1
|
Rizvi AA, Abbas M, Verma S, Verma S, Khan A, Raza ST, Mahdi F. Determinants in Tailoring Antidiabetic Therapies: A Personalized Approach. Glob Med Genet 2022; 9:63-71. [PMID: 35707783 PMCID: PMC9192178 DOI: 10.1055/s-0041-1741109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/20/2021] [Indexed: 11/02/2022] Open
Abstract
AbstractDiabetes 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.
Collapse
Affiliation(s)
- Aliya A. Rizvi
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
| | - Mohammad Abbas
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
| | - Sushma Verma
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
| | - Shrikant Verma
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
| | - Almas Khan
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
| | - Syed T. Raza
- Department of Biochemistry, Era University, Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh, India
| | - Farzana Mahdi
- Department of Personalized and Molecular Medicine, Era University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
2
|
Jamalizadeh M, Hasanzad M, Sarhangi N, Sharifi F, Nasli-Esfahani E, Larijani B. Pilot study in pharmacogenomic management of empagliflozin in type 2 diabetes mellitus patients. J Diabetes Metab Disord 2021; 20:1407-1413. [PMID: 34900792 DOI: 10.1007/s40200-021-00874-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022]
Abstract
Background Type 2 diabetes mellitus (T2DM) is a metabolic disorder in which the patients with high blood sugar develop insufficient insulin secretion or insulin resistance. The solute carrier family, 5 member 2 (SLC5A2) gene is a member of sodium/glucose transporter family which can reduce heart and kidney problems. The current study aims to look into any association between rs11646054 variant in SLC5A2 gene and the anti-diabetic efficacy and safety of empagliflozin. Methods 14 T2DM who failed to respond to previous treatments, empagliflozin 10 mg was added for 6 months. Genotyping of the rs11646054 variant of SLC5A2 gene was performed by polymerase chain reaction (PCR) followed by Sanger sequencing. Results Although hemoglobin A1c (HbA1c) and low-density lipoprotein (LDL) were not significantly different, but the mean fasting blood sugar (FBS), 2-h post prandial (2hpp), albumin-to-creatinine ratio (ACR), and total cholesterol (TC) were significantly decreased after 6 months empagliflozin treatment. There was a significant difference in the mean final reductions in FBS level among genotypes. It's important to mention that those who were GG homozygotes had a tendency to have more decrements. Conclusions The study results indicate that effects of variation in SLC5A2 (rs11646054) on the clinical efficacy of empagliflozin were negligible.
Collapse
Affiliation(s)
- Mahdieh Jamalizadeh
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negar Sarhangi
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Sharifi
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, No.10-Jalal-e-Ale-Ahmad Street, Chamran Highway, 1411713119 Tehran, Iran
| |
Collapse
|
3
|
Genetic and Epigenomic Modifiers of Diabetic Neuropathy. Int J Mol Sci 2021; 22:ijms22094887. [PMID: 34063061 PMCID: PMC8124699 DOI: 10.3390/ijms22094887] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/18/2022] Open
Abstract
Diabetic neuropathy (DN), the most common chronic and progressive complication of diabetes mellitus (DM), strongly affects patients’ quality of life. DN could be present as peripheral, autonomous or, clinically also relevant, uremic neuropathy. The etiopathogenesis of DN is multifactorial, and genetic components play a role both in its occurrence and clinical course. A number of gene polymorphisms in candidate genes have been assessed as susceptibility factors for DN, and most of them are linked to mechanisms such as reactive oxygen species production, neurovascular impairments and modified protein glycosylation, as well as immunomodulation and inflammation. Different epigenomic mechanisms such as DNA methylation, histone modifications and non-coding RNA action have been studied in DN, which also underline the importance of “metabolic memory” in DN appearance and progression. In this review, we summarize most of the relevant data in the field of genetics and epigenomics of DN, hoping they will become significant for diagnosis, therapy and prevention of DN.
Collapse
|
4
|
Sirdah MM, Reading NS. Genetic predisposition in type 2 diabetes: A promising approach toward a personalized management of diabetes. Clin Genet 2020; 98:525-547. [PMID: 32385895 DOI: 10.1111/cge.13772] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus, also known simply as diabetes, has been described as a chronic and complex endocrine metabolic disorder that is a leading cause of death across the globe. It is considered a key public health problem worldwide and one of four important non-communicable diseases prioritized for intervention through world health campaigns by various international foundations. Among its four categories, Type 2 diabetes (T2D) is the commonest form of diabetes accounting for over 90% of worldwide cases. Unlike monogenic inherited disorders that are passed on in a simple pattern, T2D is a multifactorial disease with a complex etiology, where a mixture of genetic and environmental factors are strong candidates for the development of the clinical condition and pathology. The genetic factors are believed to be key predisposing determinants in individual susceptibility to T2D. Therefore, identifying the predisposing genetic variants could be a crucial step in T2D management as it may ameliorate the clinical condition and preclude complications. Through an understanding the unique genetic and environmental factors that influence the development of this chronic disease individuals can benefit from personalized approaches to treatment. We searched the literature published in three electronic databases: PubMed, Scopus and ISI Web of Science for the current status of T2D and its associated genetic risk variants and discus promising approaches toward a personalized management of this chronic, non-communicable disorder.
Collapse
Affiliation(s)
- Mahmoud M Sirdah
- Division of Hematology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA.,Biology Department, Al Azhar University-Gaza, Gaza, Palestine
| | - N Scott Reading
- Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah, USA.,Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
5
|
Kalra S, Das AK, Bajaj S, Priya G, Ghosh S, Mehrotra RN, Das S, Shah P, Deshmukh V, Sanyal D, Chandrasekaran S, Khandelwal D, Joshi A, Nair T, Eliana F, Permana H, Fariduddin MD, Shrestha PK, Shrestha D, Kahandawa S, Sumanathilaka M, Shaheed A, Rahim AAA, Orabi A, Al-Ani A, Hussein W, Kumar D, Shaikh K. Utility of Precision Medicine in the Management of Diabetes: Expert Opinion from an International Panel. Diabetes Ther 2020; 11:411-422. [PMID: 31916214 PMCID: PMC6995789 DOI: 10.1007/s13300-019-00753-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Indexed: 12/16/2022] Open
Abstract
AIM The primary objective of this review is to develop a practice-based expert group opinion on the role of precision medicine with a specific focus on sulfonylureas (SUs) in diabetes management. BACKGROUND The clinical etiology, presentation and complications of diabetes vary from one patient to another, making the management of the disease challenging. The pre-eminent feature of diabetes mellitus (DM) are chronically elevated blood glucose concentrations; however, in clinical practice, the exclusion of autoimmunity, pregnancy, pancreatic disease or injury and rare genetic forms of diabetes is crucial. Within this framework, precision medicine provides unique insights into the risk factors and natural history of DM. Precision medicine goes beyond genomics and encompasses patient-centered care, molecular technologies and data sharing. Precision medicine has evolved in the field of diabetology. It has helped improve the efficacy of SUs, a class of drugs, which have been effectively used in the management of diabetes mellitus for decades, and it has enabled the expansion of SUs use in diabetes patients with genetic mutations. REVIEW RESULTS After due discussions, the expert group analyzed studies that focused on the use of SUs in diabetes patients with genomic variations and rare mutations. The expert group opined that SUs are important glucose-lowering drugs and that precision medicine helps in improving the efficacy of SUs by matching them to those patients who will benefit most. CONCLUSION Precision medicine opens new vistas for the effective use of SUs in unexpected patient populations, such as those with genetic mutations.
Collapse
Affiliation(s)
- Sanjay Kalra
- Department of Endocrinology, Bharti Hospital and BRIDE, Karnal, Haryana, India.
| | - A K Das
- Department of Endocrinology and Medicine, Pondicherry Institute of Medical Sciences, Puducherry, India
| | - Sarita Bajaj
- Department of Endocrinology, MLN Medical College, Allahabad, Uttar Pradesh, India
| | - Gagan Priya
- Department of Endocrinology, Fortis Hospital, Chandigarh, Punjab, India
| | - Sujoy Ghosh
- Department of Endocrinology and Metabolism, Institute of Post-Graduate Medical Education and Research (IPGMER), Kolkata, West Bengal, India
| | - R N Mehrotra
- Department of Endocrinology, Apollo Hospitals, Jubilee Hills, Hyderabad, Telangana, India
| | - Sambit Das
- Department of Endocrinology, Apollo Hospitals, Bhubaneswar, Odisha, India
| | - Parag Shah
- Department of Endocrinology and Diabetes, Gujarat Endocrine Centre, Ahmedabad, Gujarat, India
| | - Vaishali Deshmukh
- Department of Endocrinology, Deshmukh Clinic and Research Centre, Pune, Maharashtra, India
| | - Debmalya Sanyal
- Department of Endocrinology, KPC Medical College, Kolkata, West Bengal, India
| | - Sruti Chandrasekaran
- Department of Endocrinology and Diabetes, Dr Rela Institute of Medical Science (RIMC), Chennai, Tamil Nadu, India
| | - Deepak Khandelwal
- Department of Endocrinology and Diabetes, Maharaja Agrasen Hospital, New Delhi, India
| | - Amaya Joshi
- Department of Endocrinology and Diabetes, Bhaktivedanta Hospital and Research Institute, Mumbai, Maharashtra, India
| | - Tiny Nair
- Department of Cardiology, PRS Hospital, Trivandrum, Kerala, India
| | - Fatimah Eliana
- Department of Internal Medicine, Faculty of Medicine, YARSI University, Jakarta, Indonesia
| | - Hikmat Permana
- Department of Internal Medicine, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - M D Fariduddin
- Department of Endocrinology of Bangabandhu Sheikh, Mujib Medical University, Dhaka, Bangladesh
| | | | - Dina Shrestha
- Department of Endocrinology, Norvic International Hospital, Kathmandu, Nepal
| | - Shayaminda Kahandawa
- Department of Endocrinology, Teaching Hospital Karapitiya, Karapitiya, Galle, Sri Lanka
| | | | - Ahamed Shaheed
- Department of Internal Medicine, Indira Gandhi Memorial Hospital, Malé, Republic of Maldives
| | | | - Abbas Orabi
- Department of Internal Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed Al-Ani
- Department of Internal Medicine, Hamad General Hospital, Doha, Qatar
| | - Wiam Hussein
- Department of Endocrinology and Diabetes, Royal Hospital, Manama, Bahrain
| | - Dinesh Kumar
- Department of Endocrinology, NMC Specialty Hospital, Abu Dhabi, United Arab Emirates
| | - Khalid Shaikh
- Department of Diabetes, Faculty of Internal Medicine, Royal Oman Police Hospital, Muscat, Oman
| |
Collapse
|
6
|
Altall RM, Qusti SY, Filimban N, Alhozali AM, Alotaibi NA, Dallol A, Chaudhary AG, Bakhashab S. SLC22A1 And ATM Genes Polymorphisms Are Associated With The Risk Of Type 2 Diabetes Mellitus In Western Saudi Arabia: A Case-Control Study. APPLICATION OF CLINICAL GENETICS 2019; 12:213-219. [PMID: 31814751 PMCID: PMC6863135 DOI: 10.2147/tacg.s229952] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 11/03/2019] [Indexed: 01/05/2023]
Abstract
Introduction Type 2 diabetes mellitus (T2DM) is a major global health problem that is progressively affected by genetic and environmental factors. The aim of this study is to determine the influence of solute carrier family 22 member 1 (SLC22A1) rs628031 and rs461473, and ataxia telangiectasia mutated (ATM) rs11212617 polymorphisms on the risk of T2DM in Saudi Arabia by considering many parameters associated with glycemic control of T2DM, such as body mass index (BMI), fasting blood glucose, glycated hemoglobin (HbA1c), and triglyceride. Methods In a case-control study, genomic DNA from controls and diabetic groups was isolated and genotyped for each single-nucleotide polymorphism. Results There were significant correlations between T2DM and both BMI and HbA1c. Significant associations between G/G and A/G genotypes of rs628031 and rs461473 variants of SLC22A1 and high levels of HbA1c were detected. Therefore, G was predicted to be the risk allele among the assessed SLC22A1 variants. A significant correlation was observed between A/A and A/C genotypes of the rs11212617 polymorphism of ATM and elevated HbA1c. Relative risk calculation confirmed A to be the risk allele in the T2DM population. Conclusion Our study showed the risk of the assessed SLC22A1 and ATM variants on glycemic control parameters in diabetic patients.
Collapse
Affiliation(s)
- Rana M Altall
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Safaa Y Qusti
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Najlaa Filimban
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Amani M Alhozali
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Najat A Alotaibi
- Department of Family and Community Medicine, Faculty of Medicine, King Abdulaziz University Hospital, Jeddah 21589, Kingdom of Saudi Arabia
| | - Ashraf Dallol
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Adeel G Chaudhary
- KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| | - Sherin Bakhashab
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia.,KACST Technology Innovation Center in Personalized Medicine, King Abdulaziz University, Jeddah 21589, Kingdom of Saudi Arabia
| |
Collapse
|
7
|
Out M, Becker ML, van Schaik RH, Lehert P, Stehouwer CD, Kooy A. A gene variant near ATM affects the response to metformin and metformin plasma levels: a post hoc analysis of an RCT. Pharmacogenomics 2018; 19:715-726. [PMID: 29790415 DOI: 10.2217/pgs-2018-0010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
AIM To determine the influence of polymorphisms on the effects of metformin on HbA1c, daily dose of insulin and metformin plasma concentration. Methods: In a post hoc analysis of a 4.3 year placebo-controlled randomized trial with 390 patients with Type 2 diabetes already on insulin, we analyzed the influence of polymorphisms in genes coding for ATM and the transporters OCT1 and MATE1. Outcome measures were a combined HbA1c + daily dose of insulin Z score and metformin plasma concentrations. RESULTS rs11212617 (ATM) was associated with an improved Z score and a lower metformin plasma concentration. In addition, the major allele of rs2289669 (MATE1) was also associated with an improved Z score. CONCLUSION The ATM SNP rs11212617 significantly affected the effect of metformin and metformin plasma concentration. Further research is needed to determine the clinical importance of these findings, in particular the effects on metformin plasma concentration.
Collapse
Affiliation(s)
- Mattijs Out
- Department of Internal Medicine, Bethesda Hospital Hoogeveen - Care Group Treant, Hoogeveen, The Netherlands.,Bethesda Diabetes Research Center Hoogeveen, Hoogeveen, The Netherlands.,Department of Internal Medicine, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Matthijs L Becker
- Department of Clinical Chemistry, Erasmus MC Rotterdam, Rotterdam, The Netherlands.,Pharmacy Foundation of Haarlem Hospitals, Haarlem, The Netherlands
| | - Ron H van Schaik
- Department of Clinical Chemistry, Erasmus MC Rotterdam, Rotterdam, The Netherlands
| | - Philippe Lehert
- Department of Statistics, Faculty of Economics, Louvain Academy, Mons, Belgium
| | - Coen D Stehouwer
- Department of Internal Medicine & Cardiovascular Research, Maastricht University Medical Centre, The Netherlands
| | - Adriaan Kooy
- Department of Internal Medicine, Bethesda Hospital Hoogeveen - Care Group Treant, Hoogeveen, The Netherlands.,Bethesda Diabetes Research Center Hoogeveen, Hoogeveen, The Netherlands.,Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
8
|
Sanchez-Ibarra HE, Reyes-Cortes LM, Jiang XL, Luna-Aguirre CM, Aguirre-Trevino D, Morales-Alvarado IA, Leon-Cachon RB, Lavalle-Gonzalez F, Morcos F, Barrera-Saldaña HA. Genotypic and Phenotypic Factors Influencing Drug Response in Mexican Patients With Type 2 Diabetes Mellitus. Front Pharmacol 2018; 9:320. [PMID: 29681852 PMCID: PMC5898372 DOI: 10.3389/fphar.2018.00320] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
The treatment of Type 2 Diabetes Mellitus (T2DM) consists primarily of oral antidiabetic drugs (OADs) that stimulate insulin secretion, such as sulfonylureas (SUs) and reduce hepatic glucose production (e.g., biguanides), among others. The marked inter-individual differences among T2DM patients’ response to these drugs have become an issue on prescribing and dosing efficiently. In this study, fourteen polymorphisms selected from Genome-wide association studies (GWAS) were screened in 495 T2DM Mexican patients previously treated with OADs to find the relationship between the presence of these polymorphisms and response to the OADs. Then, a novel association screening method, based on global probabilities, was used to globally characterize important relationships between the drug response to OADs and genetic and clinical parameters, including polymorphisms, patient information, and type of treatment. Two polymorphisms, ABCC8-Ala1369Ser and KCNJ11-Glu23Lys, showed a significant impact on response to SUs. Heterozygous ABCC8-Ala1369Ser variant (A/C) carriers exhibited a higher response to SUs compared to homozygous ABCC8-Ala1369Ser variant (A/A) carriers (p-value = 0.029) and to homozygous wild-type genotypes (C/C) (p-value = 0.012). The homozygous KCNJ11-Glu23Lys variant (C/C) and wild-type (T/T) genotypes had a lower response to SUs compared to heterozygous (C/T) carriers (p-value = 0.039). The screening of OADs response related genetic and clinical factors could help improve the prescribing and dosing of OADs for T2DM patients and thus contribute to the design of personalized treatments.
Collapse
Affiliation(s)
| | | | - Xian-Li Jiang
- Evolutionary Information Laboratory, Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, United States
| | | | | | | | - Rafael B Leon-Cachon
- Departamento de Ciencias Básicas, Centro de Diagnóstico Molecular y Medicina Personalizada, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Monterrey, Mexico
| | - Fernando Lavalle-Gonzalez
- Servicio de Endocrinología, Hospital Universitario Dr. José E. González, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Faruck Morcos
- Evolutionary Information Laboratory, Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, United States.,Center for Systems Biology, University of Texas at Dallas, Richardson, TX, United States
| | - Hugo A Barrera-Saldaña
- Molecular Genetics Laboratory, Vitagénesis, S.A. de C.V., Monterrey, Mexico.,Tecnológico de Monterrey, Monterrey, Mexico
| |
Collapse
|
9
|
Niu N, Liu T, Cairns J, Ly RC, Tan X, Deng M, Fridley BL, Kalari KR, Abo RP, Jenkins G, Batzler A, Carlson EE, Barman P, Moran S, Heyn H, Esteller M, Wang L. Metformin pharmacogenomics: a genome-wide association study to identify genetic and epigenetic biomarkers involved in metformin anticancer response using human lymphoblastoid cell lines. Hum Mol Genet 2018; 25:4819-4834. [PMID: 28173075 DOI: 10.1093/hmg/ddw301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 08/18/2016] [Accepted: 08/26/2016] [Indexed: 12/18/2022] Open
Abstract
Metformin is currently considered as a promising anticancer agent in addition to its anti-diabetic effect. To better individualize metformin therapy and explore novel molecular mechanisms in cancer treatment, we conducted a pharmacogenomic study using 266 lymphoblastoid cell lines (LCLs). Metformin cytotoxicity assay was performed using the MTS assay. Genome-wide association (GWA) analyses were performed in LCLs using 1.3 million SNPs, 485k DNA methylation probes, 54k mRNA expression probe sets, and metformin cytotoxicity (IC50s). Top candidate genes were functionally validated using siRNA screening, followed by MTS assay in breast cancer cell lines. Further study of one top candidate, STUB1, was performed to elucidate the mechanisms by which STUB1 might contribute to metformin action. GWA analyses in LCLs identified 198 mRNA expression probe sets, 12 SNP loci, and 5 DNA methylation loci associated with metformin IC50 with P-values <10−4 or <10−5. Integrated SNP/methylation loci-expression-IC50 analyses found 3 SNP loci or 5 DNA methylation loci associated with metformin IC50 through trans-regulation of expression of 11 or 26 genes with P-value <10−4. Functional validation of top 61 candidate genes in 4 IPA networks indicated down regulation of 14 genes significantly altered metformin sensitivity in two breast cancer cell lines. Mechanistic studies revealed that the E3 ubiquitin ligase, STUB1, could influence metformin response by facilitating proteasome-mediated degradation of cyclin A. GWAS using a genomic data-enriched LCL model system, together with functional and mechanistic studies using cancer cell lines, help us to identify novel genetic and epigenetic biomarkers involved in metformin anticancer response.
Collapse
Affiliation(s)
- Nifang Niu
- Division of Clinical Pharmacology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tongzheng Liu
- Division of Oncology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Junmei Cairns
- Division of Clinical Pharmacology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Reynold C Ly
- Division of Clinical Pharmacology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Xianglin Tan
- UMDNJ/The Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Min Deng
- Division of Oncology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Brooke L Fridley
- University of Kansas Medical Center, Kansas City, Kansas City, KS, USA
| | - Krishna R Kalari
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Ryan P Abo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gregory Jenkins
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Anthony Batzler
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Erin E Carlson
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Poulami Barman
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sebastian Moran
- Bellvitge Biomedical Research Institute (IDIBELL), L Hospitalet de Llobregat, Barcelona, Spain
| | - Holger Heyn
- Bellvitge Biomedical Research Institute (IDIBELL), L Hospitalet de Llobregat, Barcelona, Spain
| | - Manel Esteller
- Bellvitge Biomedical Research Institute (IDIBELL), L Hospitalet de Llobregat, Barcelona, Spain.,Institucio Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain,Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain
| | - Liewei Wang
- Division of Clinical Pharmacology, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
10
|
Dawed AY, Ali A, Zhou K, Pearson ER, Franks PW. Evidence-based prioritisation and enrichment of genes interacting with metformin in type 2 diabetes. Diabetologia 2017; 60:2231-2239. [PMID: 28842730 PMCID: PMC6448905 DOI: 10.1007/s00125-017-4404-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS There is an extensive body of literature suggesting the involvement of multiple loci in regulating the action of metformin; most findings lack replication, without which distinguishing true-positive from false-positive findings is difficult. To address this, we undertook evidence-based, multiple data integration to determine the validity of published evidence. METHODS We (1) built a database of published data on gene-metformin interactions using an automated text-mining approach (n = 5963 publications), (2) generated evidence scores for each reported locus, (3) from which a rank-ordered gene set was generated, and (4) determined the extent to which this gene set was enriched for glycaemic response through replication analyses in a well-powered independent genome-wide association study (GWAS) dataset from the Genetics of Diabetes and Audit Research Tayside Study (GoDARTS). RESULTS From the literature search, seven genes were identified that are related to the clinical outcomes of metformin. Fifteen genes were linked with either metformin pharmacokinetics or pharmacodynamics, and the expression profiles of a further 51 genes were found to be responsive to metformin. Gene-set enrichment analysis consisting of the three sets and two more composite sets derived from the above three showed no significant enrichment in four of the gene sets. However, we detected significant enrichment of genes in the least prioritised category (a gene set in which their expression is affected by metformin) with glycaemic response to metformin (p = 0.03). This gene set includes novel candidate genes such as SLC2A4 (p = 3.24 × 10-04) and G6PC (p = 4.77 × 10-04). CONCLUSIONS/INTERPRETATION We have described a semi-automated text-mining and evidence-scoring algorithm that facilitates the organisation and extraction of useful information about gene-drug interactions. We further validated the output of this algorithm in a drug-response GWAS dataset, providing novel candidate loci for gene-metformin interactions.
Collapse
Affiliation(s)
- Adem Y Dawed
- Division of Molecular and Clinical Medicine, Medical Research Institute, Ninewells Hospital and Medical School, Level 5, Mailbox 12, University of Dundee, Dundee, DD1 9SY, UK.
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden.
| | - Ashfaq Ali
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Kaixin Zhou
- Division of Molecular and Clinical Medicine, Medical Research Institute, Ninewells Hospital and Medical School, Level 5, Mailbox 12, University of Dundee, Dundee, DD1 9SY, UK
| | - Ewan R Pearson
- Division of Molecular and Clinical Medicine, Medical Research Institute, Ninewells Hospital and Medical School, Level 5, Mailbox 12, University of Dundee, Dundee, DD1 9SY, UK
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
11
|
Influence of common polymorphisms in the SLC5A2 gene on metabolic traits in subjects at increased risk of diabetes and on response to empagliflozin treatment in patients with diabetes. Pharmacogenet Genomics 2017; 27:135-142. [PMID: 28134748 DOI: 10.1097/fpc.0000000000000268] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Inhibition of the renal sodium-glucose cotransporter 2 (SGLT2) is a novel concept in the therapy of diabetes mellitus. In this study, we first assessed whether common single nucleotide polymorphisms (SNPs) in the SGLT2-encoding gene SLC5A2 affect diabetes-related metabolic traits in subjects at risk for type 2 diabetes and, second, whether these have pharmacogenetic relevance by interfering with the response to empagliflozin treatment in patients with type 2 diabetes. PATIENTS AND METHODS Samples from a metabolically well-phenotyped cross-sectional study population (total N=2600) at increased risk for type 2 diabetes and pooled pharmacogenetic samples from patients from four phase III trials of empagliflozin (in total: 603 receiving empagliflozin, 305 receiving placebo) were genotyped for five common SNPs (minor allele frequencies ≥5%) present in the SLC5A2 gene locus. RESULTS In the cross-sectional study, none of the SLC5A2 SNPs significantly influenced metabolic traits such as body fat, insulin sensitivity/resistance, insulin release, HbA1c, plasma glucose, or systolic blood pressure when multiple testing was taken into account (all P≥0.0083). Further, no relevant effect on response to treatment with empagliflozin on HbA1c, fasting glucose, weight, or systolic blood pressure was observed for the SNPs tested in the pharmacogenetic study. CONCLUSION Common genetic variants in the SLC5A2 gene neither affects diabetes-related metabolic traits nor have a clinically relevant impact on response to treatment with the SGLT2 inhibitor empagliflozin.
Collapse
|
12
|
Elk N, Iwuchukwu OF. Using Personalized Medicine in the Management of Diabetes Mellitus. Pharmacotherapy 2017; 37:1131-1149. [PMID: 28654165 DOI: 10.1002/phar.1976] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a worldwide problem with an immense pharmacoeconomic burden. The multifactorial and complex nature of the disease lends itself to personalized pharmacotherapeutic approaches to treatment. Variability in individual risk and subsequent development of diabetes has been reported in addition to differences in response to the many oral glucose lowering therapies currently available for diabetes pharmacotherapy. Pharmacogenomic studies have attempted to uncover the heritable components of individual variability in risk susceptibility and response to pharmacotherapy. We review the current pharmacogenomics evidence as it relates to common oral glucose lowering therapies and how they can be utilized in the management of polygenic and monogenic forms of diabetes. Evidence supports the use of genetic testing and personalized approaches to the treatment of monogenic diabetes of the young. The data are not as robust for the current application of pharmacogenetic approaches to the treatment of polygenic type 2 diabetes mellitus, but there are suggestions as to future applications in this regard. We reviewed pertinent primary literature sources as well as current evidence-based guidelines on diabetes management.
Collapse
Affiliation(s)
- Nina Elk
- Division of Pharmacy Practice, Fairleigh Dickinson University School of Pharmacy, Florham Park, New Jersey
| | - Otito F Iwuchukwu
- Division of Pharmaceutical Sciences, Fairleigh Dickinson University School of Pharmacy, Florham Park, New Jersey
| |
Collapse
|
13
|
Brunetti A, Chiefari E, Foti DP. Pharmacogenetics in type 2 diabetes: still a conundrum in clinical practice. Expert Rev Endocrinol Metab 2017; 12:155-158. [PMID: 30063457 DOI: 10.1080/17446651.2017.1316192] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Antonio Brunetti
- a Department of Health Sciences , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| | - Eusebio Chiefari
- a Department of Health Sciences , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| | - Daniela Patrizia Foti
- a Department of Health Sciences , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| |
Collapse
|
14
|
Hizel C, Tremblay J, Bartlett G, Hamet P. Introduction. PROGRESS AND CHALLENGES IN PRECISION MEDICINE 2017:1-34. [DOI: 10.1016/b978-0-12-809411-2.00001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Floyd JS, Psaty BM. The Application of Genomics in Diabetes: Barriers to Discovery and Implementation. Diabetes Care 2016; 39:1858-1869. [PMID: 27926887 PMCID: PMC5079615 DOI: 10.2337/dc16-0738] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/16/2016] [Indexed: 02/03/2023]
Abstract
The emerging availability of genomic and electronic health data in large populations is a powerful tool for research that has drawn interest in bringing precision medicine to diabetes. In this article, we discuss the potential application of genomics to the prediction, prevention, and treatment of diabetes, and we use examples from other areas of medicine to illustrate some of the challenges involved in conducting genomics research in human populations and implementing findings in practice. At this time, a major barrier to the application of genomics in diabetes care is the lack of actionable genomic findings. Whether genomic information should be used in clinical practice requires a framework for evaluating the validity and clinical utility of this approach, an improved integration of genomic data into electronic health records, and the clinical decision support and educational resources for clinicians to use these data. Efforts to identify optimal approaches in all of these domains are in progress and may help to bring diabetes into the era of genomic medicine.
Collapse
Affiliation(s)
- James S Floyd
- Cardiovascular Health Research Unit and Departments of Epidemiology and Medicine, University of Washington, Seattle, WA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit and Departments of Epidemiology and Medicine, University of Washington, Seattle, WA
- Department of Health Services, University of Washington, Seattle, WA
- Group Health Research Institute, Seattle, WA
| |
Collapse
|
16
|
Santoro AB, Stage TB, Struchiner CJ, Christensen MMH, Brosen K, Suarez-Kurtz G. Limited sampling strategy for determining metformin area under the plasma concentration-time curve. Br J Clin Pharmacol 2016; 82:1002-10. [PMID: 27324407 DOI: 10.1111/bcp.13049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 12/17/2022] Open
Abstract
AIM The aim was to develop and validate limited sampling strategy (LSS) models to predict the area under the plasma concentration-time curve (AUC) for metformin. METHODS Metformin plasma concentrations (n = 627) at 0-24 h after a single 500 mg dose were used for LSS development, based on all subsets linear regression analysis. The LSS-derived AUC(0,24 h) was compared with the parameter 'best estimate' obtained by non-compartmental analysis using all plasma concentration data points. Correlation between the LSS-derived and the best estimated AUC(0,24 h) (r(2) ), bias and precision of the LSS estimates were quantified. The LSS models were validated in independent cohorts. RESULTS A two-point (3 h and 10 h) regression equation with no intercept estimated accurately the individual AUC(0,24 h) in the development cohort: r(2) = 0.927, bias (mean, 95% CI) -0.5, -2.7-1.8% and precision 6.3, 4.9-7.7%. The accuracy of the two point LSS model was verified in study cohorts of individuals receiving single 500 or 1000 mg (r(2) = -0.933-0.934) or seven 1000 mg daily doses (r(2) = 0.918), as well as using data from 16 published studies covering a wide range of metformin doses, demographics, clinical and experimental conditions (r(2) = 0.976). The LSS model reproduced previously reported results for effects of polymorphisms in OCT2 and MATE1 genes on AUC(0,24 h) and renal clearance of metformin. CONCLUSIONS The two point LSS algorithm may be used to assess the systemic exposure to metformin under diverse conditions, with reduced costs of sampling and analysis, and saving time for both subjects and investigators.
Collapse
Affiliation(s)
- Ana Beatriz Santoro
- Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro.,Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, Brazil
| | - Tore Bjerregaard Stage
- Clinical Pharmacology, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | | | | | - Kim Brosen
- Clinical Pharmacology, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
17
|
Costa V, Federico A, Pollastro C, Ziviello C, Cataldi S, Formisano P, Ciccodicola A. Computational Analysis of Single Nucleotide Polymorphisms Associated with Altered Drug Responsiveness in Type 2 Diabetes. Int J Mol Sci 2016; 17:ijms17071008. [PMID: 27347941 PMCID: PMC4964384 DOI: 10.3390/ijms17071008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/07/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022] Open
Abstract
Type 2 diabetes (T2D) is one of the most frequent mortality causes in western countries, with rapidly increasing prevalence. Anti-diabetic drugs are the first therapeutic approach, although many patients develop drug resistance. Most drug responsiveness variability can be explained by genetic causes. Inter-individual variability is principally due to single nucleotide polymorphisms, and differential drug responsiveness has been correlated to alteration in genes involved in drug metabolism (CYP2C9) or insulin signaling (IRS1, ABCC8, KCNJ11 and PPARG). However, most genome-wide association studies did not provide clues about the contribution of DNA variations to impaired drug responsiveness. Thus, characterizing T2D drug responsiveness variants is needed to guide clinicians toward tailored therapeutic approaches. Here, we extensively investigated polymorphisms associated with altered drug response in T2D, predicting their effects in silico. Combining different computational approaches, we focused on the expression pattern of genes correlated to drug resistance and inferred evolutionary conservation of polymorphic residues, computationally predicting the biochemical properties of polymorphic proteins. Using RNA-Sequencing followed by targeted validation, we identified and experimentally confirmed that two nucleotide variations in the CAPN10 gene—currently annotated as intronic—fall within two new transcripts in this locus. Additionally, we found that a Single Nucleotide Polymorphism (SNP), currently reported as intergenic, maps to the intron of a new transcript, harboring CAPN10 and GPR35 genes, which undergoes non-sense mediated decay. Finally, we analyzed variants that fall into non-coding regulatory regions of yet underestimated functional significance, predicting that some of them can potentially affect gene expression and/or post-transcriptional regulation of mRNAs affecting the splicing.
Collapse
Affiliation(s)
- Valerio Costa
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy.
| | - Antonio Federico
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy.
- DiST, Department of Science and Technology, University of Naples "Parthenope", 80134 Naples, Italy.
| | - Carla Pollastro
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy.
- DiST, Department of Science and Technology, University of Naples "Parthenope", 80134 Naples, Italy.
| | - Carmela Ziviello
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy.
| | - Simona Cataldi
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy.
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples "Federico II", 80131 Naples, Italy.
| | - Alfredo Ciccodicola
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy.
- DiST, Department of Science and Technology, University of Naples "Parthenope", 80134 Naples, Italy.
| |
Collapse
|
18
|
Zhou K, Pedersen HK, Dawed AY, Pearson ER. Pharmacogenomics in diabetes mellitus: insights into drug action and drug discovery. Nat Rev Endocrinol 2016; 12:337-46. [PMID: 27062931 DOI: 10.1038/nrendo.2016.51] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Genomic studies have greatly advanced our understanding of the multifactorial aetiology of type 2 diabetes mellitus (T2DM) as well as the multiple subtypes of monogenic diabetes mellitus. In this Review, we discuss the existing pharmacogenetic evidence in both monogenic diabetes mellitus and T2DM. We highlight mechanistic insights from the study of adverse effects and the efficacy of antidiabetic drugs. The identification of extreme sulfonylurea sensitivity in patients with diabetes mellitus owing to heterozygous mutations in HNF1A represents a clear example of how pharmacogenetics can direct patient care. However, pharmacogenomic studies of response to antidiabetic drugs in T2DM has yet to be translated into clinical practice, although some moderate genetic effects have now been described that merit follow-up in trials in which patients are selected according to genotype. We also discuss how future pharmacogenomic findings could provide insights into treatment response in diabetes mellitus that, in addition to other areas of human genetics, facilitates drug discovery and drug development for T2DM.
Collapse
Affiliation(s)
- Kaixin Zhou
- School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Helle Krogh Pedersen
- Department of Systems Biology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Adem Y Dawed
- School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Ewan R Pearson
- School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| |
Collapse
|
19
|
Stage TB, Brøsen K, Christensen MMH. A Comprehensive Review of Drug-Drug Interactions with Metformin. Clin Pharmacokinet 2016; 54:811-24. [PMID: 25943187 DOI: 10.1007/s40262-015-0270-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metformin is the world's most commonly used oral glucose-lowering drug for type 2 diabetes, and this is mainly because it protects against diabetes-related mortality and all-cause mortality. Although it is an old drug, its mechanism of action has not yet been clarified and its pharmacokinetic pathway is still not fully understood. There is considerable inter-individual variability in the response to metformin, and this has led to many drug-drug interaction (DDI) studies of metformin. In this review, we describe both in vitro and human interaction studies of metformin both as a victim and as a perpetrator. We also clarify the importance of including pharmacodynamic end points in DDI studies of metformin and taking pharmacogenetic variation into account when performing these studies to avoid hidden pitfalls in the interpretation of DDIs with metformin. This evaluation of the literature has revealed holes in our knowledge and given clues as to where future DDI studies should be focused and performed.
Collapse
Affiliation(s)
- Tore Bjerregaard Stage
- Clinical Pharmacology, Department of Public Health, University of Southern Denmark, J.B. Winsloews vej 19, 2nd Floor, 5000, Odense, Denmark,
| | | | | |
Collapse
|
20
|
Dawed AY, Zhou K, Pearson ER. Pharmacogenetics in type 2 diabetes: influence on response to oral hypoglycemic agents. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2016; 9:17-29. [PMID: 27103840 PMCID: PMC4827904 DOI: 10.2147/pgpm.s84854] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes is one of the leading causes of morbidity and mortality, consuming a significant proportion of public health spending. Oral hypoglycemic agents (OHAs) are the frontline treatment approaches after lifestyle changes. However, huge interindividual variation in response to OHAs results in unnecessary treatment failure. In addition to nongenetic factors, genetic factors are thought to contribute to much of such variability, highlighting the importance of the potential of pharmacogenetics to improve therapeutic outcome. Despite the presence of conflicting results, significant progress has been made in an effort to identify the genetic markers associated with pharmacokinetics, pharmacodynamics, and ultimately therapeutic response and/or adverse outcomes to OHAs. As such, this article presents a comprehensive review of current knowledge on pharmacogenetics of OHAs and provides insights into knowledge gaps and future directions.
Collapse
Affiliation(s)
- Adem Yesuf Dawed
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| | - Kaixin Zhou
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| | - Ewan Robert Pearson
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
21
|
Joint effects of diabetic-related genomic loci on the therapeutic efficacy of oral anti-diabetic drugs in Chinese type 2 diabetes patients. Sci Rep 2016; 6:23266. [PMID: 26983698 PMCID: PMC4794654 DOI: 10.1038/srep23266] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 03/02/2016] [Indexed: 12/11/2022] Open
Abstract
Previous pharmacogenomic studies of oral anti-diabetic drugs have primarily focused on the effect of a single site. This study aimed to examine the joint effects of multiple loci on repaglinide or rosiglitazone efficacy in newly diagnosed type 2 diabetes mellitus (T2DM) patients. A total of 209 newly diagnosed T2DM patients were randomly assigned to treatment with repaglinide or rosiglitazone for 48 weeks. The reductions in fasting glucose (ΔFPG), 2h glucose (Δ2hPG) and glycated hemoglobin (ΔHbA1c) levels were significantly associated with genetic score that was constructed using the sum of the effect alleles both in the repaglinide (P = 0.0011, 0.0002 and 0.0067, respectively) and rosiglitazone cohorts (P = 0.0002, 0.0014 and 0.0164, respectively) after adjusting for age, gender, body mass index and dosage. Survival analyses showed a trend towards a greater attainment rate of target HbA1c level in individuals with a high genetic score in the repaglinide cohort and rosiglitazone cohort (Plog-rank = 0.0815 and 0.0867, respectively) when the attainment of treatment targets were defined as more than 20% decrease of FPG, 2hPG, and HbA1c levels after treatment. In conclusion, we identified the joint effects of several T2DM-related loci on the efficacy of oral anti-diabetic drugs; moreover, we built a model to predict the drug efficacy.
Collapse
|
22
|
Shokri F, Ghaedi H, Ghafouri Fard S, Movafagh A, Abediankenari S, Mahrooz A, Kashi Z, Omrani MD. Impact of ATM and SLC22A1 Polymorphisms on Therapeutic Response to Metformin in Iranian Diabetic Patients. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2016; 5:1-7. [PMID: 27386433 PMCID: PMC4916778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Metabolic syndrome and its pathological sequel, type 2 diabetes are considered as important global health problems. Metformin is the most common drug prescribed for patients with this disorder. Consequently, understanding the genetic pathways involved in pharmacokinetics and pharmacodynamics of this drug can have a considerable effect on the personalized treatment of type 2 diabetes. In this study, we evaluated the association between rs11212617 polymorphism of ATM gene and rs628031 of SLC22A1 gene with response to treatment in newly diagnosed type 2 diabetes patients. We genotyped rs11212617 and rs628031 polymorphism by PCR based restriction fragment length polymorphism (RFLP) and assessed the role of this polymorphisms on response to treatment in 140 patients who have been recently diagnosed with type 2 diabetes and were under monotherapy with metformin for 6 months. Response to metformin was defined by HbA1c and fasting blood sugar (FBS) values. Based on such evaluations, patients were divided into two groups: responders (n= 63) and non-responders (n= 77). No significant association was found between these polymorphisms and response to treatment (OR= 0.86, [95% CI 0.52-1.41], P= 0.32) for rs11212617 and (OR= 0.45, [95% CI 0.64-1.76], P= 0.45) for rs 628031. The reported gene variants in ATM and SLC22A1 are not significantly associated with metformin treatment response in type 2 diabetic patients in an Iranian population.
Collapse
Affiliation(s)
- Fazlollah Shokri
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamid Ghaedi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolfazl Movafagh
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Abediankenari
- Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Abdolkarim Mahrooz
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Zahra Kashi
- Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mir Davood Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Corresponding author: Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. E-mail:
| |
Collapse
|
23
|
Abstract
The introduction of several new drug groups into the treatment of type 2 diabetes in the past few decades leads to an increased requirement for an individualized treatment approach. A personalized treatment is important from the point of view of both efficacy and safety. Recent guidelines are based mainly on entirely phenotypic characteristics such as diabetes duration, presence of macrovascular complications, or risk of hypoglycemia with the use of individual drugs. So far, genetic knowledge is used to guide treatment in the monogenic forms of diabetes. With the accumulating pharmacogenetic evidence in type 2 diabetes, there are reasonable expectations that genetics might help in the adjustment of drug doses to reduce severe side effects, as well as to make better therapeutic choices among the drugs available for the treatment of diabetes.
Collapse
Affiliation(s)
- Ivan Tkáč
- Department of Internal Medicine 4, P. J. Šafárik University, Faculty of Medicine, L. Pasteur University Hospital, Rastislavova 43, 041 90, Košice, Slovakia,
| |
Collapse
|
24
|
Pollastro C, Ziviello C, Costa V, Ciccodicola A. Pharmacogenomics of Drug Response in Type 2 Diabetes: Toward the Definition of Tailored Therapies? PPAR Res 2015; 2015:415149. [PMID: 26161088 PMCID: PMC4486250 DOI: 10.1155/2015/415149] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/24/2015] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes is one of the major causes of mortality with rapidly increasing prevalence. Pharmacological treatment is the first recommended approach after failure in lifestyle changes. However, a significant number of patients shows-or develops along time and disease progression-drug resistance. In addition, not all type 2 diabetic patients have the same responsiveness to drug treatment. Despite the presence of nongenetic factors (hepatic, renal, and intestinal), most of such variability is due to genetic causes. Pharmacogenomics studies have described association between single nucleotide variations and drug resistance, even though there are still conflicting results. To date, the most reliable approach to investigate allelic variants is Next-Generation Sequencing that allows the simultaneous analysis, on a genome-wide scale, of nucleotide variants and gene expression. Here, we review the relationship between drug responsiveness and polymorphisms in genes involved in drug metabolism (CYP2C9) and insulin signaling (ABCC8, KCNJ11, and PPARG). We also highlight the advancements in sequencing technologies that to date enable researchers to perform comprehensive pharmacogenomics studies. The identification of allelic variants associated with drug resistance will constitute a solid basis to establish tailored therapeutic approaches in the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Carla Pollastro
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
- DiST, Department of Science and Technology, “Parthenope” University of Naples, Centro Direzionale, Isola C4, 80143 Naples, Italy
| | - Carmela Ziviello
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Alfredo Ciccodicola
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
- DiST, Department of Science and Technology, “Parthenope” University of Naples, Centro Direzionale, Isola C4, 80143 Naples, Italy
| |
Collapse
|
25
|
Emami-Riedmaier A, Schaeffeler E, Nies AT, Mörike K, Schwab M. Stratified medicine for the use of antidiabetic medication in treatment of type II diabetes and cancer: where do we go from here? J Intern Med 2015; 277:235-247. [PMID: 25418285 DOI: 10.1111/joim.12330] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
At present, the global diabetes epidemic is affecting 347 million individuals, 90% of whom are diagnosed with type II diabetes mellitus (T2DM). T2DM is commonly treated with more than one type of therapy, including oral antidiabetic drugs (OADs) and agents used in the treatment of diabetic complications. Several pharmacological classes of OADs are currently available for the treatment of T2DM, of which insulin secretagogues (i.e. sulphonylureas and meglitinides), insulin sensitizers [thiazolidinediones (TZDs)] and biguanides are the most commonly prescribed. Although many of these OADs have been used for more than half a century in the treatment of T2DM, the pharmacogenomic characteristics of these compounds have only recently been investigated, primarily in retrospective studies. Recent advances in pharmacogenomics have led to the identification of polymorphisms that affect the expression and function of drug-metabolizing enzymes and drug transporters, as well as drug targets and receptors. These polymorphisms have been shown to affect the therapeutic response to and side effects associated with OADs. The aim of this review was to provide an up-to-date summary of some of the pharmacogenomic data obtained from studies of T2DM treatment, with a focus on polymorphisms in genes affecting pharmacokinetics, pharmacodynamics and treatment outcome of the most commonly prescribed OADs. In addition, the implications of pharmacogenomics in the use of the OAD metformin in cancer will be briefly discussed. Finally, we will focus on recent advances in novel 'omics' technologies and discuss how these might aid in the personalized management of T2DM.
Collapse
Affiliation(s)
- A Emami-Riedmaier
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tübingen, Tübingen, Germany
| | - E Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tübingen, Tübingen, Germany
| | - A T Nies
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tübingen, Tübingen, Germany
| | - K Mörike
- Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
| | - M Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
26
|
Ametov AS, Kamynina LL, Akhmedova ZG. Type 2 diabetes mellitus: Clinical aspects of genetics, nutrigenetics, and pharmacogenetics. TERAPEVT ARKH 2015. [DOI: 10.17116/terarkh2015878124-131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
27
|
Brunetti A, Brunetti FS, Chiefari E. Pharmacogenetics of type 2 diabetes mellitus: An example of success in clinical and translational medicine. World J Transl Med 2014; 3:141-149. [DOI: 10.5528/wjtm.v3.i3.141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/25/2014] [Accepted: 11/03/2014] [Indexed: 02/05/2023] Open
Abstract
The pharmacological interventions currently available to control type 2 diabetes mellitus (T2DM) show a wide interindividual variability in drug response, emphasizing the importance of a personalized, more effective medical treatment for each individual patient. In this context, a growing interest has emerged in recent years and has focused on pharmacogenetics, a discipline aimed at understanding the variability in patients’ drug response, making it possible to predict which drug is best for each patient and at what doses. Recent pharmacological and clinical evidences indicate that genetic polymorphisms (or genetic variations) of certain genes can adversely affect drug response and therapeutic efficacy of oral hypoglycemic agents in patients with T2DM, through pharmacokinetic- and/or pharmacodynamic-based mechanisms that may reduce the therapeutic effects or increase toxicity. For example, genetic variants in genes encoding enzymes of the cytochrome P-450 superfamily, or proteins of the ATP-sensitive potassium channel on the beta-cell of the pancreas, are responsible for the interindividual variability of drug response to sulfonylureas in patients with T2DM. Instead, genetic variants in the genes that encode for the organic cation transporters of metformin have been related to changes in both pharmacodynamic and pharmacokinetic responses to metformin in metformin-treated patients. Thus, based on the individual’s genotype, the possibility, in these subjects, of a personalized therapy constitutes the main goal of pharmacogenetics, directly leading to the development of the right medicine for the right patient. Undoubtedly, this represents an integral part of the translational medicine network.
Collapse
|
28
|
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 2014; 15:511-7. [PMID: 25492374 DOI: 10.1007/s10238-014-0322-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/20/2014] [Indexed: 12/12/2022]
Abstract
Metformin is an oral antidiabetic drug, commonly used for treating type 2 diabetes mellitus (T2DM) patients. It is transported into the hepatocytes by polyspecific organic cation transporter 1, which is encoded by the gene SLC22A1. It has been hypothesized that genetic variations of SLC22A1 gene will influence inter-individual variation in glucose lowering efficacy of metformin. Previous studies have demonstrated this in other populations with conflicting results, but it remains to be elucidated in Indian population. Henceforth, the objective of the study was to evaluate the impact of SLC22A1 rs622342 gene polymorphism on the clinical efficacy of metformin in South Indian T2DM patients. A total of 122 newly detected, treatment naive T2DM patients of either sex were included in this study. The patients were started on metformin monotherapy and followed up for 12 weeks. Genotype was determined using qRT-PCR. Before and after treatment with metformin, body mass index (BMI), serum lipid profile, glycated hemoglobin (HbA1c), fasting and postprandial glucose level, and blood pressure (BP) were measured. The study cohort mean age was 49.57 ± 9.88 years. Of the 122 T2DM patients, 93 were classified as responders and 29 as non-responders based on fall in HbA1c levels. Interestingly, carriers of one variant allele 'C' (AC) of rs622342 polymorphism were less among the responders than those who did not (44.8 vs. 22.6 %). The response was even lesser (13.8 vs. 4.3 %) in carriers of two copies of "C" allele (CC). On the contrary, patients with two copies of allele 'A' (AA) had 5.6 times greater chance of responding to metformin treatment. A similar trend was observed when the proportion was analyzed under different genetic models (OR 3.85, 95 % CI 1.61-9.19 for dominant; OR 3.56, 95 % CI 0.83-15.26 for recessive; OR 0.35, 95 % CI 0.14-0.86 for over-dominant; and OR 4.10, 95 % CI 1.78-9.43 for additive). Further, metformin showed significant beneficial effects on BMI, HbA1c, FPG, PPG, lipid parameters and BP. These data suggest that the allele and genotypes of SLC22A1 rs622342 gene polymorphism were associated with the therapeutic efficacy of metformin in South Indian patients with T2DM.
Collapse
Affiliation(s)
- Gurusamy Umamaheswaran
- Department of Pharmacology, ICMR Centre for Advanced Research in Pharmacogenomics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605006, India.
| | | | - Solai Elango Damodaran
- Department of Pharmacology, ICMR Centre for Advanced Research in Pharmacogenomics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605006, India
| | - Ashok Kumar Das
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605006, India.,Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605006, India
| | - Chandrasekaran Adithan
- Department of Pharmacology, ICMR Centre for Advanced Research in Pharmacogenomics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605006, India
| |
Collapse
|
29
|
Groop L, Storm P, Rosengren A. Can genetics improve precision of therapy in diabetes? Trends Endocrinol Metab 2014; 25:440-3. [PMID: 25028244 DOI: 10.1016/j.tem.2014.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 06/11/2014] [Indexed: 11/25/2022]
Abstract
Diabetes mellitus is a lifelong, incapacitating disease affecting multiple organs. Presently, type 2 diabetes (T2D) can neither be prevented nor cured and the disease is associated with devastating chronic complications. These complications impose an immense burden on the quality of life of patients and account for about 12% of direct health care costs in Europe. Genetic analysis will increase our understanding of this heterogeneous disease and may help offer more personalized treatment.
Collapse
Affiliation(s)
- Leif Groop
- Lund University Diabetes Centre, Department of Clinical Science, Lund University, 20502 Malmö, Sweden.
| | - Petter Storm
- Lund University Diabetes Centre, Department of Clinical Science, Lund University, 20502 Malmö, Sweden
| | - Anders Rosengren
- Lund University Diabetes Centre, Department of Clinical Science, Lund University, 20502 Malmö, Sweden
| |
Collapse
|