1
|
Jiang Q, Hu Y, Ma JH. Various classification methods for diabetes mellitus in the management of blood glucose control. World J Diabetes 2025; 16:103316. [DOI: 10.4239/wjd.v16.i5.103316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 04/25/2025] Open
Abstract
In the era of precision medicine, the classification of diabetes mellitus has evolved beyond the traditional categories. Various classification methods now account for a multitude of factors, including variations in specific genes, type of β-cell impairment, degree of insulin resistance, and clinical characteristics of metabolic profiles. Improved classification methods enable healthcare providers to formulate blood glucose management strategies more precisely. Applying these updated classification systems, will assist clinicians in further optimising treatment plans, including targeted drug therapies, personalized dietary advice, and specific exercise plans. Ultimately, this will facilitate stricter blood glucose control, minimize the risks of hypoglycaemia and hyperglycaemia, and reduce long-term complications associated with diabetes.
Collapse
Affiliation(s)
- Qing Jiang
- Department of Endocrinology, The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| | - Yun Hu
- Department of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214000, Jiangsu Province, China
| | - Jian-Hua Ma
- Department of Endocrinology, Nanjing First Hospital, Nanjing 210000, Jiangsu Province, China
| |
Collapse
|
2
|
Wang Y, Chen H. Clinical application of cluster analysis in patients with newly diagnosed type 2 diabetes. Hormones (Athens) 2025; 24:109-122. [PMID: 39230795 DOI: 10.1007/s42000-024-00593-4] [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: 04/25/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024]
Abstract
AIMS Early prevention and treatment of type 2 diabetes mellitus (T2DM) is still a huge challenge for patients and clinicians. Recently, a novel cluster-based diabetes classification was proposed which may offer the possibility to solve this problem. In this study, we report our performance of cluster analysis of individuals newly diagnosed with T2DM, our exploration of each subtype's clinical characteristics and medication treatment, and the comparison carried out concerning the risk for diabetes complications and comorbidities among subtypes by adjusting for influencing factors. We hope to promote the further application of cluster analysis in individuals with early-stage T2DM. METHODS In this study, a k-means cluster algorithm was applied based on five indicators, namely, age, body mass index (BMI), glycosylated hemoglobin (HbA1c), homeostasis model assessment-2 insulin resistance (HOMA2-IR), and homeostasis model assessment-2 β-cell function (HOMA2-β), in order to perform the cluster analysis among 567 newly diagnosed participants with T2DM. The clinical characteristics and medication of each subtype were analyzed. The risk for diabetes complications and comorbidities in each subtype was compared by logistic regression analysis. RESULTS The 567 patients were clustered into four subtypes, as follows: severe insulin-deficient diabetes (SIDD, 24.46%), age-related diabetes (MARD, 30.86%), mild obesity-related diabetes (MOD, 25.57%), and severe insulin-resistant diabetes (SIRD, 20.11%). According to the results of the oral glucose tolerance test (OGTT) and biochemical indices, fasting blood glucose (FBG), 2-hour postprandial blood glucose (2hBG), HbA1c, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and triglyceride-glucose index (TyG) were higher in SIDD and SIRD than in MARD and MOD. MOD had the highest fasting C-peptide (FCP), 2-hour postprandial C-peptide (2hCP), fasting insulin (FINS), 2-hour postprandial insulin (2hINS), serum creatinine (SCr), and uric acid (UA), while SIRD had the highest triglycerides (TGs) and TyG-BMI. Albumin transaminase (ALT) and albumin transaminase (AST) were higher in MOD and SIRD. As concerms medications, compared to the other subtypes, SIDD had a lower rate of metformin use (39.1%) and a higher rate of α-glucosidase inhibitor (AGI, 61.7%) and insulin (74.4%) use. SIRD showed the highest frequency of use of sodium-glucose cotransporter-2 inhibitors (SGLT-2i, 36.0%) and glucagon-like peptide-1 receptor agonists (GLP-1RA, 19.3%). Concerning diabetic complications and comorbidities, the prevalence of diabetic kidney disease (DKD), cardiovascular disease (CVD), non-alcoholic fatty liver disease (NAFLD), dyslipidemia, and hypertension differed significantly among subtypes. Employing logistic regression analysis, after adjusting for unmodifiable (sex and age) and modifiable related influences (e.g., BMI, HbA1c, and smoking), it was found that SIRD had the highest risk of developing DKD (odds ratio, OR = 2.001, 95% confidence interval (CI): 1.125-3.559) and dyslipidemia (OR = 3.550, 95% CI: 1.534-8.215). MOD was more likely to suffer from NAFLD (OR = 3.301, 95%CI: 1.586-6.870). CONCLUSIONS Patients with newly diagnosed T2DM can be successfully clustered into four subtypes with different clinical characteristics, medication treatment, and risks for diabetes-related complications and comorbidities, the cluster-based diabetes classification possibly being beneficial both for prevention of secondary diabetes and for establishment of a theoretical basis for precision medicine.
Collapse
Affiliation(s)
- Yazhi Wang
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, 730000, China
- Department of Endocrinology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China
| | - Hui Chen
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, 730000, China.
- Department of Endocrinology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
3
|
Franks PW, Rich SS, Linder B, Zaghloul NA, Cefalu WT. A Research Roadmap to Address the Heterogeneity of Diabetes and Advance Precision Medicine. J Clin Endocrinol Metab 2025; 110:601-610. [PMID: 39657245 PMCID: PMC12063085 DOI: 10.1210/clinem/dgae844] [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: 09/12/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
The current classification of diabetes had its genesis over 85 years ago, when individuals with diabetes were first subclassified into insulin sensitive and insulin insensitive states based on the response to an oral glucose tolerance test. About 35 years later, the contemporary classifications of type 1 and type 2 diabetes were coined. Today's evidence, however, suggests that multiple etiologic and pathogenic processes lead to both type 1 and type 2 diabetes, reflecting significant heterogeneity in factors associated with initiation, progression, and clinical presentation of each disorder of glucose homeostasis. Further, the current classification fails to recognize what is currently defined as "atypical" diabetes. Heterogeneity of diabetes continues through the life-course of an individual, with modification of prognosis risk (eg, diabetic complications) altered by genetics, life experience, comorbidities, and therapy. Understanding the sources of heterogeneity in diabetes will likely improve diagnosis, prevention, treatment, and prediction of complications in both the medical and public health settings. Such knowledge will help inform progress in the emerging era of precision diabetes medicine. This article presents NIDDK's Heterogeneity of Diabetes Initiative and a corresponding roadmap for future research in type 2 diabetes heterogeneity.
Collapse
Affiliation(s)
- Paul W Franks
- Department of Clinical Sciences, Lund University, Helsingborg Hospital, Helsingborg 251 87, Sweden
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA 22908, USA
| | - Barbara Linder
- Division of Diabetes, Endocrinology & Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Norann A Zaghloul
- Division of Diabetes, Endocrinology & Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - William T Cefalu
- Division of Diabetes, Endocrinology & Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Obirikorang C, Adu EA, Afum-Adjei Awuah A, Darko SN, Ghartey FN, Ametepe S, Nyarko ENY, Anto EO, Owiredu WKBA. Differential risk of cardiovascular complications in patients with type-2 diabetes mellitus in Ghana: A hospital-based cross-sectional study. PLoS One 2025; 20:e0302912. [PMID: 39913381 PMCID: PMC11801548 DOI: 10.1371/journal.pone.0302912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/31/2024] [Indexed: 02/09/2025] Open
Abstract
AIM To characterize clinically relevant subgroups of patients with type-2 diabetes mellitus (T2DM) based on adiposity, insulin secretion, and resistance indices. METHODS A cross-sectional study was conducted at Eastern Regional Hospital in Ghana from July to October 2021 to investigate long-term patients with T2DM. To select participants, a systematic random sampling method was employed. Demographic data was collected using a structured questionnaire and fasting blood samples were taken to measure glycemic and lipid levels. Blood pressure and adiposity indices were measured during recruitment. The risk of cardiovascular disease (CVD) was defined using Framingham scores and standard low-density lipoprotein thresholds. To analyze the data, k-means clustering algorithms and regression analysis were used. RESULTS The study identified three groups in female patients according to body mass index, relative fat mass, glycated hemoglobin, and triglyceride-glucose index. These groups included the obesity-related phenotype, the severe insulin resistance phenotype, and the normal weight phenotype with improved insulin resistance. Among male patients with T2DM, two groups were identified, including the obesity-related phenotype with severe insulin resistance and the normal weight phenotype with improved insulin sensitivity. The severe insulin resistance phenotype in female patients was associated with an increased risk of high CVD (OR = 5.34, 95%CI:2.11-13.55) and metabolic syndrome (OR = 7.07; 95%CI:3.24-15.42). Among male patients, the obesity-related phenotype with severe insulin resistance was associated with an increased intermediate (OR = 21.78, 95%CI:4.17-113.78) and a high-risk CVD (OR = 6.84, 95%CI:1.45-32.12). CONCLUSIONS The findings highlight significant cardiometabolic heterogeneity among T2DM patients. The subgroups of T2DM patients characterized by obesity and/or severe insulin resistance with or without poor glycemic control, have increased risk of CVD. This underscores the importance of considering differences in adiposity, insulin secretion, and sensitivity indices when making clinical decisions for patients with T2DM.
Collapse
Affiliation(s)
- Christian Obirikorang
- Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Global Health and Infectious Disease, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Evans Asamoah Adu
- Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Global Health and Infectious Disease, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Anthony Afum-Adjei Awuah
- Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Global Health and Infectious Disease, Kumasi Centre for Collaborative Research in Tropical Medicine, Kumasi, Ghana
| | - Samuel Nkansah Darko
- Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Frank Naku Ghartey
- Department of Chemical Pathology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Samuel Ametepe
- Department of Molecular Medicine, School of Medical Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Medical Laboratory Sciences, Koforidua Technical University, Koforidua, Ghana
| | - Eric N. Y. Nyarko
- Department of Chemical Pathology, University of Ghana Medical School, University of Ghana, Accra, Ghana
| | - Enoch Odame Anto
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | | |
Collapse
|
5
|
Smeijer JD, Gomez MF, Rossing P, Heerspink HJL. The effect of the endothelin receptor antagonist atrasentan on insulin resistance in phenotypic clusters of patients with type 2 diabetes and chronic kidney disease. Diabetes Obes Metab 2025; 27:511-518. [PMID: 39503150 PMCID: PMC11701200 DOI: 10.1111/dom.16041] [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: 06/02/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 01/07/2025]
Abstract
AIMS Type 2 diabetes (T2D) patients with a clinical phenotype characterized by a high degree of insulin resistance are at increased risk of chronic kidney disease (CKD). We previously demonstrated that the endothelin receptor antagonist (ERA) atrasentan reduced insulin resistance in T2D. In this study, we compared the effect of atrasentan on insulin resistance across different phenotypic clusters of patients with T2D. MATERIALS AND METHODS We performed a post hoc analysis of the SONAR trial, a randomized, placebo-controlled trial of the ERA atrasentan in patients with T2D and CKD. Patients were stratified into four previously identified phenotypic clusters: severe insulin-deficient diabetes (SIDD), severe insulin-resistant diabetes (SIRD), mild obesity-related diabetes (MOD) and mild age-related diabetes (MARD). Changes in insulin resistance, assessed by HOMA-IR, were compared between the phenotypic clusters using a mixed effects model. RESULTS In total, 931 patients were included in the analysis. In the overall population, atrasentan compared to placebo reduced HOMA-IR by 12.9% [95%CI 3.5,21.4]. This effect of atrasentan was more pronounced in clusters characterized by insulin resistance or deficiency: (SIRD cluster 26.2% [95% CI 3.8,43.3] and SIDD cluster 18.5% [95%CI -3.8,35.9]), although the latter did not reach statistical significance. The effect of atrasentan compared to placebo was less pronounced in the other two clusters (MARD 12.2% [95% CI -1.7,24.12] and MOD -5.3% [95% CI -28.9,13.9]). CONCLUSIONS Atrasentan significantly improved insulin sensitivity in patients with T2D and CKD, especially in those characterized by high insulin resistance (SIRD cluster). Further studies are warranted to investigate the long-term clinical outcomes of atrasentan treatment in these distinct phenotypic clusters.
Collapse
Affiliation(s)
- Johannes David Smeijer
- Department of Clinical Pharmacy and Pharmacology, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Maria F. Gomez
- Lund University Diabetes Centre, Department of Clinical SciencesLund UniversityMalmöSweden
| | - Peter Rossing
- Steno Diabetes Center CopenhagenHerlevDenmark
- Department of Clinical Medicine University of CopenhagenCopenhagenDenmark
| | - Hiddo J. L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- George Institute for Global HealthSydneyNew South WalesAustralia
| |
Collapse
|
6
|
Kang M, Son K, Hwang YC, Lee S, Sang H, Kim S, Yon DK, Rhee SY, Lim H. Identification of Metabolic Patterns in Korean Patients With Type 2 Diabetes and Their Association With Diabetes-Related Complications. Diabetes 2025; 74:199-211. [PMID: 39546744 DOI: 10.2337/db23-0798] [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: 10/04/2023] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
ARTICLE HIGHLIGHTS Identifying patterns of metabolic heterogeneity in type 2 diabetes (T2D) can help in the development of optimal treatment strategies. We aimed to identify metabolic patterns in patients with T2D in the Republic of Korea and analyze the risk of developing diabetes-related complications according to patterns. We identified three distinct metabolic patterns and observed that each pattern was associated with a heightened risk of developing various cardiovascular diseases. These findings highlight the necessity of devising treatment strategies based on these patterns to prevent diabetes-related complications.
Collapse
Affiliation(s)
- Minji Kang
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin, Republic of Korea
- Research Institute of Medical Nutrition, Kyung Hee University, Seoul, Republic of Korea
| | - Kumhee Son
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin, Republic of Korea
- Research Institute of Medical Nutrition, Kyung Hee University, Seoul, Republic of Korea
| | - You-Cheol Hwang
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University Hospital at Gangdong,Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Sihoon Lee
- Laboratory of Genomics and Translational Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Hyunji Sang
- Department of Endocrinology and Metabolism, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Sunyoung Kim
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Family Medicine, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Dong Keon Yon
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Sang Youl Rhee
- Department of Endocrinology and Metabolism, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Republic of Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Hyunjung Lim
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin, Republic of Korea
- Research Institute of Medical Nutrition, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Fernandez-Camins B, Vlacho B, Canudas A, Ortega M, Granado-Casas M, Perera-LLuna A, Boluda-Sanson A, El-Khattabi-Ofkir Y, Franch-Nadal J, Mauricio D. Characterisation of type 2 diabetes subgroups at diagnosis: the COPERNICAN prospective observational cohort study protocol. BMJ Open 2024; 14:e083825. [PMID: 39675821 DOI: 10.1136/bmjopen-2023-083825] [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] [Indexed: 12/17/2024] Open
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2DM) is a highly heterogeneous and complex metabolic disease harbouring different metabolic characteristics. Adequate characterisation of subjects is essential to allow the implementation of precision medicine for the prevention, diagnosis, prognosis and treatment of this condition. METHODS AND ANALYSIS This prospective observational cohort study aims to identify and characterise relevant clinical clusters that are reproducibly associated with various clinical outcomes in T2DM in our Mediterranean region. The COPERNICAN study will include 1200 subjects with newly diagnosed T2DM from 28 primary care centres from the city of Barcelona and the healthcare district of Lleida in Catalonia (Spain). Participants will undergo a comprehensive phenotypic evaluation including, among others, six relevant variables: age, antibodies against glutamic acid decarboxylase, body mass index, glycated haemoglobin (HbA1c), indexes of insulin sensibility (HOMA2-IR) and secretion (HOMA2-beta). We will collect additional comprehensive data on glucose-lowering and other drug treatments, clinical evaluation (including complications), laboratory parameters, advanced lipoprotein profile, dietary habits and physical activity. The linkage with the population database will be done to perform a pragmatic follow-up of participants as part of their usual clinical care. A state-of-the-art cluster analysis (k-means and hierarchical clustering) will be performed. ETHICS AND DISSEMINATION The present study complies with all the ethical aspects and protection of participant subjects complying with all current local and European Union legislation. All Ethics Committees from the institutions involved in the study (IR Sant Pau Ethics Committee, Ethics Committee for Drug Research at IDIAP Jordi Gol and University Hospital of Bellvitge Ethics Committee for Research) approved this protocol. Confidentiality and anonymity of the data are ensured according to the current Spanish Organic Law 3/2018 of 05 December. TRIAL REGISTRATION NUMBER ClinicalTrials.gov. registration number NCT05333718, 27 January 2023.
Collapse
Affiliation(s)
- Berta Fernandez-Camins
- Department of Endocrinology and Nutrition, IR Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- DAP_CAT group, Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol, IDIAP Jordi Gol, Barcelona, Spain
| | - Bogdan Vlacho
- DAP_CAT group, Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol, IDIAP Jordi Gol, Barcelona, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Albert Canudas
- Escola de Doctorat. Departament de Medicina, Infermeria i ciències de la Salut, University of Lleida, Lleida, Spain
| | - Marta Ortega
- Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina, IDIAP Jordi Gol, Barcelona, Spain
| | - Minerva Granado-Casas
- DAP_CAT group, Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol, IDIAP Jordi Gol, Barcelona, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Barcelona, Spain
- Department of Nursing and Physiotherapy, University of Lleida, Lleida, Catalunya, Spain
- Health Care Research Group (GRECS), Lleida Institute of Biomedical Research Foundation Dr Pifarré, Lleida, Spain
| | - Alexandre Perera-LLuna
- Networking Biomedical Research Centre in the subject area of Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
- B2SLab, Departament d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Polytechnic University of Catalonia, Barcelona, Spain
| | - Alejandro Boluda-Sanson
- Department of Endocrinology and Nutrition, IR Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Yesmina El-Khattabi-Ofkir
- Health Care Research Group (GRECS), Lleida Institute of Biomedical Research Foundation Dr Pifarré, Lleida, Spain
| | - Josep Franch-Nadal
- Department of Medicine, University of Barcelona, Barcelona, Spain
- DAP_CAT group, Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol, IDIAP Jordi Gol, Barcelona, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Didac Mauricio
- Department of Endocrinology and Nutrition, IR Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- DAP_CAT group, Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol, IDIAP Jordi Gol, Barcelona, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Instituto de Salud Carlos III, Barcelona, Spain
- Faculty of Medicine, University of Vic - Central University of Catalonia, Vic, Spain
| |
Collapse
|
8
|
Herder C, Rizzo M, Roden M. Precision diabetology: Where do we stand now? J Diabetes Complications 2024; 38:108899. [PMID: 39477695 DOI: 10.1016/j.jdiacomp.2024.108899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024]
Affiliation(s)
- Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Manfredi Rizzo
- Unit of Diabetes and Cardiometabolic Prevention, University Hospital of Palermo, Palermo, Italy; School of Medicine, Department of Health Promotion, Mother and Child Care Internal Medicine and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
9
|
Bjarkø VV, Haug EB, Langhammer A, Ruiz PLD, Carlsson S, Birkeland KI, Berg TJ, Sørgjerd EP, Lyssenko V, Åsvold BO. Clinical utility of novel diabetes subgroups in predicting vascular complications and mortality: up to 25 years of follow-up of the HUNT Study. BMJ Open Diabetes Res Care 2024; 12:e004493. [PMID: 39577876 PMCID: PMC11590787 DOI: 10.1136/bmjdrc-2024-004493] [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: 07/25/2024] [Accepted: 09/30/2024] [Indexed: 11/24/2024] Open
Abstract
INTRODUCTION Cluster analysis has previously revealed five reproducible subgroups of diabetes, differing in risks of diabetic complications. We aimed to examine the clusters' predictive ability for vascular complications as compared with established risk factors in a general adult diabetes population. RESEARCH DESIGN AND METHODS Participants from the second (HUNT2, 1995-1997) and third (HUNT3, 2006-2008) surveys of the Norwegian population-based Trøndelag Health Study (HUNT Study) with adult-onset diabetes were included (n=1899). To identify diabetes subgroups, we used the same variables (age at diagnosis, body mass index, HbA1c, homeostasis model assessment estimates of beta cell function and insulin resistance, and glutamic acid decarboxylase antibodies) and the same data-driven clustering technique as in previous studies. We used Cox proportional hazards models to investigate associations between clusters and risks of vascular complications and mortality. We estimated the C-index and R2 to compare predictive abilities of the clusters to those of established risk factors as continuous variables. All models included adjustment for age, sex, diabetes duration and time of inclusion. RESULTS We reproduced five subgroups with similar key characteristics as identified in previous studies. During median follow-up of 9-13 years (differing between outcomes), the clusters were associated with different risks of vascular complications and all-cause mortality. However, in prediction models, individual established risk factors were at least as good predictors as cluster assignment for all outcomes. For example, for retinopathy, the C-index for the model including clusters (0.65 (95% CI 0.63 to 0.68)) was similar to that of HbA1c (0.65 (95% CI 0.63 to 0.68)) or fasting C-peptide (0.66 (95% CI 0.63 to 0.68)) alone. For chronic kidney disease, the C-index for clusters (0.74 (95% CI 0.72 to 0.76)) was similar to that of triglyceride/high-density lipoprotein ratio (0.74 (95% CI 0.71 to 0.76)) or fasting C-peptide (0.74 (95% CI 0.72 to 0.76)), and baseline estimated glomerular filtration rate yielded a C-index of 0.76 (95% CI 0.74 to 0.78). CONCLUSIONS Cluster assignment did not provide better prediction of vascular complications or all-cause mortality compared with established risk factors.
Collapse
Affiliation(s)
- Vera Vik Bjarkø
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St Olavs Hospital Trondheim University Hospital, Trondheim, Norway
| | - Eirin Beate Haug
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Arnulf Langhammer
- HUNT Research Center, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | | | - Sofia Carlsson
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Kare I Birkeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tore Julsrud Berg
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Elin Pettersen Sørgjerd
- HUNT Research Center, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Valeriya Lyssenko
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, Bergen, Norway
| | - Bjørn Olav Åsvold
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St Olavs Hospital Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
10
|
Lay AC, Tran VDT, Nair V, Betin V, Hurcombe JA, Barrington AF, Pope RJ, Burdet F, Mehl F, Kryvokhyzha D, Ahmad A, Sinton MC, Lewis P, Wilson MC, Menon R, Otto E, Heesom KJ, Ibberson M, Looker HC, Nelson RG, Ju W, Kretzler M, Satchell SC, Gomez MF, Coward RJM. Profiling of insulin-resistant kidney models and human biopsies reveals common and cell-type-specific mechanisms underpinning Diabetic Kidney Disease. Nat Commun 2024; 15:10018. [PMID: 39562547 PMCID: PMC11576882 DOI: 10.1038/s41467-024-54089-1] [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] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end stage kidney failure worldwide, of which cellular insulin resistance is a major driver. Here, we study key human kidney cell types implicated in DKD (podocytes, glomerular endothelial, mesangial and proximal tubular cells) in insulin sensitive and resistant conditions, and perform simultaneous transcriptomics and proteomics for integrated analysis. Our data is further compared with bulk- and single-cell transcriptomic kidney biopsy data from early- and advanced-stage DKD patient cohorts. We identify several consistent changes (individual genes, proteins, and molecular pathways) occurring across all insulin-resistant kidney cell types, together with cell-line-specific changes occurring in response to insulin resistance, which are replicated in DKD biopsies. This study provides a rich data resource to direct future studies in elucidating underlying kidney signalling pathways and potential therapeutic targets in DKD.
Collapse
Affiliation(s)
- Abigail C Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Van Du T Tran
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Virginie Betin
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | | | | | - Robert Jp Pope
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Frédéric Burdet
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Florence Mehl
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Dmytro Kryvokhyzha
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Abrar Ahmad
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Matthew C Sinton
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Philip Lewis
- Proteomics Facility, University of Bristol, Bristol, UK
| | | | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Edgar Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kate J Heesom
- Proteomics Facility, University of Bristol, Bristol, UK
| | - Mark Ibberson
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Helen C Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Phoenix, AZ, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Phoenix, AZ, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Simon C Satchell
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Maria F Gomez
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Richard J M Coward
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
11
|
Wang J, Gao B, Wang J, Liu W, Yuan W, Chai Y, Ma J, Ma Y, Kong G, Liu M. Identifying subtypes of type 2 diabetes mellitus based on real-world electronic medical record data in China. Diabetes Res Clin Pract 2024; 217:111872. [PMID: 39332534 DOI: 10.1016/j.diabres.2024.111872] [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: 06/21/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
AIMS To replicate the European subtypes of type 2 diabetes mellitus (T2DM) in the Chinese diabetes population and investigate the risk of complications in different subtypes. METHODS A diabetes cohort using real-world patient data was constructed, and clustering was employed to subgroup the T2DM patients. Kaplan-Meier analysis and the Cox models were used to analyze the association between diabetes subtypes and the risk of complications. RESULTS A total of 2,652 T2DM patients with complete clustering data were extracted. Among them, 466 (17.57 %) were classified as severe insulin-deficient diabetes (SIDD), 502 (18.93 %) as severe insulin-resistant diabetes (SIRD), 672 (25.34 %) as mild obesity-related diabetes (MOD), and 1,012 (38.16 %) as mild age-related diabetes (MARD). The risk of chronic kidney disease (CKD) and diabetic retinopathy (DR) were different in the four subtypes. Compared with MARD, SIRD had a higher risk of CKD (HR 2.40 [1.16, 4.96]), and SIDD had a higher risk of DR (HR 2.16 [1.11, 4.20]). The risk of stroke and coronary events had no difference. CONCLUSIONS The European T2DM subtypes can be replicated in the Chinese diabetes population. The risk of CKD and DR varied among different subtypes, indicating that proper interventions can be taken to prevent specific complications in different subtypes.
Collapse
Affiliation(s)
- Jiayu Wang
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Bixia Gao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Jinwei Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Wenwen Liu
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Weijia Yuan
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Yangfan Chai
- Peking University Chongqing Research Institute of Big Data, Chongqing 100871, China
| | - Jun Ma
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Yangyang Ma
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Guilan Kong
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China.
| | - Minchao Liu
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
12
|
Capoccia D, Leonetti F, Natali A, Tricò D, Perrini S, Sbraccia P, Guglielmi V. Remission of type 2 diabetes: position statement of the Italian society of diabetes (SID). Acta Diabetol 2024; 61:1309-1326. [PMID: 38942960 PMCID: PMC11486812 DOI: 10.1007/s00592-024-02317-x] [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: 05/09/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
The primary cause of the pandemic scale of type 2 diabetes (T2D) is the excessive and/or abnormal accumulation of adiposity resulting from a chronic positive energy balance. Any form of weight loss dramatically affects the natural history of T2D, favoring prevention, treatment, and even remission in the case of significant weight loss. However, weight regain, which is often accompanied by the recurrence or worsening of obesity complications such as T2D, is an inevitable biological phenomenon that is an integral part of the pathophysiology of obesity. This can occur not only after weight loss, but also during obesity treatment if it is not effective enough to counteract the physiological responses aimed at restoring adiposity to its pre-weight-loss equilibrium state. Over the past few years, many controlled and randomized studies have suggested a superior efficacy of bariatric surgery compared to conventional therapy in terms of weight loss, glycemic control, and rates of T2D remission. Recently, the therapeutic armamentarium in the field of diabetology has been enriched with new antihyperglycemic drugs with considerable efficacy in reducing body weight, which could play a pathogenetic role in the remission of T2D, not through the classical incretin effect, but by improving adipose tissue functions. All these concepts are discussed in this position statement, which aims to deepen the pathogenetic links between obesity and T2D, shift the paradigm from a "simple" interaction between insulin resistance and insulin deficiency, and evaluate the efficacy of different therapeutic interventions to improve T2D management and induce diabetes remission whenever still possible.
Collapse
Affiliation(s)
- Danila Capoccia
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Frida Leonetti
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sebastio Perrini
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, Unit of Internal Medicine - Obesity Center, Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, Unit of Internal Medicine - Obesity Center, Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
13
|
Landgraf W, Owens DR, Frier BM, Bolli GB. Responses to Basal Insulin Glargine (300 U/mL and 100 U/mL) with or Without Pre-prandial Insulin in Pre-treated Subphenotypes of Type 2 Diabetes: Insights from a Post Hoc Analysis. Diabetes Ther 2024; 15:1769-1784. [PMID: 38879736 PMCID: PMC11263304 DOI: 10.1007/s13300-024-01608-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/03/2024] [Indexed: 07/24/2024] Open
Abstract
INTRODUCTION This study aimed to evaluate glycemic outcomes in subphenotypes of type 2 diabetes (T2D) with HbA1c > 7.0%, previously on basal insulin (pre-BI) alone (≥ 42 U/day) or on basal-bolus therapy (pre-BB), and who were switched to either basal insulin glargine 300 U/mL (IGlar-300) or 100 U/mL (IGlar-100), with or without pre-prandial insulin. METHODS Participants from EDITION 2 (pre-BI, n = 785), and EDITION 1 (pre-BB, n = 792) trials were assigned retrospectively to subphenotypes of T2D: severe insulin deficient diabetes (SIDD), mild age-related diabetes (MARD), mild obesity diabetes (MOD), and severe insulin resistant diabetes (SIRD). Key efficacy and safety parameters were analyzed at baseline, and after 26 weeks, for IGlar-300 and IGlar-100 pooled groups according to subphenotypes. Outcomes were also compared with insulin-naïve subphenotypes on oral antihyperglycemic drugs (OADs) from the EDITION 3 trial (pre-OAD, n = 858). RESULTS Pre-BI and pre-BB treated subphenotypes with SIDD had a higher mean HbA1c (8.9% and 9.1%) at baseline compared to those of MARD (7.7% and 7.8%) and MOD (8.1% and 8.2%) and after 26 weeks remained above target HbA1c (7.7% and 8.0%) despite mean glargine doses of 0.7 to 1.0 U/kg/day and pre-prandial insulin use in the pre-BB SIDD subgroup. Pre-BB treated individuals with MARD and MOD achieved lower HbA1c levels (6.9% and 7.2%) than the pre-BI groups (7.3% and 7.5%) despite similar mean FPG levels (123-130 mg/dL). Only 19-22% of participants with SIDD achieved HbA1c < 7.0% compared to 33-51% with MARD and MOD, respectively. Pre-BI and pre-BB treated subphenotypes experienced more hypoglycemia than pre-OAD treated subphenotypes. CONCLUSION Individuals with T2D assigned post hoc to the SIDD subphenotype achieved suboptimal glycemic control with glargine regimens including basal-bolus therapy, alerting clinicians to improve further diabetes treatment, particularly post-prandial glycemic control, in individuals with SIDD.
Collapse
Affiliation(s)
- Wolfgang Landgraf
- Medical Department, Diabetes Franchise General Medicines, Sanofi, Frankfurt, Germany.
- Sanofi-Aventis Deutschland GmbH, c/o Oskar Helene Park 33, 14195, Berlin, Germany.
| | - David R Owens
- Diabetes Research Group Cymru, College of Medicine, Swansea University, Swansea, UK
| | - Brian M Frier
- The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Geremia B Bolli
- Department of Medicine & Surgery, University of Perugia School of Medicine, Perugia, Italy
| |
Collapse
|
14
|
Kurgan N, Kjærgaard Larsen J, Deshmukh AS. Harnessing the power of proteomics in precision diabetes medicine. Diabetologia 2024; 67:783-797. [PMID: 38345659 DOI: 10.1007/s00125-024-06097-5] [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: 11/14/2023] [Accepted: 12/20/2023] [Indexed: 03/21/2024]
Abstract
Precision diabetes medicine (PDM) aims to reduce errors in prevention programmes, diagnosis thresholds, prognosis prediction and treatment strategies. However, its advancement and implementation are difficult due to the heterogeneity of complex molecular processes and environmental exposures that influence an individual's disease trajectory. To address this challenge, it is imperative to develop robust screening methods for all areas of PDM. Innovative proteomic technologies, alongside genomics, have proven effective in precision cancer medicine and are showing promise in diabetes research for potential translation. This narrative review highlights how proteomics is well-positioned to help improve PDM. Specifically, a critical assessment of widely adopted affinity-based proteomic technologies in large-scale clinical studies and evidence of the benefits and feasibility of using MS-based plasma proteomics is presented. We also present a case for the use of proteomics to identify predictive protein panels for type 2 diabetes subtyping and the development of clinical prediction models for prevention, diagnosis, prognosis and treatment strategies. Lastly, we discuss the importance of plasma and tissue proteomics and its integration with genomics (proteogenomics) for identifying unique type 2 diabetes intra- and inter-subtype aetiology. We conclude with a call for action formed on advancing proteomics technologies, benchmarking their performance and standardisation across sites, with an emphasis on data sharing and the inclusion of diverse ancestries in large cohort studies. These efforts should foster collaboration with key stakeholders and align with ongoing academic programmes such as the Precision Medicine in Diabetes Initiative consortium.
Collapse
Affiliation(s)
- Nigel Kurgan
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe Kjærgaard Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
15
|
Landgraf W, Owens DR, Frier BM, Bolli GB. Treatment responses to basal insulin glargine 300 U/ml and glargine 100 U/ml in newly defined subphenotypes of type 2 diabetes: A post hoc analysis of the EDITION 3 randomized clinical trial. Diabetes Obes Metab 2024; 26:503-511. [PMID: 37860918 DOI: 10.1111/dom.15336] [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: 08/01/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION To compare responses to basal insulin glargine 300 U/ml (IGlar-300) and 100 U/ml (IGlar-100) in newly defined subphenotypes of type 2 diabetes. METHODS Insulin-naive participants (n = 858) from the EDITION 3 trial were assigned to subphenotypes 'Mild Age-Related Diabetes (MARD)', 'Mild Obesity Diabetes (MOD)', 'Severe Insulin Resistant Diabetes (SIRD)' and 'Severe Insulin Deficient Diabetes (SIDD)'. Key variables were analysed at baseline and 26 weeks. RESULTS Participants were comprised of MOD 56.1% (n = 481), SIDD 22.1% (n = 190), MARD 18.2% (n = 156) and SIRD 3.0% (n = 26). After 26 weeks a similar decrease in glycated haemoglobin (HbA1c) and fasting plasma glucose (FPG) of 16-19 mmol/mol and 1.4-1.7 mmol/L, respectively, occurred in MARD and MOD with both insulins. SIDD had the most elevated HbA1c and FPG (80-83 mmol/mol/11.1-11.4 mmol/L) and reduction in both HbA1c and FPG was greater with IGlar-100 than with IGlar-300 (-18 vs. -15 mmol/mol and -1.6 vs. -1.3 mmol/L, respectively; each p = .03). In SIDD, despite receiving the highest basal insulin doses, HbA1c decline (57-60 mmol/mol/7.3-7.6%) was suboptimal at week 26. In MOD and SIDD lower incidences with IGlar-300 were found for level 1 nocturnal hypoglycaemia [odds ratio (OR) 0.59, 95% confidence intervals (CI) 0.36-0.97; OR 0.49, 95% CI 0.24-0.99]. In addition, fewer level 2 hypoglycaemia episodes occurred at any time with IGlar-300 in SIDD (OR 0.31, 95% CI 0.13-0.77). CONCLUSION Both insulins produce comparable outcomes in type 2 diabetes subphenotypes, but in SIDD, add-on treatment to basal insulin is required to achieve glycaemic targets.
Collapse
Affiliation(s)
- Wolfgang Landgraf
- Medical Department, Diabetes Franchise General Medicines, Sanofi, Paris, France
| | - David R Owens
- Diabetes Research Group Cymru, College of Medicine, Swansea University, Swansea, UK
| | - Brian M Frier
- The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Geremia B Bolli
- Section of Endocrinology and Metabolism, Department of Medicine, University of Perugia School of Medicine, Perugia, Italy
| |
Collapse
|
16
|
Schön M, Prystupa K, Mori T, Zaharia OP, Bódis K, Bombrich M, Möser C, Yurchenko I, Kupriyanova Y, Strassburger K, Bobrov P, Nair ATN, Bönhof GJ, Strom A, Delgado GE, Kaya S, Guthoff R, Stefan N, Birkenfeld AL, Hauner H, Seissler J, Pfeiffer A, Blüher M, Bornstein S, Szendroedi J, Meyhöfer S, Trenkamp S, Burkart V, Schrauwen-Hinderling VB, Kleber ME, Niessner A, Herder C, Kuss O, März W, Pearson ER, Roden M, Wagner R. Analysis of type 2 diabetes heterogeneity with a tree-like representation: insights from the prospective German Diabetes Study and the LURIC cohort. Lancet Diabetes Endocrinol 2024; 12:119-131. [PMID: 38142707 DOI: 10.1016/s2213-8587(23)00329-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Heterogeneity in type 2 diabetes can be represented by a tree-like graph structure by use of reversed graph-embedded dimensionality reduction. We aimed to examine whether this approach can be used to stratify key pathophysiological components and diabetes-related complications during longitudinal follow-up of individuals with recent-onset type 2 diabetes. METHODS For this cohort analysis, 927 participants aged 18-69 years from the German Diabetes Study (GDS) with recent-onset type 2 diabetes were mapped onto a previously developed two-dimensional tree based on nine simple clinical and laboratory variables, residualised for age and sex. Insulin sensitivity was assessed by a hyperinsulinaemic-euglycaemic clamp, insulin secretion was assessed by intravenous glucose tolerance test, hepatic lipid content was assessed by 1 H magnetic resonance spectroscopy, serum interleukin (IL)-6 and IL-18 were assessed by ELISA, and peripheral and autonomic neuropathy were assessed by functional and clinical measures. Participants were followed up for up to 16 years. We also investigated heart failure and all-cause mortality in 794 individuals with type 2 diabetes undergoing invasive coronary diagnostics from the Ludwigshafen Risk and Cardiovascular Health (LURIC) cohort. FINDINGS There were gradients of clamp-measured insulin sensitivity (both dimensions: p<0·0001) and insulin secretion (pdim1<0·0001, pdim2=0·00097) across the tree. Individuals in the region with the lowest insulin sensitivity had the highest hepatic lipid content (n=205, pdim1<0·0001, pdim2=0·037), pro-inflammatory biomarkers (IL-6: n=348, pdim1<0·0001, pdim2=0·013; IL-18: n=350, pdim1<0·0001, pdim2=0·38), and elevated cardiovascular risk (nevents=143, pdim1=0·14, pdim2<0·00081), whereas individuals positioned in the branch with the lowest insulin secretion were more prone to require insulin therapy (nevents=85, pdim1=0·032, pdim2=0·12) and had the highest risk of diabetic sensorimotor polyneuropathy (nevents=184, pdim1=0·012, pdim2=0·044) and cardiac autonomic neuropathy (nevents=118, pdim1=0·0094, pdim2=0·06). In the LURIC cohort, all-cause mortality was highest in the tree branch showing insulin resistance (nevents=488, pdim1=0·12, pdim2=0·0032). Significant gradients differentiated individuals having heart failure with preserved ejection fraction from those who had heart failure with reduced ejection fraction. INTERPRETATION These data define the pathophysiological underpinnings of the tree structure, which has the potential to stratify diabetes-related complications on the basis of routinely available variables and thereby expand the toolbox of precision diabetes diagnosis. FUNDING German Diabetes Center, German Federal Ministry of Health, Ministry of Culture and Science of the state of North Rhine-Westphalia, German Federal Ministry of Education and Research, German Diabetes Association, German Center for Diabetes Research, European Community, German Research Foundation, and Schmutzler Stiftung.
Collapse
Affiliation(s)
- Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katsiaryna Prystupa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Tim Mori
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Oana P Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maria Bombrich
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Clara Möser
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Iryna Yurchenko
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Pavel Bobrov
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Anand T N Nair
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Gidon J Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Graciela E Delgado
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Center for Preventive Medicine and Digital Health Baden-Württemberg, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sema Kaya
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Rainer Guthoff
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | - Hans Hauner
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, München, Germany
| | - Jochen Seissler
- Diabetes Research Group, Medical Department 4, Ludwig-Maximilians University Munich, München, Germany
| | - Andreas Pfeiffer
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Matthias Blüher
- Department of Medicine, Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Stefan Bornstein
- Department of Internal Medicine III, Dresden University of Technology, Dresden, Germany
| | - Julia Szendroedi
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
| | - Svenja Meyhöfer
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany; Department of Internal Medicine 1, Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany
| | - Sandra Trenkamp
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Vera B Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Marcus E Kleber
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Alexander Niessner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Austria
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Oliver Kuss
- German Center for Diabetes Research, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; Centre for Health and Society, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Winfried März
- 5th Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
| | - Ewan R Pearson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research, München-Neuherberg, Germany; Division of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
17
|
Dwibedi C, Ekström O, Brandt J, Adiels M, Franzén S, Abrahamsson B, Rosengren AH. Randomized open-label trial of semaglutide and dapagliflozin in patients with type 2 diabetes of different pathophysiology. Nat Metab 2024; 6:50-60. [PMID: 38177805 PMCID: PMC10822775 DOI: 10.1038/s42255-023-00943-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/08/2023] [Indexed: 01/06/2024]
Abstract
The limited understanding of the heterogeneity in the treatment response to antidiabetic drugs contributes to metabolic deterioration and cardiovascular complications1,2, stressing the need for more personalized treatment1. Although recent attempts have been made to classify diabetes into subgroups, the utility of such stratification in predicting treatment response is unknown3. We enrolled participants with type 2 diabetes (n = 239, 74 women and 165 men) and features of severe insulin-deficient diabetes (SIDD) or severe insulin-resistant diabetes (SIRD). Participants were randomly assigned to treatment with the glucagon-like peptide 1 receptor agonist semaglutide or the sodium-glucose cotransporter 2 inhibitor dapagliflozin for 6 months (open label). The primary endpoint was the change in glycated haemoglobin (HbA1c). Semaglutide induced a larger reduction in HbA1c levels than dapagliflozin (mean difference, 8.2 mmol mol-1; 95% confidence interval, -10.0 to -6.3 mmol mol-1), with a pronounced effect in those with SIDD. No difference in adverse events was observed between participants with SIDD and those with SIRD. Analysis of secondary endpoints showed greater reductions in fasting and postprandial glucose concentrations in response to semaglutide in participants with SIDD than in those with SIRD and a more pronounced effect on postprandial glucose by dapagliflozin in participants with SIDD than in those with SIRD. However, no significant interaction was found between drug assignment and the SIDD or SIRD subgroup. In contrast, continuous measures of body mass index, blood pressure, insulin secretion and insulin resistance were useful in identifying those likely to have the largest improvements in glycaemic control and cardiovascular risk factors by adding semaglutide or dapagliflozin. Thus, systematic evaluation of continuous pathophysiological variables can guide the prediction of the treatment response to these drugs and provide more information than stratified subgroups ( NCT04451837 ).
Collapse
Affiliation(s)
- Chinmay Dwibedi
- Department of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ola Ekström
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Jasmine Brandt
- Department of Clinical Chemistry and Pharmacology, Skåne University Hospital, Lund, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Martin Adiels
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Stefan Franzén
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- AstraZeneca, Gothenburg, Sweden
| | - Birgitta Abrahamsson
- Department of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anders H Rosengren
- Department of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
18
|
Franks PW, Cefalu WT, Dennis J, Florez JC, Mathieu C, Morton RW, Ridderstråle M, Sillesen HH, Stehouwer CDA. Precision medicine for cardiometabolic disease: a framework for clinical translation. Lancet Diabetes Endocrinol 2023; 11:822-835. [PMID: 37804856 DOI: 10.1016/s2213-8587(23)00165-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic disease is a major threat to global health. Precision medicine has great potential to help to reduce the burden of this common and complex disease cluster, and to enhance contemporary evidence-based medicine. Its key pillars are diagnostics; prediction (of the primary disease); prevention (of the primary disease); prognosis (prediction of complications of the primary disease); treatment (of the primary disease or its complications); and monitoring (of risk exposure, treatment response, and disease progression or remission). To contextualise precision medicine in both research and clinical settings, and to encourage the successful translation of discovery science into clinical practice, in this Series paper we outline a model (the EPPOS model) that builds on contemporary evidence-based approaches; includes precision medicine that improves disease-related predictions by stratifying a cohort into subgroups of similar characteristics, or using participants' characteristics to model treatment outcomes directly; includes personalised medicine with the use of a person's data to objectively gauge the efficacy, safety, and tolerability of therapeutics; and subjectively tailors medical decisions to the individual's preferences, circumstances, and capabilities. Precision medicine requires a well functioning system comprised of multiple stakeholders, including health-care recipients, health-care providers, scientists, health economists, funders, innovators of medicines and technologies, regulators, and policy makers. Powerful computing infrastructures supporting appropriate analysis of large-scale, well curated, and accessible health databases that contain high-quality, multidimensional, time-series data will be required; so too will prospective cohort studies in diverse populations designed to generate novel hypotheses, and clinical trials designed to test them. Here, we carefully consider these topics and describe a framework for the integration of precision medicine in cardiometabolic disease.
Collapse
Affiliation(s)
- Paul W Franks
- Department of Translational Medicine, Medical Science, Novo Nordisk Foundation, Hellerup, Denmark; Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, 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.
| | - William T Cefalu
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John Dennis
- Institute of Biomedical and Clinical Science, Royal Devon and Exeter Hospital, University of Exeter, Exeter, UK
| | - Jose C Florez
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Robert W Morton
- Department of Translational Medicine, Medical Science, Novo Nordisk Foundation, Hellerup, Denmark
| | | | - Henrik H Sillesen
- Department of Clinical Medicine, Medical Science, Novo Nordisk Foundation, Hellerup, Denmark
| | - Coen D A Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands; Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
19
|
Misra S, Aguilar-Salinas CA, Chikowore T, Konradsen F, Ma RCW, Mbau L, Mohan V, Morton RW, Nyirenda MJ, Tapela N, Franks PW. The case for precision medicine in the prevention, diagnosis, and treatment of cardiometabolic diseases in low-income and middle-income countries. Lancet Diabetes Endocrinol 2023; 11:836-847. [PMID: 37804857 DOI: 10.1016/s2213-8587(23)00164-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/08/2023] [Accepted: 06/01/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic diseases are the leading preventable causes of death in most geographies. The causes, clinical presentations, and pathogenesis of cardiometabolic diseases vary greatly worldwide, as do the resources and strategies needed to prevent and treat them. Therefore, there is no single solution and health care should be optimised, if not to the individual (ie, personalised health care), then at least to population subgroups (ie, precision medicine). This optimisation should involve tailoring health care to individual disease characteristics according to ethnicity, biology, behaviour, environment, and subjective person-level characteristics. The capacity and availability of local resources and infrastructures should also be considered. Evidence needed for equitable precision medicine cannot be generated without adequate data from all target populations, and the idea that research done in high-income countries will transfer adequately to low-income and middle-income countries (LMICs) is problematic, as many migration studies and transethnic comparisons have shown. However, most data for precision medicine research are derived from people of European ancestry living in high-income countries. In this Series paper, we discuss the case for precision medicine for cardiometabolic diseases in LMICs, the barriers and enablers, and key considerations for implementation. We focus on three propositions: first, failure to explore and implement precision medicine for cardiometabolic disease in LMICs will enhance global health disparities. Second, some LMICs might already be placed to implement cardiometabolic precision medicine under appropriate circumstances, owing to progress made in treating infectious diseases. Third, improvements in population health from precision medicine are most probably asymptotic; the greatest gains are more likely to be obtained in countries where health-care systems are less developed. We outline key recommendations for implementation of precision medicine approaches in LMICs.
Collapse
Affiliation(s)
- Shivani Misra
- Division of Metabolism, Digestion and Reproduction, Imperial College London, London, UK; Department of Diabetes and Endocrinology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Carlos A Aguilar-Salinas
- Dirección de Nutricion, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| | - Tinashe Chikowore
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Flemming Konradsen
- Novo Nordisk Foundation, Copenhagen, Denmark; Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, 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 Special Administrative Region, China
| | | | - Viswanathan Mohan
- Madras Diabetes Research Foundation, ICMR Centre for Advanced Research in Diabetes, Chennai, India; Dr Mohan's Diabetes Specialties Centre, IDF Centre of Excellence in Diabetes Care, Chennai, India
| | | | - Moffat J Nyirenda
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; London School of Hygiene and Tropical Medicine, London, UK
| | - Neo Tapela
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana; International Consortium for Health Outcomes Measurement, Oxford, UK
| | - Paul W Franks
- Novo Nordisk Foundation, Copenhagen, Denmark; Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmö, 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.
| |
Collapse
|
20
|
Sormunen-Harju H, Huvinen E, Girchenko PV, Kajantie E, Villa PM, Hämäläinen EK, Lahti-Pulkkinen M, Laivuori H, Räikkönen K, Koivusalo SB. Metabolomic Profiles of Nonobese and Obese Women With Gestational Diabetes. J Clin Endocrinol Metab 2023; 108:2862-2870. [PMID: 37220084 PMCID: PMC10584006 DOI: 10.1210/clinem/dgad288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 05/25/2023]
Abstract
CONTEXT In non-pregnant population, nonobese individuals with obesity-related metabolome have increased risk for type 2 diabetes and cardiovascular diseases. The risk of these diseases is also increased after gestational diabetes. OBJECTIVE This work aimed to examine whether nonobese (body mass index [BMI] < 30) and obese (BMI ≥ 30) women with gestational diabetes mellitus (GDM) and obese non-GDM women differ in metabolomic profiles from nonobese non-GDM controls. METHODS Levels of 66 metabolic measures were assessed in early (median 13, IQR 12.4-13.7 gestation weeks), and across early, mid (20, 19.3-23.0), and late (28, 27.0-35.0) pregnancy blood samples in 755 pregnant women from the PREDO and RADIEL studies. The independent replication cohort comprised 490 pregnant women. RESULTS Nonobese and obese GDM, and obese non-GDM women differed similarly from the controls across early, mid, and late pregnancy in 13 measures, including very low-density lipoprotein-related measures, and fatty acids. In 6 measures, including fatty acid (FA) ratios, glycolysis-related measures, valine, and 3-hydroxybutyrate, the differences between obese GDM women and controls were more pronounced than the differences between nonobese GDM or obese non-GDM women and controls. In 16 measures, including HDL-related measures, FA ratios, amino acids, and inflammation, differences between obese GDM or obese non-GDM women and controls were more pronounced than the differences between nonobese GDM women and controls. Most differences were evident in early pregnancy, and in the replication cohort were more often in the same direction than would be expected by chance alone. CONCLUSION Differences between nonobese and obese GDM, or obese non-GDM women and controls in metabolomic profiles may allow detection of high-risk women for timely targeted preventive interventions.
Collapse
Affiliation(s)
- Heidi Sormunen-Harju
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, FI-00270 Helsinki, Finland
| | - Emilia Huvinen
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, FI-00270 Helsinki, Finland
| | - Polina V Girchenko
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, FI-00270 Helsinki, Finland
| | - Eero Kajantie
- Clinical Medicine Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, FI-90220 Oulu, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, FI-00300 Helsinki, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491, Trondheim, Norway
- Children’s Hospital, Helsinki University Hospital and University of Helsinki, FI-00290 Helsinki, Finland
| | - Pia M Villa
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, FI-00270 Helsinki, Finland
| | - Esa K Hämäläinen
- Department of Clinical Chemistry, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, FI-00270 Helsinki, Finland
- Finnish National Institute for Health and Welfare, FI-00300 Helsinki, Finland
- University of Edinburgh, Edinburgh EH8 9YL, UK
| | - Hannele Laivuori
- Medical and Clinical Genetics, Helsinki University Hospital and University of Helsinki, FI-00270 Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, FI-00270 Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, FI-33520 Tampere, Finland
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, FI-00270 Helsinki, Finland
| | - Saila B Koivusalo
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, FI-00270 Helsinki, Finland
- Department of Obstetrics and Gynaecology, Turku University Hospital and University of Turku, FI-20520 Turku, Finland
| |
Collapse
|
21
|
Misra S, Wagner R, Ozkan B, Schön M, Sevilla-Gonzalez M, Prystupa K, Wang CC, Kreienkamp RJ, Cromer SJ, Rooney MR, Duan D, Thuesen ACB, Wallace AS, Leong A, Deutsch AJ, Andersen MK, Billings LK, Eckel RH, Sheu WHH, Hansen T, Stefan N, Goodarzi MO, Ray D, Selvin E, Florez JC, Meigs JB, Udler MS. Precision subclassification of type 2 diabetes: a systematic review. COMMUNICATIONS MEDICINE 2023; 3:138. [PMID: 37798471 PMCID: PMC10556101 DOI: 10.1038/s43856-023-00360-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Heterogeneity in type 2 diabetes presentation and progression suggests that precision medicine interventions could improve clinical outcomes. We undertook a systematic review to determine whether strategies to subclassify type 2 diabetes were associated with high quality evidence, reproducible results and improved outcomes for patients. METHODS We searched PubMed and Embase for publications that used 'simple subclassification' approaches using simple categorisation of clinical characteristics, or 'complex subclassification' approaches which used machine learning or 'omics approaches in people with established type 2 diabetes. We excluded other diabetes subtypes and those predicting incident type 2 diabetes. We assessed quality, reproducibility and clinical relevance of extracted full-text articles and qualitatively synthesised a summary of subclassification approaches. RESULTS Here we show data from 51 studies that demonstrate many simple stratification approaches, but none have been replicated and many are not associated with meaningful clinical outcomes. Complex stratification was reviewed in 62 studies and produced reproducible subtypes of type 2 diabetes that are associated with outcomes. Both approaches require a higher grade of evidence but support the premise that type 2 diabetes can be subclassified into clinically meaningful subtypes. CONCLUSION Critical next steps toward clinical implementation are to test whether subtypes exist in more diverse ancestries and whether tailoring interventions to subtypes will improve outcomes.
Collapse
Affiliation(s)
- Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK.
| | - Robert Wagner
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Bige Ozkan
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Magdalena Sevilla-Gonzalez
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Katsiaryna Prystupa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Caroline C Wang
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Raymond J Kreienkamp
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| | - Sara J Cromer
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary R Rooney
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Daisy Duan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne Cathrine Baun Thuesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amelia S Wallace
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Aaron Leong
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, 100 Cambridge St 16th Floor, Boston, MA, USA
| | - Aaron J Deutsch
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Liana K Billings
- Division of Endocrinology, Diabetes and Metabolism, NorthShore University Health System, Skokie, IL, USA
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wayne Huey-Herng Sheu
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli County, Taiwan, ROC
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Division of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Norbert Stefan
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- University Hospital of Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, Neuherberg, Germany
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Debashree Ray
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth Selvin
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - James B Meigs
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, 100 Cambridge St 16th Floor, Boston, MA, USA
| | - Miriam S Udler
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
22
|
Ordoñez-Guillen NE, Gonzalez-Compean JL, Lopez-Arevalo I, Contreras-Murillo M, Aldana-Bobadilla E. Machine learning based study for the classification of Type 2 diabetes mellitus subtypes. BioData Min 2023; 16:24. [PMID: 37608329 PMCID: PMC10463725 DOI: 10.1186/s13040-023-00340-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/07/2023] [Indexed: 08/24/2023] Open
Abstract
PURPOSE Data-driven diabetes research has increased its interest in exploring the heterogeneity of the disease, aiming to support in the development of more specific prognoses and treatments within the so-called precision medicine. Recently, one of these studies found five diabetes subgroups with varying risks of complications and treatment responses. Here, we tackle the development and assessment of different models for classifying Type 2 Diabetes (T2DM) subtypes through machine learning approaches, with the aim of providing a performance comparison and new insights on the matter. METHODS We developed a three-stage methodology starting with the preprocessing of public databases NHANES (USA) and ENSANUT (Mexico) to construct a dataset with N = 10,077 adult diabetes patient records. We used N = 2,768 records for training/validation of models and left the remaining (N = 7,309) for testing. In the second stage, groups of observations -each one representing a T2DM subtype- were identified. We tested different clustering techniques and strategies and validated them by using internal and external clustering indices; obtaining two annotated datasets Dset A and Dset B. In the third stage, we developed different classification models assaying four algorithms, seven input-data schemes, and two validation settings on each annotated dataset. We also tested the obtained models using a majority-vote approach for classifying unseen patient records in the hold-out dataset. RESULTS From the independently obtained bootstrap validation for Dset A and Dset B, mean accuracies across all seven data schemes were [Formula: see text] ([Formula: see text]) and [Formula: see text] ([Formula: see text]), respectively. Best accuracies were [Formula: see text] and [Formula: see text]. Both validation setting results were consistent. For the hold-out dataset, results were consonant with most of those obtained in the literature in terms of class proportions. CONCLUSION The development of machine learning systems for the classification of diabetes subtypes constitutes an important task to support physicians for fast and timely decision-making. We expect to deploy this methodology in a data analysis platform to conduct studies for identifying T2DM subtypes in patient records from hospitals.
Collapse
Affiliation(s)
- Nelson E Ordoñez-Guillen
- Cinvestav Tamaulipas, Carretera Victoria-Soto la Marina km 5.5, Victoria, 87130, Tamaulipas, Mexico
| | | | - Ivan Lopez-Arevalo
- Cinvestav Tamaulipas, Carretera Victoria-Soto la Marina km 5.5, Victoria, 87130, Tamaulipas, Mexico
| | - Miguel Contreras-Murillo
- Cinvestav Tamaulipas, Carretera Victoria-Soto la Marina km 5.5, Victoria, 87130, Tamaulipas, Mexico
| | - Edwin Aldana-Bobadilla
- CONAHCYT-Centro de Investigación y de Estudios Avanzados del IPN, Unidad Tamaulipas, Carretera Victoria-Soto la Marina km 5.5, Victoria, Tamaulipas, 87130, Mexico
| |
Collapse
|
23
|
Prystupa K, Delgado GE, Moissl AP, Kleber ME, Birkenfeld AL, Heni M, Fritsche A, März W, Wagner R. Clusters of prediabetes and type 2 diabetes stratify all-cause mortality in a cohort of participants undergoing invasive coronary diagnostics. Cardiovasc Diabetol 2023; 22:211. [PMID: 37592260 PMCID: PMC10436494 DOI: 10.1186/s12933-023-01923-3] [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: 02/09/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Heterogeneous metabolic clusters have been identified in diabetic and prediabetic states. It is not known whether such pathophysiologic clusters impact survival in at-risk persons being evaluated for coronary heart disease. METHODS The LURIC Study recruited patients referred for coronary angiography at a median age of 63 (IQR 56-70) with a follow-up of 16.1 (IQR 9.6, 17.7) years. Clustering of 1269 subjects without diabetes was performed with oGTT-derived glucose and insulin; fasting triglyceride, high-density lipoprotein, BMI, waist and hip circumference. Patients with T2D (n = 794) were clustered using age, BMI, glycemia, homeostasis model assessment, and islet autoantibodies. Associations of clusters with mortality were analysed using Cox regression. RESULTS Individuals without diabetes were classified into six subphenotypes, with 884 assigned to subjects at low-risk (cluster 1,2,4) and 385 at high-risk (cluster 3,5,6) for diabetes. We found significantly increased mortality in clusters 3 (hazard ratio (HR)1.42), 5 (HR 1.43), and 6 (HR 1.46) after adjusting for age, BMI, HbA1c and sex. In the T2D group, 508 were assigned to mild age-related diabetes (MARD), 183 to severe insulin-resistant diabetes (SIRD), 84 to mild obesity-related diabetes (MOD), 19 to severe insulin-deficient diabetes (SIDD). Compared to the low-risk non-diabetes group, crude mortality was not different in MOD. Increased mortality was found for MARD (HR 2.2), SIRD (HR 2.2), and SIDD (HR 2.5). CONCLUSIONS Metabolic clustering successfully stratifies survival even among persons undergoing invasive coronary diagnostics. Novel clustering approaches based on glucose metabolism can identify persons who require special attention as they are at risk of increased mortality.
Collapse
Affiliation(s)
- Katsiaryna Prystupa
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany.
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Angela P Moissl
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Division of Endocrinology and Diabetology, Internal Medicine 1, University Hospital Ulm, Ulm, Germany
| | - Andreas Fritsche
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
| | - Robert Wagner
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
| |
Collapse
|
24
|
Abbasi M, Tosur M, Astudillo M, Refaey A, Sabharwal A, Redondo MJ. Clinical Characterization of Data-Driven Diabetes Clusters of Pediatric Type 2 Diabetes. Pediatr Diabetes 2023; 2023:6955723. [PMID: 38694145 PMCID: PMC11062019 DOI: 10.1155/2023/6955723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/04/2024] Open
Abstract
Background Pediatric Type 2 diabetes (T2D) is highly heterogeneous. Previous reports on adult-onset diabetes demonstrated the existence of diabetes clusters. Therefore, we set out to identify unique diabetes subgroups with distinct characteristics among youth with T2D using commonly available demographic, clinical, and biochemical data. Methods We performed data-driven cluster analysis (K-prototypes clustering) to characterize diabetes subtypes in pediatrics using a dataset with 722 children and adolescents with autoantibody-negative T2D. The six variables included in our analysis were sex, race/ethnicity, age, BMI Z-score and hemoglobin A1c at the time of diagnosis, and non-HDL cholesterol within first year of diagnosis. Results We identified five distinct clusters of pediatric T2D, with different features, treatment regimens and risk of diabetes complications: Cluster 1 was characterized by higher A1c; Cluster 2, by higher non-HDL; Cluster 3, by lower age at diagnosis and lower A1c; Cluster 4, by lower BMI and higher A1c; and Cluster 5, by lower A1c and higher age. Youth in Cluster 1 had the highest rate of diabetic ketoacidosis (DKA) (p = 0.0001) and were most prescribed metformin (p = 0.06). Those in Cluster 2 were most prone to polycystic ovarian syndrome (p = 0.001). Younger individuals with lowest family history of diabetes were least frequently diagnosed with diabetic ketoacidosis (p = 0.001) and microalbuminuria (p = 0.06). Low-BMI individuals with higher A1c had the lowest prevalence of acanthosis nigricans (p = 0.0003) and hypertension (p = 0.03). Conclusions Utilizing clinical measures gathered at the time of diabetes diagnosis can be used to identify subgroups of pediatric T2D with prognostic value. Consequently, this advancement contributes to the progression and wider implementation of precision medicine in diabetes management.
Collapse
Affiliation(s)
- Mahsan Abbasi
- Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Mustafa Tosur
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
- Children’s Nutrition Research Center, USDA/ARS, Houston, TX, USA
| | - Marcela Astudillo
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - Ahmad Refaey
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | | | - Maria J. Redondo
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
25
|
Danquah I, Mank I, Hampe CS, Meeks KAC, Agyemang C, Owusu-Dabo E, Smeeth L, Klipstein-Grobusch K, Bahendeka S, Spranger J, Mockenhaupt FP, Schulze MB, Rolandsson O. Subgroups of adult-onset diabetes: a data-driven cluster analysis in a Ghanaian population. Sci Rep 2023; 13:10756. [PMID: 37402743 DOI: 10.1038/s41598-023-37494-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
Adult-onset diabetes mellitus (here: aDM) is not a uniform disease entity. In European populations, five diabetes subgroups have been identified by cluster analysis using simple clinical variables; these may elucidate diabetes aetiology and disease prognosis. We aimed at reproducing these subgroups among Ghanaians with aDM, and establishing their importance for diabetic complications in different health system contexts. We used data of 541 Ghanaians with aDM (age: 25-70 years; male sex: 44%) from the multi-center, cross-sectional Research on Obesity and Diabetes among African Migrants (RODAM) Study. Adult-onset DM was defined as fasting plasma glucose (FPG) ≥ 7.0 mmol/L, documented use of glucose-lowering medication or self-reported diabetes, and age of onset ≥ 18 years. We derived subgroups by cluster analysis using (i) a previously published set of variables: age at diabetes onset, HbA1c, body mass index, HOMA-beta, HOMA-IR, positivity of glutamic acid decarboxylase autoantibodies (GAD65Ab), and (ii) Ghana-specific variables: age at onset, waist circumference, FPG, and fasting insulin. For each subgroup, we calculated the clinical, treatment-related and morphometric characteristics, and the proportions of objectively measured and self-reported diabetic complications. We reproduced the five subgroups: cluster 1 (obesity-related, 73%) and cluster 5 (insulin-resistant, 5%) with no dominant diabetic complication patterns; cluster 2 (age-related, 10%) characterized by the highest proportions of coronary artery disease (CAD, 18%) and stroke (13%); cluster 3 (autoimmune-related, 5%) showing the highest proportions of kidney dysfunction (40%) and peripheral artery disease (PAD, 14%); and cluster 4 (insulin-deficient, 7%) characterized by the highest proportion of retinopathy (14%). The second approach yielded four subgroups: obesity- and age-related (68%) characterized by the highest proportion of CAD (9%); body fat-related and insulin-resistant (18%) showing the highest proportions of PAD (6%) and stroke (5%); malnutrition-related (8%) exhibiting the lowest mean waist circumference and the highest proportion of retinopathy (20%); and ketosis-prone (6%) with the highest proportion of kidney dysfunction (30%) and urinary ketones (6%). With the same set of clinical variables, the previously published aDM subgroups can largely be reproduced by cluster analysis in this Ghanaian population. This method may generate in-depth understanding of the aetiology and prognosis of aDM, particularly when choosing variables that are clinically relevant for the target population.
Collapse
Affiliation(s)
- Ina Danquah
- Heidelberg Institute of Global Health (HIGH), Faculty of Medicine and University Hospital, Heidelberg University, Heidelberg, Germany.
| | - Isabel Mank
- Heidelberg Institute of Global Health (HIGH), Faculty of Medicine and University Hospital, Heidelberg University, Heidelberg, Germany
- German Institute for Development Evaluation (DEval), Bonn, Germany
| | | | - Karlijn A C Meeks
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Public Health, Amsterdam UMC, location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Charles Agyemang
- Department of Public Health, Amsterdam UMC, location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ellis Owusu-Dabo
- Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Liam Smeeth
- London School of Hygiene and Tropical Medicine (LSHTM), London, UK
| | - Kerstin Klipstein-Grobusch
- Julius Global Health, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Division of Epidemiology and Biostatistics, School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité - Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Frank P Mockenhaupt
- Institute of Tropical Medicine and International Health, Charité - Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Olov Rolandsson
- Department of Public Health and Clinical Medicine, Section of Family Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
26
|
Zhao S, Li H, Jing X, Zhang X, Li R, Li Y, Liu C, Chen J, Li G, Zheng W, Li Q, Wang X, Wang L, Sun Y, Xu Y, Wang S. Identifying subgroups of patients with type 2 diabetes based on real-world traditional chinese medicine electronic medical records. Front Pharmacol 2023; 14:1210667. [PMID: 37456755 PMCID: PMC10339739 DOI: 10.3389/fphar.2023.1210667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction: Type 2 diabetes (T2D) is a multifactorial complex chronic disease with a high prevalence worldwide, and Type 2 diabetes patients with different comorbidities often present multiple phenotypes in the clinic. Thus, there is a pressing need to improve understanding of the complexity of the clinical Type 2 diabetes population to help identify more accurate disease subtypes for personalized treatment. Methods: Here, utilizing the traditional Chinese medicine (TCM) clinical electronic medical records (EMRs) of 2137 Type 2 diabetes inpatients, we followed a heterogeneous medical record network (HEMnet) framework to construct heterogeneous medical record networks by integrating the clinical features from the electronic medical records, molecular interaction networks and domain knowledge. Results: Of the 2137 Type 2 diabetes patients, 1347 were male (63.03%), and 790 were female (36.97%). Using the HEMnet method, we obtained eight non-overlapping patient subgroups. For example, in H3, Poria, Astragali Radix, Glycyrrhizae Radix et Rhizoma, Cinnamomi Ramulus, and Liriopes Radix were identified as significant botanical drugs. Cardiovascular diseases (CVDs) were found to be significant comorbidities. Furthermore, enrichment analysis showed that there were six overlapping pathways and eight overlapping Gene Ontology terms among the herbs, comorbidities, and Type 2 diabetes in H3. Discussion: Our results demonstrate that identification of the Type 2 diabetes subgroup based on the HEMnet method can provide important guidance for the clinical use of herbal prescriptions and that this method can be used for other complex diseases.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hengfei Li
- Department of Infectious Diseases, Hubei Provincial Hospital of Traditional Chinese Medicine (Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Province Academy of Traditional Chinese Medicine), Wuhan, China
| | - Xuan Jing
- Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Xuebin Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ronghua Li
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yinghao Li
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenguang Liu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Chen
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoxia Li
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Zheng
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Li
- Department of Nursing, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue Wang
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Letian Wang
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanyuan Sun
- Department of Obstetrics and Gynecology, Weifang Fangzi District People’s Hospital, Weifang, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shihua Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Landgraf W, Bigot G, Frier BM, Bolli GB, Owens DR. Response to insulin glargine 100 U/mL treatment in newly-defined subgroups of type 2 diabetes: Post hoc pooled analysis of insulin-naïve participants from nine randomised clinical trials. Prim Care Diabetes 2023:S1751-9918(23)00093-1. [PMID: 37142540 DOI: 10.1016/j.pcd.2023.04.010] [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: 01/17/2023] [Revised: 04/13/2023] [Accepted: 04/29/2023] [Indexed: 05/06/2023]
Abstract
AIMS To assess insulin glargine 100 U/mL (IGlar-100) treatment outcomes according to newly-defined subgroups of type 2 diabetes mellitus (T2DM). METHODS Insulin-naïve T2DM participants (n = 2684) from nine randomised clinical trials initiating IGlar-100 were pooled and assigned to subgroups "Mild Age-Related Diabetes (MARD)", "Mild Obesity Diabetes (MOD)", "Severe Insulin Resistant Diabetes (SIRD)", and "Severe Insulin Deficient Diabetes (SIDD)", according to age at onset of diabetes, baseline HbA1c, BMI, and fasting C-peptide using sex-specific nearest centroid approach. HbA1c, FPG, hypoglycemia, insulin dose, and body weight were analysed at baseline and 24 weeks. RESULTS Subgroup distribution was MARD 15.3 % (n = 411), MOD 39.8 % (n = 1067), SIRD 10.5 % (n = 283), SIDD 34.4 % (n = 923). From baseline HbA1c 8.0-9.6% adjusted least square mean reductions after 24 weeks were similar between subgroups (1.4-1.5 %). SIDD was less likely to achieve HbA1c < 7.0 % (OR: 0.40 [0.29, 0.55]) than MARD. While the final IGlar-100 dose (0.36 U/kg) in MARD was lower than in other subgroups (0.46-0.50 U/kg), it had the highest hypoglycemia risk. SIRD had lowest hypoglycemia risk and SIDD exhibited greatest body weight gain. CONCLUSIONS IGlar-100 lowered hyperglycemia similarly in all T2DM subgroups, but level of glycemic control, insulin dose, and hypoglycemia risk differed between subgroups.
Collapse
Affiliation(s)
| | | | - Brian M Frier
- The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Geremia B Bolli
- University of Perugia School of Medicine, Department of Medicine, Section of Endocrinology and Metabolism, Perugia, Italy
| | - David R Owens
- Swansea University, Diabetes Research Group Cymru, College of Medicine, Swansea, UK
| |
Collapse
|
28
|
Misra S, Wagner R, Ozkan B, Schön M, Sevilla-Gonzalez M, Prystupa K, Wang CC, Kreienkamp RJ, Cromer SJ, Rooney MR, Duan D, Thuesen ACB, Wallace AS, Leong A, Deutsch AJ, Andersen MK, Billings LK, Eckel RH, Sheu WHH, Hansen T, Stefan N, Goodarzi MO, Ray D, Selvin E, Florez JC, Meigs JB, Udler MS. Systematic review of precision subclassification of type 2 diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.19.23288577. [PMID: 37131632 PMCID: PMC10153304 DOI: 10.1101/2023.04.19.23288577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Heterogeneity in type 2 diabetes presentation, progression and treatment has the potential for precision medicine interventions that can enhance care and outcomes for affected individuals. We undertook a systematic review to ascertain whether strategies to subclassify type 2 diabetes are associated with improved clinical outcomes, show reproducibility and have high quality evidence. We reviewed publications that deployed 'simple subclassification' using clinical features, biomarkers, imaging or other routinely available parameters or 'complex subclassification' approaches that used machine learning and/or genomic data. We found that simple stratification approaches, for example, stratification based on age, body mass index or lipid profiles, had been widely used, but no strategy had been replicated and many lacked association with meaningful outcomes. Complex stratification using clustering of simple clinical data with and without genetic data did show reproducible subtypes of diabetes that had been associated with outcomes such as cardiovascular disease and/or mortality. Both approaches require a higher grade of evidence but support the premise that type 2 diabetes can be subclassified into meaningful groups. More studies are needed to test these subclassifications in more diverse ancestries and prove that they are amenable to interventions.
Collapse
|
29
|
Hwang YC, Ahn HY, Jun JE, Jeong IK, Ahn KJ, Chung HY. Subtypes of type 2 diabetes and their association with outcomes in Korean adults - A cluster analysis of community-based prospective cohort. Metabolism 2023; 141:155514. [PMID: 36746321 DOI: 10.1016/j.metabol.2023.155514] [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: 11/16/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/06/2023]
Abstract
BACKGROUND Little is known about the subtypes of type 2 diabetes (T2D) and their association with clinical outcomes in Asians. METHODS We performed data-driven cluster analysis in patients with newly diagnosed drug-naive T2D (n = 756) from the Korean Genome and Epidemiology Study. Clusters were based on five variables (age at diagnosis, BMI, HbA1c, and HOMA2 β-cell function, and insulin resistance). RESULTS We identified four clusters of patients with T2D according to k-means clustering: cluster 1 (22.4 %, severe insulin-resistant diabetes [SIRD]), cluster 2 (32.7 %, mild age-related diabetes [MARD]), cluster 3 (32.7 %, mild obesity-related diabetes [MOD]), and cluster 4 (12.3 %, severe insulin-deficient diabetes [SIDD]). During 14 years of follow-up, individuals in the SIDD cluster had the highest risk of initiation of glucose-lowering therapy compared to individuals in the other three clusters. Individuals in the MARD and SIDD clusters showed the highest risk of chronic kidney disease and cardiovascular disease, and individuals in the MOD clusters showed the lowest risk after adjusting for other risk factors (P < 0.05). CONCLUSIONS Patients with T2D can be categorized into four subgroups with different glycemic deterioration and risks of diabetes complications. Individualized management might be helpful for better clinical outcomes in Asian patients with different T2D subgroups.
Collapse
Affiliation(s)
- You-Cheol Hwang
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea.
| | - Hong-Yup Ahn
- Department of Statistics, Dongguk University, Seoul, Republic of Korea
| | - Ji Eun Jun
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - In-Kyung Jeong
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Kyu Jeung Ahn
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Ho Yeon Chung
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| |
Collapse
|
30
|
Schnell O, Battelino T, Bergenstal R, Birkenfeld AL, Ceriello A, Cheng A, Davies M, Edelman S, Forst T, Giorgino F, Green J, Groop PH, Hadjadj S, J L Heerspink H, Hompesch M, Izthak B, Ji L, Kanumilli N, Mankovsky B, Mathieu C, Miszon M, Mustafa R, Nauck M, Pecoits-Filho R, Pettus J, Ranta K, Rodbard HW, Rossing P, Ryden L, Schumm-Draeger PM, Solomon SD, Škrha J, Topsever P, Vilsbøll T, Wilding J, Standl E. CVOT Summit 2022 Report: new cardiovascular, kidney, and glycemic outcomes. Cardiovasc Diabetol 2023; 22:59. [PMID: 36927451 PMCID: PMC10019427 DOI: 10.1186/s12933-023-01788-6] [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: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
The 8th Cardiovascular Outcome Trial (CVOT) Summit on Cardiovascular, Kidney, and Glycemic Outcomes was held virtually on November 10-12, 2022. Following the tradition of previous summits, this reference congress served as a platform for in-depth discussion and exchange on recently completed outcomes trials as well as key trials important to the cardiovascular (CV) field. This year's focus was on the results of the DELIVER, EMPA-KIDNEY and SURMOUNT-1 trials and their implications for the treatment of heart failure (HF) and chronic kidney disease (CKD) with sodium-glucose cotransporter-2 (SGLT2) inhibitors and obesity with glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) receptor agonists. A broad audience of primary care physicians, diabetologists, endocrinologists, cardiologists, and nephrologists participated online in discussions on new consensus recommendations and guideline updates on type 2 diabetes (T2D) and CKD management, overcoming clinical inertia, glycemic markers, continuous glucose monitoring (CGM), novel insulin preparations, combination therapy, and reclassification of T2D. The impact of cardiovascular outcomes on the design of non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) trials, as well as the impact of real-world evidence (RWE) studies on the confirmation of CVOT outcomes and clinical trial design, were also intensively discussed. The 9th Cardiovascular Outcome Trial Summit will be held virtually on November 23-24, 2023 ( http://www.cvot.org ).
Collapse
Affiliation(s)
- Oliver Schnell
- Forschergruppe Diabetes e. V., Helmholtz Center Munich, Ingolstaedter Landstraße 1, Neuherberg, 85764, (Munich), Germany.
| | - Tadej Battelino
- University Medical Center, Ljubljana, Slovenia
- University Children's Hospital, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Richard Bergenstal
- International Diabetes Center at Park Nicollet, Health Partners, Minneapolis, MN, USA
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, University Clinic Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases at the Eberhard-Karls-University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany
| | | | - Alice Cheng
- Credit Valley Hospital, Mississauga, ON, Canada
| | - Melanie Davies
- Diabetes Research Centre, University of Leicester, Leicester, UK
- NIHR Biomedical Research Centre, Leicester, UK
| | - Steve Edelman
- Taking Control of Your Diabetes, Solana Beach, CA, USA
| | - Thomas Forst
- CRS Clinical Research Services Mannheim GmbH, Mannheim, Germany
| | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Jennifer Green
- Division of Endocrinology, Department of Medicine and Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Per-Henrik Groop
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
- Department of Diabetes, Central Medical School, Monash University, Melbourne, Australia
| | - Samy Hadjadj
- Thorax Institute, University Hospital of Nantes, Nantes, France
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Baruch Izthak
- Clalit Health Services and Technion Faculty of Medicine, Haifa, Israel
| | - Linong Ji
- Peking University People's Hospital, Xicheng District, Beijing, China
| | | | - Boris Mankovsky
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| | - Chantal Mathieu
- Department of Endocrinology, Catholic University Leuven, Leuven, Belgium
| | | | - Reem Mustafa
- Division of Nephrology and Hypertension, Medical Center, University of Kansas, Kansas City, KS, USA
| | - Michael Nauck
- Diabetes Division, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | | | - Jeremy Pettus
- Altman Clinical and Translational Research Institute (ACTRI), La Jolla, CA, USA
| | - Kari Ranta
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lars Ryden
- Department of Medicine K2, Karolinska Institute, Stockholm, Sweden
| | | | - Scott D Solomon
- Cardiovascular division, Brigham and Women's Hospital, Boston, MA, USA
| | - Jan Škrha
- Third Medical Department and Laboratory for Endocrinology and Metabolism, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pinar Topsever
- Department of Family Medicine, Acıbadem Mehmet Ali Aydınlar University School of Medicine, Istanbul, Turkey
| | - Tina Vilsbøll
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - John Wilding
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Eberhard Standl
- Forschergruppe Diabetes e. V., Helmholtz Center Munich, Ingolstaedter Landstraße 1, Neuherberg, 85764, (Munich), Germany
| |
Collapse
|
31
|
Pigeyre M, Gerstein H, Ahlqvist E, Hess S, Paré G. Identifying blood biomarkers for type 2 diabetes subtyping: a report from the ORIGIN trial. Diabetologia 2023; 66:1045-1051. [PMID: 36854916 DOI: 10.1007/s00125-023-05887-7] [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] [Received: 10/05/2022] [Accepted: 01/18/2023] [Indexed: 03/02/2023]
Abstract
AIMS/HYPOTHESIS Individuals with diabetes can be clustered into five subtypes using up to six routinely measured clinical variables. We hypothesised that circulating protein levels might be used to distinguish between these subtypes. We recently used five of these six variables to categorise 7017 participants from the Outcome Reduction with an Initial Glargine Intervention (ORIGIN) trial into these subtypes: severe autoimmune diabetes (SAID, n=241), severe insulin-deficient diabetes (SIDD, n=1594), severe insulin-resistant diabetes (SIRD, n=914), mild obesity-related diabetes (MOD, n=1595) and mild age-related diabetes (MARD, n=2673). METHODS Forward-selection logistic regression models were used to identify a subset of 233 cardiometabolic protein biomarkers that were independent determinants of one subtype vs the others. We then assessed the performance of adding identified biomarkers (one after one, from the most discriminant to the least) to predict each subtype vs the others using area under the receiver operating characteristic curve (AUC ROC). Models were adjusted for age, sex, ethnicity, C-peptide level, diabetes duration and glucose-lowering medication usage at blood collection. RESULTS A total of 25 biomarkers were independent determinants of subtypes, including 13 for SIDD, 2 for SIRD, 7 for MOD and 11 for MARD (all p<4.3 × 10-5). The performance of the biomarker sets (comprising 1 to 25 biomarkers), assessed through the AUC ROC, ranged from 0.611 to 0.734, 0.723 to 0.861, 0.672 to 0.742, and 0.651 to 0.751, for SIDD, SIRD, MOD and MARD, respectively. No biomarkers other than GAD antibodies were determinants of SAID. CONCLUSIONS/INTERPRETATION We identified 25 serum biomarkers, as independent determinants of type 2 diabetes subtypes, that could be combined into a diagnostic test for subtyping. TRIAL REGISTRATION ORIGIN trial, ClinicalTrials.gov NCT00069784.
Collapse
Affiliation(s)
- Marie Pigeyre
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada.
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada.
| | - Hertzel Gerstein
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada
- Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON, Canada
| | - Emma Ahlqvist
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Sibylle Hess
- Global Medical Diabetes, Sanofi, Frankfurt, Germany
| | - Guillaume Paré
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON, Canada
- Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, ON, Canada
| |
Collapse
|
32
|
Abstract
Diet-related diseases are the leading cause of death globally and strategies to tailor effective nutrition advice are required. Personalised nutrition advice is increasingly recognised as more effective than population-level advice to improve dietary intake and health outcomes. A potential tool to deliver personalised nutrition advice is metabotyping which groups individuals into homogeneous subgroups (metabotypes) using metabolic profiles. In summary, metabotyping has been successfully employed in human nutrition research to identify subgroups of individuals with differential responses to dietary challenges and interventions and diet–disease associations. The suitability of metabotyping to identify clinically relevant subgroups is corroborated by other fields such as diabetes research where metabolic profiling has been intensely used to identify subgroups of patients that display patterns of disease progression and complications. However, there is a paucity of studies examining the efficacy of the approach to improve dietary intake and health parameters. While the application of metabotypes to tailor and deliver nutrition advice is very promising, further evidence from randomised controlled trials is necessary for further development and acceptance of the approach.
Collapse
|
33
|
Zou X, Liu Y, Ji L. Review: Machine learning in precision pharmacotherapy of type 2 diabetes-A promising future or a glimpse of hope? Digit Health 2023; 9:20552076231203879. [PMID: 37786401 PMCID: PMC10541760 DOI: 10.1177/20552076231203879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023] Open
Abstract
Precision pharmacotherapy of diabetes requires judicious selection of the optimal therapeutic agent for individual patients. Artificial intelligence (AI), a swiftly expanding discipline, holds substantial potential to transform current practices in diabetes diagnosis and management. This manuscript provides a comprehensive review of contemporary research investigating drug responses in patient subgroups, stratified via either supervised or unsupervised machine learning approaches. The prevalent algorithmic workflow for investigating drug responses using machine learning involves cohort selection, data processing, predictor selection, development and validation of machine learning methods, subgroup allocation, and subsequent analysis of drug response. Despite the promising feature, current research does not yet provide sufficient evidence to implement machine learning algorithms into routine clinical practice, due to a lack of simplicity, validation, or demonstrated efficacy. Nevertheless, we anticipate that the evolving evidence base will increasingly substantiate the role of machine learning in molding precision pharmacotherapy for diabetes.
Collapse
Affiliation(s)
- Xiantong Zou
- Xiantong Zou, Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, 100044, China.
| | | | - Linong Ji
- Linong Ji, Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
34
|
Davies MJ, Aroda VR, Collins BS, Gabbay RA, Green J, Maruthur NM, Rosas SE, Del Prato S, Mathieu C, Mingrone G, Rossing P, Tankova T, Tsapas A, Buse JB. Management of hyperglycaemia in type 2 diabetes, 2022. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 2022; 65:1925-1966. [PMID: 36151309 PMCID: PMC9510507 DOI: 10.1007/s00125-022-05787-2] [Citation(s) in RCA: 447] [Impact Index Per Article: 149.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/18/2022] [Indexed: 01/11/2023]
Abstract
The American Diabetes Association and the European Association for the Study of Diabetes convened a panel to update the previous consensus statements on the management of hyperglycaemia in type 2 diabetes in adults, published since 2006 and last updated in 2019. The target audience is the full spectrum of the professional healthcare team providing diabetes care in the USA and Europe. A systematic examination of publications since 2018 informed new recommendations. These include additional focus on social determinants of health, the healthcare system and physical activity behaviours including sleep. There is a greater emphasis on weight management as part of the holistic approach to diabetes management. The results of cardiovascular and kidney outcomes trials involving sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists, including assessment of subgroups, inform broader recommendations for cardiorenal protection in people with diabetes at high risk of cardiorenal disease. After a summary listing of consensus recommendations, practical tips for implementation are provided.
Collapse
Affiliation(s)
- Melanie J Davies
- Leicester Diabetes Research Centre, University of Leicester, Leicester, UK.
- Leicester National Institute for Health Research (NIHR) Biomedical Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK.
| | - Vanita R Aroda
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Billy S Collins
- National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | | | - Jennifer Green
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Nisa M Maruthur
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sylvia E Rosas
- Kidney and Hypertension Unit, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Stefano Del Prato
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Geltrude Mingrone
- Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Division of Diabetes and Nutritional Sciences, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tsvetalina Tankova
- Department of Endocrinology, Medical University - Sofia, Sofia, Bulgaria
| | - Apostolos Tsapas
- Diabetes Centre, Clinical Research and Evidence-based Medicine Unit, Aristotle University Thessaloniki, Thessaloniki, Greece
- Harris Manchester College, University of Oxford, Oxford, UK
| | - John B Buse
- University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Wang J, Liu JJ, Gurung RL, Liu S, Lee J, M Y, Ang K, Shao YM, Tang JIS, Benke PI, Torta F, Wenk MR, Tavintharan S, Tang WE, Sum CF, Lim SC. Clinical variable-based cluster analysis identifies novel subgroups with a distinct genetic signature, lipidomic pattern and cardio-renal risks in Asian patients with recent-onset type 2 diabetes. Diabetologia 2022; 65:2146-2156. [PMID: 35763031 PMCID: PMC9630229 DOI: 10.1007/s00125-022-05741-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/25/2022] [Indexed: 01/11/2023]
Abstract
AIMS/HYPOTHESIS We sought to subtype South East Asian patients with type 2 diabetes by de novo cluster analysis on clinical variables, and to determine whether the novel subgroups carry distinct genetic and lipidomic features as well as differential cardio-renal risks. METHODS Analysis by k-means algorithm was performed in 687 participants with recent-onset diabetes in Singapore. Genetic risk for beta cell dysfunction was assessed by polygenic risk score. We used a discovery-validation approach for the lipidomics study. Risks for cardio-renal complications were studied by survival analysis. RESULTS Cluster analysis identified three novel diabetic subgroups, i.e. mild obesity-related diabetes (MOD, 45%), mild age-related diabetes with insulin insufficiency (MARD-II, 36%) and severe insulin-resistant diabetes with relative insulin insufficiency (SIRD-RII, 19%). Compared with the MOD subgroup, MARD-II had a higher polygenic risk score for beta cell dysfunction. The SIRD-RII subgroup had higher levels of sphingolipids (ceramides and sphingomyelins) and glycerophospholipids (phosphatidylethanolamine and phosphatidylcholine), whereas the MARD-II subgroup had lower levels of sphingolipids and glycerophospholipids but higher levels of lysophosphatidylcholines. Over a median of 7.3 years follow-up, the SIRD-RII subgroup had the highest risks for incident heart failure and progressive kidney disease, while the MARD-II subgroup had moderately elevated risk for kidney disease progression. CONCLUSIONS/INTERPRETATION Cluster analysis on clinical variables identified novel subgroups with distinct genetic, lipidomic signatures and varying cardio-renal risks in South East Asian participants with type 2 diabetes. Our study suggests that this easily actionable approach may be adapted in other ethnic populations to stratify the heterogeneous type 2 diabetes population for precision medicine.
Collapse
Affiliation(s)
- Jiexun Wang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Resham L Gurung
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Sylvia Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Janus Lee
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Yiamunaa M
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Keven Ang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Yi Ming Shao
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Justin I-Shing Tang
- Department of Medicine, Khoo Teck Puat Hospital, Singapore, Republic of Singapore
| | - Peter I Benke
- Lipidomics Incubator, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Federico Torta
- Lipidomics Incubator, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Markus R Wenk
- Lipidomics Incubator, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | | | - Wern Ee Tang
- National Healthcare Group Polyclinic, Singapore, Republic of Singapore
| | - Chee Fang Sum
- Diabetes Centre, Admiralty Medical Centre, Singapore, Republic of Singapore
| | - Su Chi Lim
- Diabetes Centre, Admiralty Medical Centre, Singapore, Republic of Singapore.
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Republic of Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore.
| |
Collapse
|
36
|
Davies MJ, Aroda VR, Collins BS, Gabbay RA, Green J, Maruthur NM, Rosas SE, Del Prato S, Mathieu C, Mingrone G, Rossing P, Tankova T, Tsapas A, Buse JB. Management of Hyperglycemia in Type 2 Diabetes, 2022. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2022; 45:2753-2786. [PMID: 36148880 PMCID: PMC10008140 DOI: 10.2337/dci22-0034] [Citation(s) in RCA: 763] [Impact Index Per Article: 254.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 02/07/2023]
Abstract
The American Diabetes Association and the European Association for the Study of Diabetes convened a panel to update the previous consensus statements on the management of hyperglycemia in type 2 diabetes in adults, published since 2006 and last updated in 2019. The target audience is the full spectrum of the professional health care team providing diabetes care in the U.S. and Europe. A systematic examination of publications since 2018 informed new recommendations. These include additional focus on social determinants of health, the health care system, and physical activity behaviors, including sleep. There is a greater emphasis on weight management as part of the holistic approach to diabetes management. The results of cardiovascular and kidney outcomes trials involving sodium-glucose cotransporter 2 inhibitors and glucagon-like peptide 1 receptor agonists, including assessment of subgroups, inform broader recommendations for cardiorenal protection in people with diabetes at high risk of cardiorenal disease. After a summary listing of consensus recommendations, practical tips for implementation are provided.
Collapse
Affiliation(s)
- Melanie J. Davies
- Leicester Diabetes Research Centre, University of Leicester, Leicester, U.K
- Leicester National Institute for Health Research Biomedical Research Centre, University Hospitals of Leicester NHS Trust, Leicester, U.K
| | - Vanita R. Aroda
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | | | - Jennifer Green
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Nisa M. Maruthur
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sylvia E. Rosas
- Kidney and Hypertension Unit, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Stefano Del Prato
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Geltrude Mingrone
- Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Division of Diabetes and Nutritional Sciences, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, U.K
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Apostolos Tsapas
- Diabetes Centre, Clinical Research and Evidence-Based Medicine Unit, Aristotle University Thessaloniki, Thessaloniki, Greece
- Harris Manchester College, University of Oxford, Oxford, U.K
| | - John B. Buse
- University of North Carolina School of Medicine, Chapel Hill, NC
| |
Collapse
|
37
|
Abstract
The historical subclassification of diabetes into predominantly types 1 and 2 is well appreciated to inadequately capture the heterogeneity seen in patient presentations, disease course, response to therapy and disease complications. This review summarises proposed data-driven approaches to further refine diabetes subtypes using clinical phenotypes and/or genetic information. We highlight the benefits as well as the limitations of these subclassification schemas, including practical barriers to their implementation that would need to be overcome before incorporation into clinical practice.
Collapse
Affiliation(s)
- Aaron J Deutsch
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical & Population Genetics, Broad Institute, Boston, MA, USA
- Program in Metabolism, Broad Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Emma Ahlqvist
- Genomics, Diabetes and Endocrinology, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden.
| | - Miriam S Udler
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical & Population Genetics, Broad Institute, Boston, MA, USA.
- Program in Metabolism, Broad Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Landgraf W, Bigot G, Hess S, Asplund O, Groop L, Ahlqvist E, Käräjämäki A, Owens DR, Frier BM, Bolli GB. Distribution and characteristics of newly-defined subgroups of type 2 diabetes in randomised clinical trials: Post hoc cluster assignment analysis of over 12,000 study participants. Diabetes Res Clin Pract 2022; 190:110012. [PMID: 35863553 DOI: 10.1016/j.diabres.2022.110012] [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: 06/09/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/28/2022]
Abstract
AIMS Newly-defined subgroups of type 2 diabetes mellitus (T2DM) have been reported from real-world cohorts but not in detail from randomised clinical trials (RCTs). METHODS T2DM participants, uncontrolled on different pre-study therapies (n = 12.738; 82 % Caucasian; 44 % with diabetes duration > 10 years) from 14 RCTs, were assigned to new subgroups according to age at onset of diabetes, HbA1c, BMI, and fasting C-peptide using the nearest centroid approach. Subgroup distribution, characteristics and influencing factors were analysed. RESULTS In both, pooled and single RCTs, "mild-obesity related diabetes" predominated (45 %) with mean BMI of 35 kg/m2. "Severe insulin-resistant diabetes" was found least often (4.6 %) and prevalence of "mild age-related diabetes" (23.9 %) was mainly influenced by age at onset of diabetes and age cut-offs. Subgroup characteristics were widely comparable to those from real-world cohorts, but all subgroups showed higher frequencies of diabetes-related complications which were associated with longer diabetes duration. A high proportion of "severe insulin-deficient diabetes" (25.4 %) was identified with poor pre-study glycaemic control. CONCLUSIONS Classification of RCT participants into newly-defined diabetes subgroups revealed the existence of a heterogeneous population of T2DM. For future RCTs, subgroup-based randomisation of T2DM will better define the target population and relevance of the outcomes by avoiding clinical heterogeneity.
Collapse
Affiliation(s)
| | | | | | - Olof Asplund
- Lund University Diabetes Centre, Department of Clinical Sciences, Skåne University Hospital, Malmö, Sweden
| | - Leif Groop
- Institute of Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Emma Ahlqvist
- Lund University Diabetes Centre, Department of Clinical Sciences, Skåne University Hospital, Malmö, Sweden
| | - Annemari Käräjämäki
- Department of Primary Health Care, Vaasa Central Hospital, and Diabetes Center, Vaasa Health Care Center, Vaasa, Finland
| | - David R Owens
- Swansea University, Diabetes Research Group Cymru, College of Medicine, Swansea, UK
| | - Brian M Frier
- The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Geremia B Bolli
- University of Perugia School of Medicine, Department of Medicine, Section of Endocrinology and Metabolism, Perugia, Italy
| |
Collapse
|
39
|
Schrader S, Perfilyev A, Ahlqvist E, Groop L, Vaag A, Martinell M, García-Calzón S, Ling C. Novel Subgroups of Type 2 Diabetes Display Different Epigenetic Patterns That Associate With Future Diabetic Complications. Diabetes Care 2022; 45:1621-1630. [PMID: 35607770 PMCID: PMC9274219 DOI: 10.2337/dc21-2489] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/05/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 2 diabetes (T2D) was recently reclassified into severe insulin-deficient diabetes (SIDD), severe insulin-resistant diabetes (SIRD), mild obesity-related diabetes (MOD), and mild age-related diabetes (MARD), which have different risk of complications. We explored whether DNA methylation differs between these subgroups and whether subgroup-unique methylation risk scores (MRSs) predict diabetic complications. RESEARCH DESIGN AND METHODS Genome-wide DNA methylation was analyzed in blood from subjects with newly diagnosed T2D in discovery and replication cohorts. Subgroup-unique MRSs were built, including top subgroup-unique DNA methylation sites. Regression models examined whether MRSs associated with subgroups and future complications. RESULTS We found epigenetic differences between the T2D subgroups. Subgroup-unique MRSs were significantly different in those patients allocated to each respective subgroup compared with the combined group of all other subgroups. These associations were validated in an independent replication cohort, showing that subgroup-unique MRSs associate with individual subgroups (odds ratios 1.6-6.1 per 1-SD increase, P < 0.01). Subgroup-unique MRSs were also associated with future complications. Higher MOD-MRS was associated with lower risk of cardiovascular (hazard ratio [HR] 0.65, P = 0.001) and renal (HR 0.50, P < 0.001) disease, whereas higher SIRD-MRS and MARD-MRS were associated with an increased risk of these complications (HR 1.4-1.9 per 1-SD increase, P < 0.01). Of 95 methylation sites included in subgroup-unique MRSs, 39 were annotated to genes previously linked to diabetes-related traits, including TXNIP and ELOVL2. Methylation in the blood of 18 subgroup-unique sites mirrors epigenetic patterns in tissues relevant for T2D, muscle and adipose tissue. CONCLUSIONS We identified differential epigenetic patterns between T2D subgroups that associated with future diabetic complications. These data support a reclassification of diabetes and the need for precision medicine in T2D subgroups.
Collapse
Affiliation(s)
- Silja Schrader
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Alexander Perfilyev
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Emma Ahlqvist
- Genomics, Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Leif Groop
- Genomics, Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Allan Vaag
- Type 2 Diabetes Biology Research, Steno Diabetes Center, Copenhagen, Denmark
| | - Mats Martinell
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden.,Academic Primary Care Centre, Uppsala, Sweden
| | - Sonia García-Calzón
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden.,Department of Food Science and Physiology, University of Navarra, Pamplona, Spain
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
40
|
Christensen DH, Nicolaisen SK, Ahlqvist E, Stidsen JV, Nielsen JS, Hojlund K, Olsen MH, García-Calzón S, Ling C, Rungby J, Brandslund I, Vestergaard P, Jessen N, Hansen T, Brøns C, Beck-Nielsen H, Sørensen HT, Thomsen RW, Vaag A. Type 2 diabetes classification: a data-driven cluster study of the Danish Centre for Strategic Research in Type 2 Diabetes (DD2) cohort. BMJ Open Diabetes Res Care 2022; 10:10/2/e002731. [PMID: 35428673 PMCID: PMC9014045 DOI: 10.1136/bmjdrc-2021-002731] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/19/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION A Swedish data-driven cluster study identified four distinct type 2 diabetes (T2D) clusters, based on age at diagnosis, body mass index (BMI), hemoglobin A1c (HbA1c) level, and homeostatic model assessment 2 (HOMA2) estimates of insulin resistance and beta-cell function. A Danish study proposed three T2D phenotypes (insulinopenic, hyperinsulinemic, and classical) based on HOMA2 measures only. We examined these two new T2D classifications using the Danish Centre for Strategic Research in Type 2 Diabetes cohort. RESEARCH DESIGN AND METHODS In 3529 individuals, we first performed a k-means cluster analysis with a forced k-value of four to replicate the Swedish clusters: severe insulin deficient (SIDD), severe insulin resistant (SIRD), mild age-related (MARD), and mild obesity-related (MOD) diabetes. Next, we did an analysis open to alternative k-values (ie, data determined the optimal number of clusters). Finally, we compared the data-driven clusters with the three Danish phenotypes. RESULTS Compared with the Swedish findings, the replicated Danish SIDD cluster included patients with lower mean HbA1c (86 mmol/mol vs 101 mmol/mol), and the Danish MOD cluster patients were less obese (mean BMI 32 kg/m2 vs 36 kg/m2). Our data-driven alternative k-value analysis suggested the optimal number of T2D clusters in our data to be three, rather than four. When comparing the four replicated Swedish clusters with the three proposed Danish phenotypes, 81%, 79%, and 69% of the SIDD, MOD, and MARD patients, respectively, fitted the classical T2D phenotype, whereas 70% of SIRD patients fitted the hyperinsulinemic phenotype. Among the three alternative data-driven clusters, 60% of patients in the most insulin-resistant cluster constituted 76% of patients with a hyperinsulinemic phenotype. CONCLUSION Different HOMA2-based approaches did not classify patients with T2D in a consistent manner. The T2D classes characterized by high insulin resistance/hyperinsulinemia appeared most distinct.
Collapse
Affiliation(s)
| | - Sia K Nicolaisen
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| | - Emma Ahlqvist
- Genomics, Diabetes and Endocrinology Unit, Department of Clinical Sciences, Lund University Diabetes Center, Malmö, Sweden
| | - Jacob V Stidsen
- The Danish Centre for Strategic Research in Type 2 Diabetes (DD2), Odense University Hospital, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Jens Steen Nielsen
- The Danish Centre for Strategic Research in Type 2 Diabetes (DD2), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kurt Hojlund
- The Danish Centre for Strategic Research in Type 2 Diabetes (DD2), Odense University Hospital, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Michael H Olsen
- Department of Internal Medicine and Steno Diabetes Center Zealand, Holbæk Hospital, Holbæk, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Sonia García-Calzón
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain
- Epigenetic and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Center, Scania University Hospital, Malmö, Sweden
| | - Charlotte Ling
- Epigenetic and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Center, Scania University Hospital, Malmö, Sweden
| | - Jørgen Rungby
- Department of Endocrinology IC, Bispebjerg University Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Ivan Brandslund
- Department of Clinical Biochemistry, University Hospital of Southern Denmark, Vejle, Denmark
| | - Peter Vestergaard
- Steno Diabetes Center Aalborg, Aalborg University Hospital, Aalborg, Denmark
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Brøns
- Steno Diabetes Center Copenhagen, Gentofte Hospital, Gentofte, Denmark
| | - Henning Beck-Nielsen
- The Danish Centre for Strategic Research in Type 2 Diabetes (DD2), Odense University Hospital, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Henrik T Sørensen
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| | - Reimar W Thomsen
- Department of Clinical Epidemiology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| | - Allan Vaag
- Steno Diabetes Center Copenhagen, Gentofte Hospital, Gentofte, Denmark
| |
Collapse
|
41
|
Physical activity and health-related quality of life among high-risk women for type 2 diabetes in the early years after pregnancy. BMC Womens Health 2022; 22:84. [PMID: 35313870 PMCID: PMC8939162 DOI: 10.1186/s12905-022-01664-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/11/2022] [Indexed: 11/16/2022] Open
Abstract
Background Previous studies have shown that physical activity (PA) correlates positively with health-related quality of life (HRQoL) in the general population. Few studies have investigated associations between device-measured PA and HRQoL among premenopausal women at risk for type 2 diabetes (T2D). In addition to physical well-being, general well-being improved by PA has been suggested to strengthen PA’s benefits in reducing metabolic diseases. The aim of this study was to examine the associations between PA and HRQoL (general and dimensions) among high-risk women in the early post-pregnancy years when T2D risk is highest and to estimate whether current obesity or prior gestational diabetes (GDM) modified these associations. Methods This cross-sectional study of high-risk women [body mass index (BMI) ≥ 30 kg/m2 and/or prior GDM)]4–6 years after delivery measured sleep, sedentary time, daily steps, and light (LPA), moderate-to-vigorous (MVPA), and vigorous PA (VPA) with the SenseWear ArmbandTM accelerometer for seven days and HRQoL with the 15D instrument. Results The analyses included 204 women with a median (IQR) age of 39 (6.0) years and a median BMI of 31.1 kg/m2 (10.9). 54% were currently obese (BMI ≥ 30 kg/m2), and 70% had prior gestational diabetes (GDM+). Women with obesity had lower PA levels than women with normal weight or overweight (p < 0.001) but there was no difference between the GDM+ or GDM− women. Women with both current obesity and GDM+ had highest sedentary time and lowest PA levels. The whole sample’s median 15D score was 0.934 (IQR 0.092), lower among women with obesity compared to the others (p < 0.001), but not different between GDM+ or GDM−. There was a positive correlation between VPA (adjusted rs = 0.262 p = 0.001) and the 15D score. After grouping according to BMI (< and ≥ 30 kg/m2), the associations remained significant only in women without obesity. Among them, sleep, total steps, MVPA, and VPA were positively associated with 15D. Conclusions Higher PA levels are associated with better HRQoL among high-risk women with normal weight and overweight but no differences were found among women affected by obesity in the early years after pregnancy. Trial registration Ethics committees of Helsinki University Hospital (Dnro 300/e9/06) and South Karelian Central Hospital (Dnro 06/08).
Collapse
|
42
|
Diabetes-Subtypen unterscheiden sich in Bezug auf Komplikationen. DIABETOL STOFFWECHS 2022. [DOI: 10.1055/a-1723-3705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|