101
|
Dutta D, Kadian J, Maisnam I, Kumar A, Bhattacharya S, Sharma M. Efficacy and Safety of Novel Thiazolidinedione Rivoglitazone in Type-2 Diabetes a Meta-Analysis. Indian J Endocrinol Metab 2023; 27:286-295. [PMID: 37867986 PMCID: PMC10586552 DOI: 10.4103/ijem.ijem_17_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/11/2023] [Accepted: 03/24/2023] [Indexed: 10/24/2023] Open
Abstract
No meta-analysis has analyzed the safety and efficacy of rivoglitazone in type-2 diabetes (T2DM). We undertook this meta-analysis to address this knowledge gap. Electronic databases were searched for RCTs involving T2DM patients receiving rivoglitazone in the intervention arm, and placebo/active comparator in the control arm. The primary outcome was to evaluate changes in HbA1c. Secondary outcomes were to evaluate alterations in glucose, lipids, and adverse events. From initially screened 24 articles, data from 3 RCTs (3591 patients) that fulfilled all criteria was analzsed. HbA1c was significantly lower with standard-dose (1 mg/d) [MD-0.86% (95%CI:-1.11--0.61); P < 0.01; I2 = 87%] and high-dose (1.5-2 mg/d) [MD-0.97%(95%CI:-1.03--0.90); P < 0.01; I2 = 19%] rivoglitazone compared to placebo. When compared to pioglitazone (30-45 mg/d), HbA1c lowering was comparable with standard-dose [MD 0.05%(95%CI:-0.01 - 0.11); P = 0.08; I2 = 11%], but superior with high-dose [MD -0.11%(95%CI:-0.18- -0.04); P < 0.01; I2 = 0%] rivoglitazone. Triglycerides were significantly lower with standard-dose [MD-17.95 mg/dl (95%CI:-34.23--1.66); P = 0.03; I2 = 0%] and high-dose [MD-40.41 mg/dl (95%CI:-72.90- -7.93);P = 0.01;I2 = 71%] rivoglitazone compared to placebo. Adiponectin significantly improved with standard-dose [MD 7.94 ng/ml (95%CI: 5.48-10.39); P < 0.01;I2 = 98%] and high-dose [MD 13.82 ng/ml (95%CI: 8.16-19.48); P < 0.01; I2 = 100%] rivoglitazone compared to placebo. hsCRP was significantly lower with standard-dose [MD -1.00 mg/L (95% CI: -1.20 - -0.80); P < 0.01; I2 = 6%] and high-dose [MD -1.50 mg/L (95%CI:-1.59- -1.40); P < 0.01; I2 = 0%] rivoglitazone compared to placebo. Treatment-emergent adverse events with standard-dose [Risk ratio (RR) 1.16 (95%CI: 0.84 -1.60); P = 0.38; I2 = 0%] and high-dose [RR1.34 (95%CI: 0.99-1.83); P = 0.06; I2 = 0%] rivoglitazone was comparable to placebo. Severe adverse events with standard-dose [RR1.88 (95%CI: 0.69-5.12);P = 0.22;I2 = 0%] and high-dose [RR 1.27 (95% CI: 0.45 - 3.59); P = 0.68; I2 = 0%] rivoglitazone was comparable to placebo. This meta-analysis highlights the good glycaemic efficacy and safety of both standard and high-dose rivoglitazone, and appears to be better than lobeglitazone in T2DM.
Collapse
Affiliation(s)
- Deep Dutta
- Department of Endocrinology, CEDAR Superspeciality Healthcare, Dwarka, New Delhi, India
| | - Jyoti Kadian
- Department of Medicine, Kalpana Chawla Government Medical College, Karnal, Haryana, India
| | - Indira Maisnam
- Department of Endocrinology, Institute of Post-graduate Medical Education and Research (IPGMER), Kolkata, West Bengal, India
| | - Ashok Kumar
- Department of Endocrinology, CEDAR Superspeciality Healthcare, Panipat, Haryana, India
| | | | - Meha Sharma
- Department of Rheumatology, CEDAR Superspeciality Healthcare, Dwarka, New Delhi, India
| |
Collapse
|
102
|
Pelligra A, Mrugala J, Griess K, Kirschner P, Nortmann O, Bartosinska B, Köster A, Krupenko NI, Gebel D, Westhoff P, Steckel B, Eberhard D, Herebian D, Belgardt BF, Schrader J, Weber APM, Krupenko SA, Lammert E. Pancreatic islet protection at the expense of secretory function involves serine-linked mitochondrial one-carbon metabolism. Cell Rep 2023; 42:112615. [PMID: 37294632 PMCID: PMC10592470 DOI: 10.1016/j.celrep.2023.112615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 06/11/2023] Open
Abstract
Type 2 diabetes is characterized by insulin hypersecretion followed by reduced glucose-stimulated insulin secretion (GSIS). Here we show that acute stimulation of pancreatic islets with the insulin secretagogue dextrorphan (DXO) or glibenclamide enhances GSIS, whereas chronic treatment with high concentrations of these drugs reduce GSIS but protect islets from cell death. Bulk RNA sequencing of islets shows increased expression of genes for serine-linked mitochondrial one-carbon metabolism (OCM) after chronic, but not acute, stimulation. In chronically stimulated islets, more glucose is metabolized to serine than to citrate, and the mitochondrial ATP/ADP ratio decreases, whereas the NADPH/NADP+ ratio increases. Activating transcription factor-4 (Atf4) is required and sufficient to activate serine-linked mitochondrial OCM genes in islets, with gain- and loss-of-function experiments showing that Atf4 reduces GSIS and is required, but not sufficient, for full DXO-mediated islet protection. In sum, we identify a reversible metabolic pathway that provides islet protection at the expense of secretory function.
Collapse
Affiliation(s)
- Angela Pelligra
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jessica Mrugala
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Kerstin Griess
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Philip Kirschner
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Oliver Nortmann
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Barbara Bartosinska
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Andrea Köster
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Natalia I Krupenko
- University of North Carolina (UNC) Nutrition Research Institute, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Dominik Gebel
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Philipp Westhoff
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany; Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Bodo Steckel
- Department of Molecular Cardiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Bengt-Frederik Belgardt
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jürgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Andreas P M Weber
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany; Cluster of Excellence on Plant Science (CEPLAS), Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Sergey A Krupenko
- University of North Carolina (UNC) Nutrition Research Institute, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
| |
Collapse
|
103
|
Dutta S, Shah RB, Singhal S, Dutta SB, Bansal S, Sinha S, Haque M. Metformin: A Review of Potential Mechanism and Therapeutic Utility Beyond Diabetes. Drug Des Devel Ther 2023; 17:1907-1932. [PMID: 37397787 PMCID: PMC10312383 DOI: 10.2147/dddt.s409373] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/10/2023] [Indexed: 07/04/2023] Open
Abstract
Metformin has been designated as one of the most crucial first-line therapeutic agents in the management of type 2 diabetes mellitus. Primarily being an antihyperglycemic agent, metformin also has a plethora of pleiotropic effects on various systems and processes. It acts majorly by activating AMPK (Adenosine Monophosphate-Activated Protein Kinase) in the cells and reducing glucose output from the liver. It also decreases advanced glycation end products and reactive oxygen species production in the endothelium apart from regulating the glucose and lipid metabolism in the cardiomyocytes, hence minimizing the cardiovascular risks. Its anticancer, antiproliferative and apoptosis-inducing effects on malignant cells might prove instrumental in the malignancy of organs like the breast, kidney, brain, ovary, lung, and endometrium. Preclinical studies have also shown some evidence of metformin's neuroprotective role in Parkinson's disease, Alzheimer's disease, multiple sclerosis and Huntington's disease. Metformin exerts its pleiotropic effects through varied pathways of intracellular signalling and exact mechanism in the majority of them remains yet to be clearly defined. This article has extensively reviewed the therapeutic benefits of metformin and the details of its mechanism for a molecule of boon in various conditions like diabetes, prediabetes, obesity, polycystic ovarian disease, metabolic derangement in HIV, various cancers and aging.
Collapse
Affiliation(s)
- Siddhartha Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Rima B Shah
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Shubha Singhal
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Sudeshna Banerjee Dutta
- Department of Medical Surgical Nursing, Shri Anand Institute of Nursing, Rajkot, Gujarat, 360005, India
| | - Sumit Bansal
- Department of Anaesthesiology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Susmita Sinha
- Department of Physiology, Khulna City Medical College and Hospital, Khulna, Bangladesh
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kuala Lumpur, 57000, Malaysia
| |
Collapse
|
104
|
Bielinski SJ, Yanes Cardozo LL, Takahashi PY, Larson NB, Castillo A, Podwika A, De Filippis E, Hernandez V, Mahajan GJ, Gonzalez C, Shubhangi, Decker PA, Killian JM, Olson JE, St. Sauver JL, Shah P, Vella A, Ryu E, Liu H, Marshall GD, Cerhan JR, Singh D, Summers RL. Predictors of Metformin Failure: Repurposing Electronic Health Record Data to Identify High-Risk Patients. J Clin Endocrinol Metab 2023; 108:1740-1746. [PMID: 36617249 PMCID: PMC10271218 DOI: 10.1210/clinem/dgac759] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023]
Abstract
CONTEXT Metformin is the first-line drug for treating diabetes but has a high failure rate. OBJECTIVE To identify demographic and clinical factors available in the electronic health record (EHR) that predict metformin failure. METHODS A cohort of patients with at least 1 abnormal diabetes screening test that initiated metformin was identified at 3 sites (Arizona, Mississippi, and Minnesota). We identified 22 047 metformin initiators (48% female, mean age of 57 ± 14 years) including 2141 African Americans, 440 Asians, 962 Other/Multiracial, 1539 Hispanics, and 16 764 non-Hispanic White people. We defined metformin failure as either the lack of a target glycated hemoglobin (HbA1c) (<7%) within 18 months of index or the start of dual therapy. We used tree-based extreme gradient boosting (XGBoost) models to assess overall risk prediction performance and relative contribution of individual factors when using EHR data for risk of metformin failure. RESULTS In this large diverse population, we observed a high rate of metformin failure (43%). The XGBoost model that included baseline HbA1c, age, sex, and race/ethnicity corresponded to high discrimination performance (C-index of 0.731; 95% CI 0.722, 0.740) for risk of metformin failure. Baseline HbA1c corresponded to the largest feature performance with higher levels associated with metformin failure. The addition of other clinical factors improved model performance (0.745; 95% CI 0.737, 0.754, P < .0001). CONCLUSION Baseline HbA1c was the strongest predictor of metformin failure and additional factors substantially improved performance suggesting that routinely available clinical data could be used to identify patients at high risk of metformin failure who might benefit from closer monitoring and earlier treatment intensification.
Collapse
Affiliation(s)
- Suzette J Bielinski
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Paul Y Takahashi
- Division of Community Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicholas B Larson
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Alexandra Castillo
- Center for Informatics and Analytics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | - Eleanna De Filippis
- Division of Endocrinology, Diabetes, and Metabolism Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | | | - Gouri J Mahajan
- UMMC Biobank-School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | - Shubhangi
- Mountain Park Health Center, Phoenix, AZ 85012, USA
| | - Paul A Decker
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Jill M Killian
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Janet E Olson
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jennifer L St. Sauver
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN 55905, USA
| | - Pankaj Shah
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Euijung Ryu
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Gailen D Marshall
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - James R Cerhan
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Richard L Summers
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
105
|
Diaz-Thomas AM, Golden SH, Dabelea DM, Grimberg A, Magge SN, Safer JD, Shumer DE, Stanford FC. Endocrine Health and Health Care Disparities in the Pediatric and Sexual and Gender Minority Populations: An Endocrine Society Scientific Statement. J Clin Endocrinol Metab 2023; 108:1533-1584. [PMID: 37191578 PMCID: PMC10653187 DOI: 10.1210/clinem/dgad124] [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: 02/24/2023] [Indexed: 05/17/2023]
Abstract
Endocrine care of pediatric and adult patients continues to be plagued by health and health care disparities that are perpetuated by the basic structures of our health systems and research modalities, as well as policies that impact access to care and social determinants of health. This scientific statement expands the Society's 2012 statement by focusing on endocrine disease disparities in the pediatric population and sexual and gender minority populations. These include pediatric and adult lesbian, gay, bisexual, transgender, queer, intersex, and asexual (LGBTQIA) persons. The writing group focused on highly prevalent conditions-growth disorders, puberty, metabolic bone disease, type 1 (T1D) and type 2 (T2D) diabetes mellitus, prediabetes, and obesity. Several important findings emerged. Compared with females and non-White children, non-Hispanic White males are more likely to come to medical attention for short stature. Racially and ethnically diverse populations and males are underrepresented in studies of pubertal development and attainment of peak bone mass, with current norms based on European populations. Like adults, racial and ethnic minority youth suffer a higher burden of disease from obesity, T1D and T2D, and have less access to diabetes treatment technologies and bariatric surgery. LGBTQIA youth and adults also face discrimination and multiple barriers to endocrine care due to pathologizing sexual orientation and gender identity, lack of culturally competent care providers, and policies. Multilevel interventions to address these disparities are required. Inclusion of racial, ethnic, and LGBTQIA populations in longitudinal life course studies is needed to assess growth, puberty, and attainment of peak bone mass. Growth and development charts may need to be adapted to non-European populations. In addition, extension of these studies will be required to understand the clinical and physiologic consequences of interventions to address abnormal development in these populations. Health policies should be recrafted to remove barriers in care for children with obesity and/or diabetes and for LGBTQIA children and adults to facilitate comprehensive access to care, therapeutics, and technological advances. Public health interventions encompassing collection of accurate demographic and social needs data, including the intersection of social determinants of health with health outcomes, and enactment of population health level interventions will be essential tools.
Collapse
Affiliation(s)
- Alicia M Diaz-Thomas
- Department of Pediatrics, Division of Endocrinology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sherita Hill Golden
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Dana M Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Adda Grimberg
- Department of Pediatrics, Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sheela N Magge
- Department of Pediatrics, Division of Pediatric Endocrinology and Diabetes, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joshua D Safer
- Department of Medicine, Division of Endocrinology, Diabetes, and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10001, USA
| | - Daniel E Shumer
- Department of Pediatric Endocrinology, C.S. Mott Children's Hospital, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Fatima Cody Stanford
- Massachusetts General Hospital, Department of Medicine-Division of Endocrinology-Neuroendocrine, Department of Pediatrics-Division of Endocrinology, Nutrition Obesity Research Center at Harvard (NORCH), Boston, MA 02114, USA
| |
Collapse
|
106
|
Goyal S, Rani J, Bhat MA, Vanita V. Genetics of diabetes. World J Diabetes 2023; 14:656-679. [PMID: 37383588 PMCID: PMC10294065 DOI: 10.4239/wjd.v14.i6.656] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/13/2023] [Accepted: 04/17/2023] [Indexed: 06/14/2023] Open
Abstract
Diabetes mellitus is a complicated disease characterized by a complex interplay of genetic, epigenetic, and environmental variables. It is one of the world's fastest-growing diseases, with 783 million adults expected to be affected by 2045. Devastating macrovascular consequences (cerebrovascular disease, cardiovascular disease, and peripheral vascular disease) and microvascular complications (like retinopathy, nephropathy, and neuropathy) increase mortality, blindness, kidney failure, and overall quality of life in individuals with diabetes. Clinical risk factors and glycemic management alone cannot predict the development of vascular problems; multiple genetic investigations have revealed a clear hereditary component to both diabetes and its related complications. In the twenty-first century, technological advancements (genome-wide association studies, next-generation sequencing, and exome-sequencing) have led to the identification of genetic variants associated with diabetes, however, these variants can only explain a small proportion of the total heritability of the condition. In this review, we address some of the likely explanations for this "missing heritability", for diabetes such as the significance of uncommon variants, gene-environment interactions, and epigenetics. Current discoveries clinical value, management of diabetes, and future research directions are also discussed.
Collapse
Affiliation(s)
- Shiwali Goyal
- Department of Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Rockville, MD 20852, United States
| | - Jyoti Rani
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Mohd Akbar Bhat
- Department of Ophthalmology, Georgetown University Medical Center, Washington DC, DC 20057, United States
| | - Vanita Vanita
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| |
Collapse
|
107
|
Ahn D, Kwon J, Song S, Lee J, Yoon S, Chung SJ. Methyl Syringate Stimulates Glucose Uptake by Inhibiting Protein Tyrosine Phosphatases Relevant to Insulin Resistance. Life (Basel) 2023; 13:1372. [PMID: 37374154 DOI: 10.3390/life13061372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/31/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Several protein tyrosine phosphatases (PTPs), particularly PTPN1, PTPN2, PTPN6, PTPN9, PTPN11, PTPRS, and DUSP9, are involved in insulin resistance. Therefore, these PTPs could be promising targets for the treatment of type 2 diabetes. Our previous studies revealed that PTPN2 and PTPN6 are potential antidiabetic targets. Therefore, the identification of dual-targeting inhibitors of PTPN2 and PTPN6 could be a potential therapeutic strategy for the treatment or prevention of type 2 diabetes. In this study, we demonstrate that methyl syringate inhibits the catalytic activity of PTPN2 and PTPN6 in vitro, indicating that methyl syringate acts as a dual-targeting inhibitor of PTPN2 and PTPN6. Furthermore, methyl syringate treatment significantly increased glucose uptake in mature 3T3-L1 adipocytes. Additionally, methyl syringate markedly enhanced phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) in 3T3L1 adipocytes. Taken together, our results suggest that methyl syringate, a dual-targeting inhibitor of PTPN2 and PTPN6, is a promising therapeutic candidate for the treatment or prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Dohee Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jihee Kwon
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Songyi Song
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jooyoung Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sunyoung Yoon
- Department of Cosmetic Science, Kwangju Women's University, Gwangju 62396, Republic of Korea
| | - Sang J Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
108
|
Husain KH, Sarhan SF, AlKhalifa HKAA, Buhasan A, Moin ASM, Butler AE. Dementia in Diabetes: The Role of Hypoglycemia. Int J Mol Sci 2023; 24:9846. [PMID: 37372995 DOI: 10.3390/ijms24129846] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Hypoglycemia, a common consequence of diabetes treatment, is associated with severe morbidity and mortality and has become a major barrier to intensifying antidiabetic therapy. Severe hypoglycemia, defined as abnormally low blood glucose requiring the assistance of another person, is associated with seizures and comas, but even mild hypoglycemia can cause troubling symptoms such as anxiety, palpitations, and confusion. Dementia generally refers to the loss of memory, language, problem-solving, and other cognitive functions, which can interfere with daily life, and there is growing evidence that diabetes is associated with an increased risk of both vascular and non-vascular dementia. Neuroglycopenia resulting from a hypoglycemic episode in diabetic patients can lead to the degeneration of brain cells, with a resultant cognitive decline, leading to dementia. In light of new evidence, a deeper understating of the relationship between hypoglycemia and dementia can help to inform and guide preventative strategies. In this review, we discuss the epidemiology of dementia among patients with diabetes, and the emerging mechanisms thought to underlie the association between hypoglycemia and dementia. Furthermore, we discuss the risks of various pharmacological therapies, emerging therapies to combat hypoglycemia-induced dementia, as well as risk minimization strategies.
Collapse
Affiliation(s)
- Khaled Hameed Husain
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Saud Faisal Sarhan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | | | - Asal Buhasan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Abu Saleh Md Moin
- Research Department, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| |
Collapse
|
109
|
Várkonyi TT, Pósa A, Pávó N, Pavo I. Perspectives on weight control in diabetes - Tirzepatide. Diabetes Res Clin Pract 2023:110770. [PMID: 37279858 DOI: 10.1016/j.diabres.2023.110770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/08/2023]
Abstract
Tirzepatide, a once-weekly glucose-dependent insulinotropic polypeptide (GIP)/glucagon-like peptide-1 (GLP-1) receptor agonist (GIP/GLP-1 RA) improves glycemic control. Besides improvement of glycemic control, tirzepatide treatment is associated with significantly more weight loss as compared to potent selective GLP-1 receptor agonists as well as other beneficial changes in cardio-metabolic parameters, such as reduced fat mass, blood pressure, improved insulin sensitivity, lipoprotein concentrations, and circulating metabolic profile in individuals with type 2 diabetes (T2D). Some of these changes are partially associated with weight reduction. We review here the putative mechanisms of GIP receptor agonism contributing to GLP-1 receptor agonism-induced weight loss and respective findings with GIP/GLP-1 RAs, including tirzepatide in T2D preclinical models and clinical studies. Subsequently, we summarize the clinical data on weight loss and related non-glycemic metabolic changes of tirzepatide in T2D. These findings suggest that the robust weight loss and associated changes are important contributors to the clinical profile of tirzepatide for the treatment of T2D diabetes and serve as the basis for further investigations including clinical outcomes.
Collapse
Affiliation(s)
- Tamas T Várkonyi
- Department of Internal Medicine, University of Szeged, Kálvária sgt. 57, H-6725 Szeged, Hungary.
| | - Anikó Pósa
- Department of Oral Biology and Experimental Dental Research, University of Szeged, Tisza Lajos krt. 64-66, H-6720 Szeged, Hungary.
| | - Noémi Pávó
- Department of Internal Medicine II, Clinical Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Imre Pavo
- Eli Lilly Regional Operations GmbH, Erdberger Lände 26/A, A-1030 Vienna, Austria.
| |
Collapse
|
110
|
Pulleyblank R, Larsen NB. Cost-Effectiveness of Semaglutide vs. Empagliflozin, Canagliflozin, and Sitagliptin for Treatment of Patients with Type 2 Diabetes in Denmark: A Decision-Analytic Modelling Study. PHARMACOECONOMICS - OPEN 2023:10.1007/s41669-023-00416-z. [PMID: 37178435 DOI: 10.1007/s41669-023-00416-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/10/2023] [Indexed: 05/15/2023]
Abstract
OBJECTIVE The aim was to evaluate the cost-effectiveness of oral and subcutaneous semaglutide versus other oral glucose-lowering drugs (i.e., empagliflozin, canagliflozin, and sitagliptin) for the management of type 2 diabetes (T2D) in Denmark using clinically relevant treatment intensification rules. METHODS A Markov-type cohort model for evaluating the cost-effectiveness of treatment pathways for T2D was used to produce cost-effectiveness estimates based on four head-to-head trials. Evidence from PIONEER 2 and 3 trials was used to evaluate the cost-effectiveness of oral semaglutide vs. empagliflozin and sitagliptin. Evidence from SUSTAIN 2 and 8 trials was used to evaluate the cost-effectiveness of subcutaneous semaglutide vs. sitagliptin and canagliflozin. Base case analyses used trial product estimands of treatment efficacy to avoid the confounding effects of rescue medication use during trials. Deterministic scenario analyses and probabilistic sensitivity analyses were conducted to assess robustness of cost-effectiveness estimates. RESULTS Semaglutide-based treatment regimens were consistently associated with higher lifetime diabetes treatment costs, lower costs of complications, and higher lifetime accumulated QALYs. The PIONEER 2 analysis estimated the cost-effectiveness of oral semaglutide vs. empagliflozin was DKK 150,618/QALY (€20,189). The PIONEER 3 analysis estimated the cost-effectiveness of oral semaglutide vs. sitagliptin was DKK 95,093/QALY (€12,746). The SUSTAIN 2 analysis estimated the cost-effectiveness of subcutaneous semaglutide vs. sitagliptin was DKK 79,982/QALY (€10,721). The SUSTAIN 8 analysis estimated the cost-effectiveness of subcutaneous semaglutide vs. canagliflozin was DKK 167,664/QALY (€22,474). CONCLUSIONS Daily oral and weekly subcutaneous semaglutide are likely to both increase cost and health benefits, but are likely to do so under commonly considered cost-effectiveness thresholds. TRIAL REGISTRATIONS Clinicaltrials.gov: NCT02863328 (PIONEER 2; registered August 11, 2016); NCT02607865 (PIONEER 3; registered November 18, 2015); NCT01930188 (SUSTAIN 2; registered August 28, 2013); NCT03136484 (SUSTAIN 8; registered May 2, 2017).
Collapse
|
111
|
Urakami T. Treatment strategy for children and adolescents with type 2 diabetes-based on ISPAD Clinical Practice Consensus Guidelines 2022. Clin Pediatr Endocrinol 2023; 32:125-136. [PMID: 37362170 PMCID: PMC10288292 DOI: 10.1297/cpe.2023-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/07/2023] [Indexed: 11/03/2023] Open
Abstract
The principles of treatment for children and adolescents with type 2 diabetes include dietary and exercise management. For dietary management, a relatively modest dietary regimen with an appropriate energy source composition is recommended. Moderate- to vigorous-intensity aerobic activity is recommended for at least 60 min/d. Family members are encouraged to modify their lifestyles. Some patients fail to improve hyperglycemia through dietary and exercise management and eventually require pharmacological treatment. If the patient is metabolically stable (HbA1c level < 8.5% [69 mmol/mol]), metformin is the first-line treatment of first choice. In a case with ketosis or HbA1c of more than 8.5% (69 mmol/mol), insulin will be required initially with once daily basal insulin (0.25-0.5 units/kg). The goal of the initial treatment is to attain an HbA1c level < 7.0% (53 mmol/mol). If the glycemic goal is not attained, the addition of a second agent should be considered. However, the use of antihyperglycemic drugs in pediatric patients is limited in most countries. Therefore, the efficacy and safety of these drugs used in adult patients, including GLP-1 receptor agonists and SGLT2 inhibitors, should be evaluated in pediatric patients worldwide.
Collapse
Affiliation(s)
- Tatsuhiko Urakami
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
112
|
Hasebe M, Yoshiji S, Keidai Y, Minamino H, Murakami T, Tanaka D, Fujita Y, Harada N, Hamasaki A, Inagaki N. Efficacy of antihyperglycemic therapies on cardiovascular and heart failure outcomes: an updated meta-analysis and meta-regression analysis of 35 randomized cardiovascular outcome trials. Cardiovasc Diabetol 2023; 22:62. [PMID: 36935489 PMCID: PMC10024854 DOI: 10.1186/s12933-023-01773-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/14/2023] [Indexed: 03/21/2023] Open
Abstract
BACKGROUND Effects of antihyperglycemic therapies on cardiovascular and heart failure (HF) risks have varied widely across cardiovascular outcome trials (CVOTs), and underlying factors remain incompletely understood. We aimed to determine the relationships of glycated hemoglobin (HbA1c) or bodyweight changes with these outcomes in all CVOTs of antihyperglycemic therapies. METHODS We searched PubMed and EMBASE up to 25 January 2023 for all randomized controlled CVOTs of antihyperglycemic therapies reporting both major adverse cardiovascular events (MACE) and HF outcomes in patients with type 2 diabetes or prediabetes. We performed meta-regression analyses following random-effects meta-analyses to evaluate the effects of HbA1c or bodyweight reductions on each outcome. RESULTS Thirty-five trials comprising 256,524 patients were included. Overall, antihyperglycemic therapies reduced MACE by 9% [risk ratio (RR): 0.91; 95% confidence interval (CI) 0.88-0.94; P < 0.001; I2 = 36.5%]. In meta-regression, every 1% greater reduction in HbA1c was associated with a 14% reduction in the RR of MACE (95% CI 4-24; P = 0.010), whereas bodyweight change was not associated with the RR of MACE. The magnitude of the reduction in MACE risk associated with HbA1c reduction was greater in trials with a higher baseline prevalence of atherosclerotic cardiovascular disease. On the other hand, antihyperglycemic therapies showed no overall significant effect on HF (RR: 0.95; 95% CI 0.87-1.04; P = 0.28; I2 = 75.9%). In a subgroup analysis based on intervention type, sodium-glucose cotransporter-2 inhibitors (SGLT2i) conferred the greatest HF risk reduction (RR: 0.68; 95% CI 0.62-0.75; P < 0.001; I2 = 0.0%). In meta-regression, every 1 kg bodyweight reduction, but not HbA1c reduction, was found to reduce the RR of HF by 7% (95% CI 4-10; P < 0.001); however, significant residual heterogeneity (P < 0.001) was observed, and SGLT2i reduced HF more than could be explained by HbA1c or bodyweight reductions. CONCLUSIONS Antihyperglycemic therapies reduce MACE in an HbA1c-dependent manner. These findings indicate that HbA1c can be a useful marker of MACE risk reduction across a wide range of antihyperglycemic therapies, including drugs with pleiotropic effects. In contrast, HF is reduced not in an HbA1c-dependent but in a bodyweight-dependent manner. Notably, SGLT2i have shown class-specific benefits for HF beyond HbA1c or bodyweight reductions.
Collapse
Affiliation(s)
- Masashi Hasebe
- Department of Diabetes and Endocrinology, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan
| | - Satoshi Yoshiji
- Department of Diabetes and Endocrinology, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan.
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Human Genetics, McGill University, Montréal, QC, Canada.
- Kyoto-McGill International Collaborative Program in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Yamato Keidai
- Department of Diabetes and Endocrinology, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroto Minamino
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takaaki Murakami
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Daisuke Tanaka
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshihito Fujita
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Norio Harada
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akihiro Hamasaki
- Department of Diabetes and Endocrinology, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan
| | - Nobuya Inagaki
- Department of Diabetes and Endocrinology, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan.
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
113
|
Chrysohoou C, Fragoulis C, Leontsinis I, Gastouniotis I, Fragouli D, Georgopoulos M, Mantzouranis E, Noutsou M, Tsioufis KP. Cardiometabolic Care: Assessing Patients with Diabetes Mellitus with No Overt Cardiovascular Disease in the Light of Heart Failure Development Risk. Nutrients 2023; 15:1384. [PMID: 36986114 PMCID: PMC10056430 DOI: 10.3390/nu15061384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/02/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
The mechanisms leading to the development of heart failure (HF) in diabetes mellitus (DM) patients are multifactorial. Assessing the risk of HF development in patients with DM is valuable not only for the identification of a high-risk subgroup, but also equally important for defining low-risk subpopulations. Nowadays, DM and HF have been recognized as sharing similar metabolic pathways. Moreover, the clinical manifestation of HF can be independent of LVEF classification. Consequently, approaching HF should be through structural, hemodynamic and functional evaluation. Thus, both imaging parameters and biomarkers are important tools for the recognition of diabetic patients at risk of HF manifestation and HF phenotypes, and arrhythmogenic risk, and eventually for prognosis, aiming to improve patients' outcomes utilizing drugs and non-pharmaceutical cardioprotective tools such as diet modification.
Collapse
Affiliation(s)
- Christina Chrysohoou
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| | - Christos Fragoulis
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| | - Ioannis Leontsinis
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| | - Ioannis Gastouniotis
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| | - Dimitra Fragouli
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| | - Maximos Georgopoulos
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| | - Emmanouil Mantzouranis
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| | - Marina Noutsou
- Diabetes Center, 2nd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokration General Hospital of Athens, 11528 Athens, Greece
| | - Konstantinos P. Tsioufis
- 1st Cardiology Clinic, Hippokration Hospital, National and Kapodistrian University of Athens, 11528 Attica, Greece
| |
Collapse
|
114
|
Nilsson K, Andersson E, Persson S, Karlsdotter K, Skogsberg J, Gustavsson S, Jendle J, Steen Carlsson K. Model-based predictions on health benefits and budget impact of implementing empagliflozin in people with type 2 diabetes and established cardiovascular disease. Diabetes Obes Metab 2023; 25:748-757. [PMID: 36371543 PMCID: PMC10107920 DOI: 10.1111/dom.14921] [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: 08/10/2022] [Revised: 10/31/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
Abstract
AIM To perform a model-based analysis of the short- and long-term health benefits and costs of further increased implementation of empagliflozin for people with type 2 diabetes and established cardiovascular disease (eCVD) in Sweden. MATERIALS AND METHODS The validated Institute for Health Economics Diabetes Cohort Model (IHE-DCM) was used to estimate health benefits and a 3-year budget impact, and lifetime costs per quality-adjusted life years (QALY) gained of increased implementation of adding empagliflozin to standard of care (SoC) for people with type 2 diabetes and eCVD in a Swedish setting. Scenarios with 100%/75%/50% implementation were explored. Analyses were based on 30 model cohorts with type 2 diabetes and eCVD (n = 131 412 at baseline) from national health data registers. Sensitivity analyses explored the robustness of results. RESULTS Over 3 years, SoC with empagliflozin (100% implementation) versus SoC before empagliflozin resulted in 7700 total life years gained and reductions in cumulative incidence of cardiovascular deaths by 30% and heart failures by 28%. Annual costs increased by 15% from higher treatment costs and increased survival. Half of these benefits and costs are not yet reached with current implementation below 50%. SoC with empagliflozin yielded 0.37 QALYs per person, with an incremental cost-effectiveness ratio of 16 000 EUR per QALY versus SoC before empagliflozin. CONCLUSIONS Model simulations using real-world data and trial treatment effects indicated that a broader implementation of empagliflozin, in line with current guidelines for treatment of people with type 2 diabetes and eCVD, would lead to further benefits even from a short-term perspective.
Collapse
Affiliation(s)
| | | | - Sofie Persson
- The Swedish Institute for Health Economics, Lund, Sweden
- Department of Clinical Sciences, Malmö, Health Economics, Lund University, Lund, Sweden
| | | | | | | | - Johan Jendle
- School of Medical Science, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Katarina Steen Carlsson
- The Swedish Institute for Health Economics, Lund, Sweden
- Department of Clinical Sciences, Malmö, Health Economics, Lund University, Lund, Sweden
| |
Collapse
|
115
|
Evaluation the food safety of cultured fat via detection of residues of adipogenic differentiation cocktail in cultured fat with high performance liquid chromatography and enzyme-linked immunosorbent assay. Food Res Int 2023; 165:112486. [PMID: 36869499 DOI: 10.1016/j.foodres.2023.112486] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/14/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Cultured fat is inducing adipose progenitor cells (APCs) to differentiate into mature adipocytes for consumption. The traditional adipogenic differentiation cocktail, including insulin, dexamethasone, indomethacin, isobutylmethylxanthine and rosiglitazone, has potential food safety problems in cultured fat. Therefore, the detection of these residues is necessary to ensure food safety. In this research, a method of high performance liquid chromatography (HPLC) was established to quantitatively analyze the potential residual content of dexamethasone, indomethacin, isobutylmethylxanthine and rosiglitazone in cultured fat and medium. Quantitative analysis showed that the content of four residues in cultured fat decreased to zero on Day 10. Subsequently, enzyme-linked immunosorbent assay (ELISA) was performed to detect the insulin content in the cultured fat and found that the insulin content in the cultured fat on Day 10 was 2.78 ± 0.21 μg/kg. After soaking with phosphate buffered saline (PBS), the insulin content decreased to 1.88 ± 0.54 μg/kg. In conclusion, this research provided an effective approach to clarify the content of potential residual components in cultured fat and it will provide reference for the safety of cultured fat in the future.
Collapse
|
116
|
Dixon SA, Mishra S, Dietsche KB, Jain S, Mabundo L, Stagliano M, Krenek A, Courville A, Yang S, Turner SA, Meyers AG, Estrada DE, Yadav H, Chung ST. The effects of prebiotics on gastrointestinal side effects of metformin in youth: A pilot randomized control trial in youth-onset type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1125187. [PMID: 36909343 PMCID: PMC9996666 DOI: 10.3389/fendo.2023.1125187] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Disclosure summary Dr. Yadav is Chief Scientific Officer and Co-Founder of Postbiotics Inc and has no conflict of interest with this work. All other authors have no conflicts of interest to disclose. Background Metformin is the only approved first-line oral glucose lowering agent for youth with type 2 diabetes mellitus (Y-T2DM) but often causes gastrointestinal (GI) side effects, which may contribute to reduced treatment adherence and efficacy. Prebiotic intake may reduce metformin's side effects by shifting microbiota composition and activity. Objective The aims of this study were to determine the feasibility and tolerability of a prebiotic supplement to improve metformin-induced GI symptoms and explore the changes in glycemia and shifts in the microbiota diversity. Methods In a two-phase pilot clinical trial, we compared, stool frequency and stool form every 1-2 days, and composite lower GI symptoms (weekly) at initiation of daily metformin combined with either a daily prebiotic or a placebo shake in a 1-week randomized double-blind crossover design (Phase 1), followed by a 1-month open-labeled extension (Phase 2). Plasma glycemic markers and stool samples were collected before and after each phase. Results Six Y-T2DM (17.2 ± 1.7y (mean ± SD), 67% male, BMI (42 ± 9 kg/m2), HbA1c (6.4 ± 0.6%)) completed the intervention. Stool frequency, stool composition, and GI symptom scores did not differ by group or study phase. There were no serious or severe adverse events reported, and no differences in metabolic or glycemic markers. After one week Phase 1metformin/placebo Proteobacteria, Enterobacteriaceae, and Enterobacteriales were identified as candidate biomarkers of metformin effects. Principle coordinate analyses of beta diversity suggested that the metformin/prebiotic intervention was associated with distinct shifts in the microbiome signatures at one week and one month. Conclusion Administration of a prebiotic fiber supplement during short-term metformin therapy was well tolerated in Y-T2DM and associated with modest shifts in microbial composition. This study provides a proof-of-concept for feasibility exploring prebiotic-metformin-microbiome interactions as a basis for adjunctive metformin therapy. Clinical trial registration https://clinicaltrials.gov/, identifier NCT04209075.
Collapse
Affiliation(s)
- Sydney A. Dixon
- National Institute of Diabetes & Digestive & Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
| | - Sidharth Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, United States
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Katrina B. Dietsche
- National Institute of Diabetes & Digestive & Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, United States
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Lilian Mabundo
- National Institute of Diabetes & Digestive & Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
| | - Michael Stagliano
- National Institute of Diabetes & Digestive & Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
| | - Andrea Krenek
- National Institute of Diabetes & Digestive & Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
| | - Amber Courville
- National Institute of Diabetes & Digestive & Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
| | - Shanna Yang
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sara A. Turner
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Abby G. Meyers
- Children’s National Hospital (CNH), Washington, DC, United States
| | - Doris E. Estrada
- Children’s National Hospital (CNH), Washington, DC, United States
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, United States
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Stephanie T. Chung
- National Institute of Diabetes & Digestive & Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, United States
- Children’s National Hospital (CNH), Washington, DC, United States
| |
Collapse
|
117
|
Remedi MS, Nichols CG. Glucokinase Inhibition: A Novel Treatment for Diabetes? Diabetes 2023; 72:170-174. [PMID: 36669001 PMCID: PMC9871191 DOI: 10.2337/db22-0731] [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: 08/25/2022] [Accepted: 11/12/2022] [Indexed: 01/21/2023]
Abstract
Chronic hyperglycemia increases pancreatic β-cell metabolic activity, contributing to glucotoxicity-induced β-cell failure and loss of functional β-cell mass, potentially in multiple forms of diabetes. In this perspective we discuss the novel paradoxical and counterintuitive concept of inhibiting glycolysis, particularly by targeted inhibition of glucokinase, the first enzyme in glycolysis, as an approach to maintaining glucose sensing and preserving functional β-cell mass, thereby improving insulin secretion, in the treatment of diabetes.
Collapse
Affiliation(s)
- Maria S. Remedi
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
| | - Colin G. Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
118
|
Rosenstock J, Kolkailah AA, McGuire DK, Espeland MA, Mattheus M, Pfarr E, Lund SS, Marx N. Incident and recurrent hypoglycaemia with linagliptin and glimepiride over a median of 6 years in the CAROLINA cardiovascular outcome trial. Diabetes Obes Metab 2023; 25:1453-1463. [PMID: 36700416 DOI: 10.1111/dom.14991] [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: 09/23/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
AIM The CAROLINA trial established non-inferiority of linagliptin versus glimepiride for major adverse cardiovascular events in patients with relatively early type 2 diabetes at increased cardiovascular risk. In pre-specified and post-hoc analyses, we investigated treatment effects on total hypoglycaemic burden in CAROLINA. MATERIALS AND METHODS Patients were randomized and treated with 5 mg linagliptin (n = 3014) or 1-4 mg glimepiride (n = 3000) once daily added to standard care. Hypoglycaemia captured from investigator-reported adverse events was analysed with Poisson and negative binomial regressions for the first and total (first plus recurrent) events, respectively. The influence of insulin initiation and glycated haemoglobin (HbA1c) change on the treatment effect for hypoglycaemia was also explored. RESULTS Over 6.3 years median follow-up, average HbA1c over time did not differ between linagliptin versus glimepiride (weighted mean difference [95% confidence interval]: 0.00%, [-0.05, 0.05]), nor did insulin initiation (18.6% vs. 19.2% of patients, respectively), whereas body weight was lower with linagliptin (-1.54 kg, [-1.80, -1.28]). Hypoglycaemia frequency was lower with linagliptin across all hypoglycaemia categories, including severe episodes. Rate ratios (95% confidence interval) for first and total events for investigator-reported hypoglycaemia were 0.21 (0.19-0.24) and 0.12 (0.10-0.14), respectively, with 8.7 first and 60.8 total estimated events prevented/100 patient-years with linagliptin versus glimepiride. These differences occurred during night-time and daytime, and in subgroup analyses of total events. Treatment differences in hypoglycaemia were neither impacted by HbA1c changes nor insulin initiation. CONCLUSIONS Across the severity spectrum, linagliptin substantially reduced the hypoglycaemic burden versus glimepiride in patients with relatively early type 2 diabetes at increased cardiovascular risk.
Collapse
Affiliation(s)
| | - Ahmed A Kolkailah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Parkland Health and Hospital System, Dallas, Texas, USA
| | - Darren K McGuire
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Parkland Health and Hospital System, Dallas, Texas, USA
| | - Mark A Espeland
- Departments of Internal Medicine and Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Egon Pfarr
- Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Søren S Lund
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
119
|
Wagner N, Wagner KD. Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease. Int J Mol Sci 2023; 24:ijms24032345. [PMID: 36768666 PMCID: PMC9916802 DOI: 10.3390/ijms24032345] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Peroxisome proliferator activated receptors, including PPARα, PPARβ/δ, and PPARγ, are ligand-activated transcription factors belonging to the nuclear receptor superfamily. They play important roles in glucose and lipid metabolism and are also supposed to reduce inflammation and atherosclerosis. All PPARs are involved in angiogenesis, a process critically involved in cardiovascular pathology. Synthetic specific agonists exist for all PPARs. PPARα agonists (fibrates) are used to treat dyslipidemia by decreasing triglyceride and increasing high-density lipoprotein (HDL) levels. PPARγ agonists (thiazolidinediones) are used to treat Type 2 diabetes mellitus by improving insulin sensitivity. PPARα/γ (dual) agonists are supposed to treat both pathological conditions at once. In contrast, PPARβ/δ agonists are not in clinical use. Although activators of PPARs were initially considered to have favorable effects on the risk factors for cardiovascular disease, their cardiovascular safety is controversial. Here, we discuss the implications of PPARs in vascular biology regarding cardiac pathology and focus on the outcomes of clinical studies evaluating their benefits in cardiovascular diseases.
Collapse
|
120
|
Wu H, Lau ESH, Yang A, Fan B, Ma RCW, Kong APS, Chow E, So WY, Chan JCN, Luk AOY. Real world evidence of clinical predictors of glycaemic response to glucose-lowering drugs among Chinese with type 2 diabetes. Diabetes Metab Res Rev 2023; 39:e3615. [PMID: 36652944 DOI: 10.1002/dmrr.3615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/03/2022] [Accepted: 12/15/2022] [Indexed: 01/20/2023]
Abstract
AIMS To examine whether simple clinical features can predict the 1-year glycaemic response to glucose-lowering drugs (GLDs) among Chinese with type 2 diabetes. MATERIALS AND METHODS We used data from a diabetes risk assessment and complication screening programme and electronic medical records. We used linear regression models to examine the association between clinical features and 1-year glycaemic response to GLDs. RESULTS Use of metformin (n = 15,433), sulphonylureas (SU) (n = 15,190), dipeptidyl peptidase-4 inhibitor (DPP-4i) (n = 7947), thiazolidinedione (TZD) (n = 4107), and sodium-glucose cotransporter 2 inhibitors (SGLT-2i) (n = 1883) were associated with a mean reduction of HbA1c ranging from 0.7% to 1.3% at one year. Men had a greater response to SU but a poorer response to metformin and TZD. Older age predicted a better response to all GLDs but not SGLT-2i, whereas increasing diabetes duration was associated with a poorer response to all GLDs except for DPP-4i. Obese patients responded greater to TZD and SGLT-2i but poorer to SU than those with normal weight. Patients with a higher level of triglycerides to high-density lipoprotein cholesterol ratio had a greater glycaemic response to TZD but a smaller response to SU and DPP-4i. CONCLUSIONS Glycaemic response to GLDs differed considerably by clinical features among Chinese patients with type 2 diabetes.
Collapse
Affiliation(s)
- Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Eric S H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Aimin Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing-Yee So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Hospital Authority, Hong Kong, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
121
|
Synthesis, Characterization, and Pharmacokinetic Studies of Thiazolidine-2,4-Dione Derivatives. J CHEM-NY 2023. [DOI: 10.1155/2023/9462176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Various derivatives of thiazolidine-2,4-dione (C1–C5) were designed and synthesized by chemical reaction with 4-nitrobenzaldehyde using Knoevenagel reaction conditions which results in the reduction of nitro group to amine and further modification results in target compounds. The chemical structures of all the 2,4-thiazolidinedione derivatives have been elucidated by 1H and 13C NMR spectroscopy. These compounds were further characterized by in silico ADME (absorption, distribution, metabolism, and excretion) studies. The pharmacokinetic properties were assessed by SwissADME software. The in silico ADME (absorption, distribution, metabolism, and excretion) assessment reveals that all derivatives (C1 to C5) have 5 to 7 rotatable bonds. Lipophilicity and water solubility showed that C1, C2, and C4 are water soluble except for C3 and C5 which are moderately soluble. All the compounds have high GI absorption except C3. None of the derivatives are blood-brain barrier permeant. Drug metabolism of TZDs derivatives showed that C3 was identified as an inhibitor of CYP2C9 and C5 as an inhibitor of CYP1A2 and CYP2C19. Drug likeness properties indicate that C1 has only one violation of the Ghose rule while C3 has violations in the Ghose and Egan rules. The in silico pharmacokinetic studies revealed high GI absorption and the inability to pass blood-brain barrier which can be further assessed by in vitro and in vivo antihyperglycemic activity. This study will contribute to providing TZDs derivatives with an improved pharmacokinetic profile and decreased toxicity.
Collapse
|
122
|
Salam M, Rana M, Baral P, Rahman M, Ahmed S, Rahman R, Jahan N, Mazumder T, Islam M, Hussain M. Glipizide has Low Influences on Lipid Index and Major Organs Weight Variation and Considerable Anxiolytic Properties: An in vivo Investigation. JOURNAL OF MEDICAL SCIENCES 2023. [DOI: 10.3923/jms.2023.7.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
123
|
Grether U, Benz J, Hartung T, Roth D. Scintillation Proximity Assay (SPA)-Based Radioligand Binding for PPARα, PPARγ, and PPARδ Receptors. Methods Mol Biol 2023; 2576:145-153. [PMID: 36152183 DOI: 10.1007/978-1-0716-2728-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been exploited as drug targets for combating multiple diseases. Several activators with different selectivity for the PPAR α, γ, and δ subtypes have been introduced into the market or have reached advanced clinical trials. Binding assays are of utmost importance for the discovery and profiling of such PPAR ligands. Binding assays are often based on radioligands, in particular, tritiated molecules are applied. We developed synthetic procedures for tritiating various PPAR agonists and applied these radioligands for setting up a scintillation proximity assay (SPA) for PPAR α, γ, and δ. These SPAs allow to assess the binding affinities of PPAR α, γ, and δ ligands, along with their respective subtype selectivity profiles. Therefore, SPA is an important tool for hit discovery and lead optimization campaigns aimed at identifying next-generation PPAR ligands.
Collapse
Affiliation(s)
- Uwe Grether
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Jörg Benz
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Thomas Hartung
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Doris Roth
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
124
|
Dutta D, Bhattacharya S, Kumar M, Datta PK, Mohindra R, Sharma M. Efficacy and safety of novel thiazolidinedione lobeglitazone for managing type-2 diabetes a meta-analysis. Diabetes Metab Syndr 2023; 17:102697. [PMID: 36580702 DOI: 10.1016/j.dsx.2022.102697] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS No meta-analysis has analysed the safety and efficacy of lobeglitazone in type-2 diabetes (T2DM). We undertook this meta-analysis to address this knowledge-gap. METHODS Electronic databases were searched for RCTs involving type-2 diabetes patients receiving lobeglitazone in intervention arm, and placebo/active comparator in control arm. Primary outcome was to evaluate changes in HbA1c. Secondary outcomes were to evaluate alterations in glucose, lipids and adverse events. RESULTS From initially screened 65 articles, data from 4 RCTs (828 patients) which fulfilled all criteria was analysed. Over 24 weeks, when compared to sitagliptin 100 mg/d and half maximal pioglitazone dose (15 mg/d), lobeglitazone 0.5 mg/day had comparable impact on HbA1c [MD 0.03% (95%CI: 0.11-0.17); P = 0.65; I2 = 0%], fasting glucose [MD 1.47 mg/dl (95%CI: 4.66-7.60); P = 0.64; I2 = 0%], triglycerides [MD-9.96 mg/dl (95%CI: 43.55-23.62); P = 0.56; I2 = 81%], LDL-cholesterol [MD0.74 mg/dl (95%CI: 4.60-6.09); P = 0.79; I2 = 0%] and HDL-cholesterol [MD1.55 mg/dl (95%CI: 3.72-6.82); P = 0.56]. Occurrence of treatment-emergent adverse events (AEs) [RR 1.07 (95% CI:0.78-1.47); P = 0.67; I2 = 0%] and severe AEs [RR 1.05(95%CI: 0.42-2.65); P = 0.91; I2 = 0%] were similar. Edema and weight gain were significantly higher with lobeglitazone compared to controls [RR 2.58 (95%CI: 1.08-6.17); P = 0.03; I2 = 0%]. Lobeglitazone 0.5 mg/d compared to half-maximal pioglitazone (15 mg/d), had similar edema and weight gain [RR 1.65 95% CI: 0.78-1.47)]. BMD percent changes at neck of femur was comparable in both groups [MD 0.07% (95%CI: 0.19-0.33); P = 0.60; I2 = 91%]. Low dose lobeglitazone (0.25 mg/d) was inferior to high dose lobeglitazone (0.5 mg/d) with regards to glycaemic efficacy with advantage of lower weight gain and edema. CONCLUSION The current evidence makes lobeglitazone unlikely to replace pioglitazone as the preferred thiazolidinedione in T2DM.
Collapse
Affiliation(s)
- Deep Dutta
- Department of Endocrinology, CEDAR Superspeciality Healthcare, Dwarka, New Delhi, India.
| | | | - Manoj Kumar
- Department of Endocrinology, CEDAR Superspeciality Healthcare, Zirakpur, Punjab, India.
| | - Priyankar K Datta
- Department of Anaesthesiology, Critical Care and Pain Medicine, All India Institute of Medical Sciences, New Delhi, India.
| | - Ritin Mohindra
- Department of Medicine, Post-graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Meha Sharma
- Department of Rheumatology, CEDAR Superspeciality Healthcare, Dwarka, New Delhi, India.
| |
Collapse
|
125
|
Arioglu-Inan E, Kayki-Mutlu G. Sex Differences in Glucose Homeostasis. Handb Exp Pharmacol 2023; 282:219-239. [PMID: 37439847 DOI: 10.1007/164_2023_664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Sexual dimorphism has been demonstrated to have an effect on various physiological functions. In this regard, researchers have investigated its impact on glucose homeostasis in both preclinical and clinical studies. Sex differences mainly arise from physiological factors such as sex hormones, body fat and muscle distribution, and sex chromosomes. The sexual dimorphism has also been studied in the context of diabetes. Reflecting the prevalence of the disease among the population, studies focusing on the sex difference in type 1 diabetes (T1D) are not common as the ones in type 2 diabetes (T2D). T1D is reported as the only major specific autoimmune disease that exhibits a male predominance. Clinical studies have demonstrated that impaired fasting glucose is more frequent in men whereas women more commonly exhibit impaired glucose tolerance. Understanding the sex difference in glucose homeostasis becomes more attractive when focusing on the findings that highlight sexual dimorphism on the efficacy or adverse effect profile of antidiabetic medications. Thus, in this chapter, we aimed to discuss the impact of sex on the glucose homeostasis both in health and in diabetes.
Collapse
Affiliation(s)
- Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey.
| | - Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
126
|
Liu L, Ruan Z, Ung COL, Zhang Y, Shen Y, Han S, Jia R, Qiao J, Hu H, Guo L. Long-Term Cost-Effectiveness of Subcutaneous Once-Weekly Semaglutide Versus Polyethylene Glycol Loxenatide for Treatment of Type 2 Diabetes Mellitus in China. Diabetes Ther 2023; 14:93-107. [PMID: 36414806 PMCID: PMC9880095 DOI: 10.1007/s13300-022-01336-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE This study aimed to evaluate the long-term cost-effectiveness of once-weekly subcutaneous semaglutide versus polyethylene glycol loxenatide (PEG-loxenatide) in patients with type 2 diabetes uncontrolled on metformin, from a Chinese healthcare systems perspective. METHODS The study applied the Swedish Institute of Health Economics Diabetes Cohort Model to evaluate the long-term clinical and economic outcomes of once-weekly treatment of semaglutide at 0.5 mg and 1.0 mg, respectively, versus PEG-loxenatide 0.2 mg, over a 40-year time horizon. Baseline cohort characteristics were collected from the SUSTAIN China trial. A network meta-analysis was conducted to obtain comparative treatment effects of once-weekly semaglutide and PEG-loxenatide based on two phase 3a clinical trials. Drug costs were sourced from the national bidding price of China. Outcomes were discounted at 5.0% per annum. One-way sensitivity analysis and probabilistic sensitivity analysis were conducted to assess the uncertainty of the base-case results. RESULTS When compared with PEG-loxenatide 0.2 mg, the projections of outcomes over the 40-year time horizon in patients with type 2 diabetes uncontrolled on metformin showed that treatment with once-weekly semaglutide 0.5 mg and 1.0 mg were associated with improved discounted life expectancy by 0.08 and 0.12 years, and improved discounted quality-adjusted life expectancy by 0.16 and 0.22 quality-adjusted life-years, respectively. Once-weekly semaglutide 0.5 mg and 1.0 mg were achieved at lifetime cost savings of 19,309 China Yuan (CNY) and 10,179 CNY, respectively. Sensitivity analyses verified the robustness of the results. CONCLUSION From the perspective of Chinese healthcare systems, treatment with once-weekly subcutaneous semaglutide represents a dominant option versus PEG-loxenatide for patients with type 2 diabetes uncontrolled on metformin.
Collapse
Affiliation(s)
- Lei Liu
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhen Ruan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Carolina Oi Lam Ung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Yawen Zhang
- Novo Nordisk (China) Pharmaceuticals Co., Ltd., Beijing, China
| | - Yang Shen
- School of Public Health, Peking University, Beijing, China
| | - Sheng Han
- International Research Center for Medicinal Administration, Peking University, Beijing, China
| | - Ruxu Jia
- Global Business School for Health, University College London, Gower Street, London, WC1E 6BT UK
| | - Jingtao Qiao
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hao Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
127
|
Combined Effects of Lycopene and Metformin on Decreasing Oxidative Stress by Triggering Endogenous Antioxidant Defenses in Diet-Induced Obese Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238503. [PMID: 36500596 PMCID: PMC9737677 DOI: 10.3390/molecules27238503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022]
Abstract
Since lycopene has antioxidant activity, its combination with metformin may be useful to contrast diabetic complications related to oxidative stress. This study aimed to investigate the effects of metformin combined with lycopene on high-fat diet (HFD)-induced obese mice. Seventy-two C57BL-6J mice were divided into six groups: C (control diet-fed mice), H (HFD-fed mice for 17 weeks), H-V (HFD-fed mice treated with vehicle), H-M (HFD-fed mice treated with 50 mg/kg metformin), H-L (HFD-fed mice treated with 45 mg/kg lycopene), and H-ML (HFD-fed mice treated with 50 mg/kg metformin + 45 mg/kg lycopene). Treatments were administered for 8 weeks. Glucose tolerance, insulin sensitivity, fluorescent AGEs (advanced glycation end products), TBARS (thiobarbituric acid-reactive substances), and activities of antioxidant enzymes paraoxonase-1 (PON-1; plasma), superoxide dismutase, catalase and glutathione peroxidase (liver and kidneys) were determined. Metformin plus lycopene reduced body weight; improved insulin sensitivity and glucose tolerance; and decreased AGEs and TBARS in plasma, liver and kidneys. Combined therapy significantly increased the activities of antioxidant enzymes, mainly PON-1. Lycopene combined with metformin improved insulin resistance and glucose tolerance, and caused further increases in endogenous antioxidant defenses, arising as a promising therapeutic strategy for combating diabetic complications resulting from glycoxidative stress.
Collapse
|
128
|
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: 453] [Impact Index Per Article: 151.0] [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
|
129
|
Preechasuk L, Khaedon N, Lapinee V, Tangjittipokin W, Srivanichakorn W, Sriwijitkamol A, Plengvidhya N, Likitmaskul S, Thongtang N. Cluster analysis of Thai patients with newly diagnosed type 2 diabetes mellitus to predict disease progression and treatment outcomes : A prospective cohort study. BMJ Open Diabetes Res Care 2022; 10:10/6/e003145. [PMID: 36581330 PMCID: PMC9806077 DOI: 10.1136/bmjdrc-2022-003145] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2D) is highly heterogeneous in disease progression and risk of complications. This study aimed to categorize Thai T2D into subgroups using variables that are commonly available based on routine clinical parameters to predict disease progression and treatment outcomes. RESEARCH DESIGN AND METHODS This was a cohort study. Data-driven cluster analysis was performed using a Python program in patients with newly diagnosed T2D (n=721) of the Siriraj Diabetes Registry using five variables (age, body mass index (BMI), glycated hemoglobin (HbA1c), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C)). Disease progression and risk of diabetic complications among clusters were compared using the Χ2 and Kruskal-Wallis test. Cox regression and the Kaplan-Meier curve were used to compare the time to diabetic complications and the time to insulin initiation. RESULTS The mean age was 53.4±11.3 years, 58.9% were women. The median follow-up time was 21.1 months (9.2-35.2). Four clusters were identified: cluster 1 (18.6%): high HbA1c, low BMI (insulin-deficiency diabetes); cluster 2 (11.8%): high TG, low HDL-C, average age and BMI (metabolic syndrome group); cluster 3 (23.3%): high BMI, low HbA1c, young age (obesity-related diabetes); cluster 4 (46.3%): older age and low HbA1c at diagnosis (age-related diabetes). Patients in cluster 1 had the highest prevalence of insulin treatment. Patients in cluster 2 had the highest risk of diabetic kidney disease and diabetic retinopathy. Patients in cluster 4 had the lowest prevalence of diabetic retinopathy, nephropathy, and insulin use. CONCLUSIONS We were able to categorize Thai patients with newly diagnosed T2D into four clusters using five routine clinical parameters. This clustering method can help predict disease progression and risk of diabetic complications similar to previous studies using parameters including insulin resistance and insulin sensitivity markers.
Collapse
Affiliation(s)
- Lukana Preechasuk
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Naichanok Khaedon
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Varisara Lapinee
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Watip Tangjittipokin
- Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Weerachai Srivanichakorn
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Apiradee Sriwijitkamol
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nattachet Plengvidhya
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supawadee Likitmaskul
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nuntakorn Thongtang
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
130
|
DeMarsilis A, Reddy N, Boutari C, Filippaios A, Sternthal E, Katsiki N, Mantzoros C. Pharmacotherapy of type 2 diabetes: An update and future directions. Metabolism 2022; 137:155332. [PMID: 36240884 DOI: 10.1016/j.metabol.2022.155332] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Type 2 diabetes (T2D) is a widely prevalent disease with substantial economic and social impact for which multiple conventional and novel pharmacotherapies are currently available; however, the landscape of T2D treatment is constantly changing as new therapies emerge and the understanding of currently available agents deepens. This review aims to provide an updated summary of the pharmacotherapeutic approach to T2D. Each class of agents is presented by mechanism of action, details of administration, side effect profile, cost, and use in certain populations including heart failure, non-alcoholic fatty liver disease, obesity, chronic kidney disease, and older individuals. We also review targets of novel therapeutic T2D agent development. Finally, we outline an up-to-date treatment approach that starts with identification of an individualized goal for glycemic control then selection, initiation, and further intensification of a personalized therapeutic plan for T2D.
Collapse
Affiliation(s)
- Antea DeMarsilis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Niyoti Reddy
- Department of Medicine, School of Medicine, Boston University, Boston, USA
| | - Chrysoula Boutari
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Andreas Filippaios
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Elliot Sternthal
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Sindos, Greece; School of Medicine, European University Cyprus, Nicosia, Cyprus.
| | - Christos Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
131
|
Kim BK, Seo KW. Changes of Guidelines in the Management of Obese Patients With Diabetes in the Metabolic Surgery Perspective. JOURNAL OF METABOLIC AND BARIATRIC SURGERY 2022; 11:13-19. [PMID: 36926674 PMCID: PMC10011678 DOI: 10.17476/jmbs.2022.11.2.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 03/12/2023]
Abstract
Recently, metabolic surgery was recommended for patients with a BMI of 30 or higher and 27.5 or higher for Asians, as evidence level A. Until 2008, bariatric surgery was not introduced for the treatment of diabetes. Bariatric surgery was first recommended for adults with body mass index ≥35 kg/m2 and type 2 diabetes in the American Diabetes Association (ADA) guidelines as evidence level B in 2009. In 2017, the terminology was changed from bariatric surgery to metabolic surgery. How such large changes could have occurred in the ADA guidelines? Because many patients have reached diabetes remission through metabolic surgery, and the long-term benefit and cost-effectiveness have been strongly proven by historical randomized controlled trials and high-quality studies. This review demonstrates how the recommendations for the treatment of obesity in patients with diabetes have changed in diabetes treatment guidelines and summarizes the evidence behind this change.
Collapse
Affiliation(s)
- Bu Kyung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Kyung Won Seo
- Department of Surgery, Kosin University College of Medicine, Busan, Korea
| |
Collapse
|
132
|
Lim LL, Lau ESH, Ozaki R, So TTY, Wong RYM, Chow EYK, Ma RCW, Luk AOY, Chan JCN, Kong APS. Team-based multicomponent care improved and sustained glycaemic control in obese people with type 2 diabetes (T2D) in a Diabetes Centre setting: A quality improvement program with quasi-experimental design. Diabetes Res Clin Pract 2022; 194:110138. [PMID: 36328212 DOI: 10.1016/j.diabres.2022.110138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To evaluate the effect of a team-based multi-component intervention care (MIC) program in obese patients with type 2 diabetes (T2D) and poor glycemic control. METHODS Patients with T2D and HbA1c ≥ 8 % and body mass index (BMI) ≥ 27 kg/m2 and/or waist circumference ≥ 80 cm in women and ≥90 cm in men were recruited. The intervention in Diabetes Centre included 1) nurse-led, group-based workshops; 2) review by endocrinologists; 3) telephone reminders by healthcare assistants and 4) peer support during visits. The usual care (UC) group received consultations at outpatient clinic without workshops or peer support. The MIC group received UC after 1-year of intervention. The primary outcome was change of HbA1c from baseline at 1- and 3-year. RESULTS Of 207 eligible patients [age (mean ± standard deviation): 56.9 ± 8.8 years, 47.4 % men, disease duration: 13.5 ± 8.2 years, HbA1c: 9.6 ± 1.3 %, BMI: 28.8 ± 4.3 kg/m2, waist circumference: 101.5 ± 9.9 cm (men), 95.3 ± 9.8 cm (women)], 104 received MIC and 103 received UC. 95 % patients had repeat assessments at 1- and 3-year. After adjustment for confounders, MIC had greater HbA1c reduction (β -0.51, 95 % confidence interval [CI] -1.00 to -0.01; P = 0.045) than UC at 1-year, with sustained improvement at 3-year (β -0.56, CI -1.10 to -0.02; P = 0.044). CONCLUSION Team-based MIC for 1 year improved glycemic control in obese T2D which was sustained at 3-year.
Collapse
Affiliation(s)
- Lee-Ling Lim
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region; Asia Diabetes Foundation, Hong Kong Special Administrative Region; Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Eric S H Lau
- Asia Diabetes Foundation, Hong Kong Special Administrative Region
| | - Risa Ozaki
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region
| | - Tammy T Y So
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region
| | - Rebecca Y M Wong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region
| | - Elaine Y K Chow
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, CUHK, Hong Kong Special Administrative Region
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, CUHK, Hong Kong Special Administrative Region
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region; Asia Diabetes Foundation, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, CUHK, Hong Kong Special Administrative Region
| | - Alice P S Kong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, CUHK, Hong Kong Special Administrative Region.
| |
Collapse
|
133
|
Tsai CJ, Tsao CF. Comparison of Glucose Lowering Efficacy of Human GLP-1 Agonist in Taiwan Type 2 Diabetes Patients after Switching from DPP-4 Inhibitor Use or Non-Use. J Pers Med 2022; 12:jpm12111915. [PMID: 36422091 PMCID: PMC9698961 DOI: 10.3390/jpm12111915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
To determine the efficacy of glucose control in type 2 diabetes patients who switch from dipeptidyl peptidase-4 (DPP-4) inhibitors use or non-use to GLP-1 receptor agonists (GLP-1 RAs). We conducted a cohort study using data from the Chang Gung Research Database. Patients aged ≥18 years using newly initiated GLP-1 RAs between 1 January 2009, and 31 December 2016, were included. Cox proportional hazards models were used to adjust for treatment selection bias. The primary outcome was changes in the glycated hemoglobin (HbA1c) level. The HbA1c level fell substantially after initiating GLP-1 RAs in DPP-4 inhibitor users and nonusers. A mean HbA1c reduction of −0.42% was found in patients who received DPP-4 inhibitors. Those who were DPP-4 inhibitor nonusers had a reduction in HbA1c of −0.99%. The degree of reduction in HbA1c was significantly greater in patients who were DPP-4 inhibitor nonusers (p value < 0.01), compared to the DPP-4 inhibitor users. In routine care, DPP-4 inhibitor nonusers had better efficacy in glucose control than DPP-4 inhibitor users after switching to a GLP-1 agonist.
Collapse
|
134
|
Causal deep learning reveals the comparative effectiveness of antihyperglycemic treatments in poorly controlled diabetes. Nat Commun 2022; 13:6921. [PMID: 36376286 PMCID: PMC9663714 DOI: 10.1038/s41467-022-33732-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Type-2 diabetes is associated with severe health outcomes, the effects of which are responsible for approximately 1/4th of the total healthcare spending in the United States (US). Current treatment guidelines endorse a massive number of potential anti-hyperglycemic treatment options in various combinations. Strategies for optimizing treatment selection are lacking. Real-world data from a nationwide population of over one million high-risk diabetic patients (HbA1c ≥ 9%) in the US is analyzed to evaluate the comparative effectiveness for HbA1c reduction in this population of more than 80 different treatment strategies ranging from monotherapy up to combinations of five concomitant classes of drugs across each of 10 clinical cohorts defined by age, insulin dependence, and a number of other chronic conditions. A causal deep learning approach developed on such data allows for more personalized evaluation of treatment selection. An average confounder-adjusted reduction in HbA1c of 0.69% [-0.75, -0.65] is observed between patients receiving high vs low ranked treatments across cohorts for which the difference was significant. This method can be extended to explore treatment optimization for other chronic conditions.
Collapse
|
135
|
Predicting Factors for Metabolic Non-Response to a Complex Lifestyle Intervention-A Replication Analysis to a Randomized-Controlled Trial. Nutrients 2022; 14:nu14224721. [PMID: 36432409 PMCID: PMC9699496 DOI: 10.3390/nu14224721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND T2DM heterogeneity affects responsiveness to lifestyle treatment. Beta-cell failure and nonalcoholic fatty liver disease (NAFLD) independently predict T2DM, but NAFLD inconsistently predicts metabolic response to lifestyle intervention. AIM We attempt to replicate a prediction model deducted from the Tübinger Lifestyle Intervention Program by assessing similar metabolic factors to predict conversion to normal glucose regulation (NGR) in a comparable lifestyle intervention trial. METHODS In the Optimal Fiber Trial (OptiFiT), 131 Caucasian participants with prediabetes completed a one-year lifestyle intervention program and received a fiber or placebo supplement. We compared baseline parameters for responders and non-responders, assessed correlations of major metabolic changes and conducted a logistic regression analysis for predictors of remission to NGR. RESULTS NGR was achieved by 33 participants, respectively. At baseline, for the placebo group only, 1 h and 2 h glucose levels, glucose AUC and Cederholm index predicted conversion to NGR. HOMA-beta, HOMA-IR or liver fat indices did not differ between responders and non-responders of the placebo or the fiber group. Changes in waist circumference or fatty liver index correlated with changes in glycemia and insulin resistance, but not with changes in insulin secretion. Insulin-resistant NAFLD did not predict non-response. Differences in compliance did not explain the results. CONCLUSIONS Higher post-challenge glucose levels strongly predicted the metabolic non-response to complex lifestyle intervention in our cohort. Depending on the specific intervention and the investigated cohort, fasting glucose levels and insulin sensitivity might contribute to the risk pattern. Beta-cell function did not improve in accordance with other metabolic improvements, qualifying as a potential risk factor for non-response. We could not replicate previous data suggesting that an insulin-resistant fatty liver is a specific risk factor for treatment failure. Replication studies are required.
Collapse
|
136
|
Alassad Z, AboRaed A, Mizrachi MS, Pérez-Temprano MH, Milo A. Metal-Free Multicomponent Strategy for Amidine Synthesis. J Am Chem Soc 2022; 144:20672-20679. [DOI: 10.1021/jacs.2c07918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Zayed Alassad
- Department of Chemistry, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Anas AboRaed
- Department of Chemistry, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Meital Shema Mizrachi
- Department of Chemistry, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Mónica H. Pérez-Temprano
- Institute of Chemical Research of Catalonia (ICIQ), Barcelona Institute of Science and Technology (BIST), Tarragona 43007, Spain
| | - Anat Milo
- Department of Chemistry, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
137
|
Affiliation(s)
- Eun-Hee Cho
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
- Corresponding author: Eun-Hee Cho https://orcid.org/0000-0002-1349-8894 Department of Internal Medicine, Kangwon National University School of Medicine, 1 Gangwondaehak-gil, Chuncheon 24341, Korea E-mail:
| |
Collapse
|
138
|
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: 775] [Impact Index Per Article: 258.3] [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
|
139
|
Abstract
Data generated over nearly two decades clearly demonstrate the importance of epigenetic modifications and mechanisms in the pathogenesis of type 2 diabetes. However, the role of pharmacoepigenetics in type 2 diabetes is less well established. The field of pharmacoepigenetics covers epigenetic biomarkers that predict response to therapy, therapy-induced epigenetic alterations as well as epigenetic therapies including inhibitors of epigenetic enzymes. Not all individuals with type 2 diabetes respond to glucose-lowering therapies in the same way, and there is therefore a need for clinically useful biomarkers that discriminate responders from non-responders. Blood-based epigenetic biomarkers may be useful for this purpose. There is also a need for a better understanding of whether existing glucose-lowering therapies exert their function partly through therapy-induced epigenetic alterations. Finally, epigenetic enzymes may be drug targets for type 2 diabetes. Here, I discuss whether pharmacoepigenetics is clinically relevant for type 2 diabetes based on studies addressing this topic.
Collapse
Affiliation(s)
- Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden.
| |
Collapse
|
140
|
Florez JC, Pearson ER. A roadmap to achieve pharmacological precision medicine in diabetes. Diabetologia 2022; 65:1830-1838. [PMID: 35748917 PMCID: PMC9522818 DOI: 10.1007/s00125-022-05732-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
Abstract
Current pharmacological treatment of diabetes is largely algorithmic. Other than for cardiovascular disease or renal disease, where sodium-glucose cotransporter 2 inhibitors and/or glucagon-like peptide-1 receptor agonists are indicated, the choice of treatment is based upon overall risks of harm or side effect and cost, and not on probable benefit. Here we argue that a more precise approach to treatment choice is necessary to maximise benefit and minimise harm from existing diabetes therapies. We propose a roadmap to achieve precision medicine as standard of care, to discuss current progress in relation to monogenic diabetes and type 2 diabetes, and to determine what additional work is required. The first step is to identify robust and reliable genetic predictors of response, recognising that genotype is static over time and provides the skeleton upon which modifiers such as clinical phenotype and metabolic biomarkers can be overlaid. The second step is to identify these metabolic biomarkers (e.g. beta cell function, insulin sensitivity, BMI, liver fat, metabolite profile), which capture the metabolic state at the point of prescribing and may have a large impact on drug response. Third, we need to show that predictions that utilise these genetic and metabolic biomarkers improve therapeutic outcomes for patients, and fourth, that this is cost-effective. Finally, these biomarkers and prediction models need to be embedded in clinical care systems to enable effective and equitable clinical implementation. Whilst this roadmap is largely complete for monogenic diabetes, we still have considerable work to do to implement this for type 2 diabetes. Increasing collaborations, including with industry, and access to clinical trial data should enable progress to implementation of precision treatment in type 2 diabetes in the near future.
Collapse
Affiliation(s)
- Jose C Florez
- Center for Genomic Medicine and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard & MIT, Cambridge, MA, USA.
| | - Ewan R Pearson
- Department of Population Health & Genomics, School of Medicine, University of Dundee, Dundee, Scotland, UK.
| |
Collapse
|
141
|
Shah AS, Zeitler PS, Wong J, Pena AS, Wicklow B, Arslanian S, Chang N, Fu J, Dabadghao P, Pinhas-Hamiel O, Urakami T, Craig ME. ISPAD Clinical Practice Consensus Guidelines 2022: Type 2 diabetes in children and adolescents. Pediatr Diabetes 2022; 23:872-902. [PMID: 36161685 DOI: 10.1111/pedi.13409] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022] Open
Abstract
Since the 2018 ISPAD guidelines on this topic, follow-up of large cohorts from around the globe have continued informing the current incidence and prevalence of co-morbidities and complications in young adults with youth-onset type 2 diabetes (T2D). This chapter focuses on the risk factors, diagnosis and presentation of youth-onset T2D, the initial and subsequent management of youth-onset T2D, and management of co-morbidities and complications. We include key updates from the observational phase of the multi-center Treatment Options for Type 2 Diabetes in Adolescents and Youth (TODAY) clinical trial, the SEARCH for Diabetes in Youth (SEARCH) study and new data from the Restoring Insulin Secretion (RISE) study, a head-to-head comparison of youth onset vs adult-onset T2D. We also include an expanded section on risk factors associated with T2D, algorithms and tables for treatment, management, and assessment of co-morbidities and complications, and sections on recently approved pharmacologic therapies for the treatment of youth-onset T2D, social determinants of health, and settings of care given COVID-19 pandemic.
Collapse
Affiliation(s)
- Amy S Shah
- Division of Pediatric Endocrinology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, Ohio, USA
| | - Philip S Zeitler
- Division of Pediatric Endocrinology, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jencia Wong
- Department of Endocrinology, Royal Prince Alfred Hospital and Central Clinical School, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Alexia S Pena
- The University of Adelaide, Robinson Research Institute, North Adelaide, South Australia, Australia
| | - Brandy Wicklow
- Division of Endocrinology, Winnipeg Children's Hospital and University of Manitoba, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Silva Arslanian
- Division of Pediatric Endocrinology, Metabolism, and Diabetes Mellitus, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nancy Chang
- Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Junfen Fu
- Division of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Preeti Dabadghao
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Orit Pinhas-Hamiel
- Edmond and Lily Safra Children's Hospital, Sackler School of Medicine, Tel-Aviv, Israel
| | - Tatsuhiko Urakami
- Department of Pediatrics, Nihon University School of Medicine, Tokyo, Japan
| | - Maria E Craig
- The Children's Hospital at Westmead, University of Sydney, Sydney, New South Wales, Australia
- Discipline of Pediatrics & Child Health, School of Clinical Medicine, University of NSW Medicine and Health, Sydney, New South Wales, Australia
| |
Collapse
|
142
|
Volke V, Katus U, Johannson A, Toompere K, Heinla K, Rünkorg K, Uusküla A. Systematic review and meta-analysis of head-to-head trials comparing sulfonylureas and low hypoglycaemic risk antidiabetic drugs. BMC Endocr Disord 2022; 22:251. [PMID: 36261824 PMCID: PMC9580135 DOI: 10.1186/s12902-022-01158-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Safety of sulfonylurea drugs in the treatment of Type 2 Diabetes is still under debate. The aim of this study was to compare the all-cause mortality and cardiovascular adverse events of sulfonylureas and drugs with a low risk for hypoglycaemia in adults with type 2 diabetes. METHODS Systematic review and meta-analysis of randomised controlled trials. DATA SOURCES MEDLINE (PubMed, OVID), Embase, Cochrane Central Register of Controlled Trials, CINAHL, WOS and Lilacs. STUDY SELECTION Randomised controlled head-to-head trials that compared sulfonylureas with active control with low hypoglycaemic potential in adults (≥ 18 years old) with type 2 diabetes published up to August 2015. The drug classes involved in the analysis were metformin, dipeptidyl peptidase-4 (DPP-4) inhibitors, sodium-glucose co-transporter-2 (SGLT-2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists. OUTCOMES The primary endpoint was all-cause mortality. The secondary endpoints were MACE, cardiovascular events and severe hypoglycaemia. SYNTHESIS OF RESULTS Two reviewers checked study eligibility, independently extracted data and assessed quality with disagreements resolved through discussion. We assessed the risk of bias of the included studies using the Cochrane risk of bias tool for randomized trials v2. Pooled odds ratios (ORs) were estimated by using fixed effects model. The study is registered on PROSPERO (26/05/2016 CRD42016038780). RESULTS Our final analysis comprised 31 studies (26,204 patients, 11,711 patients given sulfonylureas and 14,493 given comparator drugs). In comparison to drugs with low hypoglycaemic potential, sulfonylureas had higher odds for all-cause mortality (OR 1.32, 95% CI 1.00-1.75), MACE (OR 1.32, 95% CI 1.07-1.61), myocardial infarction (fatal and non-fatal) (OR 1.67, 95% CI 1.17-2.38) and hypoglycaemia (OR 5.24, 95% CI 4.20-6.55). Subsequent sensitivity analysis revealed differences in the effect of sulfonylureas, with an increased risk of all-cause mortality with glipizide but not the other molecules. CONCLUSION Our meta-analysis raises concern about the safety of SUs compared to alternative drugs involved in current analysis. Important differences may exist within the drug class, and glimepiride seems to have best safety profile.
Collapse
Affiliation(s)
- Vallo Volke
- Department of Physiology, Institute of Biomedicine and Translational Medicine, Centre of excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia.
- Endocrinology Unit, Tartu University Hospital, 8 L. Puusepa Street, 51014, Tartu, Estonia.
| | - Urmeli Katus
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Annika Johannson
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Karolin Toompere
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Keiu Heinla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, Centre of excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Kertu Rünkorg
- Department of Physiology, Institute of Biomedicine and Translational Medicine, Centre of excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Anneli Uusküla
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| |
Collapse
|
143
|
Giglio RV, Papanas N, Rizvi AA, Ciaccio M, Patti AM, Ilias I, Pantea Stoian A, Sahebkar A, Janez A, Rizzo M. An Update on the Current and Emerging Use of Thiazolidinediones for Type 2 Diabetes. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1475. [PMID: 36295635 PMCID: PMC9609741 DOI: 10.3390/medicina58101475] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/09/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Guidelines have increasingly stressed the concept that adequate glycemic control is required to prevent or decrease the macro- and microvascular complications of type 2 diabetes mellitus (T2DM). PPAR-gamma agonists ("glitazones") are no longer prioritized due to their effects on heart failure. However, the association between these drugs and innovative therapies could be a valuable tool to attenuate the risk factors of the metabolic syndrome. Glitazones are used for the treatment of diabetes and associated comorbidities. There is substantial scientific evidence demonstrating the effect of glitazones at a cardiometabolic level, as well as on hematological and neurological pathologies that point to their usefulness. The use of glitazones has always been controversial both for the type of patients who must take these drugs and for the side effects associated with them. Unfortunately, the recent guidelines do not include them among the preferred drugs for the treatment of hyperglycemia and rosiglitazone is out of the market in many countries due to an adverse cardiovascular risk profile. Even though real-life studies have proven otherwise, and their pleiotropic effects have been highlighted, they have been unable to achieve primacy in the choice of antihyperglycemic drugs. It would be appropriate to demonstrate the usefulness of pioglitazone and its therapeutic benefit with further cardiovascular safety studies.
Collapse
Affiliation(s)
- Rosaria Vincenza Giglio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “Paolo Giaccone”, 90127 Palermo, Italy
| | - Nikolaos Papanas
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, 68132 Alexandroupoli, Greece
| | - Ali Abbas Rizvi
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “Paolo Giaccone”, 90127 Palermo, Italy
| | - Angelo Maria Patti
- Promise Department, School of Medicine, University of Palermo, 90133 Palermo, Italy
| | - Ioannis Ilias
- Department of Endocrinology, Diabetes and Metabolism, Elena Venizelou Hospital, 11521 Athens, Greece
| | - Anca Pantea Stoian
- Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, 050474 Bucharest, Romania
| | - Amirhossein Sahebkar
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 1696700, Iran
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Manfredi Rizzo
- Promise Department, School of Medicine, University of Palermo, 90133 Palermo, Italy
- Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, 050474 Bucharest, Romania
| |
Collapse
|
144
|
Syringic Acid Ameliorates Cardiac, Hepatic, Renal and Neuronal Damage Induced by Chronic Hyperglycaemia in Wistar Rats: A Behavioural, Biochemical and Histological Analysis. Molecules 2022; 27:molecules27196722. [PMID: 36235257 PMCID: PMC9573038 DOI: 10.3390/molecules27196722] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
This study investigated the effects of syringic acid (SA) on renal, cardiac, hepatic, and neuronal diabetic complications in streptozotocin-induced neonatal (nSTZ) diabetic rats. STZ (110 mg/kg i.p) was injected into Wistar rat neonates as a split dose (second and third postnatal day). Diabetes mellitus was diagnosed in adults by measuring fasting blood glucose levels, urine volume, and food and water intake. The treatment of SA (25 mg/kg, 50 mg/kg p.o) was given from the 8th to 18th postnatal week. To assess the development of diabetic complications and the effect of therapy, biochemical indicators in serum and behavioural parameters were recorded at specific intervals during the study period. SA (25 mg/kg, 50 mg/kg p.o) treatment reduced hyperglycaemia, polydipsia, polyphagia, polyuria, relative organ weight, cardiac hypertrophic indices, inflammatory markers, cell injury markers, glycated haemoglobin, histopathological score, and oxidative stress, and increased Na/K ATPase activity. These findings suggest that SA might significantly alleviate diabetic complications and/or renal, neuronal, cardiac, and hepatic damage in nSTZ diabetic rats.
Collapse
|
145
|
Patel O, Muller CJF, Joubert E, Rosenkranz B, Louw J, Awortwe C. Aspalathin-rich green rooibos tea in combination with glyburide and atorvastatin enhances lipid metabolism in a db/db mouse model. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:963489. [PMID: 36992750 PMCID: PMC10012079 DOI: 10.3389/fcdhc.2022.963489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022]
Abstract
Rooibos (Aspalathus linearis), an indigenous South African plant and its major flavonoid, aspalathin, exhibited positive effects on glycemia and dyslipidemia in animal studies. Limited evidence exists on the effects of rooibos extract taken in combination with oral hypoglycemic and lipid-lowering medications. This study investigated the combined effects of a pharmaceutical grade aspalathin-rich green rooibos extract (GRT) with the sulfonylurea, glyburide, and atorvastatin in a type 2 diabetic (db/db) mouse model. Six-week-old male db/db mice and their nondiabetic lean db+ littermates were divided into 8 experimental groups (n=6/group). Db/db mice were treated orally with glyburide (5 mg/kg bodyweight), atorvastatin (80 mg/kg bodyweight) and GRT (100 mg/kg bodyweight) as mono- and combination therapies respectively, for 5 weeks. An intraperitoneal glucose tolerance test was conducted at 3 weeks of treatment. Serum was collected for lipid analyses and liver tissues for histological examination and gene expression. A significant increase in the fasting plasma glucose (FPG) of the db/db mice compared to their lean counterparts (from 7.98 ± 0.83 to 26.44 ± 1.84, p < 0.0001) was observed. Atorvastatin reduced cholesterol (from 4.00 ± 0.12 to 2.93 ± 0.13, p < 0.05) and triglyceride levels (from 2.77 ± 0.50 to 1.48 ± 0.23, p < 0.05). In db/db mice, the hypotriglyceridemic effect of atorvastatin was enhanced when combined with both GRT and glyburide (from 2.77 ± 0.50 to 1.73 ± 0.35, p = 0.0002). Glyburide reduced the severity and pattern of steatotic lipid droplet accumulation from a mediovesicular type across all lobular areas, whilst combining GRT with glyburide reduced the abundance and severity of lipid droplet accumulation in the centri- and mediolobular areas. The combination of GRT, glyburide and atorvastatin reduced the abundance and severity of lipid accumulation and the intensity score compared to the administered drugs alone. The addition of either GRT or glyburide in combination with atorvastatin had no effect on blood glucose or lipid profiles, but significantly reduced lipid droplet accumulation.
Collapse
Affiliation(s)
- Oelfah Patel
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (MRC), Tygerberg, South Africa
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (MRC), Tygerberg, South Africa
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
- *Correspondence: Christo J. F. Muller,
| | - Elizabeth Joubert
- Department of Food Science, Stellenbosch University, Matieland, South Africa
- Post-Harvest and Agro-Processing Technologies, Agricultural Research Council, Infruitec-Nietvoorbij, Stellenbosch, South Africa
| | - Bernd Rosenkranz
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (MRC), Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Charles Awortwe
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (MRC), Tygerberg, South Africa
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| |
Collapse
|
146
|
Ruan Z, Ung COL, Shen Y, Zhang Y, Wang W, Luo J, Zou H, Xue Y, Wang Y, Hu H, Guo L. Long-Term Cost-Effectiveness Analysis of Once-Weekly Semaglutide versus Dulaglutide in Patients with Type 2 Diabetes with Inadequate Glycemic Control in China. Diabetes Ther 2022; 13:1737-1753. [PMID: 35934763 PMCID: PMC9500126 DOI: 10.1007/s13300-022-01301-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/14/2022] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION The objective of the current study was to assess the long-term cost-effectiveness of once-weekly semaglutide 0.5 mg and 1.0 mg versus dulaglutide 1.5 mg for the treatment of patients with type 2 diabetes uncontrolled on metformin in the Chinese setting. METHODS The Swedish Institute of Health Economics Diabetes Cohort Model (IHE-DCM) was used to evaluate the long-term health and economic outcomes of once-weekly semaglutide and dulaglutide. Analysis was conducted from the perspective of the Chinese healthcare systems over a time horizon of 40 years. Data on baseline cohort characteristics and treatment effects were sourced from the SUSTAIN 7 clinical trial. Costs included treatment costs and costs of complications. Projected health and economic outcomes were discounted at a rate of 5% annually. The robustness of the results was evaluated through one-way sensitivity analyses and probabilistic sensitivity analyses. RESULTS Compared with dulaglutide 1.5 mg, once-weekly semaglutide 0.5 mg and 1.0 mg were associated with improvements in discounted life expectancy of 0.04 and 0.10 years, respectively, and improvements in discounted quality-adjusted life expectancy of 0.08 and 0.19 quality-adjusted life years (QALYs), respectively. Clinical benefits were achieved at reduced costs, with lifetime cost savings of 8355 Chinese Yuan (CNY) with once-weekly semaglutide 0.5 mg and 11,553 CNY with once-weekly semaglutide 1.0 mg. Sensitivity analyses verified the robustness of the research results. CONCLUSIONS Once-weekly semaglutide was suggested to be dominant (more effective and less costly) versus dulaglutide 1.5 mg in patients with type 2 diabetes uncontrolled on metformin treatment in China.
Collapse
Affiliation(s)
- Zhen Ruan
- Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Yang Shen
- Novo Nordisk (China) Pharmaceuticals Co., Ltd., Beijing, China
| | - Yawen Zhang
- Novo Nordisk (China) Pharmaceuticals Co., Ltd., Beijing, China
| | - Weihao Wang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyi Luo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Huimin Zou
- Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Yan Xue
- Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Yao Wang
- Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Hao Hu
- Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
147
|
Blonde L, Umpierrez GE, Reddy SS, McGill JB, Berga SL, Bush M, Chandrasekaran S, DeFronzo RA, Einhorn D, Galindo RJ, Gardner TW, Garg R, Garvey WT, Hirsch IB, Hurley DL, Izuora K, Kosiborod M, Olson D, Patel SB, Pop-Busui R, Sadhu AR, Samson SL, Stec C, Tamborlane WV, Tuttle KR, Twining C, Vella A, Vellanki P, Weber SL. American Association of Clinical Endocrinology Clinical Practice Guideline: Developing a Diabetes Mellitus Comprehensive Care Plan-2022 Update. Endocr Pract 2022; 28:923-1049. [PMID: 35963508 PMCID: PMC10200071 DOI: 10.1016/j.eprac.2022.08.002] [Citation(s) in RCA: 234] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The objective of this clinical practice guideline is to provide updated and new evidence-based recommendations for the comprehensive care of persons with diabetes mellitus to clinicians, diabetes-care teams, other health care professionals and stakeholders, and individuals with diabetes and their caregivers. METHODS The American Association of Clinical Endocrinology selected a task force of medical experts and staff who updated and assessed clinical questions and recommendations from the prior 2015 version of this guideline and conducted literature searches for relevant scientific papers published from January 1, 2015, through May 15, 2022. Selected studies from results of literature searches composed the evidence base to update 2015 recommendations as well as to develop new recommendations based on review of clinical evidence, current practice, expertise, and consensus, according to established American Association of Clinical Endocrinology protocol for guideline development. RESULTS This guideline includes 170 updated and new evidence-based clinical practice recommendations for the comprehensive care of persons with diabetes. Recommendations are divided into four sections: (1) screening, diagnosis, glycemic targets, and glycemic monitoring; (2) comorbidities and complications, including obesity and management with lifestyle, nutrition, and bariatric surgery, hypertension, dyslipidemia, retinopathy, neuropathy, diabetic kidney disease, and cardiovascular disease; (3) management of prediabetes, type 2 diabetes with antihyperglycemic pharmacotherapy and glycemic targets, type 1 diabetes with insulin therapy, hypoglycemia, hospitalized persons, and women with diabetes in pregnancy; (4) education and new topics regarding diabetes and infertility, nutritional supplements, secondary diabetes, social determinants of health, and virtual care, as well as updated recommendations on cancer risk, nonpharmacologic components of pediatric care plans, depression, education and team approach, occupational risk, role of sleep medicine, and vaccinations in persons with diabetes. CONCLUSIONS This updated clinical practice guideline provides evidence-based recommendations to assist with person-centered, team-based clinical decision-making to improve the care of persons with diabetes mellitus.
Collapse
Affiliation(s)
| | | | - S Sethu Reddy
- Central Michigan University, Mount Pleasant, Michigan
| | | | | | | | | | | | - Daniel Einhorn
- Scripps Whittier Diabetes Institute, La Jolla, California
| | | | | | - Rajesh Garg
- Lundquist Institute/Harbor-UCLA Medical Center, Torrance, California
| | | | | | | | | | | | - Darin Olson
- Colorado Mountain Medical, LLC, Avon, Colorado
| | | | | | - Archana R Sadhu
- Houston Methodist; Weill Cornell Medicine; Texas A&M College of Medicine; Houston, Texas
| | | | - Carla Stec
- American Association of Clinical Endocrinology, Jacksonville, Florida
| | | | - Katherine R Tuttle
- University of Washington and Providence Health Care, Seattle and Spokane, Washington
| | | | | | | | - Sandra L Weber
- University of South Carolina School of Medicine-Greenville, Prisma Health System, Greenville, South Carolina
| |
Collapse
|
148
|
Pal R, Bhadada SK. Glycemic Control in Youth-onset Type 2 Diabetes: Predicting the Tomorrow Based on Lessons Learnt from TODAY. J Clin Endocrinol Metab 2022; 107:e4252-e4253. [PMID: 35640436 PMCID: PMC9516040 DOI: 10.1210/clinem/dgac338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Indexed: 11/19/2022]
Affiliation(s)
- Rimesh Pal
- Department of Endocrinology, Postgraduate Education of Medical Education and Research (PGIMER), Chandigarh, India-160012
| | - Sanjay K Bhadada
- Correspondence: Sanjay Kumar Bhadada, MD, DM, Department of Endocrinology, PGIMER, Chandigarh, India-160012.
| |
Collapse
|
149
|
Nathan DM, Lachin JM, Balasubramanyam A, Burch HB, Buse JB, Butera NM, Cohen RM, Crandall JP, Kahn SE, Krause-Steinrauf H, Larkin ME, Rasouli N, Tiktin M, Wexler DJ, Younes N. Glycemia Reduction in Type 2 Diabetes - Glycemic Outcomes. N Engl J Med 2022; 387:1063-1074. [PMID: 36129996 PMCID: PMC9829320 DOI: 10.1056/nejmoa2200433] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND The comparative effectiveness of glucose-lowering medications for use with metformin to maintain target glycated hemoglobin levels in persons with type 2 diabetes is uncertain. METHODS In this trial involving participants with type 2 diabetes of less than 10 years' duration who were receiving metformin and had glycated hemoglobin levels of 6.8 to 8.5%, we compared the effectiveness of four commonly used glucose-lowering medications. We randomly assigned participants to receive insulin glargine U-100 (hereafter, glargine), the sulfonylurea glimepiride, the glucagon-like peptide-1 receptor agonist liraglutide, or sitagliptin, a dipeptidyl peptidase 4 inhibitor. The primary metabolic outcome was a glycated hemoglobin level, measured quarterly, of 7.0% or higher that was subsequently confirmed, and the secondary metabolic outcome was a confirmed glycated hemoglobin level greater than 7.5%. RESULTS A total of 5047 participants (19.8% Black and 18.6% Hispanic or Latinx) who had received metformin for type 2 diabetes were followed for a mean of 5.0 years. The cumulative incidence of a glycated hemoglobin level of 7.0% or higher (the primary metabolic outcome) differed significantly among the four groups (P<0.001 for a global test of differences across groups); the rates with glargine (26.5 per 100 participant-years) and liraglutide (26.1) were similar and lower than those with glimepiride (30.4) and sitagliptin (38.1). The differences among the groups with respect to a glycated hemoglobin level greater than 7.5% (the secondary outcome) paralleled those of the primary outcome. There were no material differences with respect to the primary outcome across prespecified subgroups defined according to sex, age, or race or ethnic group; however, among participants with higher baseline glycated hemoglobin levels there appeared to be an even greater benefit with glargine, liraglutide, and glimepiride than with sitagliptin. Severe hypoglycemia was rare but significantly more frequent with glimepiride (in 2.2% of the participants) than with glargine (1.3%), liraglutide (1.0%), or sitagliptin (0.7%). Participants who received liraglutide reported more frequent gastrointestinal side effects and lost more weight than those in the other treatment groups. CONCLUSIONS All four medications, when added to metformin, decreased glycated hemoglobin levels. However, glargine and liraglutide were significantly, albeit modestly, more effective in achieving and maintaining target glycated hemoglobin levels. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases and others; GRADE ClinicalTrials.gov number, NCT01794143.).
Collapse
Affiliation(s)
- David M Nathan
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - John M Lachin
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Ashok Balasubramanyam
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Henry B Burch
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - John B Buse
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Nicole M Butera
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Robert M Cohen
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Jill P Crandall
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Steven E Kahn
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Heidi Krause-Steinrauf
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Mary E Larkin
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Neda Rasouli
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Margaret Tiktin
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Deborah J Wexler
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| | - Naji Younes
- From the Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston (D.M.N., M.E.L., D.J.W.); the Biostatistics Center, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Rockville (J.M.L., N.M.B., H.K.-S., N.Y.), and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda (H.B.B.) - both in Maryland; the Section of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston (A.B.); the Division of Endocrinology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill (J.B.B.); the Cincinnati Veterans Affairs (VA) Medical Center, University of Cincinnati College of Medicine, Cincinnati (R.M.C.); the Division of Endocrinology and Diabetes and the Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY (J.P.C.); the Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, VA Puget Sound Health Care System, University of Washington, Seattle (S.E.K.); the Division of Endocrinology, Metabolism, and Diabetes, University of Colorado School of Medicine, and the VA Eastern Colorado Health Care System - both in Aurora (N.R.); and the Louis Stokes Cleveland VA Medical Center, Case Western Reserve University, Cleveland (M.T.)
| |
Collapse
|
150
|
The Progression of Prediabetes to Type 2 Diabetes in Children and Adolescents in the United States: Current Challenges and Solutions. ENDOCRINES 2022. [DOI: 10.3390/endocrines3030045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Prediabetes, the precursor of type 2 diabetes (T2D), is on the rise among children and adolescents in the United States. The natural history of prediabetes is poorly characterized in children compared to adults. The available data indicate a phenotype of an accelerated β-cell failure in youth with prediabetes. Data from randomized controlled trials showed no benefit on β-cell preservation or A1c in youth with prediabetes from therapeutic agents such as metformin and insulin. As a result, the American Diabetes Association recommends only lifestyle intervention, but not therapeutic agents, for the management of prediabetes in children and adolescents. These recommendations for lifestyle modification in youth, largely derived from data in adults, lack the precision necessary for efficacy in youth. However, a recent 4-year real-world study on youth reported that adherence to nutrition visits was associated with a 4-fold reduction in the likelihood of progressing from prediabetes to T2D. The finding that this reversal is associated with reduced insulin resistance (IR) and not with decreased body weight is novel and provides the foundation for trialing investigational products that may protect β-cells and reduce IR and/or body weight. This study provides the much-needed foundation for further exploration of the impact of lifestyle modification in conjunction with other approaches for the reversal of prediabetes in youth. The systematization of the protocol for medical nutrition therapy for the reversal of prediabetes in youth will ensure optimal and consistent results from adherent patients. This communication provides updates on the pathobiology of prediabetes in youth and a clear direction for efficacious studies in the field.
Collapse
|