22/2024] [Accepted: 09/14/2024] [Indexed: 01/31/2025]
Abstract
AIMS Multiple studies have addressed the association between detectable levels of C-peptide and glycemic control, as well as the development of chronic complications of type 1 diabetes mellitus (T1DM), including both macrovascular and microvascular diseases. We aimed to summarize the available evidence on the clinical significance of detectable levels of C-peptide in T1DM. METHOD A systematic search was performed on online databases using the following key terms: T1DM, C-peptide, diabetes mellitus complications, and glycemic parameters. We pooled standardized mean difference (SMD) and odds ratios (OR). RESULTS Of the 1519 articles retrieved from the initial search, 38 (12 cohort and 26 cross-sectional studies) met our eligibility criteria. Individuals with T1DM in the detectable C-peptide group, compared with the undetectable C-peptide group, had lower mean HbA1c [pooled SMD (95 % confidence interval (95 % CI)): -0.08 (-0.13 to -0.02), I2 = 0 %, p. VALUE 0.005] and daily insulin dose [-0.41 (-0.65 to -0.18), I2 = 83 %, p.value < 0.001]. They also showed lower odds for retinopathy [pooled crude OR (95 % CI): 0.53 (0.41 to 0.69), I2 = 65 %, p.value < 0.001] and nephropathy complications [0.62 (0.55 to 0.70), I2 = 19 %, p.value < 0.001]; however, the two groups were similar regarding neuropathy [0.92 (0.65 to 1.31), I2 = 0 %, p. VALUE 0.31]. CONCLUSIONS The available evidence suggests that individuals with T1DM in the detectable C-peptide group may experience better clinical outcomes.
Collapse
Affiliation(s)
- Mahin Seifi Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Amirhossein Tayebi
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran; Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Elmira Jafari Afshar
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Sanaz Seifi Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mahnaz Seifi Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ramina Fazeli
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Tooba Sohbatzade
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Parham Samimisedeh
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| | - Hadith Rastad
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran; Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
9
|
Kokori E, Olatunji G, Ogieuhi IJ, Aboje JE, Olatunji D, Aremu SA, Igwe SC, Moradeyo A, Ajayi YI, Aderinto N. Teplizumab's immunomodulatory effects on pancreatic β-cell function in type 1 diabetes mellitus. Clin Diabetes Endocrinol 2024; 10:23. [PMID: 39123252 PMCID: PMC11316332 DOI: 10.1186/s40842-024-00181-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/09/2024] [Indexed: 08/12/2024] Open
Abstract
This review explores the immunomodulatory potential of Teplizumab and its impact on pancreatic β-cell function in T1D. Characterized by the autoimmune destruction of insulin-producing beta cells, T1D's management involves maintaining glycemic control through exogenous insulin. Teplizumab, a humanized monoclonal antibody targeting the CD3 antigen, has shown promise in delaying T1D onset and preserving residual β-cell function. The review employs a narrative approach, synthesizing evidence from diverse clinical trials and studies gathered through a meticulous literature search. It scrutinizes Teplizumab's mechanisms of action, including its influence on autoreactive CD8 + T cells and regulatory T cells, offering insights into its immunological pathways. The synthesis of findings from various trials demonstrates Teplizumab's efficacy in preserving C-peptide levels and reducing exogenous insulin requirements, particularly in recent-onset T1D. Considering Teplizumab's real-world implications, the paper addresses potential obstacles, including side effects, patient selection criteria, and logistical challenges. It also emphasizes exploring combination therapies and personalized treatment strategies to maximize Teplizumab's benefits. The review contributes a nuanced perspective on Teplizumab's clinical implications and future directions in T1D management, bridging theoretical understanding with practical considerations.
Collapse
Affiliation(s)
- Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - John Ehi Aboje
- Department of Medicine, College of Health Sciences, Benue State University, Benue, Nigeria
| | - Doyin Olatunji
- Department of Health Sciences, Western Illinois University, Macomb, USA
| | | | | | - Abdulrahmon Moradeyo
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Yusuf Ismaila Ajayi
- Department of Medicine and Surgery, Obafemi Awolowo University, Ife, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| |
Collapse
|
10
|
Mellor J, Kuznetsov D, Heller S, Gall MA, Rosilio M, Amiel SA, Ibberson M, McGurnaghan S, Blackbourn L, Berthon W, Salem A, Qu Y, McCrimmon RJ, de Galan BE, Pedersen-Bjergaard U, Leaviss J, McKeigue PM, Colhoun HM. Risk factors and prediction of hypoglycaemia using the Hypo-RESOLVE cohort: a secondary analysis of pooled data from insulin clinical trials. Diabetologia 2024; 67:1588-1601. [PMID: 38795153 PMCID: PMC11343909 DOI: 10.1007/s00125-024-06177-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/28/2024] [Indexed: 05/27/2024]
Abstract
AIMS/HYPOTHESIS The objective of the Hypoglycaemia REdefining SOLutions for better liVES (Hypo-RESOLVE) project is to use a dataset of pooled clinical trials across pharmaceutical and device companies in people with type 1 or type 2 diabetes to examine factors associated with incident hypoglycaemia events and to quantify the prediction of these events. METHODS Data from 90 trials with 46,254 participants were pooled. Analyses were done for type 1 and type 2 diabetes separately. Poisson mixed models, adjusted for age, sex, diabetes duration and trial identifier were fitted to assess the association of clinical variables with hypoglycaemia event counts. Tree-based gradient-boosting algorithms (XGBoost) were fitted using training data and their predictive performance in terms of area under the receiver operating characteristic curve (AUC) evaluated on test data. Baseline models including age, sex and diabetes duration were compared with models that further included a score of hypoglycaemia in the first 6 weeks from study entry, and full models that included further clinical variables. The relative predictive importance of each covariate was assessed using XGBoost's importance procedure. Prediction across the entire trial duration for each trial (mean of 34.8 weeks for type 1 diabetes and 25.3 weeks for type 2 diabetes) was assessed. RESULTS For both type 1 and type 2 diabetes, variables associated with more frequent hypoglycaemia included female sex, white ethnicity, longer diabetes duration, treatment with human as opposed to analogue-only insulin, higher glucose variability, higher score for hypoglycaemia across the 6 week baseline period, lower BP, lower lipid levels and treatment with psychoactive drugs. Prediction of any hypoglycaemia event of any severity was greater than prediction of hypoglycaemia requiring assistance (level 3 hypoglycaemia), for which events were sparser. For prediction of level 1 or worse hypoglycaemia during the whole follow-up period, the AUC was 0.835 (95% CI 0.826, 0.844) in type 1 diabetes and 0.840 (95% CI 0.831, 0.848) in type 2 diabetes. For level 3 hypoglycaemia, the AUC was lower at 0.689 (95% CI 0.667, 0.712) for type 1 diabetes and 0.705 (95% CI 0.662, 0.748) for type 2 diabetes. Compared with the baseline models, almost all the improvement in prediction could be captured by the individual's hypoglycaemia history, glucose variability and blood glucose over a 6 week baseline period. CONCLUSIONS/INTERPRETATION Although hypoglycaemia rates show large variation according to sociodemographic and clinical characteristics and treatment history, looking at a 6 week period of hypoglycaemia events and glucose measurements predicts future hypoglycaemia risk.
Collapse
Affiliation(s)
- Joseph Mellor
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.
| | | | - Simon Heller
- Division of Clinical Medicine, University of Sheffield, Sheffield, UK
| | - Mari-Anne Gall
- Medical & Science, Insulin, Clinical Drug Development, Novo Nordisk A/S, Soeberg, Denmark
| | - Myriam Rosilio
- Eli Lilly and Company, Diabetes Medical Unit, Neuilly sur seine, France
| | - Stephanie A Amiel
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mark Ibberson
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Stuart McGurnaghan
- Institute of Genetics and Cancer, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Luke Blackbourn
- Institute of Genetics and Cancer, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - William Berthon
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Adel Salem
- RW Data Assets, AI & Analytics (AIA), Novo Nordisk A/S, Soeberg, Denmark
| | - Yongming Qu
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Rory J McCrimmon
- Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Bastiaan E de Galan
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center, Maastricht, the Netherlands
| | | | - Joanna Leaviss
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Paul M McKeigue
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Helen M Colhoun
- Institute of Genetics and Cancer, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
11
|
Alqahtani YA, Shati AA, Alhawyan FS, Alhanshani AA, Al-Garni AM, Al-Qahtani SM, Alshehri MA. Assessment of depression in children and adolescents with Type 1 diabetes mellitus: Impact and intervention strategies. Medicine (Baltimore) 2024; 103:e38868. [PMID: 39029074 PMCID: PMC11398734 DOI: 10.1097/md.0000000000038868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Depression is a common comorbidity in children and adolescents with type 1 diabetes mellitus (T1DM), yet its prevalence, impact, and intervention strategies remain underexplored. This study aims to assess the prevalence of depression among children and adolescents with T1DM, investigate its impact on health outcomes, and explore potential intervention strategies. A convenient sampling method was employed to recruit 229 participants aged 6 to 18 years from a single center. Data collection involved validated assessments, demographic surveys, and diabetes-related factor examinations during routine clinic visits. The patient health questionnaire-9 was utilized to evaluate the severity of depressive symptoms. Associations between depression and sociodemographic variables, diabetes management factors, and health behaviors were analyzed using chi-squared tests and logistic regression analysis. The prevalence of depression among participants was 43.23%. Older age, lower parental education levels, lower household income, smoking, and comorbidities were identified as significant risk factors for depression. Associations were found between depression and diabetes management factors, including glycemic control and frequency of glucose monitoring. Depression is highly prevalent among children and adolescents with T1DM and is associated with sociodemographic factors, health behaviors, and diabetes management. Integrated approaches to care that address both physical and mental health aspects are crucial for improving outcomes in this population.
Collapse
Affiliation(s)
- Youssef A Alqahtani
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ayed A Shati
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Fatimah S Alhawyan
- Department of Internal Medicine, Armed Forces Hospital Southern Region, Khamis Mushayt, Saudi Arabia
| | - Ahmad A Alhanshani
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Abdulaziz M Al-Garni
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Saleh M Al-Qahtani
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohammed A Alshehri
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
12
|
Mellor J, Jeyam A, Beulens JW, Bhandari S, Broadhead G, Chew E, Fickweiler W, van der Heijden A, Gordin D, Simó R, Snell-Bergeon J, Tynjälä A, Colhoun H. Role of Systemic Factors in Improving the Prognosis of Diabetic Retinal Disease and Predicting Response to Diabetic Retinopathy Treatment. OPHTHALMOLOGY SCIENCE 2024; 4:100494. [PMID: 38694495 PMCID: PMC11061755 DOI: 10.1016/j.xops.2024.100494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 05/04/2024]
Abstract
Topic To review clinical evidence on systemic factors that might be relevant to update diabetic retinal disease (DRD) staging systems, including prediction of DRD onset, progression, and response to treatment. Clinical relevance Systemic factors may improve new staging systems for DRD to better assess risk of disease worsening and predict response to therapy. Methods The Systemic Health Working Group of the Mary Tyler Moore Vision Initiative reviewed systemic factors individually and in multivariate models for prediction of DRD onset or progression (i.e., prognosis) or response to treatments (prediction). Results There was consistent evidence for associations of longer diabetes duration, higher glycosylated hemoglobin (HbA1c), and male sex with DRD onset and progression. There is strong trial evidence for the effect of reducing HbA1c and reducing DRD progression. There is strong evidence that higher blood pressure (BP) is a risk factor for DRD incidence and for progression. Pregnancy has been consistently reported to be associated with worsening of DRD but recent studies reflecting modern care standards are lacking. In studies examining multivariate prognostic models of DRD onset, HbA1c and diabetes duration were consistently retained as significant predictors of DRD onset. There was evidence of associations of BP and sex with DRD onset. In multivariate prognostic models examining DRD progression, retinal measures were consistently found to be a significant predictor of DRD with little evidence of any useful marginal increment in prognostic information with the inclusion of systemic risk factor data apart from retinal image data in multivariate models. For predicting the impact of treatment, although there are small studies that quantify prognostic information based on imaging data alone or systemic factors alone, there are currently no large studies that quantify marginal prognostic information within a multivariate model, including both imaging and systemic factors. Conclusion With standard imaging techniques and ways of processing images rapidly evolving, an international network of centers is needed to routinely capture systemic health factors simultaneously to retinal images so that gains in prediction increment may be precisely quantified to determine the usefulness of various health factors in the prognosis of DRD and prediction of response to treatment. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Joe Mellor
- Centre for Population Health Sciences, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Anita Jeyam
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital Crewe Road, Edinburgh, Scotland
| | - Joline W.J. Beulens
- Department of Epidemiology & Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Sanjeeb Bhandari
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Geoffrey Broadhead
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Emily Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Ward Fickweiler
- Beetham Eye Institute, Joslin Diabetes Center, Boston, Massachusetts
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Amber van der Heijden
- Department of General Practice, Amsterdam Public Health Institute, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Daniel Gordin
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Department of Nephrology, Helsinki University Hospital, University of Helsinki, Finland
| | - Rafael Simó
- Endocrinology & Nutrition, Institut de Recerca Hospital Universitari Vall d’Hebron (VHIR), Barcelona, Spain
| | - Janet Snell-Bergeon
- Clinical Epidemiology Division, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Colorado
| | - Anniina Tynjälä
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Department of Nephrology, Helsinki University Hospital, University of Helsinki, Finland
| | - Helen Colhoun
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital Crewe Road, Edinburgh, Scotland
| |
Collapse
|
13
|
Aanstoot HJ, Varkevisser RDM, Mul D, Dekker P, Birnie E, Boesten LSM, Brugts MP, van Dijk PR, Duijvestijn PHLM, Dutta S, Fransman C, Gonera RK, Hoogenberg K, Kooy A, Latres E, Loves S, Nefs G, Sas T, Vollenbrock CE, Vosjan-Noeverman MJ, de Vries-Velraeds MMC, Veeze HJ, Wolffenbuttel BHR, van der Klauw MM. Cohort profile: the 'Biomarkers of heterogeneity in type 1 diabetes' study-a national prospective cohort study of clinical and metabolic phenotyping of individuals with long-standing type 1 diabetes in the Netherlands. BMJ Open 2024; 14:e082453. [PMID: 38904129 PMCID: PMC11191834 DOI: 10.1136/bmjopen-2023-082453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/30/2024] [Indexed: 06/22/2024] Open
Abstract
PURPOSE The 'Biomarkers of heterogeneity in type 1 diabetes' study cohort was set up to identify genetic, physiological and psychosocial factors explaining the observed heterogeneity in disease progression and the development of complications in people with long-standing type 1 diabetes (T1D). PARTICIPANTS Data and samples were collected in two subsets. A prospective cohort of 611 participants aged ≥16 years with ≥5 years T1D duration from four Dutch Diabetes clinics between 2016 and 2021 (median age 32 years; median diabetes duration 12 years; 59% female; mean glycated haemoglobin (HbA1c) 61 mmol/mol (7.7%); 61% on insulin pump; 23% on continuous glucose monitoring (CGM)). Physical assessments were performed, blood and urine samples were collected, and participants completed questionnaires. A subgroup of participants underwent mixed-meal tolerance tests (MMTTs) at baseline (n=169) and at 1-year follow-up (n=104). Genetic data and linkage to medical and administrative records were also available. A second cross-sectional cohort included participants with ≥35 years of T1D duration (currently n=160; median age 64 years; median diabetes duration 45 years; 45% female; mean HbA1c 58 mmol/mol (7.4%); 51% on insulin pump; 83% on CGM), recruited from five centres and measurements, samples and 5-year retrospective data were collected. FINDINGS TO DATE Stimulated residual C-peptide was detectable in an additional 10% of individuals compared with fasting residual C-peptide secretion. MMTT measurements at 90 min and 120 min showed good concordance with the MMTT total area under the curve. An overall decrease of C-peptide at 1-year follow-up was observed. Fasting residual C-peptide secretion is associated with a decreased risk of impaired awareness of hypoglycaemia. FUTURE PLANS Research groups are invited to consider the use of these data and the sample collection. Future work will include additional hormones, beta-cell-directed autoimmunity, specific immune markers, microRNAs, metabolomics and gene expression data, combined with glucometrics, anthropometric and clinical data, and additional markers of residual beta-cell function. TRIAL REGISTRATION NUMBER NCT04977635.
Collapse
Affiliation(s)
- Henk-Jan Aanstoot
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
| | | | - Dick Mul
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
| | - Pim Dekker
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
| | - Erwin Birnie
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
- Department of Genetics, UMCG, Groningen, Groningen, The Netherlands
| | - Lianne S M Boesten
- Department of Clinical Chemistry, IJsselland Ziekenhuis, Capelle aan den IJssel, The Netherlands
| | - Michael P Brugts
- Department of Internal Medicine, Ikazia Hospital, Rotterdam, The Netherlands
| | | | | | | | - Christine Fransman
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
| | - Rob K Gonera
- Department of Internal Medicine, Wilhelmina Hospital, Assen, The Netherlands
| | - Klaas Hoogenberg
- Department of Internal Medicine, Martini Ziekenhuis, Groningen, The Netherlands
| | - Adriaan Kooy
- Bethesda Diabetes Research Center & Treant, Treant Care Group, Hoogeveen, Drenthe, The Netherlands
- Department of Internal Medicine, UMCG, Groningen, Groningen, Netherlands
| | | | - Sandra Loves
- Department of Internal Medicine, Treant Care Group, Hoogeveen, Drenthe, Netherlands
| | - Giesje Nefs
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
- Department of Medical Psychology, Radboudumc, Nijmegen, The Netherlands
- Department of Medical and Clinical Psychology, Center of Research on Psychological disordersand Somatic diseases (CoRPS), Tilburg, Netherlands
| | - Theo Sas
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
- Department of Paediatrics, Division of Paediatric Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | - Henk J Veeze
- Diabeter Netherlands, Center for Type 1 Diabetes Care and Research, Rotterdam, The Netherlands
| | | | | |
Collapse
|
14
|
Baxter F, Baillie N, Dover A, Stimson RH, Gibb F, Forbes S. A cross-sectional questionnaire study: Impaired awareness of hypoglycaemia remains prevalent in adults with type 1 diabetes and is associated with the risk of severe hypoglycaemia. PLoS One 2024; 19:e0297601. [PMID: 38875308 PMCID: PMC11178233 DOI: 10.1371/journal.pone.0297601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/14/2024] [Indexed: 06/16/2024] Open
Abstract
OBJECTIVE Impaired awareness of hypoglycaemia (IAH) is a risk factor for severe hypoglycaemia (SH) in type 1 diabetes (T1D). Much of the IAH prevalence data comes from older studies where participants did not have the benefit of the latest insulins and technologies. This study surveyed the prevalence of IAH and SH in a tertiary adult clinic population and investigated the associated factors. METHODS Adults (≥18 years) attending a tertiary T1D clinic completed a questionnaire, including a Gold and Clarke score. Background information was collected from health records. RESULTS 189 people (56.1% female) with T1D (median [IQR] disease duration 19.3 [11.5, 29.1] years and age of 41.0 [29.0, 52.0] years) participated. 17.5% had IAH and 16.0% reported ≥1 episode of SH in the previous 12 months. Those with IAH were more likely to report SH (37.5% versus 11.7%, p = 0.001) a greater number of SH episodes per person (median [IQR] 0 [0,2] versus 0 [0,0] P<0.001) and be female (72.7% versus 52.6%, p = 0.036). Socio-economic deprivation was associated with IAH (p = 0.032) and SH (p = 0.005). Use of technology was the same between IAH vs aware groups, however, participants reporting SH were more likely to use multiple daily injections (p = 0.026). Higher detectable C-peptide concentrations were associated with a reduced risk of SH (p = 0.04). CONCLUSION Insulin pump and continuous glucose monitor use was comparable in IAH versus aware groups. Despite this, IAH remains a risk factor for SH and is prevalent in females and in older people. Socioeconomic deprivation was associated with IAH and SH, making this an important population to target for interventions.
Collapse
Affiliation(s)
- Faye Baxter
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola Baillie
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Dover
- Department of Diabetes and Endocrinology, Outpatient Department 2, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Roland H Stimson
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Diabetes and Endocrinology, Outpatient Department 2, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Fraser Gibb
- Department of Diabetes and Endocrinology, Outpatient Department 2, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Shareen Forbes
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Diabetes and Endocrinology, Outpatient Department 2, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Latres E, Greenbaum CJ, Oyaski ML, Dayan CM, Colhoun HM, Lachin JM, Skyler JS, Rickels MR, Ahmed ST, Dutta S, Herold KC, Marinac M. Evidence for C-Peptide as a Validated Surrogate to Predict Clinical Benefits in Trials of Disease-Modifying Therapies for Type 1 Diabetes. Diabetes 2024; 73:823-833. [PMID: 38349844 DOI: 10.2337/dbi23-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
Type 1 diabetes is a chronic autoimmune disease in which destruction of pancreatic β-cells causes life-threatening metabolic dysregulation. Numerous approaches are envisioned for new therapies, but limitations of current clinical outcome measures are significant disincentives to development efforts. C-peptide, a direct byproduct of proinsulin processing, is a quantitative biomarker of β-cell function that is not cleared by the liver and can be measured in the peripheral blood. Studies of quantitative measures of β-cell function have established a predictive relationship between stimulated C-peptide as a measure of β-cell function and clinical benefits. C-peptide levels at diagnosis are often high enough to afford glycemic control benefits associated with protection from end-organ complications of diabetes, and even lower levels offer protection from severe hypoglycemia in type 1 diabetes, as observed in large prospective cohort studies and interventional trials of islet transplantation. These observations support consideration of C-peptide not just as a biomarker of β-cell function but also as a specific, sensitive, feasible, and clinically meaningful outcome defining β-cell preservation or restoration for clinical trials of disease-modifying therapies. Regulatory acceptance of C-peptide as a validated surrogate for demonstration of efficacy would greatly facilitate development of disease-modifying therapies for type 1 diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | | | | | | | - Helen M Colhoun
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, U.K
| | - John M Lachin
- Biostatistics Center, George Washington University, Rockville, MD
| | - Jay S Skyler
- Diabetes Research Institute, University of Miami, Miami, FL
| | - Michael R Rickels
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Simi T Ahmed
- New York Stem Cell Foundation Research Institute, New York, NY
| | | | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale School of Medicine, New Haven, CT
| | | |
Collapse
|
16
|
Lebbad H, Desir C, Vesin A, Adenis A, Charpentier G, Benhamou PY. Insulin Requirements According to Sex and Weight in a Population of 9036 Adult Persons With Type 1 Diabetes Using Closed-Loop Insulin Delivery. J Diabetes Sci Technol 2024:19322968241252366. [PMID: 38804537 PMCID: PMC11571726 DOI: 10.1177/19322968241252366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND The prediction of the individual insulin needs may facilitate the initiation of insulin therapy. Our aim was to explore the relationships between body weight, sex, and daily amounts of insulin delivered by a hybrid closed-loop system. METHODS We performed a retrospective data collection of all consenting adult patients with type 1 diabetes who were equipped in Europe with the Diabeloop Generation 1 (DBLG1) hybrid closed-loop insulin delivery device between March 1, 2021 and February 28, 2023. RESULTS A total of 9036 users (59% females, age 45.6 ± 14.3 years) were included, reaching a mean follow-up of 320 ± 143 days, an overall 2 887 188 days of data. We observed a mean insulin-weight ratio of 0.617 ± 0.207 U/kg (0.665 ± 0.217 for males and 0.584 ± 0.193 for females, P < .001). Exploratory analysis of a subset of 4066 patients reaching >70% Time in Range (70-180 mg/dL) showed a mean insulin-weight ratio of 0.55 ± 0.17 U/kg (P < .001) (0.59 ± 0.18 for the 1438 males and 0.53 ± 0.16 for the 2628 females). CONCLUSION This large real-world analysis provides a quantitative estimation of the daily insulin requirements in adult patients with type 1 diabetes and shows significant differences between sex. These findings have relevant implications in the practical management of insulin therapy.
Collapse
Affiliation(s)
| | | | | | | | - Guillaume Charpentier
- Center for Study and Research for Improvement of the Treatment of Diabetes (CERITD), Bioparc-Genopole Evry-Corbeil, Evry, France
- Department of Diabetes and Endocrinology, Sud-Francilien Hospital, Corbeil, France
| | - Pierre-Yves Benhamou
- Department of Endocrinology, Grenoble University Hospital, Grenoble Alpes University, INSERM U1055, LBFA, Grenoble, France
| |
Collapse
|
17
|
Pinheiro MM, Pinheiro FMM, Garo ML, Pastore D, Pacifici F, Ricordi C, Della-Morte D, Infante M. Prevention and treatment of type 1 diabetes: in search of the ideal combination therapy targeting multiple immunometabolic pathways. METABOLISM AND TARGET ORGAN DAMAGE 2024; 4. [DOI: 10.20517/mtod.2024.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Type 1 diabetes (T1D) represents an autoimmune disease caused by the gradual immune-mediated destruction of the insulin-producing beta cells within the pancreatic islets of Langerhans, resulting in the lifelong need for exogenous insulin therapy. According to recent estimates, T1D currently affects about 8.4 million individuals worldwide. Since a definitive biological cure for this disease is not available yet, there is a great need for novel therapeutic strategies aimed at safely and effectively altering the natural history of the disease during its sequential stages. Ideal therapeutic goals in T1D include the prevention of autoimmune beta-cell destruction, the preservation of residual beta-cell mass and endogenous insulin secretion, the replacement and/or regeneration of beta cells, as well as automated insulin delivery through advanced closed-loop artificial pancreas systems. With this regard, an important research area focused on the identification of a definitive biological cure for T1D is represented by the investigation of immunotherapeutic and beta-cell-protective agents used as disease-modifying therapies to forestall or eliminate insulin dependence. In this commentary, we discuss the reasons why the use of combination therapies targeting the multiple immunometabolic dysfunctions associated with T1D (other than beta-cell autoimmunity) is likely to be more effective in preserving beta cell function in individuals at different stages of T1D, as compared to the use of single therapeutic agents.
Collapse
|
18
|
Piersanti A, Pacini G, Tura A, D'Argenio DZ, Morettini M. An in-silico modeling approach to separate exogenous and endogenous plasma insulin appearance, with application to inhaled insulin. Sci Rep 2024; 14:10936. [PMID: 38740832 PMCID: PMC11091049 DOI: 10.1038/s41598-024-61293-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
The aim of this study was to develop a dynamic model-based approach to separately quantify the exogenous and endogenous contributions to total plasma insulin concentration and to apply it to assess the effects of inhaled-insulin administration on endogenous insulin secretion during a meal test. A three-step dynamic in-silico modeling approach was developed to estimate the two insulin contributions of total plasma insulin in a group of 21 healthy subjects who underwent two equivalent standardized meal tests on separate days, one of which preceded by inhalation of a Technosphere® Insulin dose (22U or 20U). In the 30-120 min test interval, the calculated endogenous insulin component showed a divergence in the time course between the test with and without inhaled insulin. Moreover, the supra-basal area-under-the-curve of endogenous insulin in the test with inhaled insulin was significantly lower than that in the test without (2.1 ± 1.7 × 104 pmol·min/L vs 4.2 ± 1.8 × 104 pmol·min/L, p < 0.01). The percentage of exogenous insulin reaching the plasma, relative to the inhaled dose, was 42 ± 21%. The proposed in-silico approach separates exogenous and endogenous insulin contributions to total plasma insulin, provides individual bioavailability estimates, and can be used to assess the effect of inhaled insulin on endogenous insulin secretion during a meal.
Collapse
Affiliation(s)
- Agnese Piersanti
- Department of Information Engineering, Università Politecnica Delle Marche, Via Brecce Bianche 12, Ancona, Italy
| | | | | | - David Z D'Argenio
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Micaela Morettini
- Department of Information Engineering, Università Politecnica Delle Marche, Via Brecce Bianche 12, Ancona, Italy.
| |
Collapse
|
19
|
Uitbeijerse BS, Nijhoff MF, de Koning EJP. Comparison of an oral mixed meal plus arginine and intravenous glucose, GLP-1 plus arginine to unmask residual islet function in longstanding type 1 diabetes. Am J Physiol Endocrinol Metab 2024; 326:E673-E680. [PMID: 38446636 PMCID: PMC11376486 DOI: 10.1152/ajpendo.00030.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Residual beta cells are present in most patients with longstanding type 1 diabetes but it is unknown whether these beta cells react normally to different stimuli. Moreover a defect in proinsulin conversion and abnormal alpha cell response are also part of the islet dysfunction. A three-phase [euglycemia, hyperglycemia, and hyperglycemia + glucagon-like peptide 1 (GLP-1)] clamp was performed in patients with longstanding type 1 diabetes. Intravenous arginine boluses were administered at the end of each phase. On another day, a mixed meal stimulation test with a subsequent intravenous arginine bolus was performed. C-peptide was detectable in a subgroup of subjects at baseline (2/15) or only after stimulation (3/15). When detectable, C-peptide increased 2.9-fold [95% CI: 1.2-7.1] during the hyperglycemia phase and 14.1-fold [95% CI: 3.1-65.2] during the hyperglycemia + GLP-1 phase, and 22.3-fold [95% CI: 5.6-89.1] during hyperglycemia + GLP-1 + arginine phase when compared with baseline. The same subset of patients with a C-peptide response were identified during the mixed meal stimulation test as during the clamp. There was an inhibition of glucagon secretion (0.72-fold, [95% CI: 0.63-0.84]) during the glucose clamp irrespective of the presence of detectable beta cell function. Proinsulin was only present in a subset of subjects with detectable C-peptide (3/15) and proinsulin mimicked the C-peptide response to the different stimuli when detectable. Residual beta cells in longstanding type 1 diabetes respond adequately to different stimuli and could be of clinical benefit.NEW & NOTEWORTHY If beta cell function is detectable, the beta cells react relatively normal to the different stimuli except for the first phase response to intravenous glucose. An oral mixed meal followed by an intravenous arginine bolus can identify residual beta cell function/mass as well as the more commonly used glucose potentiated arginine-induced insulin secretion during a hyperglycemic clamp.
Collapse
Affiliation(s)
- Bas S Uitbeijerse
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Michiel F Nijhoff
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
Liu W, Fang Y, Cai X, Zhu Y, Zhang M, Han X, Li J, Yin S, Cai D, Chen J, Wang L, Shi D, Ji L. Preserved C-peptide is common and associated with higher time in range in Chinese type 1 diabetes. Front Endocrinol (Lausanne) 2024; 15:1335913. [PMID: 38405156 PMCID: PMC10884320 DOI: 10.3389/fendo.2024.1335913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Objective The aim of this study is to determine the residual C-peptide level and to explore the clinical significance of preserved C-peptide secretion in glycemic control in Chinese individuals with type 1 diabetes (T1D). Research design and methods A total of 534 participants with T1D were enrolled and divided into two groups, low-C-peptide group (fasting C-peptide ≤10 pmol/L) and preserved-C-peptide group (fasting C-peptide >10 pmol/L), and clinical factors were compared between the two groups. In 174 participants who were followed, factors associated with C-peptide loss were also identified by Cox regression. In addition, glucose metrics derived from intermittently scanned continuous glucose monitoring were compared between individuals with low C-peptide and those with preserved C-peptide in 178 participants. Results The lack of preserved C-peptide was associated with longer diabetes duration, glutamic acid decarboxylase autoantibody, and higher daily insulin doses, after adjustment {OR, 1.10 [interquartile range (IQR), 1.06-1.14]; OR, 0.46 (IQR, 0.27-0.77); OR, 1.04 (IQR, 1.02-1.06)}. In the longitudinal analysis, the percentages of individuals with preserved C-peptide were 71.4%, 56.8%, 71.7%, 62.5%, and 22.2% over 5 years of follow-up. Preserved C-peptide was also associated with higher time in range after adjustment of diabetes duration [62.4 (IQR, 47.3-76.6) vs. 50.3 (IQR, 36.2-63.0) %, adjusted P = 0.003]. Conclusions Our results indicate that a high proportion of Chinese patients with T1D had preserved C-peptide secretion. Meanwhile, residual C-peptide was associated with favorable glycemic control, suggesting the importance of research on adjunctive therapy to maintain β-cell function in T1D.
Collapse
Affiliation(s)
- Wei Liu
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Yayu Fang
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Yu Zhu
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Mingxia Zhang
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Juan Li
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Sai Yin
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| | - Deheng Cai
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Jing Chen
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Lei Wang
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Dawei Shi
- School of Automation, Beijing Institute of Technology, Beijing, ;China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing, ;China
| |
Collapse
|
21
|
Christen U, Pouzol L, Tunis M, Sassi A, Tondello C, Bayer M, Hintermann E, Strasser DS, Schuldes S, Mentzel U, Martinic MM. Combination treatment of a novel CXCR3 antagonist ACT-777991 with an anti-CD3 antibody synergistically increases persistent remission in experimental models of type 1 diabetes. Clin Exp Immunol 2023; 214:131-143. [PMID: 37458220 PMCID: PMC10714188 DOI: 10.1093/cei/uxad083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/29/2023] [Accepted: 07/15/2023] [Indexed: 12/18/2023] Open
Abstract
Treatment of patients with recent-onset type 1 diabetes with an anti-CD3 antibody leads to the transient stabilization of C-peptide levels in responder patients. Partial efficacy may be explained by the entry of islet-reactive T-cells spared by and/or regenerated after the anti-CD3 therapy. The CXCR3/CXCL10 axis has been proposed as a key player in the infiltration of autoreactive T cells into the pancreatic islets followed by the destruction of β cells. Combining the blockade of this axis using ACT-777991, a novel small-molecule CXCR3 antagonist, with anti-CD3 treatment may prevent further infiltration and β-cell damage and thus, preserve insulin production. The effect of anti-CD3 treatment on circulating T-cell subsets, including CXCR3 expression, in mice was evaluated by flow cytometry. Anti-CD3/ACT-777991 combination treatment was assessed in the virally induced RIP-LCMV-GP and NOD diabetes mouse models. Treatments started at disease onset. The effects on remission rate, blood glucose concentrations, insulitis, and plasma C-peptide were evaluated for the combination treatment and the respective monotherapies. Anti-CD3 treatment induced transient lymphopenia but spared circulating CXCR3+ T cells. Combination therapy in both mouse models synergistically and persistently reduced blood glucose concentrations, resulting in increased disease remission rates compared to each monotherapy. At the study end, mice in disease remission demonstrated reduced insulitis and detectable plasma C-peptide levels. When treatments were initiated in non-severely hyperglycemic NOD mice at diabetes onset, the combination treatment led to persistent disease remission in all mice. These results provide preclinical validation and rationale to investigate the combination of ACT-777991 with anti-CD3 for the treatment of patients with recent-onset diabetes.
Collapse
Affiliation(s)
- Urs Christen
- Pharmazentrum Frankfurt, Goethe University Frankfurt, Germany
| | - Laetitia Pouzol
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Mélanie Tunis
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Anna Sassi
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | | | - Monika Bayer
- Pharmazentrum Frankfurt, Goethe University Frankfurt, Germany
| | | | - Daniel S Strasser
- Translational Biomarkers Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Sabrina Schuldes
- Project Management Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Ulrich Mentzel
- Pharmacology and Preclinical Development Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| | - Marianne M Martinic
- Immunology and Pharmacology Department, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil, Switzerland
| |
Collapse
|
22
|
Waibel M, Wentworth JM, So M, Couper JJ, Cameron FJ, MacIsaac RJ, Atlas G, Gorelik A, Litwak S, Sanz-Villanueva L, Trivedi P, Ahmed S, Martin FJ, Doyle ME, Harbison JE, Hall C, Krishnamurthy B, Colman PG, Harrison LC, Thomas HE, Kay TWH. Baricitinib and β-Cell Function in Patients with New-Onset Type 1 Diabetes. N Engl J Med 2023; 389:2140-2150. [PMID: 38055252 DOI: 10.1056/nejmoa2306691] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
BACKGROUND Janus kinase (JAK) inhibitors, including baricitinib, block cytokine signaling and are effective disease-modifying treatments for several autoimmune diseases. Whether baricitinib preserves β-cell function in type 1 diabetes is unclear. METHODS In this phase 2, double-blind, randomized, placebo-controlled trial, we assigned patients with type 1 diabetes diagnosed during the previous 100 days to receive baricitinib (4 mg once per day) or matched placebo orally for 48 weeks. The primary outcome was the mean C-peptide level, determined from the area under the concentration-time curve, during a 2-hour mixed-meal tolerance test at week 48. Secondary outcomes included the change from baseline in the glycated hemoglobin level, the daily insulin dose, and measures of glycemic control assessed with the use of continuous glucose monitoring. RESULTS A total of 91 patients received baricitinib (60 patients) or placebo (31 patients). The median of the mixed-meal-stimulated mean C-peptide level at week 48 was 0.65 nmol per liter per minute (interquartile range, 0.31 to 0.82) in the baricitinib group and 0.43 nmol per liter per minute (interquartile range, 0.13 to 0.63) in the placebo group (P = 0.001). The mean daily insulin dose at 48 weeks was 0.41 U per kilogram of body weight per day (95% confidence interval [CI], 0.35 to 0.48) in the baricitinib group and 0.52 U per kilogram per day (95% CI, 0.44 to 0.60) in the placebo group. The levels of glycated hemoglobin were similar in the two trial groups. However, the mean coefficient of variation of the glucose level at 48 weeks, as measured by continuous glucose monitoring, was 29.6% (95% CI, 27.8 to 31.3) in the baricitinib group and 33.8% (95% CI, 31.5 to 36.2) in the placebo group. The frequency and severity of adverse events were similar in the two trial groups, and no serious adverse events were attributed to baricitinib or placebo. CONCLUSIONS In patients with type 1 diabetes of recent onset, daily treatment with baricitinib over 48 weeks appeared to preserve β-cell function as estimated by the mixed-meal-stimulated mean C-peptide level. (Funded by JDRF International and others; BANDIT Australian New Zealand Clinical Trials Registry number, ACTRN12620000239965.).
Collapse
Affiliation(s)
- Michaela Waibel
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - John M Wentworth
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Michelle So
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Jennifer J Couper
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Fergus J Cameron
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Richard J MacIsaac
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Gabby Atlas
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Alexandra Gorelik
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Sara Litwak
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Laura Sanz-Villanueva
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Prerak Trivedi
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Simi Ahmed
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Francis J Martin
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Madeleine E Doyle
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Jessica E Harbison
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Candice Hall
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Balasubramanian Krishnamurthy
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Peter G Colman
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Leonard C Harrison
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Helen E Thomas
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Thomas W H Kay
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| |
Collapse
|
23
|
Taylor PN, Collins KS, Lam A, Karpen SR, Greeno B, Walker F, Lozano A, Atabakhsh E, Ahmed ST, Marinac M, Latres E, Senior PA, Rigby M, Gottlieb PA, Dayan CM. C-peptide and metabolic outcomes in trials of disease modifying therapy in new-onset type 1 diabetes: an individual participant meta-analysis. Lancet Diabetes Endocrinol 2023; 11:915-925. [PMID: 37931637 DOI: 10.1016/s2213-8587(23)00267-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Metabolic outcomes in type 1 diabetes remain suboptimal. Disease modifying therapy to prevent β-cell loss presents an alternative treatment framework but the effect on metabolic outcomes is unclear. We, therefore, aimed to define the relationship between insulin C-peptide as a marker of β-cell function and metabolic outcomes in new-onset type 1 diabetes. METHODS 21 trials of disease-modifying interventions within 100 days of type 1 diabetes diagnosis comprising 1315 adults (ie, those 18 years and older) and 1396 children (ie, those younger than 18 years) were combined. Endpoints assessed were stimulated area under the curve C-peptide, HbA1c, insulin use, hypoglycaemic events, and composite scores (such as insulin dose adjusted A1c, total daily insulin, U/kg per day, and BETA-2 score). Positive studies were defined as those meeting their primary endpoint. Differences in outcomes between active and control groups were assessed using the Wilcoxon rank test. FINDINGS 6 months after treatment, a 24·8% greater C-peptide preservation in positive studies was associated with a 0·55% lower HbA1c (p<0·0001), with differences being detectable as early as 3 months. Cross-sectional analysis, combining positive and negative studies, was consistent with this proportionality: a 55% improvement in C-peptide preservation was associated with 0·64% lower HbA1c (p<0·0001). Higher initial C-peptide levels and greater preservation were associated with greater improvement in HbA1c. For HbA1c, IDAAC, and BETA-2 score, sample size predictions indicated that 2-3 times as many participants per group would be required to show a difference at 6 months as compared with C-peptide. Detecting a reduction in hypoglycaemia was affected by reporting methods. INTERPRETATION Interventions that preserve β-cell function are effective at improving metabolic outcomes in new-onset type 1 diabetes, confirming their potential as adjuncts to insulin. We have shown that improvements in HbA1c are directly proportional to the degree of C-peptide preservation, quantifying this relationship, and supporting the use of C-peptides as a surrogate endpoint in clinical trials. FUNDING JDRF and Diabetes UK.
Collapse
Affiliation(s)
- Peter N Taylor
- Department of Infection and Immunity, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | | | - Anna Lam
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | | - Simi T Ahmed
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | | | | | - Peter A Senior
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Mark Rigby
- Critical Path Institute, Tucson, AZ, USA
| | | | - Colin M Dayan
- Department of Infection and Immunity, Cardiff University School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
24
|
Sims EK, Kulkarni A, Hull A, Woerner SE, Cabrera S, Mastrandrea LD, Hammoud B, Sarkar S, Nakayasu ES, Mastracci TL, Perkins SM, Ouyang F, Webb-Robertson BJ, Enriquez JR, Tersey SA, Evans-Molina C, Long SA, Blanchfield L, Gerner EW, Mirmira RG, DiMeglio LA. Inhibition of polyamine biosynthesis preserves β cell function in type 1 diabetes. Cell Rep Med 2023; 4:101261. [PMID: 37918404 PMCID: PMC10694631 DOI: 10.1016/j.xcrm.2023.101261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/18/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023]
Abstract
In preclinical models, α-difluoromethylornithine (DFMO), an ornithine decarboxylase (ODC) inhibitor, delays the onset of type 1 diabetes (T1D) by reducing β cell stress. However, the mechanism of DFMO action and its human tolerability remain unclear. In this study, we show that mice with β cell ODC deletion are protected against toxin-induced diabetes, suggesting a cell-autonomous role of ODC during β cell stress. In a randomized controlled trial (ClinicalTrials.gov: NCT02384889) involving 41 recent-onset T1D subjects (3:1 drug:placebo) over a 3-month treatment period with a 3-month follow-up, DFMO (125-1,000 mg/m2) is shown to meet its primary outcome of safety and tolerability. DFMO dose-dependently reduces urinary putrescine levels and, at higher doses, preserves C-peptide area under the curve without apparent immunomodulation. Transcriptomics and proteomics of DFMO-treated human islets exposed to cytokine stress reveal alterations in mRNA translation, nascent protein transport, and protein secretion. These findings suggest that DFMO may preserve β cell function in T1D through islet cell-autonomous effects.
Collapse
Affiliation(s)
- Emily K Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Abhishek Kulkarni
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Audrey Hull
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Nationwide Children's Hospital Pediatric Residency Program, Columbus, OH 43205, USA
| | - Stephanie E Woerner
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Susanne Cabrera
- Department of Pediatrics, Section of Endocrinology and Diabetes, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Lucy D Mastrandrea
- Division of Pediatric Endocrinology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Batoul Hammoud
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | - Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Teresa L Mastracci
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Susan M Perkins
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fangqian Ouyang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Jacob R Enriquez
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A Tersey
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Carmella Evans-Molina
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Medicine and the Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - S Alice Long
- Benaroya Research Institute, Center for Translational Immunology, Seattle, WA 98101, USA
| | - Lori Blanchfield
- Benaroya Research Institute, Center for Translational Immunology, Seattle, WA 98101, USA
| | | | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA.
| | - Linda A DiMeglio
- Division of Pediatric Endocrinology and Diabetology, Herman B. Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
25
|
Ylescupidez A, Bahnson HT, O'Rourke C, Lord S, Speake C, Greenbaum CJ. A standardized metric to enhance clinical trial design and outcome interpretation in type 1 diabetes. Nat Commun 2023; 14:7214. [PMID: 37940642 PMCID: PMC10632453 DOI: 10.1038/s41467-023-42581-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
The use of a standardized outcome metric enhances clinical trial interpretation and cross-trial comparison. If a disease course is predictable, comparing modeled predictions with outcome data affords the precision and confidence needed to accelerate precision medicine. We demonstrate this approach in type 1 diabetes (T1D) trials aiming to preserve endogenous insulin secretion measured by C-peptide. C-peptide is predictable given an individual's age and baseline value; quantitative response (QR) adjusts for these variables and represents the difference between the observed and predicted outcome. Validated across 13 trials, the QR metric reduces each trial's variance and increases statistical power. As smaller studies are especially subject to random sampling variability, using QR as the outcome introduces alternative interpretations of previous clinical trial results. QR can provide model-based estimates that quantify whether individuals or groups did better or worse than expected. QR also provides a purer metric to associate with biomarker measurements. Using data from more than 1300 participants, we demonstrate the value of QR in advancing disease-modifying therapy in T1D. QR applies to any disease where outcome is predictable by pre-specified baseline covariates, rendering it useful for defining responders to therapy, comparing therapeutic efficacy, and understanding causal pathways in disease.
Collapse
Affiliation(s)
- Alyssa Ylescupidez
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Henry T Bahnson
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Colin O'Rourke
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Sandra Lord
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Carla J Greenbaum
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| |
Collapse
|
26
|
Krogvold L, Mynarek IM, Ponzi E, Mørk FB, Hessel TW, Roald T, Lindblom N, Westman J, Barker P, Hyöty H, Ludvigsson J, Hanssen KF, Johannesen J, Dahl-Jørgensen K. Pleconaril and ribavirin in new-onset type 1 diabetes: a phase 2 randomized trial. Nat Med 2023; 29:2902-2908. [PMID: 37789144 PMCID: PMC10667091 DOI: 10.1038/s41591-023-02576-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
Previous studies showed a low-grade enterovirus infection in the pancreatic islets of patients with newly diagnosed type 1 diabetes (T1D). In the Diabetes Virus Detection (DiViD) Intervention, a phase 2, placebo-controlled, randomized, parallel group, double-blind trial, 96 children and adolescents (aged 6-15 years) with new-onset T1D received antiviral treatment with pleconaril and ribavirin (n = 47) or placebo (n = 49) for 6 months, with the aim of preserving β cell function. The primary endpoint was the mean stimulated C-peptide area under the curve (AUC) 12 months after the initiation of treatment (less than 3 weeks after diagnosis) using a mixed linear model. The model used longitudinal log-transformed serum C-peptide AUCs at baseline, at 3 months, 6 months and 1 year. The primary endpoint was met with the serum C-peptide AUC being higher in the pleconaril and ribavirin treatment group compared to the placebo group at 12 months (average marginal effect = 0.057 in the linear mixed model; 95% confidence interval = 0.004-0.11, P = 0.037). The treatment was well tolerated. The results show that antiviral treatment may preserve residual insulin production in children and adolescent with new-onset T1D. This provides a rationale for further evaluating antiviral strategies in the prevention and treatment of T1D. European Union Drug Regulating Authorities Clinical Trials identifier: 2015-003350-41 .
Collapse
Affiliation(s)
- Lars Krogvold
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Ida Maria Mynarek
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Erica Ponzi
- Clinical Trial Unit, Oslo University Hospital, Oslo, Norway
| | - Freja Barrett Mørk
- Steno Diabetes Center Copenhagen, Herlev University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Witzner Hessel
- Steno Diabetes Center Copenhagen, Herlev University Hospital, Copenhagen, Denmark
| | - Trine Roald
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | | | | | - Peter Barker
- National Institute for Health and Care Research Cambridge Biomedical Research Centre, Core Biochemistry Assay Laboratory, Cambridge, UK
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| | | | | | - Jesper Johannesen
- Steno Diabetes Center Copenhagen, Herlev University Hospital, Copenhagen, Denmark
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway.
- Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
27
|
Leslie RD, Ma RCW, Franks PW, Nadeau KJ, Pearson ER, Redondo MJ. Understanding diabetes heterogeneity: key steps towards precision medicine in diabetes. Lancet Diabetes Endocrinol 2023; 11:848-860. [PMID: 37804855 DOI: 10.1016/s2213-8587(23)00159-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/30/2023] [Accepted: 05/27/2023] [Indexed: 10/09/2023]
Abstract
Diabetes is a highly heterogeneous condition; yet, it is diagnosed by measuring a single blood-borne metabolite, glucose, irrespective of aetiology. Although pragmatically helpful, disease classification can become complex and limit advances in research and medical care. Here, we describe diabetes heterogeneity, highlighting recent approaches that could facilitate management by integrating three disease models across all forms of diabetes, namely, the palette model, the threshold model and the gradient model. Once diabetes has developed, further worsening of established diabetes and the subsequent emergence of diabetes complications are kept in check by multiple processes designed to prevent or circumvent metabolic dysfunction. The impact of any given disease risk factor will vary from person-to-person depending on their background, diabetes-related propensity, and environmental exposures. Defining the consequent heterogeneity within diabetes through precision medicine, both in terms of diabetes risk and risk of complications, could improve health outcomes today and shine a light on avenues for novel therapy in the future.
Collapse
Affiliation(s)
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong SAR, China; Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Paul W Franks
- Novo Nordisk Foundation, Hellerup, Denmark; Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmo, Sweden; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Kristen J Nadeau
- Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
| | - Ewan R Pearson
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, UK
| | | |
Collapse
|
28
|
Bhagadurshah RR, Eagappan S, Kasthuri Santharam R, Subbiah S. The Impact of Body Mass Index, Residual Beta Cell Function and Estimated Glucose Disposal Rate on the Development of Double Diabetes and Microvascular Complications in Patients With Type 1 Diabetes Mellitus. Cureus 2023; 15:e48979. [PMID: 38111445 PMCID: PMC10726016 DOI: 10.7759/cureus.48979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 12/20/2023] Open
Abstract
Background The clinical impact of body mass index (BMI), residual beta cell function and estimated glucose disposal rate (eGDR) in the development of double diabetes (DD) and microvascular complications are largely unknown. We aimed to assess whether BMI, residual beta cell function measured by plasma "C" peptide and insulin resistance measured by eGDR have any impact on the development of DD and microvascular complications in patients with type 1 diabetes mellitus (T1DM). Methods It is a cross-sectional observational study involving 113 T1DM patients of more than five years duration who were classified into two groups: normal BMI (18.5-22.9 kg/m2) and overweight/obese group (≥ 23kg/m2) based on Asian BMI classification. Based on their eGDR values, they were grouped into four categories: ≥ 8, 6-7.99, 4-5.99, and < 4 mg/kg/min. The prevalence of DD based on eGDR values was determined. Their BMI and different eGDR categories were compared with the prevalence of diabetic retinopathy and nephropathy and their odds ratio (OR) was calculated. Results The median and interquartile range (IQR) of the eGDR of the overweight/obese group was significantly lower than the normal BMI group (5.3 [3.96-8.15] vs 8.72 [6.50-9.77 mg/kg/min], p < 0.001). The prevalence of DD in the overweight/obese T1DM group and normal BMI group was 75% and 33.3% respectively. The OR of retinopathy and nephropathy in the overweight/obese group was 3.28 (p = 0.007) and 3.01 (p = 0.015) respectively when compared to the normal BMI group. The OR of retinopathy and nephropathy in T1DM patients with eGDR < 4 mg/kg/min was 17.13 (p = 0.001) and 18.5 (p = 0.001) respectively. The lower the eGDR values, the higher the prevalence of retinopathy and nephropathy regardless of HbA1c levels. Conclusion As overweight and obesity are increasingly becoming more prevalent in T1DM, the eGDR will better predict the development of DD and microvascular complications irrespective of HbA1c levels. It is more useful as a variable and easily inducted into routine clinical practice. However, residual beta cell function was not useful in predicting the development of microvascular complications.
Collapse
Affiliation(s)
- Rameez Raja Bhagadurshah
- Department of Endocrinology and Diabetology, Madurai Medical College and Government Rajaji Hospital, Madurai, IND
| | - Subbiah Eagappan
- Department of Endocrinology and Diabetology, Madurai Medical College and Government Rajaji Hospital, Madurai, IND
| | - Raghavan Kasthuri Santharam
- Department of Endocrinology and Diabetology, Madurai Medical College and Government Rajaji Hospital, Madurai, IND
| | - Sridhar Subbiah
- Department of Endocrinology and Diabetology, Madurai Medical College and Government Rajaji Hospital, Madurai, IND
| |
Collapse
|
29
|
Jones AG, Shields BM, Oram RA, Dabelea DM, Hagopian WA, Lustigova E, Shah AS, Knupp J, Mottl AK, DÀgostino RB, Williams A, Marcovina SM, Pihoker C, Divers J, Redondo MJ. Clinical prediction models combining routine clinical measures identify participants with youth-onset diabetes who maintain insulin secretion in the range associated with type 2 diabetes: The SEARCH for Diabetes in Youth Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.27.23296128. [PMID: 37808789 PMCID: PMC10557841 DOI: 10.1101/2023.09.27.23296128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Objective With the high prevalence of pediatric obesity and overlapping features between diabetes subtypes, accurately classifying youth-onset diabetes can be challenging. We aimed to develop prediction models that, using characteristics available at diabetes diagnosis, can identify youth who will retain endogenous insulin secretion at levels consistent with type 2 diabetes (T2D). Methods We studied 2,966 youth with diabetes in the prospective SEARCH study (diagnosis age ≤19 years) to develop prediction models to identify participants with fasting c-peptide ≥250 pmol/L (≥0.75ng/ml) after >3 years (median 74 months) of diabetes duration. Models included clinical measures at baseline visit, at a mean diabetes duration of 11 months (age, BMI, sex, waist circumference, HDL-C), with and without islet autoantibodies (GADA, IA-2A) and a Type 1 Diabetes Genetic Risk Score (T1DGRS). Results Models using routine clinical measures with or without autoantibodies and T1DGRS were highly accurate in identifying participants with c-peptide ≥0.75 ng/ml (17% of participants; 2.3% and 53% of those with and without positive autoantibodies) (area under receiver operator curve [AUCROC] 0.95-0.98). In internal validation, optimism was very low, with excellent calibration (slope=0.995-0.999). Models retained high performance for predicting retained c-peptide in older youth with obesity (AUCROC 0.88-0.96), and in subgroups defined by self-reported race/ethnicity (AUCROC 0.88-0.97), autoantibody status (AUCROC 0.87-0.96), and clinically diagnosed diabetes types (AUCROC 0.81-0.92). Conclusion Prediction models combining routine clinical measures at diabetes diagnosis, with or without islet autoantibodies or T1DGRS, can accurately identify youth with diabetes who maintain endogenous insulin secretion in the range associated with type 2 diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Amy S Shah
- University of Cincinnati & Cincinnati Children's Hospital Medical Center
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gavigan C, Donner T. Predictors of Responsiveness to GLP-1 Receptor Agonists in Insulin-Treated Patients with Type 2 Diabetes. J Diabetes Res 2023; 2023:9972132. [PMID: 37589043 PMCID: PMC10427225 DOI: 10.1155/2023/9972132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 08/18/2023] Open
Abstract
Background Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are potent antihyperglycemic agents with beneficial effects on weight, cardiovascular, and renal outcomes. Physicians lack guidance as to which patients with insulin-requiring type 2 diabetes will respond best to GLP-1 RAs with respect to glycemic control, insulin dose reduction, and weight loss. This study evaluated the efficacy of GLP-1 RAs in patients with type 2 diabetes on insulin and patient factors that may predict a beneficial clinical response. Methods Adults with type 2 diabetes treated with insulin who had a GLP-1 RA added to their regimen were evaluated retrospectively. Baseline parameters and outcomes at 3, 6, and 12 months were collected. Results Among the 81 patients included, there was a mean reduction in hemoglobin A1C of 0.94% (SD, 0.26; p = 0.0007), 0.40% (SD, 0.21; p = 0.0636), and 0.58% (SD, 0.23, p = 0.0154) at 3, 6, and 12 months, respectively, following the addition of a GLP-1 RA. There was also a reduction in body weight noted at each time point. Baseline characteristics including BMI, duration of diabetes, and insulin requirement did not significantly affect A1C reduction when GLP-1 RA was added. At 3 months, patients with a random C-peptide that was normal (≥0.8 ng/ml) were significantly more likely to have discontinued insulin than those with random C-peptide that was low (<0.8 ng/ml) (11 of 23 vs. 0 of 7 patients, p = 0.029). Conclusions The addition of a GLP-1 RA reduced HbA1C, weight, and insulin requirements in this cohort of patients with type 2 diabetes on insulin. BMI, baseline insulin dose, and diabetes duration did not predict response. A C-peptide level ≥ 0.8 ng/ml predicted a beneficial response after 3 months of therapy.
Collapse
Affiliation(s)
- Colleen Gavigan
- Johns Hopkins Diabetes Center, Division of Endocrinology, Diabetes & Metabolism, Johns Hopkins University School of Medicine, 601 N Caroline Street, Baltimore, Maryland, USA 21287
| | - Thomas Donner
- Johns Hopkins Diabetes Center, Division of Endocrinology, Diabetes & Metabolism, Johns Hopkins University School of Medicine, 601 N Caroline Street, Baltimore, Maryland, USA 21287
| |
Collapse
|
31
|
Harsunen M, Haukka J, Harjutsalo V, Mars N, Syreeni A, Härkönen T, Käräjämäki A, Ilonen J, Knip M, Sandholm N, Miettinen PJ, Groop PH, Tuomi T. Residual insulin secretion in individuals with type 1 diabetes in Finland: longitudinal and cross-sectional analyses. Lancet Diabetes Endocrinol 2023; 11:465-473. [PMID: 37290465 DOI: 10.1016/s2213-8587(23)00123-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Contrary to the presumption that type 1 diabetes leads to an absolute insulin deficiency, many individuals with type 1 diabetes have circulating C-peptide years after the diagnosis. We studied factors affecting random serum C-peptide concentration in individuals with type 1 diabetes and the association with diabetic complications. METHODS Our longitudinal analysis included individuals newly diagnosed with type 1 diabetes from Helsinki University Hospital (Helsinki, Finland) with repeated random serum C-peptide and concomitant glucose measurements from within 3 months of diagnosis and at least once later. The long-term cross-sectional analysis included data from participants from 57 centres in Finland who had type 1 diabetes diagnosed after 5 years of age, initiation of insulin treatment within 1 year from diagnosis, and a C-peptide concentration of less than 1·0 nmol/L (FinnDiane study) and patients with type 1 diabetes from the DIREVA study. We tested the association of random serum C-peptide concentrations and polygenic risk scores with one-way ANOVA, and association of random serum C-peptide concentrations, polygenic risk scores, and clinical factors with logistic regression. FINDINGS The longitudinal analysis included 847 participants younger than 16 years and 110 aged 16 years or older. In the longitudinal analysis, age at diagnosis strongly correlated with the decline in C-peptide secretion. The cross-sectional analysis included 3984 participants from FinnDiane and 645 from DIREVA. In the cross-sectional analysis, at a median duration of 21·6 years (IQR 12·5-31·2), 776 (19·4%) of 3984 FinnDiane participants had residual random serum C-peptide secretion (>0·02 nmol/L), which was associated with lower type 1 diabetes polygenic risk compared with participants without random serum C-peptide (p<0·0001). Random serum C-peptide was inversely associated with hypertension, HbA1c, and cholesterol, but also independently with microvascular complications (adjusted OR 0·61 [95% CI 0·38-0·96], p=0·033, for nephropathy; 0·55 [0·34-0·89], p=0·014, for retinopathy). INTERPRETATION Although children with multiple autoantibodies and HLA risk genotypes progressed to absolute insulin deficiency rapidly, many adolescents and adults had residual random serum C-peptide decades after the diagnosis. Polygenic risk of type 1 and type 2 diabetes affected residual random serum C-peptide. Even low residual random serum C-peptide concentrations seemed to be associated with a beneficial complications profile. FUNDING Folkhälsan Research Foundation; Academy of Finland; University of Helsinki and Helsinki University Hospital; Medical Society of Finland; the Sigrid Juselius Foundation; the "Liv and Hälsa" Society; Novo Nordisk Foundation; and State Research Funding via the Helsinki University Hospital, the Vasa Hospital District, Turku University Hospital, Vasa Central Hospital, Jakobstadsnejdens Heart Foundation, and the Medical Foundation of Vaasa.
Collapse
Affiliation(s)
- Minna Harsunen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jani Haukka
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Nina Mars
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anna Syreeni
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Taina Härkönen
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Annemari Käräjämäki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Diabetes unit of Ostrobothnia, Wellbeing Services County of Ostrobothnia, Vaasa, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi Johanna Miettinen
- Translational Stem Cell Biology and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Diabetes, Central Medical School, Monash University, Melbourne, VIC, Australia
| | - Tiinamaija Tuomi
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland; Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Abdominal Center, Endocrinology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Lund, Sweden.
| |
Collapse
|
32
|
Skyler JS. Importance of residual insulin secretion in type 1 diabetes. Lancet Diabetes Endocrinol 2023; 11:443-444. [PMID: 37290467 DOI: 10.1016/s2213-8587(23)00149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023]
Affiliation(s)
- Jay S Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
33
|
Mameli C, Triolo TM, Chiarelli F, Rewers M, Zuccotti G, Simmons KM. Lessons and Gaps in the Prediction and Prevention of Type 1 Diabetes. Pharmacol Res 2023; 193:106792. [PMID: 37201589 DOI: 10.1016/j.phrs.2023.106792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
Type 1 diabetes (T1D) is a serious chronic autoimmune condition. Even though the root cause of T1D development has yet to be determined, enough is known about the natural history of T1D pathogenesis to allow study of interventions that may delay or even prevent the onset of hyperglycemia and clinical T1D. Primary prevention aims to prevent the onset of beta cell autoimmunity in asymptomatic people at high genetic risk for T1D. Secondary prevention strategies aim to preserve functional beta cells once autoimmunity is present, and tertiary prevention aims to initiate and extend partial remission of beta cell destruction after the clinical onset of T1D. The approval of teplizumab in the United States to delay the onset of clinical T1D marks an impressive milestone in diabetes care. This treatment opens the door to a paradigm shift in T1D care. People with T1D risk need to be identified early by measuring T1D related islet autoantibodies. Identifying people with T1D before they have symptoms will facilitate better understanding of pre-symptomatic T1D progression and T1D prevention strategies that may be effective.
Collapse
Affiliation(s)
- Chiara Mameli
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
| | - Taylor M Triolo
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
| | | | - Marian Rewers
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Kimber M Simmons
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
34
|
Huang M, Chen W, Wang M, Huang Y, Liu H, Ming Y, Chen Y, Tang Z, Jia B. Advanced Delivery Strategies for Immunotherapy in Type I Diabetes Mellitus. BioDrugs 2023; 37:331-352. [PMID: 37178431 PMCID: PMC10182560 DOI: 10.1007/s40259-023-00594-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2023] [Indexed: 05/15/2023]
Abstract
Type 1 diabetes mellitus (T1DM) has been defined as an autoimmune disease characterised by immune-mediated destruction of the pancreatic β cells, leading to absolute insulin deficiency and hyperglycaemia. Current research has increasingly focused on immunotherapy based on immunosuppression and regulation to rescue T-cell-mediated β-cell destruction. Although T1DM immunotherapeutic drugs are constantly under clinical and preclinical development, several key challenges remain, including low response rates and difficulty in maintaining therapeutic effects. Advanced drug delivery strategies can effectively harness immunotherapies and improve their potency while reducing their adverse effects. In this review, we briefly introduce the mechanisms of T1DM immunotherapy and focus on the current research status of the integration of the delivery techniques in T1DM immunotherapy. Furthermore, we critically analyse the challenges and future directions of T1DM immunotherapy.
Collapse
Affiliation(s)
- Mingshu Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Weixing Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Min Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yisheng Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Hongyu Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yue Ming
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yuanxin Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhengming Tang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bo Jia
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
35
|
Tatovic D, Narendran P, Dayan CM. A perspective on treating type 1 diabetes mellitus before insulin is needed. Nat Rev Endocrinol 2023; 19:361-370. [PMID: 36914759 DOI: 10.1038/s41574-023-00816-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 03/16/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a progressive autoimmune disease that starts long before a clinical diagnosis is made. The American Diabetes Association recognizes three stages: stage 1 (normoglycaemic and positive for autoantibodies to β-cell antigens); stage 2 (asymptomatic with dysglycaemia); and stage 3, which is defined by glucose levels consistent with the definition of diabetes mellitus. This Perspective focuses on the management of the proportion of individuals with early stage 3 T1DM who do not immediately require insulin; a stage we propose should be termed stage 3a. To date, this period of non-insulin-dependent T1DM has been largely unrecognized. Importantly, it represents a window of opportunity for intervention, as remaining at this stage might delay the need for insulin by months or years. Extending the insulin-free period and/or avoiding unnecessary insulin therapy are important goals, as there is no risk of hypoglycaemia during this period and the adherence burden on patients of glycaemic monitoring and daily adjustments for diet and exercise is substantially reduced. Recognizing the pressing need for guidance on adequate management of children and adults with stage 3a T1DM, we present our perspective on the subject, which needs to be tested in formal and adequately powered clinical trials.
Collapse
Affiliation(s)
- Danijela Tatovic
- Diabetes Research Group, Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Parth Narendran
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Colin M Dayan
- Diabetes Research Group, Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK.
| |
Collapse
|
36
|
Hörber S, Orth M, Fritsche A, Peter A. Comparability of C-Peptide Measurements - Current Status and Clinical Relevance. Exp Clin Endocrinol Diabetes 2023; 131:173-178. [PMID: 36630986 PMCID: PMC9998184 DOI: 10.1055/a-1998-6889] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
C-peptide is an increasingly used and established marker for beta cell function by assessing endogenous insulin secretion. Accurate and comparable C-peptide measurements are needed in clinical practice and research studies. For example, to calculate HOMA-indices, the C-peptide/glucose ratio, and the classification of recently published novel subgroups of diabetes and prediabetes have used C-peptide measurements. Although the process for standardization of C-peptide measurements is advanced, its full implementation is still missing; therefore, the current status of the comparability of C-peptide measurements using different immunoassays is unclear. Here we compared five widely used C-peptide immunoassays on different analyzers (Abbott ALINITY i, DiaSorin Liaison XL, Roche Cobas e411, Siemens Healthineers ADVIA Centaur XPT, and Immulite 2000 XPi) using serum samples covering the clinically relevant C-peptide concentration range. Although all investigated immunoassays are traceable to the international reference reagent for C-peptide (NIBSC code: 84/510), results of C-peptide measurements showed significant differences between analyzers in the entire concentration range, especially with increasing C-peptide concentrations. The mean bias was largest (36.6%) between results of the immunoassays by Roche and Siemens Healthineers (ADVIA Centaur XPT), and both assays revealed large discrepancies compared to immunoassays by Abbott, DiaSorin, and Siemens Healthineers (Immulite 2000 XPi). In contrast, the three latter assays showed similar C-peptide results (mean bias: 2.3% to 4.2%). Consequently, C-peptide discrepancies might affect clinical diagnosis and the interpretation of study results. Therefore, there is an urgent need to implement and finalize the standardization process of C-peptide measurements to improve patient care and the comparability of research studies.
Collapse
Affiliation(s)
- Sebastian Hörber
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Matthias Orth
- Institute of Laboratory Medicine, Vinzenz von Paul Kliniken gGmbH, Stuttgart, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Germany
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
37
|
Yang Q, Liu Y, Peng J, Pan J, Chen L, Cui J, Yi B. High levels of serum C-peptide are associated with a decreased risk for incident renal progression in patients with type 2 diabetes: a retrospective cohort study. BMJ Open Diabetes Res Care 2023; 11:11/2/e003201. [PMID: 36958752 PMCID: PMC10040069 DOI: 10.1136/bmjdrc-2022-003201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/01/2023] [Indexed: 03/25/2023] Open
Abstract
INTRODUCTION C-peptide has been reported to provide renoprotective effects. This study aims to explore the relationship between C-peptide and progression of renal function in patients with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS We retrospectively collected clinical data from 854 T2DM patients over a median follow-up of 5 years. Renal events included an annual decline in estimated glomerular filtration rate (eGFR), a rapid kidney function decline and a renal composite endpoint. A linear mixed-effects model and Cox regression analysis were used to investigate the effect of C-peptide on renal events, and a subgroup analysis was performed after stratification by risk factors. RESULTS The highest-level C-peptide group had a smaller annual eGFR decline compared with those in the group with the lowest level (p<0.05). Higher levels of 2 h postprandial C-peptide (2hPCP) (adjusted HR 0.53; 95% CI 0.31 to 0.92), difference between 2 h postprandial and fasting C-peptide (ΔCP) (adjusted HR 0.39; 95% CI 0.22 to 0.69), and 2 h postprandial C-peptide-to-glucose ratio (PCGR) (adjusted HR 0.44; 95% CI 0.24 to 0.82) were independently related to a decreased risk for the renal composite endpoint. 2hPCP <2.92 ng/mL, ΔCP <1.86 ng/mL, and PCGR <1.11 significantly increased the risk of progression in kidney function (adjusted HRs <0.50, p<0.05) among T2DM patients with male sex, an age of <65 years old, a disease course of <10 years, an glycosylated hemoglobin value of ≥7%, or a history of hypertension. CONCLUSIONS Higher levels of 2hPCP, ΔCP and PCGR could protect T2DM patients from renal progression, especially in the aforementioned population with diabetes.
Collapse
Affiliation(s)
- Qi Yang
- Department of Nephrology, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, Central South University, Changsha, Hunan, China
| | - Juan Peng
- Department of Nephrology, Central South University, Changsha, Hunan, China
| | - Jinting Pan
- Department of Nephrology, Central South University, Changsha, Hunan, China
| | - Li Chen
- Department of Nephrology, Central South University, Changsha, Hunan, China
| | - Jing Cui
- Department of Nephrology, Central South University, Changsha, Hunan, China
| | - Bin Yi
- Department of Nephrology, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Vollenbrock CE, Mul D, Dekker P, Birnie E, de Vries-Velraeds MMC, Boesten L, Groen J, Geelhoed-Duijvestijn PHLM, Aanstoot HJ, Wolffenbuttel BHR. Fasting and meal-stimulated serum C-peptide in long-standing type 1 diabetes mellitus. Diabet Med 2023; 40:e15012. [PMID: 36398450 PMCID: PMC10107202 DOI: 10.1111/dme.15012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
AIMS This study aims to evaluate the stability of C-peptide over time and to compare fasting C-peptide and C-peptide response after mixed-meal tolerance test (MMTT) at T90 or T120 with C-peptide area under the curve (AUC) in long-standing type 1 diabetes. METHODS We included 607 type 1 diabetes individuals with diabetes duration >5 years. C-peptide concentrations (ultrasensitive assay) were collected in the fasting state, and in a subpopulation after MMTT (T0, just prior to, T30-T60-T90-T120, 30-120 min after ingestion of mixed-meal) (n = 168). Fasting C-peptide concentrations (in n = 535) at Year 0 and Year 1 were compared. The clinical determinants associated with residual C-peptide secretion and the correspondence of C-peptide at MMTT T90 / T120 and total AUC were assessed. RESULTS A total of 153 participants (25%) had detectable fasting serum C-peptide (i.e ≥ 3.8 pmol/L). Fasting C-peptide was significantly lower at Year 1 (p < 0.001, effect size = -0.16). Participants with higher fasting C-peptide had a higher age at diagnosis and shorter disease duration and were less frequently insulin pump users. Overall, 109 of 168 (65%) participants had both non-detectable fasting and post-meal serum C-peptide concentrations. The T90 and T120 C-peptide values at MMTT were concordant with total AUC. In 17 (10%) individuals, C-peptide was only detectable at MMTT and not in the fasting state. CONCLUSIONS Stimulated C-peptide was detectable in an additional 10% of individuals compared with fasting in individuals with >5 years of diabetes duration. T90 and T120 MMTT measurements showed good concordance with the MMTT total AUC. Overall, there was a decrease of C-peptide at 1-year follow-up.
Collapse
Affiliation(s)
- Charlotte E Vollenbrock
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dick Mul
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | - Pim Dekker
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | - Erwin Birnie
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | | | - Lianne Boesten
- Department of Clinical Chemistry, IJsselland Ziekenhuis, Capelle aan den IJssel, The Netherlands
| | - Joost Groen
- Department of Clinical Chemistry, IJsselland Ziekenhuis, Capelle aan den IJssel, The Netherlands
| | | | - Henk-Jan Aanstoot
- Diabeter, Center for Paediatric and Adolescent Diabetes Care and Research, Rotterdam, The Netherlands
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
39
|
Abstract
Regular physical activity improves cardiometabolic and musculoskeletal health, helps with weight management, improves cognitive and psychosocial functioning, and is associated with reduced mortality related to cancer and diabetes mellitus. However, turnover rates of glucose in the blood increase dramatically during exercise, which often results in either hypoglycaemia or hyperglycaemia as well as increased glycaemic variability in individuals with type 1 diabetes mellitus (T1DM). A complex neuroendocrine response to an acute exercise session helps to maintain circulating levels of glucose in a fairly tight range in healthy individuals, while several abnormal physiological processes and limitations of insulin therapy limit the capacity of people with T1DM to exercise in a normoglycaemic state. Knowledge of the acute and chronic effects of exercise and regular physical activity is critical for the formulation of clinical strategies for the management of insulin and nutrition for active patients with T1DM. Emerging diabetes-related technologies, such as continuous glucose monitors, automated insulin delivery systems and the administration of solubilized glucagon, are demonstrating efficacy for preserving glucose homeostasis during and after exercise in this population of patients. This Review highlights the beneficial effects of regular exercise and details the complex endocrine and metabolic responses to different types of exercise for adults with T1DM. An overview of basic clinical strategies for the preservation of glucose homeostasis using emerging technologies is also provided.
Collapse
Affiliation(s)
- Michael C Riddell
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.
- LMC Diabetes and Endocrinology, Toronto, Ontario, Canada.
| | - Anne L Peters
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
40
|
Abstract
First envisioned by early diabetes clinicians, a person-centred approach to care was an aspirational goal that aimed to match insulin therapy to each individual's unique requirements. In the 100 years since the discovery of insulin, this goal has evolved to include personalised approaches to type 1 diabetes diagnosis, treatment, prevention and prediction. These advances have been facilitated by the recognition of type 1 diabetes as an autoimmune disease and by advances in our understanding of diabetes pathophysiology, genetics and natural history, which have occurred in parallel with advancements in insulin delivery, glucose monitoring and tools for self-management. In this review, we discuss how these personalised approaches have improved diabetes care and how improved understanding of pathogenesis and human biology might inform precision medicine in the future.
Collapse
Affiliation(s)
- Alice L J Carr
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
41
|
Ludvigsson J, Eriksson L, Nowak C, Teixeira PF, Widman M, Lindqvist A, Casas R, Lind M, Hannelius U. Phase III, randomised, double-blind, placebo-controlled, multicentre trial to evaluate the efficacy and safety of rhGAD65 to preserve endogenous beta cell function in adolescents and adults with recently diagnosed type 1 diabetes, carrying the genetic HLA DR3-DQ2 haplotype: the DIAGNODE-3 study protocol. BMJ Open 2022; 12:e061776. [PMID: 36316084 PMCID: PMC9628549 DOI: 10.1136/bmjopen-2022-061776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Type 1 diabetes (T1D) is an autoimmune disease leading to the destruction of the insulin-producing beta cells resulting in insulin deficiency and hyperglycaemic. Today, no approved therapy exists to halt this detrimental immunologic process. In a recent phase 2b study, intralymphatic administration of recombinant human glutamic acid decarboxylase 65 kDa (rhGAD65) adsorbed to Alhydrogel adjuvant to individuals recently diagnosed with T1D and carrying the HLA DR3-DQ2 haplotype showed promising results in preserving endogenous insulin secretion, confirming the results of a large meta-analysis of three randomised placebo-controlled trials of subcutaneous rhGAD65. The aim of the current precision medicine phase 3 study is to determine whether intralymphatic administration of rhGAD65 preserves insulin secretion and improves glycaemic control in presumed responder individuals with recently diagnosed T1D carrying HLA DR3-DQ2. METHODS AND ANALYSIS Individuals ≥12 and <29 years recently diagnosed with T1D (<6 months) will be screened for the HLA DR3-DQ2 haplotype, endogenous insulin production estimated by fasting C-peptide and presence of GAD65 antibodies. 330 patients are planned to be randomised to 3 monthly intralymphatic injections of rhGAD65 or placebo (both accompanied by oral vitamin D supplementation), followed by 22 months of follow-up. The study is powered to detect a treatment effect in the two coprimary endpoints; change from baseline in AUC(0-120min) C-peptide levels during a mixed meal tolerance test, and change from baseline in glycaemic control estimated by haemoglobin A1c at 24 months. Secondary endpoints include effects on glucose patterns collected by masked continuous glucose monitoring, proportion of patients in partial remission and number of episodes of severe hypoglycaemia and/or diabetic ketoacidosis. ETHICS AND DISSEMINATION The trial is approved by Ethics Committees in Poland (124/2021), the Netherlands (R21.089), Sweden (2021-05063), Czech Republic (EK-1144/21), Germany (2021361) and Spain (21/2021). Results will be published in international peer-reviewed scientific journals and presented at national and international conferences. TRIAL REGISTRATION NUMBER EudraCT identifier: 2021-002731-32, NCT identifier: NCT05018585.
Collapse
Affiliation(s)
- Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children's Hospital, Linköping, Sweden
| | | | - Christoph Nowak
- Diamyd Medical AB, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | | | | | | | - Rosaura Casas
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Marcus Lind
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- NU-Hospital Group and the Sahlgrenska University Hospital, Uddevalla and Gothenburg, Sweden
| | | |
Collapse
|
42
|
Maddaloni E, Bolli GB, Frier BM, Little RR, Leslie RD, Pozzilli P, Buzzetti R. C-peptide determination in the diagnosis of type of diabetes and its management: A clinical perspective. Diabetes Obes Metab 2022; 24:1912-1926. [PMID: 35676794 PMCID: PMC9543865 DOI: 10.1111/dom.14785] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/21/2022] [Accepted: 06/01/2022] [Indexed: 12/19/2022]
Abstract
Impaired beta-cell function is a recognized cornerstone of diabetes pathophysiology. Estimates of insulin secretory capacity are useful to inform clinical practice, helping to classify types of diabetes, complication risk stratification and to guide treatment decisions. Because C-peptide secretion mirrors beta-cell function, it has emerged as a valuable clinical biomarker, mainly in autoimmune diabetes and especially in adult-onset diabetes. Nonetheless, the lack of robust evidence about the clinical utility of C-peptide measurement in type 2 diabetes, where insulin resistance is a major confounder, limits its use in such cases. Furthermore, problems remain in the standardization of the assay for C-peptide, raising concerns about comparability of measurements between different laboratories. To approach the heterogeneity and complexity of diabetes, reliable, simple and inexpensive clinical markers are required that can inform clinicians about probable pathophysiology and disease progression, and so enable personalization of management and therapy. This review summarizes the current evidence base about the potential value of C-peptide in the management of the two most prevalent forms of diabetes (type 2 diabetes and autoimmune diabetes) to address how its measurement may assist daily clinical practice and to highlight current limitations and areas of uncertainties to be covered by future research.
Collapse
Affiliation(s)
- Ernesto Maddaloni
- Experimental Medicine DepartmentSapienza University of RomeRomeItaly
| | - Geremia B. Bolli
- Department of Medicine and Surgery, Section of Endocrinology and MetabolismUniversity of PerugiaPerugiaItaly
| | - Brian M. Frier
- The Queen's Medical Research InstituteUniversity of EdinburghEdinburghScotlandUK
| | - Randie R. Little
- Department of Pathology and Anatomical SciencesUniversity of MissouriColumbiaMissouriUSA
| | - Richard D. Leslie
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Paolo Pozzilli
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- Department of MedicineUnit of Endocrinology and Diabetes, Campus Bio‐Medico University of RomeRomeItaly
| | - Raffaela Buzzetti
- Experimental Medicine DepartmentSapienza University of RomeRomeItaly
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Epidemiological research on type 1 diabetes (T1D) has traditionally focussed on the paediatric age group, but recent data in adults has confirmed it to be a disease of all ages with a wide clinical spectrum. We review the epidemiology and clinical features of T1D across the lifespan. RECENT FINDINGS While the peak incidence of T1D is still in early adolescence, T1D is now diagnosed more commonly in adulthood than childhood due to increasing recognition of adult-onset T1D and the length of the adult lifespan. It still follows the known geographic variations in incidence, being highest in Northern Europe and lowest in Asia. The onset of T1D in adulthood is usually less acute than in childhood and confers a lower, although still substantial, risk of complications and early mortality. Interventions to delay T1D onset are emerging and screening for those at risk at birth is increasingly available. Type 1 diabetes can develop at any age and may not present with ketosis or an immediate insulin requirement in adults. Macro- and microvascular complications are the greatest cause of excess morbidity and mortality in this population.
Collapse
|
44
|
Lee VTY, Poynten A, Depczynski B. Continuous glucose monitoring to assess glucose variability in type 3c diabetes. Diabet Med 2022; 39:e14882. [PMID: 35569007 PMCID: PMC9545045 DOI: 10.1111/dme.14882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/12/2022] [Indexed: 11/28/2022]
Abstract
AIM The effectiveness of continuous glucose monitoring (CGM) in maintaining glycaemic control in type 1 diabetes mellitus and type 2 diabetes mellitus has been well demonstrated. However, the degree of glycaemic variability (GV) in people with type 3c diabetes mellitus has not been fully explored using CGM. This study aims to evaluate GV in type 3c diabetes mellitus participants and compare it to type 1 diabetes mellitus and type 2 diabetes mellitus. METHODS Participants were grouped according to type of diabetes. GV, defined as percentage coefficient of variation (%CV), and other glycaemic indices were obtained using CGM (FreeStyle Libre, Abbott, Australia) from 82 participants across all three cohorts over a 14-day period. Comparison of baseline characteristics and GV were performed across all groups. Correlation of GV with C-peptide values, and whether pancreatic supplementation had an effect on GV were also assessed in the type 3c diabetes mellitus cohort. RESULTS GV of type 3c diabetes mellitus participants was within the recommended target of less than %CV 36% (p = 0.004). Type 3c diabetes mellitus participants had the lowest GV among the three groups (p = 0.001). There was a trend for lower C-peptide levels to be associated with higher GV in type 3c diabetes mellitus participants (p = 0.22). Pancreatic enzyme supplementation in type 3c diabetes mellitus participants did not have an effect on GV (p = 0.664). CONCLUSIONS Although type 3c diabetes mellitus participants were the least variable, they had the highest mean glucose levels and estimated HbA1c , which suggests that the concept of 'brittle' diabetes in type 3c diabetes mellitus is not supported by the results of CGM in this study and may be leading to poorer glycaemic control.
Collapse
Affiliation(s)
- Victoria T. Y. Lee
- Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Ann Poynten
- Department of Endocrinology, Diabetes and MetabolismPrince of Wales HospitalSydneyNew South WalesAustralia
| | - Barbara Depczynski
- Department of Endocrinology, Diabetes and MetabolismPrince of Wales HospitalSydneyNew South WalesAustralia
| |
Collapse
|
45
|
Achenbach P, Hippich M, Zapardiel-Gonzalo J, Karges B, Holl RW, Petrera A, Bonifacio E, Ziegler AG. A classification and regression tree analysis identifies subgroups of childhood type 1 diabetes. EBioMedicine 2022; 82:104118. [PMID: 35803018 PMCID: PMC9270253 DOI: 10.1016/j.ebiom.2022.104118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 12/22/2022] Open
Abstract
Background Diabetes in childhood and adolescence includes autoimmune and non-autoimmune forms with heterogeneity in clinical and biochemical presentations. An unresolved question is whether there are subtypes, endotypes, or theratypes within these forms of diabetes. Methods The multivariable classification and regression tree (CART) analysis method was used to identify subgroups of diabetes with differing residual C-peptide levels in patients with newly diagnosed diabetes before 20 years of age (n=1192). The robustness of the model was assessed in a confirmation and prognosis cohort (n=2722). Findings The analysis selected age, haemoglobin A1c (HbA1c), and body mass index (BMI) as split parameters that classified patients into seven islet autoantibody-positive and three autoantibody-negative groups. There were substantial differences in genetics, inflammatory markers, diabetes family history, lipids, 25-OH-Vitamin D3, insulin treatment, insulin sensitivity and insulin autoimmunity among the groups, and the method stratified patients with potentially different pathogeneses and prognoses. Interferon-ɣ and/or tumour necrosis factor inflammatory signatures were enriched in the youngest islet autoantibody-positive groups and in patients with the lowest C-peptide values, while higher BMI and type 2 diabetes characteristics were found in older patients. The prognostic relevance was demonstrated by persistent differences in HbA1c at 7 years median follow-up. Interpretation This multivariable analysis revealed subgroups of young patients with diabetes that have potential pathogenetic and therapeutic relevance. Funding The work was supported by funds from the German Federal Ministry of Education and Research (01KX1818; FKZ 01GI0805; DZD e.V.), the Innovative Medicine Initiative 2 Joint Undertaking INNODIA (grant agreement No. 115797), the German Robert Koch Institute, and the German Diabetes Association.
Collapse
Affiliation(s)
- Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany; German Center for Diabetes Research (DZD), Munich, Germany; Technical University Munich, School of Medicine, Forschergruppe Diabetes at Klinikum rechts der Isar, Munich, Germany
| | - Markus Hippich
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany; German Center for Diabetes Research (DZD), Munich, Germany
| | - Jose Zapardiel-Gonzalo
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Beate Karges
- Division of Endocrinology and Diabetes, Medical Faculty, RWTH Aachen University, D 52074 Aachen, Germany
| | - Reinhard W Holl
- German Center for Diabetes Research (DZD), Munich, Germany; Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, D 89081 Ulm, Germany
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Zentrum Munich - German Research Center for Environmental Health, Neuherberg, Germany
| | - Ezio Bonifacio
- German Center for Diabetes Research (DZD), Munich, Germany; DFG Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany; German Center for Diabetes Research (DZD), Munich, Germany; Technical University Munich, School of Medicine, Forschergruppe Diabetes at Klinikum rechts der Isar, Munich, Germany.
| | | |
Collapse
|
46
|
de Leur K, Vollenbrock C, Dekker P, de Vries M, Birnie E, Mul D, Wolffenbuttel BHR, Groen J, Aanstoot H, Boesten L. How low is really low? Comparison of two C-peptide assays to establish residual C-peptide production in type 1 diabetes. Diabet Med 2022; 39:e14785. [PMID: 34989030 PMCID: PMC9303196 DOI: 10.1111/dme.14785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/04/2022] [Indexed: 01/12/2023]
Abstract
INTRODUCTION C-peptide is an important marker to assess residual insulin production in individuals with type 1 diabetes (T1D). The accuracy and detection limits of C-peptide assays are important to detect C-peptide microsecretion and to reliably observe changes over time in these people. We compared and verified two commercially available assays able to measure C-peptide in the picomolar range. METHODS The ultrasensitive Mercodia enzyme-linked immunosorbent C-peptide assay (ELISA) was compared with the Beckman immunoradiometric assay (IRMA) for C-peptide, assessing reproducibility (coefficient of variation [CV]), limit of blank (LoB), limit of detection (LoD) and limit of quantitation (LoQ). RESULTS For both assays within-run and between-run variation were high at the low (around the detection limit) C-peptide concentration range, with CVs of around 40%. LoB values for the ultrasensitive ELISA and the IRMA were 1.3 and 0.16 pmol/L respectively. LoD values were 2.4 and 0.54 pmol/L respectively. LoQ values were 9.7 and 3.8 pmol/L respectively. Only the IRMA met the specifications claimed by the manufacturer. CONCLUSIONS The IRMA provided the lowest threshold for quantification of serum C-peptide. LoQ of commercially available assays should be established in-house before applying them in research studies and clinical trials in which low C-peptide levels have clinical or scientific relevance.
Collapse
Affiliation(s)
- Kitty de Leur
- Department of Clinical ChemistryIJsselland HospitalCapelle aan den IJsselThe Netherlands
| | - Charlotte Vollenbrock
- Department of EndocrinologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Pim Dekker
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Martine de Vries
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Erwin Birnie
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
- Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Dick Mul
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Bruce H. R. Wolffenbuttel
- Department of EndocrinologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Joost Groen
- Department of Clinical ChemistryIJsselland HospitalCapelle aan den IJsselThe Netherlands
| | - Henk‐Jan Aanstoot
- Diabeter, Center for Pediatric and Adult Diabetes Care and ResearchRotterdamThe Netherlands
| | - Lianne Boesten
- Department of Clinical ChemistryIJsselland HospitalCapelle aan den IJsselThe Netherlands
| |
Collapse
|
47
|
Holdcraft RW, Graham MJ, Bemrose MA, Mutch LA, Martis PC, Janecek JL, Hall RD, Smith BH, Gazda LS. Long-term efficacy and safety of porcine islet macrobeads in nonimmunosuppressed diabetic cynomolgus macaques. Xenotransplantation 2022; 29:e12747. [PMID: 35384085 DOI: 10.1111/xen.12747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/05/2021] [Accepted: 03/10/2022] [Indexed: 11/26/2022]
Abstract
Although human islet transplantation has proven to provide clinical benefits, especially the near complete amelioration of hypoglycemia, the supply of human islets is limited and insufficient to meet the needs of all people that could benefit from islet transplantation. Porcine islets, secreting insulin nearly identical to that of human insulin, have been proposed as a viable supply of unlimited islets. Further, encapsulation of the porcine islets has been shown to reduce or eliminate the use of immunosuppressive therapy that would be required to prevent rejection of the foreign islet tissue. The goal of the current study was to determine the long-term safety and efficacy of agarose encapsulated porcine islets (macrobeads) in diabetic cynomolgus macaques, in a study emulating a proposed IND trial in which daily exogenous insulin therapy would be reduced by 50% with no loss of glucose regulation. Four of six animals implanted with macrobeads demonstrated ≥ 30% reduction in insulin requirements in year 1 of follow-up. Animals were followed for 2, 3.5, and 7.4 years with no serious adverse events, mortality or evidence of pathogen transmission. This study supports the continued pursuit of encapsulated porcine islet therapy as a promising treatment option for diabetes mellitus.
Collapse
Affiliation(s)
| | - Melanie J Graham
- Preclinical Research Center, University of Minnesota, St. Paul, Minnesota, USA
| | | | - Lucas A Mutch
- Preclinical Research Center, University of Minnesota, St. Paul, Minnesota, USA
| | | | - Jody L Janecek
- Preclinical Research Center, University of Minnesota, St. Paul, Minnesota, USA
| | | | | | | |
Collapse
|
48
|
Lal R, Leelarathna L. Insulin Delivery Hardware: Pumps and Pens. Diabetes Technol Ther 2022; 24:S21-S34. [PMID: 35475688 PMCID: PMC9206467 DOI: 10.1089/dia.2022.2502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Rayhan Lal
- Division of Endocrinology, Department of Medicine & Pediatrics, Stanford University School of Medicine, Stanford, CA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA
| | - Lalantha Leelarathna
- Manchester Diabetes Centre, Manchester University NHS Foundation Trust, Manchester, UK and Division of Diabetes, Endocrinology and Gastroenterology, University of Manchester, Manchester, UK
| |
Collapse
|
49
|
Utzschneider KM. Reduced β-cell function and risk of retinopathy: What's the connection? J Diabetes Complications 2022; 36:108045. [PMID: 34802901 DOI: 10.1016/j.jdiacomp.2021.108045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Kristina M Utzschneider
- Research and Development, Department of Medicine, VA Puget Sound Health Care System; Division of metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, United States of America.
| |
Collapse
|
50
|
Taylor GS, Shaw A, Scragg JH, Smith K, Campbell MD, McDonald TJ, Shaw JA, Ross MD, West DJ. Type 1 Diabetes Patients With Different Residual Beta-Cell Function but Similar Age, HBA1c, and Cardiorespiratory Fitness Have Differing Exercise-Induced Angiogenic Cell Mobilisation. Front Endocrinol (Lausanne) 2022; 13:797438. [PMID: 35222269 PMCID: PMC8874313 DOI: 10.3389/fendo.2022.797438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Many individuals with type 1 diabetes retain residual beta-cell function. Sustained endogenous insulin and C-peptide secretion is associated with reduced diabetes related complications, but underlying mechanisms remain unclear. Lower circulating numbers of endothelial and hematopoietic progenitor cells (EPCs and HPCs), and the inability to increase the count of these cells in response to exercise, are also associated with increased diabetes complications and cardiovascular disease. It is unknown whether residual beta-cell function influences HPCs and EPCs. Thus, this study examined the influence of residual beta-cell function in type 1 diabetes upon exercise-induced changes in haematopoietic (HPCs) and endothelial progenitor cells (EPCs). METHODS Participants with undetectable stimulated C-peptide (n=11; Cpepund), 10 high C-peptide (Cpephigh; >200 pmol/L), and 11 non-diabetes controls took part in this observational exercise study, completing 45 minutes of intensive walking at 60% V˙O2peak . Clinically significant HPCs (CD34+) and EPCs (CD34+VEGFR2+) phenotypes for predicting future adverse cardiovascular outcomes, and subsequent cell surface expression of chemokine receptor 4 (CXCR4) and 7 (CXCR7), were enumerated at rest and immediately post-exercise by flow cytometry. RESULTS Exercise increased HPCs and EPCs phenotypes similarly in the Cpephigh and control groups (+34-121% across phenotypes, p<0.04); but Cpepund group did not significantly increase from rest, even after controlling for diabetes duration. Strikingly, the post-exercise Cpepund counts were still lower than Cpephigh at rest. CONCLUSIONS Residual beta-cell function is associated with an intact exercise-induced HPCs and EPCs mobilisation. As key characteristics (age, fitness, HbA1c) were similar between groups, the mechanisms underpinning the absent mobilisation within those with negative C-peptide, and the vascular implications, require further investigation.
Collapse
Affiliation(s)
- Guy S. Taylor
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Daniel J. West, ; Guy S. Taylor,
| | - Andy Shaw
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jadine H. Scragg
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Kieran Smith
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Matthew D. Campbell
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, United Kingdom
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Timothy J. McDonald
- National Institute for Health Research Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
- Academic Department of Blood Sciences, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - James A. Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Newcastle Centre for Diabetes Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Mark D. Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Daniel J. West
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Daniel J. West, ; Guy S. Taylor,
| |
Collapse
|