1
|
Pradana AD, Kristin E, Nugrahaningsih DAA, Nugroho AK, Pinzon RT. Influence of Solute Carrier Family 22 Member 1 ( SLC22A1) Gene Polymorphism on Metformin Pharmacokinetics and HbA1c Levels: A Systematic Review. Curr Diabetes Rev 2024; 20:e070823219470. [PMID: 37550919 DOI: 10.2174/1573399820666230807145202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/23/2023] [Accepted: 06/16/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Solute Carrier Family 22 Member 1 (SLC22A1, also known as OCT1) protein has a vital role in the metabolism of metformin, a first-line anti-diabetes medication. Genetic poly-morphism in SLC22A1 influences individual response to metformin. OBJECTIVE This review aims to compile the current knowledge about the effects of SLC22A1 genetic polymorphism on metformin pharmacokinetics and HbA1c levels. METHODS We followed the PRISMA 2020 standards to conduct a systematic review. We searched the publications for all appropriate evidence on the effects of SLC22A1 genetic polymorphism on metformin pharmacokinetics and HbA1c from January 2002 to December 2022. RESULTS Initial database searches identified 7,171 relevant studies. We reviewed 155 titles and abstracts after deleting duplicates. After applying inclusion and exclusion criteria, 23 studies remained. CONCLUSION Three studies found that rs12208357, rs34059508, and G465R had a considerable impact (p < 0.05) on metformin pharmacokinetics, resulting in increased metformin plasma (Cmax), a higher active amount of drug in the blood (AUC), and lower volume of distribution (Vd) (p<0.05). SLC22A1 polymorphisms with effects on HbA1c include rs628031 (four of seven studies), rs622342 (four of six studies), rs594709 (one study), rs2297374, and rs1867351 (one of two studies), rs34130495 (one study), and rs11212617 (one study) (p < 0.05).
Collapse
Affiliation(s)
- A D Pradana
- Department of Pharmacy, Universitas Islam Indonesia, Yogyakarta 55584, Indonesia
- Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, Yogyakarta, Indonesia
| | - E Kristin
- Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, Yogyakarta, Indonesia
| | - D A A Nugrahaningsih
- Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, Yogyakarta, Indonesia
| | - A K Nugroho
- Faculty of Pharmacy, Gadjah Mada University, Yogyakarta, Indonesia
| | - R T Pinzon
- Medical Faculty, Duta Wacana Christian University, Yogyakarta, Indonesia
| |
Collapse
|
2
|
Ahmed A, Elsadek HM, Shalaby SM, Elnahas HM. Association of SLC22A1, SLC47A1, and KCNJ11 polymorphisms with efficacy and safety of metformin and sulfonylurea combination therapy in Egyptian patients with type 2 diabetes. Res Pharm Sci 2023; 18:614-625. [PMID: 39005567 PMCID: PMC11246114 DOI: 10.4103/1735-5362.389949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/08/2023] [Accepted: 09/12/2023] [Indexed: 07/16/2024] Open
Abstract
Background and purpose Multidrug and toxin extrusion transporter 1 (MATE1), encoded by the SLC47A1 gene and single nucleotide polymorphisms of organic cation transport 1, may impact metformin's responsiveness and side effects. Inward-rectifier potassium channel 6.2 (Kir 6.2) subunits encoded by KCNJ11 may affect the response to sulfonylurea. This study aimed to evaluate the association between SLC22A1 rs72552763 and rs628031, SLC47A1 rs2289669 and KCNJ11 rs5219 genetic variations with sulfonylurea and metformin combination therapy efficacy and safety in Egyptian type 2 diabetes mellitus patients. Experimental approach This study was conducted on 100 cases taking at least one year of sulfonylurea and metformin combination therapy. Patients were genotyped via the polymerase chain reaction-restriction fragment length polymorphism technique. Then, according to their glycated hemoglobin level, cases were subdivided into non-responders or responders. Depending on metformin-induced gastrointestinal tract side effects incidence, patients are classified as tolerant or intolerant. Findings/Results KCNJ11 rs5219 heterozygous and homozygous mutant genotypes, SLC47A1 rs2289669 heterozygous and homozygous mutant genotypes (AA and AG), and mutant alleles of both polymorphisms were significantly related with increased response to combined therapy. Individuals with the SLC22A1 (rs72552763) GAT/del genotype and the SLC22A1 (rs628031) AG and AA genotypes were at a higher risk for metformin-induced gastrointestinal tract adverse effects. Conclusion and implications The results implied a role for SLC47A1 rs2289669 and KCNJ11 rs5219 in the responsiveness to combined therapy. SLC22A1 (rs628031) and (rs72552763) polymorphisms may be associated with increased metformin adverse effects in type 2 diabetes mellitus patients.
Collapse
Affiliation(s)
- Aya Ahmed
- Department of Pharmacy Practice, Faculty of Pharmacy, Zagazig University, Egypt
| | - Hany M Elsadek
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Egypt
| | - Sally M Shalaby
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Egypt
| | - Hanan M Elnahas
- Department of Pharmaceutical and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Egypt
| |
Collapse
|
3
|
Degaga A, Sirgu S, Huri HZ, Sim MS, Kebede T, Tegene B, Loganadan NK, Engidawork E, Shibeshi W. Association of Met420del Variant of Metformin Transporter Gene SLC22A1 with Metformin Treatment Response in Ethiopian Patients with Type 2 Diabetes. Diabetes Metab Syndr Obes 2023; 16:2523-2535. [PMID: 37641646 PMCID: PMC10460611 DOI: 10.2147/dmso.s426632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/12/2023] [Indexed: 08/31/2023] Open
Abstract
Objective This study aimed to evaluate whether the M420del variants of SLC22A1 (rs72552763) is associated with metformin treatment response in Ethiopian patients with type 2 diabetes mellitus (T2DM). Patients and Methods A prospective observational cohort study was conducted on 86 patients with T2DM who had been receiving metformin monotherapy for <1 year. Patients showing ≥0.5% reduction in HbA1c levels from baseline within 3 months and remained low for at least another 3 months were defined as responders while those patients with <0.5% reduction in HbA1c levels and/or those whom started a new class of glucose-lowering drug(s) because of unsatisfactory reduction were defined as non-responders. In addition, good glycemic control was observed when HbA1c ≤7.0%, and the above values were regarded as poor. Genotyping of rs72552763 SNP was performed using TaqMan® Drug Metabolism Enzyme Genotyping Assay and its association with metformin response and glycemic control were assessed by measuring the change in HbA1c and fasting blood glucose levels using Chi-square, logistic regression and Mann-Whitney U-test. Statistical significance was set at p <0.05. Results The minor allele frequency of the rs72552763 SNP of SLC22A1 was 9.3%. Metformin response was significantly higher in deletion_GAT (del_G) genotypes as compared to the wild-type GAT_GAT (G_G) genotypes. Furthermore, a significantly lower median treatment HbA1 level was found in del_G genotypes as compared to G_G genotypes. However, the association of rs72552763 with metformin response was not replicated at the allele level. In contrast, the minor del_allele was significantly associated with good glycemic control compared to the G_allele, though not replicated at del_G genotypes level. Conclusion This study demonstrated that metformin response was significantly higher in study participants with a heterozygous carrier of M420del variants of SLC22A1 as compared to the wild-type G_G genotypes after 3 months of treatment.
Collapse
Affiliation(s)
- Abraham Degaga
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Clinical Pharmacy & Pharmacy Practice, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Sisay Sirgu
- Department of Internal Medicine, Diabetes and Endocrinology Unit, Saint Paul Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Hasniza Zaman Huri
- Department of Clinical Pharmacy & Pharmacy Practice, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Maw Shin Sim
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur, Malaysia
| | - Tedla Kebede
- Department of Internal Medicine, Diabetes and Endocrinology Unit, Addis Ababa University, Addis Ababa, Ethiopia
| | - Birhanemeskel Tegene
- Department of Microbiology, Saint Paul Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | | | - Ephrem Engidawork
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Workineh Shibeshi
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
4
|
Dutta S, Shah RB, Singhal S, Dutta SB, Bansal S, Sinha S, Haque M. Metformin: A Review of Potential Mechanism and Therapeutic Utility Beyond Diabetes. Drug Des Devel Ther 2023; 17:1907-1932. [PMID: 37397787 PMCID: PMC10312383 DOI: 10.2147/dddt.s409373] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/10/2023] [Indexed: 07/04/2023] Open
Abstract
Metformin has been designated as one of the most crucial first-line therapeutic agents in the management of type 2 diabetes mellitus. Primarily being an antihyperglycemic agent, metformin also has a plethora of pleiotropic effects on various systems and processes. It acts majorly by activating AMPK (Adenosine Monophosphate-Activated Protein Kinase) in the cells and reducing glucose output from the liver. It also decreases advanced glycation end products and reactive oxygen species production in the endothelium apart from regulating the glucose and lipid metabolism in the cardiomyocytes, hence minimizing the cardiovascular risks. Its anticancer, antiproliferative and apoptosis-inducing effects on malignant cells might prove instrumental in the malignancy of organs like the breast, kidney, brain, ovary, lung, and endometrium. Preclinical studies have also shown some evidence of metformin's neuroprotective role in Parkinson's disease, Alzheimer's disease, multiple sclerosis and Huntington's disease. Metformin exerts its pleiotropic effects through varied pathways of intracellular signalling and exact mechanism in the majority of them remains yet to be clearly defined. This article has extensively reviewed the therapeutic benefits of metformin and the details of its mechanism for a molecule of boon in various conditions like diabetes, prediabetes, obesity, polycystic ovarian disease, metabolic derangement in HIV, various cancers and aging.
Collapse
Affiliation(s)
- Siddhartha Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Rima B Shah
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Shubha Singhal
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Sudeshna Banerjee Dutta
- Department of Medical Surgical Nursing, Shri Anand Institute of Nursing, Rajkot, Gujarat, 360005, India
| | - Sumit Bansal
- Department of Anaesthesiology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Susmita Sinha
- Department of Physiology, Khulna City Medical College and Hospital, Khulna, Bangladesh
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kuala Lumpur, 57000, Malaysia
| |
Collapse
|
5
|
Dong Y, Qi Y, Jiang H, Mi T, Zhang Y, Peng C, Li W, Zhang Y, Zhou Y, Zang Y, Li J. The development and benefits of metformin in various diseases. Front Med 2023; 17:388-431. [PMID: 37402952 DOI: 10.1007/s11684-023-0998-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/01/2023] [Indexed: 07/06/2023]
Abstract
Metformin has been used for the treatment of type II diabetes mellitus for decades due to its safety, low cost, and outstanding hypoglycemic effect clinically. The mechanisms underlying these benefits are complex and still not fully understood. Inhibition of mitochondrial respiratory-chain complex I is the most described downstream mechanism of metformin, leading to reduced ATP production and activation of AMP-activated protein kinase (AMPK). Meanwhile, many novel targets of metformin have been gradually discovered. In recent years, multiple pre-clinical and clinical studies are committed to extend the indications of metformin in addition to diabetes. Herein, we summarized the benefits of metformin in four types of diseases, including metabolic associated diseases, cancer, aging and age-related diseases, neurological disorders. We comprehensively discussed the pharmacokinetic properties and the mechanisms of action, treatment strategies, the clinical application, the potential risk of metformin in various diseases. This review provides a brief summary of the benefits and concerns of metformin, aiming to interest scientists to consider and explore the common and specific mechanisms and guiding for the further research. Although there have been countless studies of metformin, longitudinal research in each field is still much warranted.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yingbei Qi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tian Mi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yunkai Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanchen Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongmei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Lingang Laboratory, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
6
|
Lange C, Brüggemann J, Thüner T, Jauckus J, Strowitzki T, Germeyer A. Changes in the expression of cancer- and metastasis-related genes and proteins after metformin treatment under different metabolic conditions in endometrial cancer cells. Heliyon 2023; 9:e16678. [PMID: 37313172 PMCID: PMC10258389 DOI: 10.1016/j.heliyon.2023.e16678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 05/20/2023] [Accepted: 05/24/2023] [Indexed: 06/15/2023] Open
Abstract
Research question Hyperinsulinemia and elevated estrogen levels are known risk factors for endometrial cancer (EC) development and are associated with obesity, type 2 diabetes mellitus (T2DM), insulin resistance, among others. Metformin, an insulin-sensitizing drug, displays anti-tumor effects in cancer patients, including EC, but the mechanism of action is still not completely understood. In the present study, the effects of metformin on gene and protein expression were investigated in pre- and postmenopausal EC in vitro models in order to identify candidates that are potentially involved in the drug's anti-cancer mechanism. Design After treating the cells with metformin (0.1 and 1.0 mmol/L), changes in the expression of >160 cancer- and metastasis-related gene transcripts were evaluated with RNA arrays. A total of 19 genes and 7 proteins were selected for a follow-up expression analysis, including further treatment conditions, in order to evaluate the influence of hyperinsulinemia and hyperglycemia on metformin-induced effects. Results Changes in the expression of BCL2L11, CDH1, CDKN1A, COL1A1, PTEN, MMP9 and TIMP2 were analyzed on gene and protein level. The consequences resulting from the detected expression changes as well as the influence of varying environmental influences are discussed in detail. With the presented data, we contribute to a better understanding of the direct anti-cancer activity of metformin as well as its underlying mechanism of action in EC cells. Conclusions Although further research will be necessary to confirm the data, the influence of different environmental settings on metformin-induced effects could be highlighted with the presented data. Additionally, gene and protein regulation were not similar in the pre- and postmenopausal in vitro models.
Collapse
|
7
|
Chen P, Cao Y, Chen S, Liu Z, Chen S, Guo Y. Association of SLC22A1, SLC22A2, SLC47A1, and SLC47A2 Polymorphisms with Metformin Efficacy in Type 2 Diabetic Patients. Biomedicines 2022; 10:2546. [PMID: 36289808 PMCID: PMC9599747 DOI: 10.3390/biomedicines10102546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023] Open
Abstract
Response to metformin, first-line therapy for type 2 diabetes mellitus (T2DM), exists interindividual variation. Considering that transporters belonging to the solute carrier (SLC) superfamily are determinants of metformin pharmacokinetics, we evaluated the effects of promoter variants in organic cation transporter 1 (OCT1) (SLC22A1 rs628031), OCT2 (SLC22A2 rs316019), multidrug and toxin extrusion protein 1 (MATE1) (SLC47A1 rs2289669), and MATE2 (SLC47A2 rs12943590) on the variation in metformin response. The glucose-lowering effects and improvement of insulin resistance of metformin were assessed in newly diagnosed, treatment-naive type 2 diabetic patients of Han nationality in Chaoshan China (n = 93) receiving metformin. Fasting plasma glucose (FPG), fasting insulin (FINS), glycated hemoglobin A1 (HbA1C), homeostasis model assessment-insulin sensitivity (HOMA-IS), and homeostasis model assessment-insulin resistance (HOMA-IR) were the main metformin efficacy measurements. There were significant correlations between both SLC47A1 rs2289669 and SLC47A2 rs12943590 and the efficacy of metformin in individuals with T2DM. In normal weight T2DM patients, significant associations between the AA and GG genotypes of the rs2289669 variant of SLC47A1 and a greater reduction in FINS and HOMA-IR were detected. A significant correlation was observed between the AG genotype of the rs12943590 polymorphism of SLC47A2 and a greater reduction in HOMA-IR. Gene-environment interaction analysis showed that in the FINS interaction model, the second-order of dose30_g-SLC47A2 rs12943590 was statistically significant. The variants of SLC47A1 rs2289669 and SLC47A2 rs12943590 could be predictors of insulin resistance in type 2 diabetic patients treated with metformin. The second-order interaction of dose30_g-SLC47A2 rs12943590 may have a significant effect on FINS in patients with T2DM on metformin treatment. These findings suggest that promoter variants of SLC47A1 and SLC47A2 are important determinants of metformin transport and response in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Peixian Chen
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Shantou University Medical College, Shantou 515000, China
| | - Yumin Cao
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Shantou University Medical College, Shantou 515000, China
| | - Shenren Chen
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Shantou University Medical College, Shantou 515000, China
| | - Zhike Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Shiyi Chen
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Shantou University Medical College, Shantou 515000, China
| | - Yali Guo
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Shantou University Medical College, Shantou 515000, China
| |
Collapse
|
8
|
Nies AT, Schaeffeler E, Schwab M. Hepatic solute carrier transporters and drug therapy: Regulation of expression and impact of genetic variation. Pharmacol Ther 2022; 238:108268. [DOI: 10.1016/j.pharmthera.2022.108268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
|
9
|
Triggle CR, Mohammed I, Bshesh K, Marei I, Ye K, Ding H, MacDonald R, Hollenberg MD, Hill MA. Metformin: Is it a drug for all reasons and diseases? Metabolism 2022; 133:155223. [PMID: 35640743 DOI: 10.1016/j.metabol.2022.155223] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Metformin was first used to treat type 2 diabetes in the late 1950s and in 2022 remains the first-choice drug used daily by approximately 150 million people. An accumulation of positive pre-clinical and clinical data has stimulated interest in re-purposing metformin to treat a variety of diseases including COVID-19. In polycystic ovary syndrome metformin improves insulin sensitivity. In type 1 diabetes metformin may help reduce the insulin dose. Meta-analysis and data from pre-clinical and clinical studies link metformin to a reduction in the incidence of cancer. Clinical trials, including MILES (Metformin In Longevity Study), and TAME (Targeting Aging with Metformin), have been designed to determine if metformin can offset aging and extend lifespan. Pre-clinical and clinical data suggest that metformin, via suppression of pro-inflammatory pathways, protection of mitochondria and vascular function, and direct actions on neuronal stem cells, may protect against neurodegenerative diseases. Metformin has also been studied for its anti-bacterial, -viral, -malaria efficacy. Collectively, these data raise the question: Is metformin a drug for all diseases? It remains unclear as to whether all of these putative beneficial effects are secondary to its actions as an anti-hyperglycemic and insulin-sensitizing drug, or result from other cellular actions, including inhibition of mTOR (mammalian target for rapamycin), or direct anti-viral actions. Clarification is also sought as to whether data from ex vivo studies based on the use of high concentrations of metformin can be translated into clinical benefits, or whether they reflect a 'Paracelsus' effect. The environmental impact of metformin, a drug with no known metabolites, is another emerging issue that has been linked to endocrine disruption in fish, and extensive use in T2D has also raised concerns over effects on human reproduction. The objectives for this review are to: 1) evaluate the putative mechanism(s) of action of metformin; 2) analyze the controversial evidence for metformin's effectiveness in the treatment of diseases other than type 2 diabetes; 3) assess the reproducibility of the data, and finally 4) reach an informed conclusion as to whether metformin is a drug for all diseases and reasons. We conclude that the primary clinical benefits of metformin result from its insulin-sensitizing and antihyperglycaemic effects that secondarily contribute to a reduced risk of a number of diseases and thereby enhancing healthspan. However, benefits like improving vascular endothelial function that are independent of effects on glucose homeostasis add to metformin's therapeutic actions.
Collapse
Affiliation(s)
- Chris R Triggle
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar.
| | - Ibrahim Mohammed
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Khalifa Bshesh
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Isra Marei
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Kevin Ye
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Hong Ding
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Ross MacDonald
- Distribution eLibrary, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, a Cumming School of Medicine, University of Calgary, T2N 4N1, Canada
| | - Michael A Hill
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia 65211, MO, USA
| |
Collapse
|
10
|
Nasykhova YA, Barbitoff YA, Tonyan ZN, Danilova MM, Nevzorov IA, Komandresova TM, Mikhailova AA, Vasilieva TV, Glavnova OB, Yarmolinskaya MI, Sluchanko EI, Glotov AS. Genetic and Phenotypic Factors Affecting Glycemic Response to Metformin Therapy in Patients with Type 2 Diabetes Mellitus. Genes (Basel) 2022; 13:genes13081310. [PMID: 35893047 PMCID: PMC9330240 DOI: 10.3390/genes13081310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/10/2022] Open
Abstract
Metformin is an oral hypoglycemic agent widely used in clinical practice for treatment of patients with type 2 diabetes mellitus (T2DM). The wide interindividual variability of response to metformin therapy was shown, and recently the impact of several genetic variants was reported. To assess the independent and combined effect of the genetic polymorphism on glycemic response to metformin, we performed an association analysis of the variants in ATM, SLC22A1, SLC47A1, and SLC2A2 genes with metformin response in 299 patients with T2DM. Likewise, the distribution of allele and genotype frequencies of the studied gene variants was analyzed in an extended group of patients with T2DM (n = 464) and a population group (n = 129). According to our results, one variant, rs12208357 in the SLC22A1 gene, had a significant impact on response to metformin in T2DM patients. Carriers of TT genotype and T allele had a lower response to metformin compared to carriers of CC/CT genotypes and C allele (p-value = 0.0246, p-value = 0.0059, respectively). To identify the parameters that had the greatest importance for the prediction of the therapy response to metformin, we next built a set of machine learning models, based on the various combinations of genetic and phenotypic characteristics. The model based on a set of four parameters, including gender, rs12208357 genotype, familial T2DM background, and waist–hip ratio (WHR) showed the highest prediction accuracy for the response to metformin therapy in patients with T2DM (AUC = 0.62 in cross-validation). Further pharmacogenetic studies may aid in the discovery of the fundamental mechanisms of type 2 diabetes, the identification of new drug targets, and finally, it could advance the development of personalized treatment.
Collapse
Affiliation(s)
- Yulia A. Nasykhova
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
| | - Yury A. Barbitoff
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
- St. Petersburg State University, 199034 Saint-Petersburg, Russia
| | - Ziravard N. Tonyan
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
| | - Maria M. Danilova
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
| | - Ivan A. Nevzorov
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
| | | | - Anastasiia A. Mikhailova
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
| | | | - Olga B. Glavnova
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
| | - Maria I. Yarmolinskaya
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
| | | | - Andrey S. Glotov
- D. O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, 199034 Saint-Petersburg, Russia; (Y.A.N.); (Y.A.B.); (Z.N.T.); (M.M.D.); (I.A.N.); (A.A.M.); (O.B.G.); (M.I.Y.)
- Correspondence: ; Tel.: +7-9117832003
| |
Collapse
|
11
|
Chen P, Cao Y, Guo Y, Xu Q, Wang X, Zhang L, Liu Z, Chen D, Chen S, Chen S. Association of SLC22A1 rs622342 and ATM rs11212617 polymorphisms with metformin efficacy in patients with type 2 diabetes. Pharmacogenet Genomics 2022; 32:67-71. [PMID: 34545025 DOI: 10.1097/fpc.0000000000000454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Metformin is the first-choice oral anti-hyperglycemic drug for type 2 diabetes mellitus (T2DM) patients. There are controversies about the association of SLC22A1 rs622342, which was not reported in the Chinese population, and ataxia-telangiectasia mutated (ATM) rs11212617 polymorphisms with metformin efficacy in T2DM. Our study was to investigate the effects of the two single nucleotide polymorphisms on the efficacy of metformin in T2DM of Han nationality in Chaoshan China. After enrollment, 82 newly diagnosed T2DM patients went on 2-month metformin monotherapy. According to BMI before treatment, the patients were divided into a normal weight group (≥18.5 and <25 kg/m2) and an overweight group (BMI ≥ 25 and <30 kg/m2). T-test, Pearson χ2 test, and regression analysis, which adjusted for age, BMI, sex, the dose of metformin, education, tea drink, smoking, and sweet, were used to evaluate the effects of rs622342 and rs11212617 on several variables, such as fasting plasma glucose (FPG). Compared with the AA or CC genotype, patients with AC genotype of rs622342 achieved greater reduction in Δ60FPG and Δ(60-30)FPG (P = 0.00820, 0.00089, respectively). For 11212617, the reduction in Δ30FPG and Δ60FPG was significantly different among patients with the AC genotype (P = 0.00026, 0.00820, respectively). Our results indicated that common variants of SLC22A1 rs622342 and ATM rs11212617 were associated with the efficacy of metformin in T2DM of Han nationality in Chaoshan China.
Collapse
Affiliation(s)
- Peixian Chen
- Department of Endocrinology
- Department of Infection Control
| | - Yumin Cao
- Department of Neurology, Meizhou People's Hospital, Meizhou, Guangdong Province
| | - Yali Guo
- Department of Endocrinology, Central Hospital of Shenzhen Guangming New District, Shenzhen
| | - Qi Xu
- Department of Endocrinology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province
| | - Xiaozhu Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, People's Republic of China
| | - Liuwei Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, People's Republic of China
| | - Zhike Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, People's Republic of China
| | - Dafang Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, People's Republic of China
| | - Shiyi Chen
- Department of Endocrinology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province
| | - Shenren Chen
- Department of Endocrinology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province
| |
Collapse
|
12
|
Cahua-Pablo JÁ, Gómez-Zamudio JH, Reséndiz-Abarca CA, Tello-Flores VA, Eulogio-Metodio Y, Ramírez-Vargas MA, Cruz M, Del Carmen Alarcón-Romero L, Matia-García I, Marino-Ortega LA, Zubillaga-Guerrero MI, Flores-Alfaro E. Genetic variants in SLC22A1 are related to serum lipid levels in Mexican women. Lipids 2021; 57:105-114. [PMID: 34927264 DOI: 10.1002/lipd.12334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/10/2021] [Accepted: 12/03/2021] [Indexed: 11/07/2022]
Abstract
Dyslipidemia is the main risk factor for coronary artery disease and is characterized by alterations in concentrations of lipids, including low-density lipoprotein cholesterol (LDL-c), high-density lipoprotein cholesterol (HDL-c), and triacylglycerols. The participation of several genes in the development of dyslipidemia has been evidenced. Genetic variants in SLC22A1 have been associated with elevated cholesterol and LDL-c levels. The aim of this study was to evaluate the association between single-nucleotide polymorphisms (SNPs) in the SLC22A1 gene with atherogenic risk lipid levels in Mexican women. Anthropometric and biochemical measurements were performed, and four SNPs in SLC22A1 were genotyped by real-time polymerase chain reaction. The Hardy-Weinberg equilibrium was verified, and haplotype frequencies were calculated. We found significant differences between the allele frequencies of the SNPs analyzed with those reported in Mexico and in the world, which could be due to differences in the historical admixture of the women studied. Generalized linear models were evaluated to determine the association between genotypes and haplotypes with lipids levels. We identified a significant increase in total cholesterol and LDL-c levels in women who were carriers of the GA and AG genotypes of the polymorphisms rs628031 and rs594709, respectively, significant effect that is also shown in a dominant inheritance model. Interestingly, we identified an important relationship of the AGC-GAT haplotype with the elevation in LDL-c levels and AGA-GAT haplotype with the elevation in HDL-c levels. On the other hand, we found a strong linkage disequilibrium between the polymorphisms studied. Our results show that variants in the SLC22A1 gene influence serum levels of atherogenic risk lipids, suggesting that these variants probably affect the function of organic cation transporter-1 and therefore, on the regulation of lipid metabolism.
Collapse
Affiliation(s)
- José Ángel Cahua-Pablo
- Laboratorio en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Jaime Héctor Gómez-Zamudio
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Carlos Alberto Reséndiz-Abarca
- Laboratorio en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Vianet Argelia Tello-Flores
- Laboratorio en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Yesica Eulogio-Metodio
- Laboratorio en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Marco Antonio Ramírez-Vargas
- Laboratorio en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Luz Del Carmen Alarcón-Romero
- Laboratorio en Citopatología e Histoquímica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Inés Matia-García
- Laboratorio en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Linda Anahí Marino-Ortega
- Laboratorio en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Ma Isabel Zubillaga-Guerrero
- Laboratorio en Citopatología e Histoquímica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Eugenia Flores-Alfaro
- Laboratorio en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| |
Collapse
|
13
|
Koepsell H. Update on drug-drug interaction at organic cation transporters: mechanisms, clinical impact, and proposal for advanced in vitro testing. Expert Opin Drug Metab Toxicol 2021; 17:635-653. [PMID: 33896325 DOI: 10.1080/17425255.2021.1915284] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Organic cation transporters collectively called OCTs belong to three gene families (SLC22A1 OCT1, SLC22A2 OCT2, SLC22A3 OCT3, SLC22A4 OCTN1, SLC22A5 OCTN2, SLC29A4 PMAT, SLC47A1 MATE1, and SLC47A1 MATE2-K). OCTs transport structurally diverse drugs with overlapping selectivity. Some OCTs were shown to be critically involved in pharmacokinetics and therapeutic efficacy of cationic drugs. Drug-drug interactions at individual OCTs were shown to result in clinical effects. Procedures for in vitro testing of drugs for interaction with OCT1, OCT2, MATE1, and MATE2-K have been recommended.Areas covered: An overview of functional properties, cation selectivity, location, and clinical impact of OCTs is provided. In addition, clinically relevant drug-drug interactions in OCTs are compiled. Because it was observed that the half maximal concentration of drugs to inhibit transport by OCTs (IC50) is dependent on the transported cation and its concentration, an advanced protocol for in vitro testing of drugs for interaction with OCTs is proposed. In addition, it is suggested to include OCT3 and PMAT for in vitro testing.Expert opinion: Research on clinical roles of OCTs should be reinforced including more transporters and drugs. An improvement of the in vitro testing protocol considering recent data is imperative for the benefit of patients.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology, University Würzburg, Würzburg, Germany
| |
Collapse
|
14
|
A Whole-Body Physiologically Based Pharmacokinetic Model Characterizing Interplay of OCTs and MATEs in Intestine, Liver and Kidney to Predict Drug-Drug Interactions of Metformin with Perpetrators. Pharmaceutics 2021; 13:pharmaceutics13050698. [PMID: 34064886 PMCID: PMC8151202 DOI: 10.3390/pharmaceutics13050698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022] Open
Abstract
Transmembrane transport of metformin is highly controlled by transporters including organic cation transporters (OCTs), plasma membrane monoamine transporter (PMAT), and multidrug/toxin extrusions (MATEs). Hepatic OCT1, intestinal OCT3, renal OCT2 on tubule basolateral membrane, and MATE1/2-K on tubule apical membrane coordinately work to control metformin disposition. Drug–drug interactions (DDIs) of metformin occur when co-administrated with perpetrators via inhibiting OCTs or MATEs. We aimed to develop a whole-body physiologically based pharmacokinetic (PBPK) model characterizing interplay of OCTs and MATEs in the intestine, liver, and kidney to predict metformin DDIs with cimetidine, pyrimethamine, trimethoprim, ondansetron, rabeprazole, and verapamil. Simulations showed that co-administration of perpetrators increased plasma exposures to metformin, which were consistent with clinic observations. Sensitivity analysis demonstrated that contributions of the tested factors to metformin DDI with cimetidine are gastrointestinal transit rate > inhibition of renal OCT2 ≈ inhibition of renal MATEs > inhibition of intestinal OCT3 > intestinal pH > inhibition of hepatic OCT1. Individual contributions of transporters to metformin disposition are renal OCT2 ≈ renal MATEs > intestinal OCT3 > hepatic OCT1 > intestinal PMAT. In conclusion, DDIs of metformin with perpetrators are attributed to integrated effects of inhibitions of these transporters.
Collapse
|
15
|
Wenzel C, Drozdzik M, Oswald S. Organic Cation Transporter 1 an Intestinal Uptake Transporter: Fact or Fiction? Front Pharmacol 2021; 12:648388. [PMID: 33935750 PMCID: PMC8080103 DOI: 10.3389/fphar.2021.648388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/01/2021] [Indexed: 01/11/2023] Open
Abstract
Intestinal transporter proteins are known to affect the pharmacokinetics and in turn the efficacy and safety of many orally administered drugs in a clinically relevant manner. This knowledge is especially well-established for intestinal ATP-binding cassette transporters such as P-gp and BCRP. In contrast to this, information about intestinal uptake carriers is much more limited although many hydrophilic or ionic drugs are not expected to undergo passive diffusion but probably require specific uptake transporters. A transporter which is controversially discussed with respect to its expression, localization and function in the human intestine is the organic cation transporter 1 (OCT1). This review article provides an up-to-date summary on the available data from expression analysis as well as functional studies in vitro, animal findings and clinical observations. The current evidence suggests that OCT1 is expressed in the human intestine in small amounts (on gene and protein levels), while its cellular localization in the apical or basolateral membrane of the enterocytes remains to be finally defined, but functional data point to a secretory function of the transporter at the basolateral membrane. Thus, OCT1 should not be considered as a classical uptake transporter in the intestine but rather as an intestinal elimination pathway for cationic compounds from the systemic circulation.
Collapse
Affiliation(s)
- Christoph Wenzel
- Department of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany
| | - Marek Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
16
|
Mohammed I, Hollenberg MD, Ding H, Triggle CR. A Critical Review of the Evidence That Metformin Is a Putative Anti-Aging Drug That Enhances Healthspan and Extends Lifespan. Front Endocrinol (Lausanne) 2021; 12:718942. [PMID: 34421827 PMCID: PMC8374068 DOI: 10.3389/fendo.2021.718942] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
The numerous beneficial health outcomes associated with the use of metformin to treat patients with type 2 diabetes (T2DM), together with data from pre-clinical studies in animals including the nematode, C. elegans, and mice have prompted investigations into whether metformin has therapeutic utility as an anti-aging drug that may also extend lifespan. Indeed, clinical trials, including the MILES (Metformin In Longevity Study) and TAME (Targeting Aging with Metformin), have been designed to assess the potential benefits of metformin as an anti-aging drug. Preliminary analysis of results from MILES indicate that metformin may induce anti-aging transcriptional changes; however it remains controversial as to whether metformin is protective in those subjects free of disease. Furthermore, despite clinical use for over 60 years as an anti-diabetic drug, the cellular mechanisms by which metformin exerts either its actions remain unclear. In this review, we have critically evaluated the literature that has investigated the effects of metformin on aging, healthspan and lifespan in humans as well as other species. In preparing this review, particular attention has been placed on the strength and reproducibility of data and quality of the study protocols with respect to the pharmacokinetic and pharmacodynamic properties of metformin. We conclude that despite data in support of anti-aging benefits, the evidence that metformin increases lifespan remains controversial. However, via its ability to reduce early mortality associated with various diseases, including diabetes, cardiovascular disease, cognitive decline and cancer, metformin can improve healthspan thereby extending the period of life spent in good health. Based on the available evidence we conclude that the beneficial effects of metformin on aging and healthspan are primarily indirect via its effects on cellular metabolism and result from its anti-hyperglycemic action, enhancing insulin sensitivity, reduction of oxidative stress and protective effects on the endothelium and vascular function.
Collapse
Affiliation(s)
- Ibrahim Mohammed
- Department of Medical Education, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- *Correspondence: Chris R. Triggle, ; Ibrahim Mohammed,
| | - Morley D. Hollenberg
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Hong Ding
- Department of Medical Education, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- Departments of Medical Education and Pharmacology, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
| | - Chris R. Triggle
- Department of Medical Education, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- Departments of Medical Education and Pharmacology, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- *Correspondence: Chris R. Triggle, ; Ibrahim Mohammed,
| |
Collapse
|
17
|
Li L, Guan Z, Li R, Zhao W, Hao G, Yan Y, Xu Y, Liao L, Wang H, Gao L, Wu K, Gao Y, Li Y. Population pharmacokinetics and dosing optimization of metformin in Chinese patients with type 2 diabetes mellitus. Medicine (Baltimore) 2020; 99:e23212. [PMID: 33181704 PMCID: PMC7668473 DOI: 10.1097/md.0000000000023212] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Approximately 35% of patients fail to attain ideal initial blood glucose control under metformin monotherapy. The objective of this observational study is to simulate the optimal protocol of metformin according to the different renal function.The population pharmacokinetics of metformin was performed in 125 subjects with type 2 diabetes mellitus. Plasma concentrations of metformin were quantified by high-performance liquid chromatography. A population pharmacokinetic model of metformin was developed using NONMEN (version 7.2, Icon Development Solutions, USA). Monte Carlo simulation was used to simulate the concentration-time profiles for doses of metformin for 1000 times at different stages of renal function.The mean population pharmacokinetic parameters were apparent clearance 53.0 L/h, apparent volume of distribution 438 L, absorption rate constant 1.4 hour and lag-time 0.91 hour. Covariate analyses revealed that estimated glomerular filtration rate (eGFR) and bodyweight as individual factors influencing the apparent oral clearance: CL/F = 53.0 × ( bodyweight/75) × (eGFR/102.5)EXP(0.1797). The results of the simulation showed that patients should be prescribed metformin 2550 mg/d (t.i.d.) vs 3000 mg/d (b.i.d.) as the minimum doses for patients with augmented renal clearance.eGFR had a significant impact on metformin pharmacokinetics. Patients administered metformin twice a day require higher total daily doses than those with a regimen of 3 times a day at each stage of kidney function.
Collapse
Affiliation(s)
- Ling Li
- School of Pharmaceutical Sciences, Shandong University
| | - Ziwan Guan
- School of Pharmaceutical Sciences, Shandong University
| | - Rui Li
- School of Pharmaceutical Sciences, Shandong University
| | - Wei Zhao
- School of Pharmaceutical Sciences, Shandong University
| | - Guoxiang Hao
- School of Pharmaceutical Sciences, Shandong University
| | - Yan Yan
- School of Pharmaceutical Sciences, Shandong University
| | - Yuedong Xu
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan
| | - Lin Liao
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan
| | - Huanjun Wang
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan
| | - Li Gao
- School of Pharmaceutical Sciences, Shandong University
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan
| | - Kunrong Wu
- School of Pharmaceutical Sciences, Shandong University
| | - Yuxia Gao
- Department of Pharmacy, Shengli Hospital of Shengli Oilfield, Dongying, Shandong, China
| | - Yan Li
- School of Pharmaceutical Sciences, Shandong University
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan
| |
Collapse
|
18
|
Kuhlmann I, Arnspang Pedersen S, Skov Esbech P, Bjerregaard Stage T, Hougaard Christensen MM, Brøsen K. Using a limited sampling strategy to investigate the interindividual pharmacokinetic variability in metformin: A large prospective trial. Br J Clin Pharmacol 2020; 87:1963-1969. [PMID: 33118168 DOI: 10.1111/bcp.14591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/10/2020] [Accepted: 09/27/2020] [Indexed: 01/26/2023] Open
Abstract
AIMS Recently a limited sampling strategy (LSS) for determination of metformin' pharmacokinetics was developed. The LSS utilizes the plasma concentration of metformin 3 and 10 hours after oral intake of a single dose to estimate the area under the concentration-time curve up to 24 hours (AUC0-24h ). The main purpose of this study was to support the feasibility of this strategy in a large prospective trial. METHODS Volunteers orally ingested two 500-mg tablets of metformin hydrochloride. A blood sample was drawn three and ten hours after the ingestion. Urine was collected for 0-10 and 10-24 hours and urine volumes recorded. The AUC0-24h was calculated using the equation AUC0-24h = 4.779 * C3 + 13.174 * C10 . Additionally, all participants were genotyped for the single-nucleotide polymorphism A270S in OCT2, g.-66 T > C in MATE1, R61C, G465R, G401S and the deletion M420del in OCT1. RESULTS In total, 212 healthy volunteers participated. The median (25th - 75th interquartile range) AUC0 - 24h , CLrenal , C3 and C10 , were 10 600 (8470-12 500) ng* hr* mL-1 , 29 (24-34) L* hour-1 , 1460 (1180-1770) and 260 (200-330) ng* mL-1 , respectively, which is in agreement with our previous results. GFRi was correlated with metformin AUC and CLrenal (P < .001). As expected, we found a great pharmacokinetic interindividual variability among the volunteers and no effect of the OCT1 genotype on the AUC0 - 24h . We were unable to reproduce our previous finding of a gene-gene interaction (OCT2 and MATE1) effect on CLrenal in this cohort. CONCLUSION This study further supports the use of the 2-point LSS algorithm in large pharmacokinetic trials.
Collapse
Affiliation(s)
- Ida Kuhlmann
- Clinical Pharmacology and Pharmacy, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | | | - Peter Skov Esbech
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Tore Bjerregaard Stage
- Clinical Pharmacology and Pharmacy, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Mette Marie Hougaard Christensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kim Brøsen
- Clinical Pharmacology and Pharmacy, Department of Public Health, University of Southern Denmark, Odense, Denmark.,OPEN, Odense Patient data Explorative Network, Odense University Hospital, Odense, Denmark
| |
Collapse
|
19
|
Sundelin E, Jensen JB, Jakobsen S, Gormsen LC, Jessen N. Metformin Biodistribution: A Key to Mechanisms of Action? J Clin Endocrinol Metab 2020; 105:5850036. [PMID: 32480406 DOI: 10.1210/clinem/dgaa332] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023]
Abstract
Metformin has undisputed glucose-lowering effects in diabetes and an impressive safety record. It has also shown promising effects beyond diabetes, and several hundred clinical trials involving metformin are currently planned or active. Metformin targets intracellular effectors, but exactly which remain to be established, and in an era of precision medicine, an incomplete understanding of mechanisms of action may limit the use of metformin. Distribution of metformin depends on specific organic cation transporter proteins that are organ- and species-specific. Therefore, target tissues of metformin can be identified by cellular uptake of the drug, and exploring the biodistribution of the drug in humans becomes an attractive strategy to assist the many investigations into the mechanisms of action of metformin performed in animals. In this review, we combine the emerging evidence from the use of 11C-labeled metformin in humans to discuss metformin action in liver, intestines, and kidney, which are the organs with the most avid uptake of the drug.
Collapse
Affiliation(s)
- Elias Sundelin
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jonas Brorson Jensen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine & PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Lars C Gormsen
- Department of Nuclear Medicine & PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Jessen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
20
|
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:ijms21186627. [PMID: 32927790 PMCID: PMC7554776 DOI: 10.3390/ijms21186627] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
A considerable number of drugs and/or their metabolites are excreted by the kidneys through glomerular filtration and active renal tubule secretion via transporter proteins. Uptake transporters in the proximal tubule are part of the solute carrier (SLC) superfamily, and include the organic cation transporters (OCTs). Several studies have shown that specific genetic polymorphisms in OCTs alter drug disposition and may lead to nephrotoxicity. Multiple single nucleotide polymorphisms (SNPs) have been reported for the OCT genes (SLC22A1, SLC22A2 and SLC22A3), which can influence the proteins’ structure and expression levels and affect their transport function. A gain-in-function mutation may lead to accumulation of drugs in renal proximal tubule cells, eventually leading to nephrotoxicity. This review illustrates the impact of genetic polymorphisms in OCTs on renal drug disposition and kidney injury, the clinical significances and how to personalize therapies to minimize the risk of drug toxicity.
Collapse
Affiliation(s)
- Zulfan Zazuli
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.J.H.V.); (A.H.M.-v.d.Z.)
- Department of Pharmacology-Clinical Pharmacy, School of Pharmacy, Bandung Institute of Technology, Jawa Barat 40132, Indonesia
- Correspondence: (Z.Z.); (R.M.)
| | - Naut J. C. B. Duin
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.J.C.B.D.); (K.J.)
| | - Katja Jansen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.J.C.B.D.); (K.J.)
| | - Susanne J. H. Vijverberg
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.J.H.V.); (A.H.M.-v.d.Z.)
| | - Anke H. Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.J.H.V.); (A.H.M.-v.d.Z.)
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (N.J.C.B.D.); (K.J.)
- Correspondence: (Z.Z.); (R.M.)
| |
Collapse
|
21
|
Samuel SM, Varghese E, Koklesová L, Líšková A, Kubatka P, Büsselberg D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers (Basel) 2020; 12:E2482. [PMID: 32883003 PMCID: PMC7565921 DOI: 10.3390/cancers12092482] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the leaps and bounds in achieving success in the management and treatment of breast cancers through surgery, chemotherapy, and radiotherapy, breast cancer remains the most frequently occurring cancer in women and the most common cause of cancer-related deaths among women. Systemic therapeutic approaches, such as chemotherapy, although beneficial in treating and curing breast cancer subjects with localized breast tumors, tend to fail in metastatic cases of the disease due to (a) an acquired resistance to the chemotherapeutic drug and (b) the development of intrinsic resistance to therapy. The existence of cancer stem cells (CSCs) plays a crucial role in both acquired and intrinsic chemoresistance. CSCs are less abundant than terminally differentiated cancer cells and confer chemoresistance through a unique altered metabolism and capability to evade the immune response system. Furthermore, CSCs possess active DNA repair systems, transporters that support multidrug resistance (MDR), advanced detoxification processes, and the ability to self-renew and differentiate into tumor progenitor cells, thereby supporting cancer invasion, metastasis, and recurrence/relapse. Hence, current research is focusing on targeting CSCs to overcome resistance and improve the efficacy of the treatment and management of breast cancer. Studies revealed that metformin (1, 1-dimethylbiguanide), a widely used anti-hyperglycemic agent, sensitizes tumor response to various chemotherapeutic drugs. Metformin selectively targets CSCs and improves the hypoxic microenvironment, suppresses the tumor metastasis and inflammation, as well as regulates the metabolic programming, induces apoptosis, and reverses epithelial-mesenchymal transition and MDR. Here, we discuss cancer (breast cancer) and chemoresistance, the molecular mechanisms of chemoresistance in breast cancers, and metformin as a chemo-sensitizing/re-sensitizing agent, with a particular focus on breast CSCs as a critical contributing factor to acquired and intrinsic chemoresistance. The review outlines the prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer/chemo-sensitizing drug in the treatment of breast cancer. It intends to provide a rationale for the use of metformin as a combinatory therapy in a clinical setting.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Lenka Koklesová
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Alena Líšková
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
22
|
Teufelsbauer M, Rath B, Plangger A, Staud C, Nanobashvili J, Huk I, Neumayer C, Hamilton G, Radtke C. Effects of metformin on adipose-derived stromal cell (ADSC) - Breast cancer cell lines interaction. Life Sci 2020; 261:118371. [PMID: 32882267 DOI: 10.1016/j.lfs.2020.118371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
AIMS Metformin is a clinical drug administered to patients to treat type 2 diabetes mellitus that was found to be associated with a lower risk of occurrence of cancer and cancer-related death. The present study investigated the effects of metformin on human adipose-derived stromal cells (ADSC) - breast cancer cell line interactions. MAIN METHODS ADSCs grown from lipoaspirates were tested for growth-stimulating and migration-controlling activity on breast cancer cell lines after pretreatment with metformin. Furthermore, secreted proteins of ADSCs, phosphorylation of intracellular proteins and the effect of metformin on adipocytic differentiation of ADSCs were assayed. KEY FINDINGS Compared to breast cancer cell lines (4.0 ± 3.5% reduction of proliferation), 2 mM metformin significantly inhibited the proliferation of ADSC lines (19.2 ± 8.4% reduction of proliferation). This effect on ADSCs seems to be mediated by altered phosphorylation of GSK-3, CREB and PRAS40. Furthermore, treatment with metformin abolished the induction of differentiation of three ADSC lines to adipocytes. 1 and 2 mM metformin significantly impaired the migration of breast cancer cell lines MDA-MB-231 and MDA-MB-436 in scratch assays. SIGNIFICANCE Metformin showed low direct inhibitory effects on breast cancer cell lines at physiological concentrations but exerted a significant retardation of the growth and the adipocytic differentiation of ADSCs. Thus, the anticancer activity of metformin in breast cancer at physiological drug concentrations seems to be mediated by an indirect mechanism that lowers the supportive activity of ADSCs.
Collapse
Affiliation(s)
- Maryana Teufelsbauer
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Adelina Plangger
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Josif Nanobashvili
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Ihor Huk
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Neumayer
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria.
| | - Christine Radtke
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Naja K, El Shamieh S, Fakhoury R. rs622342A>C in SLC22A1 is associated with metformin pharmacokinetics and glycemic response. Drug Metab Pharmacokinet 2020; 35:160-164. [DOI: 10.1016/j.dmpk.2019.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/19/2019] [Accepted: 10/28/2019] [Indexed: 11/15/2022]
|
24
|
Anderson JT, Huang KM, Lustberg MB, Sparreboom A, Hu S. Solute Carrier Transportome in Chemotherapy-Induced Adverse Drug Reactions. Rev Physiol Biochem Pharmacol 2020; 183:177-215. [PMID: 32761456 DOI: 10.1007/112_2020_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Members of the solute carrier (SLC) family of transporters are responsible for the cellular influx of a broad range of endogenous compounds and xenobiotics. These proteins are highly expressed in the gastrointestinal tract and eliminating organs such as the liver and kidney, and are considered to be of particular importance in governing drug absorption and elimination. Many of the same transporters are also expressed in a wide variety of organs targeted by clinically important anticancer drugs, directly affect cellular sensitivity to these agents, and indirectly influence treatment-related side effects. Furthermore, targeted intervention strategies involving the use of transport inhibitors have been recently developed, and have provided promising lead candidates for combinatorial therapies associated with decreased toxicity. Gaining a better understanding of the complex interplay between transporter-mediated on-target and off-target drug disposition will help guide the further development of these novel treatment strategies to prevent drug accumulation in toxicity-associated organs, and improve the safety of currently available treatment modalities. In this report, we provide an update on this rapidly emerging field with particular emphasis on anticancer drugs belonging to the classes of taxanes, platinum derivatives, nucleoside analogs, and anthracyclines.
Collapse
Affiliation(s)
- Jason T Anderson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Maryam B Lustberg
- Department of Medical Oncology, The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
25
|
Abstract
The organic cation transporters (OCTs) OCT1, OCT2, OCT3, novel OCT (OCTN)1, OCTN2, multidrug and toxin exclusion (MATE)1, and MATE kidney-specific 2 are polyspecific transporters exhibiting broadly overlapping substrate selectivities. They transport organic cations, zwitterions, and some uncharged compounds and operate as facilitated diffusion systems and/or antiporters. OCTs are critically involved in intestinal absorption, hepatic uptake, and renal excretion of hydrophilic drugs. They modulate the distribution of endogenous compounds such as thiamine, L-carnitine, and neurotransmitters. Sites of expression and functions of OCTs have important impact on energy metabolism, pharmacokinetics, and toxicity of drugs, and on drug-drug interactions. In this work, an overview about the human OCTs is presented. Functional properties of human OCTs, including identified substrates and inhibitors of the individual transporters, are described. Sites of expression are compiled, and data on regulation of OCTs are presented. In addition, genetic variations of OCTs are listed, and data on their impact on transport, drug treatment, and diseases are reported. Moreover, recent data are summarized that indicate complex drug-drug interaction at OCTs, such as allosteric high-affinity inhibition of transport and substrate dependence of inhibitor efficacies. A hypothesis about the molecular mechanism of polyspecific substrate recognition by OCTs is presented that is based on functional studies and mutagenesis experiments in OCT1 and OCT2. This hypothesis provides a framework to imagine how observed complex drug-drug interactions at OCTs arise. Finally, preclinical in vitro tests that are performed by pharmaceutical companies to identify interaction of novel drugs with OCTs are discussed. Optimized experimental procedures are proposed that allow a gapless detection of inhibitory and transported drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| |
Collapse
|
26
|
Samuel SM, Varghese E, Kubatka P, Triggle CR, Büsselberg D. Metformin: The Answer to Cancer in a Flower? Current Knowledge and Future Prospects of Metformin as an Anti-Cancer Agent in Breast Cancer. Biomolecules 2019; 9:E846. [PMID: 31835318 PMCID: PMC6995629 DOI: 10.3390/biom9120846] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 12/25/2022] Open
Abstract
Interest has grown in studying the possible use of well-known anti-diabetic drugs as anti-cancer agents individually or in combination with, frequently used, chemotherapeutic agents and/or radiation, owing to the fact that diabetes heightens the risk, incidence, and rapid progression of cancers, including breast cancer, in an individual. In this regard, metformin (1, 1-dimethylbiguanide), well known as 'Glucophage' among diabetics, was reported to be cancer preventive while also being a potent anti-proliferative and anti-cancer agent. While meta-analysis studies reported a lower risk and incidence of breast cancer among diabetic individuals on a metformin treatment regimen, several in vitro, pre-clinical, and clinical studies reported the efficacy of using metformin individually as an anti-cancer/anti-tumor agent or in combination with chemotherapeutic drugs or radiation in the treatment of different forms of breast cancer. However, unanswered questions remain with regards to areas such as cancer treatment specific therapeutic dosing of metformin, specificity to cancer cells at high concentrations, resistance to metformin therapy, efficacy of combinatory therapeutic approaches, post-therapeutic relapse of the disease, and efficacy in cancer prevention in non-diabetic individuals. In the current article, we discuss the biology of metformin and its molecular mechanism of action, the existing cellular, pre-clinical, and clinical studies that have tested the anti-tumor potential of metformin as a potential anti-cancer/anti-tumor agent in breast cancer therapy, and outline the future prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer drug in the treatment of breast cancer.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Chris R. Triggle
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
27
|
Mathews Samuel S, Satheesh NJ, Ghosh S, Büsselberg D, Majeed Y, Ding H, Triggle CR. Treatment with a Combination of Metformin and 2-Deoxyglucose Upregulates Thrombospondin-1 in Microvascular Endothelial Cells: Implications in Anti-Angiogenic Cancer Therapy. Cancers (Basel) 2019; 11:E1737. [PMID: 31698699 PMCID: PMC6895998 DOI: 10.3390/cancers11111737] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Metformin, the most widely used anti-diabetic drug, also exhibits anti-cancer properties; however, the true potential of metformin as an anticancer drug remains largely unknown. In this study using mouse microvascular endothelial cells (MMECs), we investigated the effects of metformin alone or in combination with the glycolytic inhibitor, 2-deoxyglucose (2DG), on angiogenesis-a process known to be an integral part of tumor growth, cancer cell survival and metastasis. MMECs were exposed to 2DG (1-10 mM) for 48 h in the absence or presence of metformin (2 mM). The status of angiogenic and anti-angiogenic marker proteins, proteins of the mTOR pathway and cell-cycle-related proteins were quantified by Western blot analysis. Assays for cell proliferation, migration and tubulogenesis were also performed. We observed robust up-regulation of anti-angiogenic thrombospondin-1 (TSP1) and increased TSP1-CD36 co-localization with a marked decrease in the levels of phosphorylated vascular endothelial growth factor receptor-2 (pVEGFR2; Y1175) in 2DG (5 mM) exposed cells treated with metformin (2 mM). Additionally, treatment with metformin and 2DG (5 mM) inhibited the Akt/mTOR pathway and down-regulated the cell-cycle-related proteins such as p-cyclin B1 (S147) and cyclins D1 and D2 when compared to cells that were treated with either 2DG or metformin alone. Treatment with a combination of 2DG (5 mM) and metformin (2 mM) also significantly decreased cell proliferation, migration and tubulogenic capacity when compared to cells that were treated with either 2DG or metformin alone. The up-regulation of TSP1, inhibition of cell proliferation, migration and tubulogenesis provides support to the argument that the combination of metformin and 2DG may prove to be an appropriate anti-proliferative and anti-angiogenic therapeutic strategy for the treatment of some cancers.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (N.J.S.); (S.G.); (Y.M.); (H.D.)
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Noothan Jyothi Satheesh
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (N.J.S.); (S.G.); (Y.M.); (H.D.)
| | - Suparna Ghosh
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (N.J.S.); (S.G.); (Y.M.); (H.D.)
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Yasser Majeed
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (N.J.S.); (S.G.); (Y.M.); (H.D.)
| | - Hong Ding
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (N.J.S.); (S.G.); (Y.M.); (H.D.)
- Department of Medical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Chris R. Triggle
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (N.J.S.); (S.G.); (Y.M.); (H.D.)
- Department of Medical Education, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| |
Collapse
|
28
|
Sundelin EIO, Gormsen LC, Heebøll S, Vendelbo MH, Jakobsen S, Munk OL, Feddersen S, Brøsen K, Hamilton-Dutoit SJ, Pedersen SB, Grønbaek H, Jessen N. Hepatic exposure of metformin in patients with non-alcoholic fatty liver disease. Br J Clin Pharmacol 2019; 85:1761-1770. [PMID: 30973968 DOI: 10.1111/bcp.13962] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 12/14/2022] Open
Abstract
AIMS Metformin is first-line treatment of type 2 diabetes mellitus and reduces cardiovascular events in patients with insulin resistance and type 2 diabetes. Target tissue for metformin action is thought to be the liver, where metformin distribution depends on facilitated transport by polyspecific transmembrane organic cation transporters (OCTs). Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in the western world with strong associations to insulin resistance and the metabolic syndrome, but whether NAFLD affects metformin biodistribution to the liver is not known. In this study, the primary aim was to investigate in vivo hepatic uptake of metformin dynamically in humans with variable degrees of liver affection. As a secondary aim, we wished to correlate hepatic metformin distribution with OCT gene transcription determined in diagnostic liver biopsies. METHODS Eighteen patients with biopsy-proven NAFLD were investigated using 11C-metformin PET/CT technique. Gene transcripts of OCTs were determined by real-time polymerase chain reaction (PCR). RESULTS We observed similar hepatic volume of distribution of metformin between patients with simple steatosis and non-alcoholic steatohepatitis (NASH) (Vd 2.38 ± 0.56 vs. 2.10 ± 0.39, P = 0.3). There was no association between hepatic exposure to metformin and the degree of inflammation or fibrosis, and no clear correlation between metformin distribution and OCT gene transcription. CONCLUSION Metformin is distributed to the liver in patients with NAFLD and the distribution is not impaired by inflammation or fibrosis. The findings imply that metformin action in liver in patients with NAFLD may be preserved.
Collapse
Affiliation(s)
| | | | - Sara Heebøll
- Department of Gastroenterology & Hepatology, Aarhus University Hospital, Denmark
| | - Mikkel Holm Vendelbo
- Department of Nuclear Medicine & PET Center, Aarhus University Hospital, Denmark.,Department of Biomedicine, Aarhus University, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine & PET Center, Aarhus University Hospital, Denmark
| | - Ole Lajord Munk
- Department of Nuclear Medicine & PET Center, Aarhus University Hospital, Denmark
| | - Søren Feddersen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kim Brøsen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark.,Department of Public Health, Clinical Pharmacology, University of Southern Denmark, Denmark
| | | | - Steen Bønløkke Pedersen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark
| | - Henning Grønbaek
- Department of Gastroenterology & Hepatology, Aarhus University Hospital, Denmark
| | - Niels Jessen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Denmark.,Department of Biomedicine, Aarhus University, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark.,Department of Clinical Pharmacology, Aarhus University Hospital, Denmark
| |
Collapse
|
29
|
Ebid AHIM, Ehab M, Ismail A, Soror S, Mahmoud MA. The influence of SLC22A1 rs622342 and ABCC8 rs757110 genetic variants on the efficacy of metformin and glimepiride combination therapy in Egyptian patients with type 2 diabetes. J Drug Assess 2019; 8:115-121. [PMID: 31231590 PMCID: PMC6566583 DOI: 10.1080/21556660.2019.1619571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/30/2019] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background: The incidence of Type 2 Diabetes Mellitus (T2DM) in Egypt is considered one of the highest in the world. Metformin and Sulfonylureas are usually prescribed together due to their efficacy and their relatively low cost. Organic cation transport 1, encoded by SLC22A1 gene, is the main transporter of metformin into hepatocytes, which is considered metformin site of action. Sulfonylureas enhance insulin release from pancreatic B-cells through binding to sulfonylurea receptor 1, encoded by ABCC8 gene. Single nucleotide polymorphisms in the SLC22A1 and ABCC8 genes might affect the response of each drug. Aims: To investigate the influence of SLC22A1 rs622342 (A>C) and ABCC8 rs757110 (A>C) genetic variants on the efficacy of metformin and glimepiride combination therapy in Egyptian T2DM patients. Methods: Observational cross-sectional study in which patients receiving metformin and glimepiride combination therapy for at least 6 months were included for genotyping and classified into either responders or non-responders, based on their HbA1C level. Results: A total of 127 patients were included and genotyped. They were divided into 93 responders (HbA1C<7%) and 34 non-responders (HbA1C≥7%). Minor allele frequencies for rs622342 and rs757110 were 0.189 and 0.271, respectively. Only SLC22A1 rs622342 variant was found to be associated with the response of combination therapy, in which AA alleles carriers were 2.7-times more responsive to metformin than C allele carriers (Recessive model, odds ratio = 2.718, p = 0.025, 95% CI = 1.112–6.385). Conclusion: Genotyping of rs622342 can be useful in predicting the response to metformin in combination therapy in Egyptian T2DM patients.
Collapse
Affiliation(s)
- Abdel-Hameed I M Ebid
- Department of Pharmacy Practice, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Moataz Ehab
- Department of Pharmacy Practice, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Ashraf Ismail
- Clinical Pathology and Head of Research and Education Center, National Institute of Diabetes and Endocrinology, Cairo, Egypt
| | - Sameh Soror
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed Adel Mahmoud
- Department of Pharmacy Practice, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
30
|
Reséndiz-Abarca CA, Flores-Alfaro E, Suárez-Sánchez F, Cruz M, Valladares-Salgado A, Del Carmen Alarcón-Romero L, Vázquez-Moreno MA, Wacher-Rodarte NA, Gómez-Zamudio JH. Altered Glycemic Control Associated With Polymorphisms in the SLC22A1 (OCT1) Gene in a Mexican Population With Type 2 Diabetes Mellitus Treated With Metformin: A Cohort Study. J Clin Pharmacol 2019; 59:1384-1390. [PMID: 31012983 DOI: 10.1002/jcph.1425] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/28/2019] [Indexed: 01/06/2023]
Abstract
The organic cation transporters OCT1 and OCT2 and the multidrug and toxin extrusion transporter MATE1, encoded by the SLC22A1, SLC22A2, and SLC47A1 genes, respectively, are responsible for the absorption of metformin in enterocytes, hepatocytes, and kidney cells. The aim of this study was to evaluate whether genetic variations in the SLC22A1, SLC22A2, and SLC47A1 genes could be associated with an altered response to metformin in patients with type 2 diabetes mellitus. A cohort study was conducted in 308 individuals with a diagnosis of type 2 diabetes mellitus of less than 3 years and who had metformin monotherapy. Three measurements of blood glycated hemoglobin (HbA1c ) were obtained at the beginning of the study and after 6 and 12 months. Five polymorphisms were analyzed in the SLC22A1 (rs622342, rs628031, rs594709), SLC22A2 (rs316019), and SLC47A1 (rs2289669) genes by real-time polymerase chain reaction. The results showed a significant association among genotypes CC-rs622342 (β = 1.36; P < .001), AA-rs628031 (β = 0.98; P = .032), and GG-rs594709 (β = 1.21; P = .016) in the SLC22A1 gene with an increase in HbA1c levels during the follow-up period. Additionally, a significant association was found in the CGA and CAG haplotypes with an increase in HbA1c levels compared to the highest-frequency haplotype (AGA). In conclusion, the genetic variation in the SLC22A1 gene was significantly related to the variation of the HbA1c levels, an important indicator of glycemic control in diabetic patients. This information may contribute to identifying patients with an altered response to metformin before starting their therapy.
Collapse
Affiliation(s)
- Carlos Alberto Reséndiz-Abarca
- Laboratorio de Investigación en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México.,Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda," Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Eugenia Flores-Alfaro
- Laboratorio de Investigación en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Fernando Suárez-Sánchez
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda," Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda," Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Adán Valladares-Salgado
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda," Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Luz Del Carmen Alarcón-Romero
- Laboratorio de Investigación en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Miguel Alexander Vázquez-Moreno
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda," Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Niels Agustín Wacher-Rodarte
- Unidad de Investigación en Epidemiología Clínica, Hospital de Especialidades "Bernardo Sepúlveda," Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Jaime Héctor Gómez-Zamudio
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades "Bernardo Sepúlveda," Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| |
Collapse
|
31
|
Chan P, Shao L, Tomlinson B, Zhang Y, Liu ZM. Metformin transporter pharmacogenomics: insights into drug disposition-where are we now? Expert Opin Drug Metab Toxicol 2018; 14:1149-1159. [PMID: 30375241 DOI: 10.1080/17425255.2018.1541981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Metformin is recommended as first-line treatment for type 2 diabetes (T2D) by all major diabetes guidelines. With appropriate usage it is safe and effective overall, but its efficacy and tolerability show considerable variation between individuals. It is a substrate for several drug transporters and polymorphisms in these transporter genes have shown effects on metformin pharmacokinetics and pharmacodynamics. Areas covered: This article provides a review of the current status of the influence of transporter pharmacogenomics on metformin efficacy and tolerability. The transporter variants identified to have an important influence on the absorption, distribution, and elimination of metformin, particularly those in organic cation transporter 1 (OCT1, gene SLC22A1), are reviewed. Expert opinion: Candidate gene studies have shown that genetic variations in SLC22A1 and other drug transporters influence the pharmacokinetics, glycemic responses, and gastrointestinal intolerance to metformin, although results are somewhat discordant. Conversely, genome-wide association studies of metformin response have identified signals in the pharmacodynamic pathways rather than the transporters involved in metformin disposition. Currently, pharmacogenomic testing to predict metformin response and tolerability may not have a clinical role, but with additional data from larger studies and availability of safe and effective alternative antidiabetic agents, this is likely to change.
Collapse
Affiliation(s)
- Paul Chan
- a Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital , Taipei Medical University , Taipei City , Taiwan
| | - Li Shao
- b The VIP Department, Shanghai East Hospital , Tongji University School of Medicine , Shanghai , China
| | - Brian Tomlinson
- c Research Center for Translational Medicine , Shanghai East Hospital Affiliated to Tongji University School of Medicine , Shanghai , China.,d Department of Medicine & Therapeutics , The Chinese University of Hong Kong , Shatin , Hong Kong
| | - Yuzhen Zhang
- c Research Center for Translational Medicine , Shanghai East Hospital Affiliated to Tongji University School of Medicine , Shanghai , China
| | - Zhong-Min Liu
- e Department of Cardiac Surgery, Shanghai East Hospital , Tongji University , Shanghai , China
| |
Collapse
|
32
|
Stage TB, Wellhagen G, Christensen MMH, Guiastrennec B, Brøsen K, Kjellsson MC. Using a semi-mechanistic model to identify the main sources of variability of metformin pharmacokinetics. Basic Clin Pharmacol Toxicol 2018; 124:105-114. [DOI: 10.1111/bcpt.13139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Tore Bjerregaard Stage
- Clinical Pharmacology and Pharmacy; Department of Public Health; University of Southern Denmark; Odense Denmark
- Pharmacometrics Group; Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
| | - Gustaf Wellhagen
- Pharmacometrics Group; Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
| | | | - Benjamin Guiastrennec
- Pharmacometrics Group; Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
| | - Kim Brøsen
- Clinical Pharmacology and Pharmacy; Department of Public Health; University of Southern Denmark; Odense Denmark
| | - Maria C. Kjellsson
- Pharmacometrics Group; Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
| |
Collapse
|
33
|
Challenges and perspectives in the treatment of diabetes associated breast cancer. Cancer Treat Rev 2018; 70:98-111. [PMID: 30130687 DOI: 10.1016/j.ctrv.2018.08.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is one of the most common chronic disease worldwide and affects all cross-sections of the society including children, women, youth and adults. Scientific evidence has linked diabetes to higher incidence, accelerated progression and increased aggressiveness of different cancers. Among the different forms of cancer, research has reinforced a link between diabetes and the risk of breast cancer. Some studies have specifically linked diabetes to the highly aggressive, triple negative breast cancers (TNBCs) which do not respond to conventional hormonal/HER2 targeted interventions, have chances of early recurrence, metastasize, tend to be more invasive in nature and develop drug resistance. Commonly used anti-diabetic drugs, such as metformin, have recently gained importance in the treatment of breast cancer due to their proposed anti-cancer properties. Here we discuss the link between diabetes and breast cancer, the metabolic disturbances in diabetes that support the development of breast cancer, the challenges involved and future perspective and directions. We link the three main metabolic disturbances (dyslipidemia, hyperinsulinemia and hyperglycemia) that occur in diabetes to potential aberrant molecular pathways that may lead to the development of an oncogenic phenotype of the breast tissue, thereby leading to acceleration of cell growth, proliferation, migration, inflammation, angiogenesis, EMT and metastasis and inhibition of apoptosis in breast cancer cells. Furthermore, managing diabetes and treating cancer using a combination of anti-diabetic and classical anti-cancer drugs should prove to be more efficient in the treatment diabetes associated cancers.
Collapse
|
34
|
Mato EPM, Guewo-Fokeng M, Faadiel Essop M, Owira PMO. Genetic polymorphisms of organic cation transporters 1 (OCT1) and responses to metformin therapy in individuals with type 2 diabetes mellitus: a systematic review protocol. Syst Rev 2018; 7:105. [PMID: 30041690 PMCID: PMC6058382 DOI: 10.1186/s13643-018-0773-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/10/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metformin is one of the most commonly used drugs for type 2 diabetes mellitus (T2DM). Despite its efficacy and safety, metformin is frequently associated with highly variable glycemic responses, which is hypothesized to be the result of genetic variations in its transport by organic cation transporters (OCTs). This systematic review aims to highlight and summarize the overall effects of OCT1 polymorphisms on therapeutic responses to metformin and to evaluate their potential role in terms of interethnic differences with metformin responses. METHODS/DESIGN We will systematically review observational studies reporting on the genetic association between OCT1 polymorphisms and metformin responses in T2DM patients. A comprehensive search strategy formulated with the help of a librarian will be used to search MEDLINE via PubMed, Embase, and CINAHL for relevant studies published between January 1990 and July 2017. Two review authors will independently screen titles and abstracts in duplicate, extract data, and assess the risk of bias with discrepancies resolved by discussion or arbitration of a third review author. Mined data will be grouped according to OCT1 polymorphisms, and their effects on therapeutic responses to metformin will be narratively synthesized. If sufficient numbers of homogeneous studies are scored, meta-analyses will be performed to obtain pooled effect estimates. Funnel plots analysis and Egger's test will be used to assess publication bias. This study will be reported according to the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. DISCUSSION This review will summarize the genetic effects of OCT1 polymorphisms associated with variabilities in glycemic responses to metformin. The findings of this study could help to develop genetic tests that could predict a person's response to metformin treatment and create personalized drugs with greater efficacy and safety. SYSTEMATIC REVIEW REGISTRATION Registration number: PROSPERO, CRD42017079978.
Collapse
Affiliation(s)
- Edith Pascale Mofo Mato
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, PO Box X5401, Durban, South Africa
| | - Magellan Guewo-Fokeng
- Laboratory of Public Health Research Biotechnology (LAPHER-Biotech), Biotechnology Centre, University of Yaounde I, PO Box 3851, Yaounde, Cameroon
- Laboratory of Molecular Medicine and Metabolism (LMMM), Biotechnology Centre, University of Yaounde I, PO Box 3851, Yaounde, Cameroon
| | - M. Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Peter Mark Oroma Owira
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, PO Box X5401, Durban, South Africa
| |
Collapse
|
35
|
McCreight LJ, Stage TB, Connelly P, Lonergan M, Nielsen F, Prehn C, Adamski J, Brøsen K, Pearson ER. Pharmacokinetics of metformin in patients with gastrointestinal intolerance. Diabetes Obes Metab 2018; 20:1593-1601. [PMID: 29457876 PMCID: PMC6033038 DOI: 10.1111/dom.13264] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/27/2022]
Abstract
AIMS To assess potential causes of metformin intolerance, including altered metformin uptake from the intestine, increased anaerobic glucose utilization and subsequent lactate production, altered serotonin uptake, and altered bile acid pool. METHODS For this pharmacokinetic study, we recruited 10 severely intolerant and 10 tolerant individuals, matched for age, sex and body mass index. A single 500-mg dose of metformin was administered, with blood sampling at 12 time points over 24 hours. Blood samples were analysed for metformin, lactate, serotonin and bile acid concentrations, and compared across the phenotypes. RESULTS The intolerant individuals were severely intolerant to 500 mg metformin. No significant difference was identified between tolerant and intolerant cohorts in metformin pharmacokinetics: median (interquartile range [IQR]) peak concentration 2.1 (1.7-2.3) mg/L and 2.0 (1.8-2.2) mg/L, respectively (P = .76); time to peak concentration 2.5 hours; median (IQR) area under the curve (AUC)0-24 16.9 (13.9-18.6) and 13.9 (12.9-16.8) mg/L*h, respectively (P = .72). Lactate concentration peaked at 3.5 hours, with mean peak concentration of 2.4 mmol/L in both cohorts (95% CI 2.0-2.8 and 1.8-3.0 mmol/L, respectively), and similar incremental AUC0-24 in each cohort: tolerant cohort 6.98 (95% CI 3.03-10.93) and intolerant cohort 4.47 (95% CI -3.12-12.06) mmol/L*h (P = .55). Neither serotonin nor bile acid concentrations were significantly different. CONCLUSIONS Despite evidence of severe intolerance in our cohort, there was no significant difference in metformin pharmacokinetics or systemic measures of lactate, serotonin or bile acids. This suggests that metformin intolerance may be attributable to local factors within the lumen or enterocyte.
Collapse
Affiliation(s)
- Laura J. McCreight
- Division of Molecular and Clinical MedicineNinewells Hospital and Medical SchoolDundeeUK
| | - Tore B. Stage
- Department of Public Health, Clinical Pharmacology and PharmacyUniversity of Southern Denmark, OdenseDenmark
| | - Paul Connelly
- Division of Molecular and Clinical MedicineNinewells Hospital and Medical SchoolDundeeUK
| | - Mike Lonergan
- Division of Molecular and Clinical MedicineNinewells Hospital and Medical SchoolDundeeUK
| | - Flemming Nielsen
- Department of Public Health, Clinical Pharmacology and PharmacyUniversity of Southern Denmark, OdenseDenmark
| | - Cornelia Prehn
- Institute of Experimental GeneticsGenome Analysis CenterMunichGermany
- German Research Centre for Environmental HealthNeuherbergGermany
| | - Jerzy Adamski
- Institute of Experimental GeneticsGenome Analysis CenterMunichGermany
- Lehrstuhl für Experimentelle GenetikTechnische Universität MünchenFreising‐Weihenstephan, MunichGermany
- German Centre for Diabetes Research (DZD)Munich‐Neuherberg, MunichGermany
| | - Kim Brøsen
- Department of Public Health, Clinical Pharmacology and PharmacyUniversity of Southern Denmark, OdenseDenmark
| | - Ewan R. Pearson
- Division of Molecular and Clinical MedicineNinewells Hospital and Medical SchoolDundeeUK
| |
Collapse
|
36
|
Mofo Mato EP, Guewo-Fokeng M, Essop MF, Owira PMO. Genetic polymorphisms of organic cation transporter 1 (OCT1) and responses to metformin therapy in individuals with type 2 diabetes: A systematic review. Medicine (Baltimore) 2018; 97:e11349. [PMID: 29979413 PMCID: PMC6076123 DOI: 10.1097/md.0000000000011349] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Metformin is one of the most commonly used drugs for the treatment of type 2 diabetes mellitus (T2DM). Despite its widespread use, there are considerable interindividual variations in metformin response, with about 35% of patients failing to achieve initial glycemic control. These variabilities that reflect phenotypic differences in drug disposition and action may indeed be due to polymorphisms in genes that regulate pharmacokinetics and pharmacodynamics of metformin. Moreover, interethnic differences in drug responses in some cases correspond to substantial differences in the frequencies of the associated pharmacogenomics risk allele. AIM This study aims to highlight and summarize the overall effects of organic cation transporter 1(OCT1) polymorphisms on therapeutic responses to metformin and to evaluate the potential role of such polymorphisms in interethnic differences in metformin therapy. METHODS We conducted a systematic review according to the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. We searched for PubMed/MEDLINE, Embase, and CINAHL, relevant studies reporting the effects of OCT1 polymorphisms on metformin therapy in T2DM individuals. Data were extracted on study design, population characteristics, relevant polymorphisms, measure of genetic association, and outcomes. The presence of gastrointestinal side effects, glycated hemoglobin A1 (HbA1c) levels, fasting plasma glucose (FPG), and postprandial plasma glucose (PPG) concentrations after treatment with metformin were chosen as measures of the metformin responses. This systematic review protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO). RESULTS According to the data extracted, a total of 34 OCT1 polymorphisms were identified in 10 ethnic groups. Significant differences in the frequencies of common alleles were observed among these groups. Met408Val (rs628031) variant was the most extensively explored with metformin responses. Although some genotypes and alleles have been associated with deleterious effects on metformin response, others indeed, exhibited positive effects. CONCLUSION Genetic effects of OCT1 polymorphisms on metformin responses were population specific. Further investigations in other populations are required to set ethnicity-specific reference for metformin responses and to obtain a solid basis to design personalized therapeutic approaches for T2DM treatment.
Collapse
Affiliation(s)
- Edith Pascale Mofo Mato
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Magellan Guewo-Fokeng
- Laboratory of Public Health Research Biotechnology (LAPHER-Biotech)
- Laboratory of Molecular Medicine and Metabolism (LMMM), Biotechnology Centre, University of Yaounde I, Yaounde, Cameroon
| | - M. Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Peter Mark Oroma Owira
- Molecular and Clinical Pharmacology Research Laboratory, Department of Pharmacology, Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
37
|
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] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 04/06/2018] [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
|
38
|
Ningrum VD, Ikawati Z, Sadewa AH, Ikhsan MR. Patient-factors associated with metformin steady-state levels in type 2 diabetes mellitus with therapeutic dosage. J Clin Transl Endocrinol 2018; 12:42-47. [PMID: 29892566 PMCID: PMC5992324 DOI: 10.1016/j.jcte.2018.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/07/2018] [Indexed: 01/08/2023] Open
Abstract
AIMS This prospective study aimed to analyze metformin steady-state concentration in repeated constant dosage and the influencing patient-factors as well as to correlate them with glycemic control. METHODS The validated HPLC-UV method was used to examine metformin steady-state concentration, while FBG and glycated albumin were used as the parameters of glycemic control during metformin administration. RESULTS A total of 82 type-2 diabetes patients were involved with 32.1% of them having metformin Cssmin and 84.1% having Cssmax of metformin within the recommended therapeutic range. One patient had metformin Css that exceeded minimum toxic concentration despite his normal renal function and administered therapeutic dosage of metformin. Higher Cssmax was found in patients with metformin monotherapy, while patients with longer duration of metformin use had significantly higher Cssmin. CONCLUSIONS Along with initial hyperglycemia and eGFR, metformin Cssmin became the only parameter that influenced FBG level (P < 0.05). Duration of previous metformin use should be considered in the strategy of optimizing metformin dosage. The type-2 diabetes patients with obesity are more suggested to take shorter interval of metformin administration (or possibly with sustained-release formulation) to keep Cssmin within the therapeutic range.
Collapse
Affiliation(s)
| | - Zullies Ikawati
- Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Ahmad H. Sadewa
- Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Mohammad R. Ikhsan
- Department of Internal Medicine, Dr. Sardjito General Hospital, Yogyakarta 55281, Indonesia
| |
Collapse
|
39
|
Santoro AB, Botton MR, Struchiner CJ, Suarez-Kurtz G. Influence of pharmacogenetic polymorphisms and demographic variables on metformin pharmacokinetics in an admixed Brazilian cohort. Br J Clin Pharmacol 2018; 84:987-996. [PMID: 29352482 DOI: 10.1111/bcp.13522] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/07/2017] [Accepted: 01/12/2018] [Indexed: 12/15/2022] Open
Abstract
AIMS To identify pharmacogenetic and demographic variables that influence the systemic exposure to metformin in an admixed Brazilian cohort. METHODS The extreme discordant phenotype was used to select 106 data sets from nine metformin bioequivalence trials, comprising 256 healthy adults. Eleven single-nucleotide polymorphisms in SLC22A1, SLC22A2, SLC47A1 SLC47A2 and in transcription factor SP1 were genotyped and a validated panel of ancestry informative markers was used to estimate the individual proportions of biogeographical ancestry. Two-step (univariate followed by multivariate) regression modelling was developed to identify covariates associated with systemic exposure to metformin, accessed by the area under the plasma concentration-time curve, between 0 and 48 h (AUC0-48h ), after single oral doses of metformin (500 or 1000 mg). RESULTS The individual proportions of African, Amerindian and European ancestry varied widely, as anticipated from the structure of the Brazilian population The dose-adjusted, log-transformed AUC0-48h 's (ng h ml-1 mg-1 ) differed largely in the two groups at the opposite ends of the distribution histogram, namely 0.82, 0.79-0.85 and 1.08, 1.06-1.11 (mean, 95% confidence interval; P = 6.10-26 , t test). Multivariate modelling revealed that metformin AUC0-48h increased with age, food and carriage of rs12208357 in SLC22A1 but was inversely associated with body surface area and individual proportions of African ancestry. CONCLUSIONS A pharmacogenetic marker in OCT1 (SLC22A1 rs12208357), combined with demographic covariates (age, body surface area and individual proportion of African ancestry) and a food effect explained 29.7% of the variability in metformin AUC0-48h .
Collapse
|
40
|
Chung H, Oh J, Yoon SH, Yu KS, Cho JY, Chung JY. A non-linear pharmacokinetic-pharmacodynamic relationship of metformin in healthy volunteers: An open-label, parallel group, randomized clinical study. PLoS One 2018; 13:e0191258. [PMID: 29342199 PMCID: PMC5771593 DOI: 10.1371/journal.pone.0191258] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/26/2017] [Indexed: 11/19/2022] Open
Abstract
Background The aim of this study was to explore the pharmacokinetic-pharmacodynamic (PK-PD) relationship of metformin on glucose levels after the administration of 250 mg and 1000 mg of metformin in healthy volunteers. Methods A total of 20 healthy male volunteers were randomized to receive two doses of either a low dose (375 mg followed by 250 mg) or a high dose (1000 mg followed by 1000 mg) of metformin at 12-h intervals. The pharmacodynamics of metformin was assessed using oral glucose tolerance tests before and after metformin administration. The PK parameters after the second dose were evaluated through noncompartmental analyses. Four single nucleotide polymorphisms in MATE1, MATE2-K, and OCT2 were genotyped, and their effects on PK characteristics were additionally evaluated. Results The plasma exposure of metformin increased as the metformin dose increased. The mean values for the area under the concentration-time curve from dosing to 12 hours post-dose (AUC0-12h) were 3160.4 and 8808.2 h·μg/L for the low- and high-dose groups, respectively. Non-linear relationships were found between the glucose-lowering effect and PK parameters with a significant inverse trend at high metformin exposure. The PK parameters were comparable among subjects with the genetic polymorphisms. Conclusions This study showed a non-linear PK-PD relationship on plasma glucose levels after the administration of metformin. The inverse relationship between systemic exposure and the glucose-lowering effect at a high exposure indicates a possible role for the intestines as an action site for metformin. Trial registration ClinicalTrials.gov NCT02712619
Collapse
Affiliation(s)
- Hyewon Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Toxicology, Korea University Guro Hospital, Seoul, Korea
| | - Jaeseong Oh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul, Korea
| | - Seo Hyun Yoon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul, Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul, Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul, Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Clinical Trials Center, Seoul National University Bundang Hospital, Seongnam, Korea
- * E-mail:
| |
Collapse
|
41
|
Al-Oanzi ZH, Fountana S, Moonira T, Tudhope SJ, Petrie JL, Alshawi A, Patman G, Arden C, Reeves HL, Agius L. Opposite effects of a glucokinase activator and metformin on glucose-regulated gene expression in hepatocytes. Diabetes Obes Metab 2017; 19:1078-1087. [PMID: 28206714 DOI: 10.1111/dom.12910] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 02/10/2017] [Accepted: 02/11/2017] [Indexed: 01/19/2023]
Abstract
AIM Small molecule activators of glucokinase (GKAs) have been explored extensively as potential anti-hyperglycaemic drugs for type 2 diabetes (T2D). Several GKAs were remarkably effective in lowering blood glucose during early therapy but then lost their glycaemic efficacy chronically during clinical trials. MATERIALS AND METHODS We used rat hepatocytes to test the hypothesis that GKAs raise hepatocyte glucose 6-phosphate (G6P, the glucokinase product) and down-stream metabolites with consequent repression of the liver glucokinase gene ( Gck). We compared a GKA with metformin, the most widely prescribed drug for T2D. RESULTS Treatment of hepatocytes with 25 mM glucose raised cell G6P, concomitantly with Gck repression and induction of G6pc (glucose 6-phosphatase) and Pklr (pyruvate kinase). A GKA mimicked high glucose by raising G6P and fructose-2,6-bisphosphate, a regulatory metabolite, causing a left-shift in glucose responsiveness on gene regulation. Fructose, like the GKA, repressed Gck but modestly induced G6pc. 2-Deoxyglucose, which is phosphorylated by glucokinase but not further metabolized caused Gck repression but not G6pc induction, implicating the glucokinase product in Gck repression. Metformin counteracted the effect of high glucose on the elevated G6P and fructose 2,6-bisphosphate and on Gck repression, recruitment of Mlx-ChREBP to the G6pc and Pklr promoters and induction of these genes. CONCLUSIONS Elevation in hepatocyte G6P and downstream metabolites, with consequent liver Gck repression, is a potential contributing mechanism to the loss of GKA efficacy during chronic therapy. Cell metformin loads within the therapeutic range attenuate the effect of high glucose on G6P and on glucose-regulated gene expression.
Collapse
Affiliation(s)
- Ziad H Al-Oanzi
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
- Medical Laboratory Science, Aljouf University, Sakaka, Saudi Arabia
| | - Sophia Fountana
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - Tabassum Moonira
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - Susan J Tudhope
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - John L Petrie
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - Ahmed Alshawi
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - Gillian Patman
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Catherine Arden
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - Helen L Reeves
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Loranne Agius
- Institute of Cellular Medicine and Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
42
|
Sundelin E, Gormsen LC, Jensen JB, Vendelbo MH, Jakobsen S, Munk OL, Christensen M, Brøsen K, Frøkiaer J, Jessen N. Genetic Polymorphisms in Organic Cation Transporter 1 Attenuates Hepatic Metformin Exposure in Humans. Clin Pharmacol Ther 2017; 102:841-848. [PMID: 28380657 DOI: 10.1002/cpt.701] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/28/2017] [Indexed: 12/17/2022]
Abstract
Metformin has been used successfully to treat type 2 diabetes for decades. However, the efficacy of the drug varies considerably from patient to patient and this may in part be due to its pharmacokinetic properties. The aim of this study was to examine if common polymorphisms in SLC22A1, encoding the transporter protein OCT1, affect the hepatic distribution of metformin in humans. We performed noninvasive 11 C-metformin positron emission tomography (PET)/computed tomography (CT) to determine hepatic exposure in 12 subjects genotyped for variants in SLC22A1. Hepatic distribution of metformin was significantly reduced after oral intake in carriers of M420del and R61C variants in SLC22A1 without being associated with changes in circulating levels of metformin. Our data show that genetic polymorphisms in transporter proteins cause variation in hepatic exposure to metformin, and it demonstrates the application of novel imaging techniques to investigate pharmacogenetic properties in humans.
Collapse
Affiliation(s)
- Eio Sundelin
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Denmark
| | - L C Gormsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Denmark
| | - J B Jensen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Denmark
| | - M H Vendelbo
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Denmark
| | - S Jakobsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Denmark
| | - O L Munk
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Denmark
| | - Mmh Christensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark
| | - K Brøsen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Denmark.,Department of Public Health, Clinical Pharmacology, University of Southern Denmark, Denmark
| | - J Frøkiaer
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Denmark
| | - N Jessen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Denmark.,Department of Clinical Pharmacology, Aarhus University Hospital, Denmark
| |
Collapse
|
43
|
Nielsen LM, Sverrisdóttir E, Stage TB, Feddersen S, Brøsen K, Christrup LL, Drewes AM, Olesen AE. Lack of genetic association between OCT1, ABCB1, and UGT2B7 variants and morphine pharmacokinetics. Eur J Pharm Sci 2017; 99:337-342. [PMID: 28063968 DOI: 10.1016/j.ejps.2016.12.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/20/2016] [Accepted: 12/31/2016] [Indexed: 12/14/2022]
Abstract
AIM A high inter-individual variation in the pharmacokinetics and pharmacodynamics of morphine has been observed. Genetic polymorphisms in genes encoding the organic cation transporter isoform 1 (OCT1), the efflux transporter p-glycoprotein (ABCB1), and the UDP-glucuronosyltransferase-2B7 (UGT2B7) may influence morphine pharmacokinetics and thus, also pharmacodynamics. The aim of this study was to evaluate the association between OCT1, ABCB1, and UGT2B7 variants, and morphine pharmacokinetics and -dynamics in healthy volunteers. METHODS Pharmacokinetic and pharmacodynamic data were collected from a double-blinded, randomized, crossover trial in 37 healthy subjects. Pharmacokinetic data were analyzed in NONMEM®, and the time-concentration relationship of morphine, morphine-3-glucuronide, and morphine-6-glucuronide was parameterized as the transit compartment rate constant (ktr), clearance (CL), and volume of distribution (VD). The area under the plasma concentration-time curve (AUC0-150min) and the maximum plasma concentration (Cmax) were also calculated. Pharmacodynamic data were measured as pain tolerance thresholds to mechanical stimulation of the rectum and muscle, as well as tonic cold pain stimulation ("the cold pressor test" where hand was immersed in cold water). Six different single nucleotide polymorphisms in three different genes (OCT1 (n=22), ABCB1 (n=37), and UGT2B (n=22)) were examined. RESULTS Neither AUC0-150min, ktr, CL, nor VD were associated with genetic variants in OCT1, ABCB1, and UGT2B7 (all P>0.05). Similarly, the antinociceptive effects of morphine on rectal, muscle, and cold pressor tests were not associated with these genetic variants (all P>0.05). CONCLUSIONS In this experimental study in healthy volunteers, we found no association between different genotypes of OCT1, ABCB1, and UGT2B7, and morphine pharmacokinetics and pharmacodynamics. Nonetheless, due to methodological limitations we cannot exclude that associations exist.
Collapse
Affiliation(s)
- L M Nielsen
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - E Sverrisdóttir
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - T B Stage
- Clinical Pharmacology, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - S Feddersen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - K Brøsen
- Clinical Pharmacology, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - L L Christrup
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - A M Drewes
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - A E Olesen
- Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
44
|
Brøsen K, Andersen SE, Borregaard J, Christensen HR, Christensen PM, Dalhoff KP, Damkier P, Hallas J, Heisterberg J, Jessen N, Jürgens G, Kampmann JPK, Laursen BE, Laursen T, Nielsen LP, Poulsen BK, Poulsen HE, Andersen LV, Senderovitz T, Sonne J. Clinical Pharmacology in Denmark in 2016 - 40 Years with the Danish Society of Clinical Pharmacology and 20 Years as a Medical Speciality. Basic Clin Pharmacol Toxicol 2016; 119:523-532. [PMID: 27685872 DOI: 10.1111/bcpt.12681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/22/2016] [Indexed: 12/01/2022]
Abstract
The Danish Society of Clinical Pharmacology was founded in 1976, and mainly thanks to the persistent efforts of the society, clinical pharmacology became an independent medical speciality in Denmark in 1996. Since then, clinical pharmacology has gone from strength to strength. In the Danish healthcare system, clinical pharmacology has established itself as an indispensible part of the efforts to promote the rational, safe and economic use of drugs. Clinical pharmacologists are active in drug committees both in hospitals and in the primary sector. All clinical pharmacology centres offer a local medicines information service. Some centres have established an adverse drug effect manager function. Only one centre offers a therapeutic drug monitoring service. Clinical pharmacologists are responsible for the toxicological advice at the Danish Poison Information Centre at Bispebjerg University Hospital in the Capital Region. The Department of Clinical Pharmacology at Aarhus University Hospital works closely together with forensic toxicologists and pathologists, covering issues regarding illicit substances, forensic pharmacology, post-mortem toxicology, expert testimony and research. Therapeutic geriatric and psychiatric teach-inns for specialist and junior doctors are among the newest initiatives organized by clinical pharmacologists. Clinical pharmacologists work also in the Danish Medicines Agency and in the Danish pharmaceutical industry, and the latter has in particular a great growth potential for creating new jobs and career opportunities for clinical pharmacologists. As of July 2016, the Danish Society of Clinical Pharmacology has 175 members, and 70 of these are specialists in clinical pharmacology corresponding to approximately 2.5 specialists per 1000 doctors (Denmark has in total 28,000 doctors) or approximately 12 specialists per one million inhabitants.
Collapse
Affiliation(s)
- Kim Brøsen
- Department of Public Health, Clinical Pharmacology and Pharmacy, University of Southern Denmark, Odense C, Denmark
| | - Stig Ejdrup Andersen
- Unit of Clinical Pharmacology, University Hospital of Sealand, Roskilde, Denmark
| | | | - Hanne Rolighed Christensen
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg University Hospital, Copenhagen, Denmark
| | | | - Kim Peder Dalhoff
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg University Hospital, Copenhagen, Denmark
| | - Per Damkier
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Jesper Hallas
- Department of Public Health, Clinical Pharmacology and Pharmacy, University of Southern Denmark, Odense C, Denmark
| | | | - Niels Jessen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Institute of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| | - Gesche Jürgens
- Unit of Clinical Pharmacology, University Hospital of Sealand, Roskilde, Denmark
| | | | - Britt Elmedal Laursen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Institute of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| | - Torben Laursen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Institute of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| | - Lars Peter Nielsen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Institute of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| | - Birgitte Klindt Poulsen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Institute of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| | | | - Ljubica Vukelic Andersen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Institute of Biomedicine, Pharmacology, Aarhus University, Aarhus, Denmark
| | | | - Jesper Sonne
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg University Hospital, Copenhagen, Denmark
| |
Collapse
|
45
|
Varma MV, El-Kattan AF. Transporter-Enzyme Interplay: Deconvoluting Effects of Hepatic Transporters and Enzymes on Drug Disposition Using Static and Dynamic Mechanistic Models. J Clin Pharmacol 2016; 56 Suppl 7:S99-S109. [DOI: 10.1002/jcph.695] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/14/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Manthena V. Varma
- Pharmacokinetics; Dynamics and Metabolism; Worldwide Research and Development; Pfizer Inc; Groton CT USA
| | - Ayman F. El-Kattan
- Pharmacokinetics; Dynamics and Metabolism; Worldwide Research and Development; Pfizer Inc; Cambridge MA USA
| |
Collapse
|
46
|
Ismaiel AAK, Espinosa-Oliva AM, Santiago M, García-Quintanilla A, Oliva-Martín MJ, Herrera AJ, Venero JL, de Pablos RM. Metformin, besides exhibiting strong in vivo anti-inflammatory properties, increases mptp-induced damage to the nigrostriatal dopaminergic system. Toxicol Appl Pharmacol 2016; 298:19-30. [PMID: 26971375 DOI: 10.1016/j.taap.2016.03.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/16/2016] [Accepted: 03/08/2016] [Indexed: 01/11/2023]
Abstract
Metformin is a widely used oral antidiabetic drug with known anti-inflammatory properties due to its action on AMPK protein. This drug has shown a protective effect on various tissues, including cortical neurons. The aim of this study was to determine the effect of metformin on the dopaminergic neurons of the substantia nigra of mice using the animal model of Parkinson's disease based on the injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, an inhibitor of the mitochondrial complex I. In vivo and in vitro experiments were used to study the activation of microglia and the damage of the dopaminergic neurons. Our results show that metformin reduced microglial activation measured both at cellular and molecular levels. Rather than protecting, metformin exacerbated dopaminergic damage in response to MPTP. Our data suggest that, contrary to other brain structures, metformin treatment could be deleterious for the dopaminergic system. Hence, metformin treatment may be considered as a risk factor for the development of Parkinson's disease.
Collapse
Affiliation(s)
- Afrah A K Ismaiel
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Ana M Espinosa-Oliva
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Martiniano Santiago
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Albert García-Quintanilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - María J Oliva-Martín
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Antonio J Herrera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - José L Venero
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Rocío M de Pablos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
| |
Collapse
|
47
|
Abstract
Metformin is an effective agent with a good safety profile that is widely used as a first-line treatment for type 2 diabetes, yet its mechanisms of action and variability in terms of efficacy and side effects remain poorly understood. Although the liver is recognised as a major site of metformin pharmacodynamics, recent evidence also implicates the gut as an important site of action. Metformin has a number of actions within the gut. It increases intestinal glucose uptake and lactate production, increases GLP-1 concentrations and the bile acid pool within the intestine, and alters the microbiome. A novel delayed-release preparation of metformin has recently been shown to improve glycaemic control to a similar extent to immediate-release metformin, but with less systemic exposure. We believe that metformin response and tolerance is intrinsically linked with the gut. This review examines the passage of metformin through the gut, and how this can affect the efficacy of metformin treatment in the individual, and contribute to the side effects associated with metformin intolerance.
Collapse
Affiliation(s)
- Laura J McCreight
- Pearson Group, Division of Cardiovascular and Diabetes Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Mailbox 12, Level 5, Dundee, DD1 9SY, UK
| | - Clifford J Bailey
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Ewan R Pearson
- Pearson Group, Division of Cardiovascular and Diabetes Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Mailbox 12, Level 5, Dundee, DD1 9SY, UK.
| |
Collapse
|
48
|
Christensen MMH, Højlund K, Hother-Nielsen O, Stage TB, Damkier P, Beck-Nielsen H, Brøsen K. Endogenous glucose production increases in response to metformin treatment in the glycogen-depleted state in humans: a randomised trial. Diabetologia 2015; 58:2494-502. [PMID: 26271344 DOI: 10.1007/s00125-015-3733-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/28/2015] [Indexed: 01/20/2023]
Abstract
AIMS/HYPOTHESIS Metformin is believed to reduce glucose levels primarily by inhibiting hepatic glucose production. Recent data indicate that metformin antagonises glucagon-dependent glucose output, suggesting that compensatory mechanisms protect against hypoglycaemia. Here, we examined the effect of metformin on glucose metabolism in humans after a glycogen-depleting fast and the role of reduced-function alleles in OCT1 (also known as SLC22A1). METHODS In a randomised, crossover trial, healthy individuals with or without reduced-function alleles in OCT1 were fasted for 42 h twice, either with or without prior treatment with 1 g metformin twice daily. Participants were recruited from the Pharmacogenomics Biobank of the University of Southern Denmark. Treatment allocation was generated by the Good Clinical Practice Unit, Odense University Hospital, Denmark. Variables of whole-body glucose metabolism were assessed using [3-(3)H]glucose, indirect calorimetry and measurement of substrates and counter-regulatory hormones. The primary outcome was endogenous glucose production (EGP). RESULTS Thirty-seven individuals were randomised. Thirty-four completed the study (12 had none, 13 had one and nine had two reduced-function alleles in OCT1). Three were excluded from the analysis because of early dropout. Metformin significantly stimulated glucose disposal rates and non-oxidative glucose metabolism with no effect on glucose oxidation. This increase in glucose utilisation was explained by a concomitant increase in glycolytic flux and accompanied by increased EGP, most likely mediated by increased plasma lactate, glucagon and cortisol levels. There was no effect of reduced-function OCT1 alleles on any of these measures. All individuals completed the glycogen-depleting fast without hypoglycaemia. CONCLUSIONS/INTERPRETATION Metformin stimulates glycolytic glucose utilisation and lactate production in the glycogen-depleted state. This may trigger a rise in glucose counter-regulatory hormones and subsequently an increase in EGP, which protects against hypoglycaemia. TRIAL REGISTRATION ClinicalTrials.gov NCT01400191 FUNDING: Danish Research Council for Health and Disease (0602-02695B) and Odense University Hospital Free Research Fund, 2012.
Collapse
Affiliation(s)
- Mette Marie H Christensen
- Department of Public Health, Clinical Pharmacology, University of Southern Denmark, J.B. Winsloews Vej 19, 5000, Odense, Denmark.
| | - Kurt Højlund
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Section of Molecular Diabetes & Metabolism, Institute of Molecular Medicine & Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ole Hother-Nielsen
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Tore B Stage
- Department of Public Health, Clinical Pharmacology, University of Southern Denmark, J.B. Winsloews Vej 19, 5000, Odense, Denmark
| | - Per Damkier
- Department of Public Health, Clinical Pharmacology, University of Southern Denmark, J.B. Winsloews Vej 19, 5000, Odense, Denmark
- Department of Clinical Chemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | | | - Kim Brøsen
- Department of Public Health, Clinical Pharmacology, University of Southern Denmark, J.B. Winsloews Vej 19, 5000, Odense, Denmark
| |
Collapse
|