CC BY 4.0 · Glob Med Genet 2022; 09(02): 063-071
DOI: 10.1055/s-0041-1741109
Review Article

Determinants in Tailoring Antidiabetic Therapies: A Personalized Approach

Aliya A. Rizvi
1   Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
,
Mohammad Abbas
1   Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
,
Sushma Verma
1   Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
,
Shrikant Verma
1   Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
,
Almas Khan
1   Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
,
Syed T. Raza
2   Department of Biochemistry, Era University, Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh, India
,
Farzana Mahdi
1   Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
› Author Affiliations
Funding The work was supported by intramural funding of Era University, Lucknow, India.

Abstract

Diabetes has become a pandemic as the number of diabetic people continues to rise globally. Being a heterogeneous disease, it has different manifestations and associated complications in different individuals like diabetic nephropathy, neuropathy, retinopathy, and others. With the advent of science and technology, this era desperately requires increasing the pace of embracing precision medicine and tailoring of drug treatment based on the genetic composition of individuals. It has been previously established that response to antidiabetic drugs, like biguanides, sulfonylureas, dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide 1 (GLP-1) agonists, and others, depending on variations in their transporter genes, metabolizing genes, genes involved in their action, etc. Responsiveness of these drugs also relies on epigenetic factors, including histone modifications, miRNAs, and DNA methylation, as well as environmental factors and the lifestyle of an individual. For precision medicine to make its way into clinical procedures and come into execution, all these factors must be reckoned with. This review provides an insight into several factors oscillating around the idea of precision medicine in type-2 diabetes mellitus.



Publication History

Received: 08 November 2021

Accepted: 20 November 2021

Article published online:
21 January 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2011; 34 (Suppl. 01) S62-S69
  • 2 International Diabetes Federation. IDF diabetes atlas, 9th ed. Accessed December 18, 2021 at: https://www.diabetesatlas.org/data/en/country/93/in.html
  • 3 Wolf AM, Conaway MR, Crowther JQ. et al; Improving Control with Activity and Nutrition (ICAN) Study. Translating lifestyle intervention to practice in obese patients with type 2 diabetes: Improving Control with Activity and Nutrition (ICAN) study. Diabetes Care 2004; 27 (07) 1570-1576
  • 4 Nathan DM, Genuth S, Lachin J. et al; Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 1993; 329 (14) 977-986
  • 5 Guidelines on second-and third-line medicines and type of insulin for the control of blood glucose levels in non-pregnant adults with diabetes mellitus. Accessed December 6, 2021 at: https://apps.who.int/iris/bitstream/handle/10665/272433/9789241550284-eng.pdf?sequence=1&isAllowed=y
  • 6 Motulsky AG. Drug reactions enzymes, and biochemical genetics. J Am Med Assoc 1957; 165 (07) 835-837
  • 7 Vogel F. Moderne probleme der humangenetik. In: Heilmeyer L, Schoen R, de Rudder B. Ergebnisse der inneren medizin und kinderheilkunde. Berlin, Germany: Springer; 1959: 52-125
  • 8 American Diabetes Association. Standards of medical care in diabetes–2013. Diabetes Care 2013; 36 (Suppl. 01) S11-S66
  • 9 Cappelletti P. Medicina di precisione e medicina di laboratorio. La Rivista Italiana della Medicina di Laboratorio-Italian Journal of Laboratory Medicine 2016; 12 (03) 129-133
  • 10 International HapMap Consortium. The International HapMap Project. Nature 2003; 426 (6968): 789-796
  • 11 Xue A, Wu Y, Zhu Z. et al; eQTLGen Consortium. Genome-wide association analyses identify 143 risk variants and putative regulatory mechanisms for type 2 diabetes. Nat Commun 2018; 9 (01) 2941
  • 12 Hattersley AT, Patel KA. Precision diabetes: learning from monogenic diabetes. Diabetologia 2017; 60 (05) 769-777
  • 13 Voight BF, Scott LJ, Steinthorsdottir V. et al; MAGIC investigators, GIANT Consortium. Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat Genet 2010; 42 (07) 579-589
  • 14 Wang-Sattler R, Yu Z, Herder C. et al. Novel biomarkers for pre-diabetes identified by metabolomics. Mol Syst Biol 2012; 8: 615
  • 15 Grant SF, Thorleifsson G, Reynisdottir I. et al. Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 2006; 38 (03) 320-323
  • 16 Tong Y, Lin Y, Zhang Y. et al. Association between TCF7L2 gene polymorphisms and susceptibility to type 2 diabetes mellitus: a large Human Genome Epidemiology (HuGE) review and meta-analysis. BMC Med Genet 2009; 10: 15
  • 17 Pearson ER. Translating TCF7L2: from gene to function. Diabetologia 2009; 52 (07) 1227-1230
  • 18 Zazuli Z, Duin NJCB, Jansen K, Vijverberg SJH, Maitland-van der Zee AH, Masereeuw R. The impact of genetic polymorphisms in organic cation transporters on renal drug disposition. Int J Mol Sci 2020; 21 (18) 6627
  • 19 Becker ML, Pearson ER, Tkáč I. Pharmacogenetics of oral antidiabetic drugs. Int J Endocrinol 2013; 2013: 686315
  • 20 Schäfer SA, Tschritter O, Machicao F. et al. Impaired glucagon-like peptide-1-induced insulin secretion in carriers of transcription factor 7-like 2 (TCF7L2) gene polymorphisms . Diabetologia 2007; 50 (12) 2443-2450
  • 21 Schäfer SA, Müssig K, Staiger H. et al. A common genetic variant in WFS1 determines impaired glucagon-like peptide-1-induced insulin secretion. Diabetologia 2009; 52 (06) 1075-1082
  • 22 Pernicova I, Korbonits M. Metformin–mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol 2014; 10 (03) 143-156
  • 23 Todd JN, Florez JC. An update on the pharmacogenomics of metformin: progress, problems and potential. Pharmacogenomics 2014; 15 (04) 529-539
  • 24 Takane H, Shikata E, Otsubo K, Higuchi S, Ieiri I. Polymorphism in human organic cation transporters and metformin action. Pharmacogenomics 2008; 9 (04) 415-422
  • 25 Choi MK, Song IS. Organic cation transporters and their pharmacokinetic and pharmacodynamic consequences. Drug Metab Pharmacokinet 2008; 23 (04) 243-253
  • 26 Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia 2017; 60 (09) 1577-1585
  • 27 Zhou Y, Ye W, Wang Y. et al. Genetic variants of OCT1 influence glycemic response to metformin in Han Chinese patients with type-2 diabetes mellitus in Shanghai. Int J Clin Exp Pathol 2015; 8 (08) 9533-9542
  • 28 Giannoudis A, Wang L, Jorgensen AL. et al. The hOCT1 SNPs M420del and M408V alter imatinib uptake and M420del modifies clinical outcome in imatinib-treated chronic myeloid leukemia. Blood 2013; 121 (04) 628-637
  • 29 Umamaheswaran G, Praveen RG, Damodaran SE, Das AK, Adithan C. Influence of SLC22A1 rs622342 genetic polymorphism on metformin response in South Indian type 2 diabetes mellitus patients. Clin Exp Med 2015; 15 (04) 511-517
  • 30 Becker ML, Visser LE, van Schaik RH, Hofman A, Uitterlinden AG, Stricker BH. Interaction between polymorphisms in the OCT1 and MATE1 transporter and metformin response. Pharmacogenet Genomics 2010; 20 (01) 38-44
  • 31 Shu Y, Leabman MK, Feng B. et al; Pharmacogenetics Of Membrane Transporters Investigators. Evolutionary conservation predicts function of variants of the human organic cation transporter, OCT1. Proc Natl Acad Sci U S A 2003; 100 (10) 5902-5907
  • 32 Takeuchi A, Motohashi H, Okuda M, Inui K. Decreased function of genetic variants, Pro283Leu and Arg287Gly, in human organic cation transporter hOCT1. Drug Metab Pharmacokinet 2003; 18 (06) 409-412
  • 33 Yee SW, Brackman DJ, Ennis EA. et al. Influence of transporter polymorphisms on drug disposition and response: a perspective from the international transporter consortium. Clin Pharmacol Ther 2018; 104 (05) 803-817
  • 34 Ito S, Kusuhara H, Yokochi M. et al. Competitive inhibition of the luminal efflux by multidrug and toxin extrusions, but not basolateral uptake by organic cation transporter 2, is the likely mechanism underlying the pharmacokinetic drug-drug interactions caused by cimetidine in the kidney. J Pharmacol Exp Ther 2012; 340 (02) 393-403
  • 35 Song IS, Shin HJ, Shim EJ. et al. Genetic variants of the organic cation transporter 2 influence the disposition of metformin. Clin Pharmacol Ther 2008; 84 (05) 559-562
  • 36 Kroetz DL, Yee SW, Giacomini KM. The pharmacogenomics of membrane transporters project: research at the interface of genomics and transporter pharmacology. Clin Pharmacol Ther 2010; 87 (01) 109-116
  • 37 Chen EC, Liang X, Yee SW. et al. Targeted disruption of organic cation transporter 3 attenuates the pharmacologic response to metformin. Mol Pharmacol 2015; 88 (01) 75-83
  • 38 Kekuda R, Prasad PD, Wu X. et al. Cloning and functional characterization of a potential-sensitive, polyspecific organic cation transporter (OCT3) most abundantly expressed in placenta. J Biol Chem 1998; 273 (26) 15971-15979
  • 39 Bleasby K, Castle JC, Roberts CJ. et al. Expression profiles of 50 xenobiotic transporter genes in humans and pre-clinical species: a resource for investigations into drug disposition. Xenobiotica 2006; 36 (10-11): 963-988
  • 40 Chen L, Pawlikowski B, Schlessinger A. et al. Role of organic cation transporter 3 (SLC22A3) and its missense variants in the pharmacologic action of metformin. Pharmacogenet Genomics 2010; 20 (11) 687-699
  • 41 Sakata T, Anzai N, Kimura T. et al. Functional analysis of human organic cation transporter OCT3 (SLC22A3) polymorphisms. J Pharmacol Sci 2010; 113 (03) 263-266
  • 42 Lozano E, Briz O, Macias RIR, Serrano MA, Marin JJG, Herraez E. Genetic heterogeneity of SLC22 family of transporters in drug disposition. J Pers Med 2018; 8 (02) 14
  • 43 Hakooz N, Jarrar YB, Zihlif M, Imraish A, Hamed S, Arafat T. Effects of the genetic variants of organic cation transporters 1 and 3 on the pharmacokinetics of metformin in Jordanians. Drug Metab Pers Ther 2017; 32 (03) 157-162
  • 44 Motohashi H, Inui K. Organic cation transporter OCTs (SLC22) and MATEs (SLC47) in the human kidney. AAPS J 2013; 15 (02) 581-588
  • 45 Becker ML, Visser LE, van Schaik RH, Hofman A, Uitterlinden AG, Stricker BH. Genetic variation in the multidrug and toxin extrusion 1 transporter protein influences the glucose-lowering effect of metformin in patients with diabetes: a preliminary study. Diabetes 2009; 58 (03) 745-749
  • 46 Mostafa-Hedeab G, Mohamed AA, Thabet G. et al. Effect of MATE 1, MATE 2 and OCT1 single nucleotide polymorphisms on metformin action in recently diagnosed egyptian type-2 diabetic patients. Biomed Pharmacol J 2018; 11 (01) 149-157
  • 47 Stocker SL, Morrissey KM, Yee SW. et al. The effect of novel promoter variants in MATE1 and MATE2 on the pharmacokinetics and pharmacodynamics of metformin. Clin Pharmacol Ther 2013; 93 (02) 186-194
  • 48 Choi JH, Yee SW, Ramirez AH. et al. A common 5′-UTR variant in MATE2-K is associated with poor response to metformin. Clin Pharmacol Ther 2011; 90 (05) 674-684
  • 49 Dawed AY, Zhou K, van Leeuwen N. et al; IMI DIRECT Consortium. Variation in the plasma membrane monoamine transporter (PMAT) (encoded by SLC29A4) and organic cation transporter 1 (OCT1) (encoded by SLC22A1) and gastrointestinal intolerance to metformin in type 2 diabetes: an IMI DIRECT study. Diabetes Care 2019; 42 (06) 1027-1033
  • 50 ICMR. ICMR guidelines for management of type 2 diabetes. . Accessed September 15, 2021: https://main.icmr.nic.in/sites/default/files/guidelines/ICMR_GuidelinesType2diabetes2018_0.pdf
  • 51 Reis AF, Velho G. Sulfonylurea receptor -1 (SUR1): genetic and metabolic evidences for a role in the susceptibility to type 2 diabetes mellitus. Diabetes Metab 2002; 28 (01) 14-19
  • 52 Flanagan SE, Clauin S, Bellanné-Chantelot C. et al. Update of mutations in the genes encoding the pancreatic beta-cell K(ATP) channel subunits Kir6.2 (KCNJ11) and sulfonylurea receptor 1 (ABCC8) in diabetes mellitus and hyperinsulinism. Hum Mutat 2009; 30 (02) 170-180
  • 53 Sesti G, Laratta E, Cardellini M. et al. The E23K variant of KCNJ11 encoding the pancreatic beta-cell adenosine 5′-triphosphate-sensitive potassium channel subunit Kir6.2 is associated with an increased risk of secondary failure to sulfonylurea in patients with type 2 diabetes. J Clin Endocrinol Metab 2006; 91 (06) 2334-2339
  • 54 Fodor A, Cozma A, Suharoschi R, Sitar-Taut A, Roman G. Clinical and genetic predictors of diabetes drug's response. Drug Metab Rev 2019; 51 (04) 408-427
  • 55 Zhang H, Liu X, Kuang H, Yi R, Xing H. Association of sulfonylurea receptor 1 genotype with therapeutic response to gliclazide in type 2 diabetes. Diabetes Res Clin Pract 2007; 77 (01) 58-61
  • 56 Holstein A, Plaschke A, Ptak M. et al. Association between CYP2C9 slow metabolizer genotypes and severe hypoglycaemia on medication with sulphonylurea hypoglycaemic agents. Br J Clin Pharmacol 2005; 60 (01) 103-106
  • 57 Huang C, Florez JC. Pharmacogenetics in type 2 diabetes: potential implications for clinical practice. Genome Med 2011; 3 (11) 76
  • 58 Ragia G, Tavridou A, Elens L, Van Schaik RH, Manolopoulos VG. CYP2C9*2 allele increases risk for hypoglycemia in POR*1/*1 type 2 diabetic patients treated with sulfonylureas. Exp Clin Endocrinol Diabetes 2014; 122 (01) 60-63
  • 59 Ragia G, Petridis I, Tavridou A, Christakidis D, Manolopoulos VG. Presence of CYP2C9*3 allele increases risk for hypoglycemia in type 2 diabetic patients treated with sulfonylureas. Pharmacogenomics 2009; 10 (11) 1781-1787
  • 60 Jin T. The WNT signalling pathway and diabetes mellitus. Diabetologia 2008; 51 (10) 1771-1780
  • 61 Sesti G, Marini MA, Cardellini M. et al. The Arg972 variant in insulin receptor substrate-1 is associated with an increased risk of secondary failure to sulfonylurea in patients with type 2 diabetes. Diabetes Care 2004; 27 (06) 1394-1398
  • 62 Becker ML, Aarnoudse AJ, Newton-Cheh C. et al. Common variation in the NOS1AP gene is associated with reduced glucose-lowering effect and with increased mortality in users of sulfonylurea. Pharmacogenet Genomics 2008; 18 (07) 591-597
  • 63 Mentlein R, Gallwitz B, Schmidt WE. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7-36)amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem 1993; 214 (03) 829-835
  • 64 Dalla Man C, Bock G, Giesler PD. et al. Dipeptidyl peptidase-4 inhibition by vildagliptin and the effect on insulin secretion and action in response to meal ingestion in type 2 diabetes. Diabetes Care 2009; 32 (01) 14-18
  • 65 Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132 (06) 2131-2157
  • 66 Kieffer TJ, McIntosh CH, Pederson RA. Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology 1995; 136 (08) 3585-3596
  • 67 Rathmann W, Bongaerts B. Pharmacogenetics of novel glucose-lowering drugs. Diabetologia 2021; 64 (06) 1201-1212
  • 68 Jamaluddin JL, Huri HZ, Vethakkan SR, Mustafa N. Pancreatic gene variants potentially associated with dipeptidyl peptidase-4 inhibitor treatment response in type 2 diabetes. Pharmacogenomics 2014; 15 (02) 235-249
  • 69 Wilson JR, Shuey MM, Brown NJ, Devin JK. Hypertension and type 2 diabetes are associated with decreased inhibition of dipeptidyl peptidase-4 by sitagliptin. J Endocr Soc 2017; 1 (09) 1168-1178
  • 70 Űrgeová A, Javorský M, Klimčáková L. et al. Genetic variants associated with glycemic response to treatment with dipeptidylpeptidase 4 inhibitors. Pharmacogenomics 2020; 21 (05) 317-323
  • 71 Han E, Park HS, Kwon O. et al. A genetic variant in GLP1R is associated with response to DPP-4 inhibitors in patients with type 2 diabetes. Medicine (Baltimore) 2016; 95 (44) e5155
  • 72 van Vliet-Ostaptchouk JV, van Haeften TW, Landman GW. et al. Common variants in the type 2 diabetes KCNQ1 gene are associated with impairments in insulin secretion during hyperglycaemic glucose clamp. PLoS One 2012; 7 (03) e32148
  • 73 Gotthardová I, Javorský M, Klimčáková L. et al. KCNQ1 gene polymorphism is associated with glycaemic response to treatment with DPP-4 inhibitors. Diabetes Res Clin Pract 2017; 130: 142-147
  • 74 Haghvirdizadeh P, Mohamed Z, Abdullah NA, Haghvirdizadeh P, Haerian MS, Haerian BS. KCNJ11: genetic polymorphisms and risk of diabetes mellitus. J Diabetes Res 2015; 2015: 908152
  • 75 Jamaluddin JL, Huri HZ, Vethakkan SR. Clinical and genetic predictors of dipeptidyl peptidase-4 inhibitor treatment response in type 2 diabetes mellitus. Pharmacogenomics 2016; 17 (08) 867-881
  • 76 Osada UN, Sunagawa H, Terauchi Y, Ueda S. A common susceptibility gene for type 2 diabetes is associated with drug response to a DPP-4 inhibitor: pharmacogenomic cohort in Okinawa Japan. PLoS One 2016; 11 (05) e0154821
  • 77 Doyle-Delgado K, Chamberlain JJ, Shubrook JH, Skolnik N, Trujillo J. Pharmacologic approaches to glycemic treatment of type 2 diabetes: synopsis of the 2020 American Diabetes Association's Standards of Medical Care in Diabetes Clinical Guideline. Ann Intern Med 2020; 173 (10) 813-821
  • 78 Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368 (9548): 1696-1705
  • 79 Lin CH, Lee YS, Huang YY, Hsieh SH, Chen ZS, Tsai CN. Polymorphisms of GLP-1 receptor gene and response to GLP-1 analogue in patients with poorly controlled type 2 diabetes. J Diabetes Res 2015; 2015: 176949
  • 80 McCarthy MI. Progress in defining the molecular basis of type 2 diabetes mellitus through susceptibility-gene identification. Hum Mol Genet 2004; 13 (Spec No 1): R33-R41
  • 81 Cannell IG, Kong YW, Bushell M. How do microRNAs regulate gene expression?. Biochem Soc Trans 2008; 36 (Pt 6): 1224-1231
  • 82 Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 2012; 13 (07) 484-492
  • 83 Barres R, Zierath JR. DNA methylation in metabolic disorders. Am J Clin Nutr 2011; 93 (04) 897S-900
  • 84 Jin B, Li Y, Robertson KD. DNA methylation: superior or subordinate in the epigenetic hierarchy?. Genes Cancer 2011; 2 (06) 607-617
  • 85 Ban N, Yamada Y, Someya Y. et al. Hepatocyte nuclear factor-1alpha recruits the transcriptional co-activator p300 on the GLUT2 gene promoter. Diabetes 2002; 51 (05) 1409-1418
  • 86 García-Calzón S, Perfilyev A, Männistö V. et al. Diabetes medication associates with DNA methylation of metformin transporter genes in the human liver. Clin Epigenetics 2017; 9: 102
  • 87 Karaglani M, Ragia G, Panagopoulou M. et al. Search for pharmacoepigenetic correlations in type 2 diabetes under sulfonylurea treatment. Exp Clin Endocrinol Diabetes 2019; 127 (04) 226-233
  • 88 Lin CH, Lee YS, Huang YY, Tsai CN. Methylation status of vault RNA 2-1 promoter is a predictor of glycemic response to glucagon-like peptide-1 analog therapy in type 2 diabetes mellitus. BMJ Open Diabetes Res Care 2021; 9 (01) e001416
  • 89 Zarkesh M, Ehsandar S, Hedayati M. Genetic and epigenetic aspects of type 2 diabetes mellitus: a review. Austin Endocrinol Diabetes Case Rep 2016; 1 (01) 1004
  • 90 Kanasaki K, Shi S, Kanasaki M. et al. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes 2014; 63 (06) 2120-2131
  • 91 Chen ZR, He FZ, Liu MZ. et al. MIR4532 gene variant rs60432575 influences the expression of KCNJ11 and the sulfonylureas-stimulated insulin secretion. Endocrine 2019; 63 (03) 489-496
  • 92 Tuomilehto J, Schwarz P, Lindström J. Long-term benefits from lifestyle interventions for type 2 diabetes prevention: time to expand the efforts. Diabetes Care 2011; 34 (Suppl. 02) S210-S214
  • 93 Colberg SR, Sigal RJ, Fernhall B. et al; American College of Sports Medicine, American Diabetes Association. Exercise and type 2 diabetes: the American College of Sports Medicine and the American Diabetes Association: joint position statement. Diabetes Care 2010; 33 (12) e147-e167
  • 94 Boulé NG, Robert C, Bell GJ. et al. Metformin and exercise in type 2 diabetes: examining treatment modality interactions. Diabetes Care 2011; 34 (07) 1469-1474
  • 95 Gulve EA. Exercise and glycemic control in diabetes: benefits, challenges, and adjustments to pharmacotherapy. Phys Ther 2008; 88 (11) 1297-1321
  • 96 Larsen JJ, Dela F, Madsbad S, Vibe-Petersen J, Galbo H. Interaction of sulfonylureas and exercise on glucose homeostasis in type 2 diabetic patients. Diabetes Care 1999; 22 (10) 1647-1654
  • 97 Shahar J, Hamdy O. Medication and exercise interactions: considering and managing hypoglycemia risk. Diabetes Spectr 2015; 28 (01) 64-67
  • 98 Khan MAB, Hashim MJ, King JK, Govender RD, Mustafa H, Al Kaabi J. Epidemiology of type 2 diabetes - global burden of disease and forecasted trends. J Epidemiol Glob Health 2020; 10 (01) 107-111