Endocrinology & Metabolic Syndrome

Endocrinology & Metabolic Syndrome
Open Access

ISSN: 2161-1017

+44 1478 350008

Review Article - (2012) Volume 0, Issue 0

Pharmacogenetics of Oral Antidiabetic Drugs: Potential Clinical Application

Ji-Ye Yin1, Yu Guo1, Qiong Huang2, Hong Sun3, Hong-Hao Zhou1 and Zhao-Qian Liu1*
1Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan 410078, People’s Republic of China
2Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education Ministry, Hefei, Anhui 230032, P.R. China
3Department of Pharmacy, Fujian Provincal Hospital, Fuzhou 350001, People’s Republic of China
*Corresponding Author: Zhao-Qian Liu, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, Hunan 410078, P.R. China, Tel: +86 731 84805380, Fax: +86-731-82354476 Email:

Abstract

Oral antidiabetic drugs (OADs) are widely used to treat type 2 diabetes mellitus (T2DM). However, inherited difference is one of the major issues that affect OADs therapeutic efficacy. In the present paper, we performed a critical review on recent advances in genetic polymorphisms that influence OADs therapeutic efficacy and metabolism. Sulfonylureas (SUs), Meglitinides, Metformin and Thiazolidinediones (TZDs) will be discussed.

Keywords: Diabetes, Oral Antidiabetic Drugs (OADs), Sulfonylureas (SUs), Meglitinides, Metformin, Thiazolidinediones, Polymorphism

Introduction

Type 2 diabetes mellitus (T2DM) is one of the most serious public health problems worldwide [1]. It is characterized by two major pathophysiological features: insulin resistance and pancreatic β-cell dysfunction [2]. Oral antidiabetic drugs (OADs) are widely used to treat T2DM. However, inherited difference is one of the major issues that affect OADs therapeutic efficacy. Present OADs can be classified into at least five groups: Sulfonylureas (SUs), Meglitinides, Metformin, Thiazolidinediones (TZDs) and α-glucosidase inhibitors. Polymorphisms in drug-metabolizing enzymes, transporters, receptors and other drug targets have been proved to be correlated with efficacy and toxicity of many medications [3,4]. Thus, genetic polymorphism involved in drug absorption, distribution, metabolism and excretion (ADME) play important role in OADs treatment [5]. In the present paper, we performed a critical review on recent advances in genetic polymorphisms that influence OADs therapeutic efficacy (Table 1).

Drug Classification Drugs Related gene polymorphisms rs number Genotype/Observation Ref
Sulfonylureas Sulfonylureas TCF7L2 G483+9017T rs12255372 GG/More sensitive to sulfonylurea [15]
  Glibenclamide KCNJ11 G635A rs5219 AA/Increased risk of secondary failure to sulfonylurea [13]
    CYP2C9*2   CYP2C9 *2/*3/Increased risk of severe hypoglycaemia [8]
    CYP2C9*3   CYP2C9 *3/*3/Increased risk of severe hypoglycaemia [8]
Meglitinides Repaglinide KCNJ11 G635A rs5219 GG/ Good drug efficacy [20]
    SLC30A8 C826T rs13266634 CC/ Poor drug efficacy. [21]
    SLC30A8 G827A rs168s89462 GG/ Poor drug efficacy [21]
    NAMPT G-3186T rs11977021 CT/ Poor drug efficacy [22]
    IGF2BP2 C239+29254G rs4402960 GG/ Good drug efficacy [23]
    IGF2BP2 A239+11861C rs1470579 AA/ Poor drug efficacy [23]
    TCF7L2 T948-1026C rs290487 TT/ Good drug efficacy [20]
    KCNQ1 C1795-10451T rs2237897 TT/ Good drug efficacy [44]
    CYP2C8*1   CYP2C8*1/*3/ Lower AUC(0-infinity) and Cmax of repaglinide [25]
    CYP3A4*1   CYP3A4*1/*18/Reduced mean elimination rate constant and increased mean half-life [26]
  Nateglinide CYP2C9*3   CYP2C9*3/*3 / Increased risk of hypoglycaemia. [27]
Metformin Metformin SRR T-4-2593C rs391300 GG/ Poor drug efficacy [58]
    SLC22A1 C181T
SLC22A1 G1201A
SLC22A1 G1393C
SLC22A1 M420del
rs12208357
rs34130495
rs34059508
Defective variants /Poor drug efficacy
Defective variants /Poor drug efficacy
Defective variants /Poor drug efficacy
Defective variants /Poor drug efficacy
[50,51]
[50,51]
[50,51]
[50,51]

Table 1: Selected studies on pharmacogenomics of oral antidiabetic drugs.

Sulfonylureas (SUs)

SUs are the first developed OAD. They close ATP-sensitive potassium channels (KATP) by targeting pancreatic β-cell membrane, which lead to an enhanced insulin secretion in a glucose independent manner. Genetic polymorphisms are critical important for the individual different sensitivity to SUs. Cytochrome P450 (CYP) 2C9 is the main enzyme that catalyzes biotransformation of Sulfonylureas, thus its polymorphisms affect SUs therapeutic efficiency [6]. In addition, polymorphisms of potassium inwardly-rectifying channel, subfamily J, member 11 (KCNJ11) and transcription factor 7-like 2 (TCF7L2) also affect SUs medication [7], thus these genetic polymorphisms will be discussed in this section.

Polymorphisms of CYP2C9: Genotyping of CYP2C9 allelic variants is a relatively easy and inexpensive test, thus its polymorphisms were widely studied [8]. In a study of 20 T2DM patients with severe hypoglycemia during sulfonylurea treatment, the *3/*3 and *2/*3 genotype carriers were significantly higher than those in control subjects and T2DM patients without hypoglycemia [8]. However, another prospective study conducted by the same group showed that CYP2C9 slow metabolizer genotypes were not correlated with hypoglycemia [9]. This study included 102 T2DM patients with sulfonylurea-induced hypoglycemia and 101 control patients. There were no significant difference of CYP2C9 *2/*2, *2/*3 and *3/*3 genotype frequencies between case and control populations. Thus, the conclusion of correlation of CYP2C9 polymorphisms and sulfonylurea treatment efficacy needs further confirmation.

Polymorphisms of KCNJ11: KCNJ11 encodes Kir6.2, which is a subunit of KATP channel. KCNJ11 polymorphisms and sulfonylurea response in T2DM patients was widely studied. The most intensively investigated polymorphism is Glu23Lys (E23K) [10-12]. It was hypothesized that K23 allele may be associated with interindividual variability of sulfonylurea response. This hypothesis was tested in a group of newly diagnosed diabetes patients treated with Sulfonylureas [10]. However, in this study, the KCNJ11 E23K variant was not significantly associated with patients’ response to sulfonylurea. In another study, Sesti et al. [13] showed that E23K variant was associated with an increased risk of secondary sulfonylurea failure in T2DM patients. Thus, the correlation between KCNJ11 E23K variant and sulfonylurea response in T2DM patients need more investigations.

Polymorphisms of TCF7L2: Among T2DM associated genes, TCF7L2 were widely studied. It correlated with an increased risk of T2DM in several populations [14]. It was reported that TCF7L2 polymorphisms influence the initial success treatment of sulfonylurea in T2DM patients. The most intensively studied polymorphisms are rs12255372 and rs7903146, which were associated with diabetes risk. In a study of large sample size T2DM patients, risk allele carriers of rs12255372 and rs7903146 were more sensitive to sulfonylurea. However, no association was detected for metformin [15].

Sulfonylurea therapy is safe and effective for a short term use in most patients and may successfully replace treatment with insulin injections [16]. It dramatically improves glycemic control and should be considered as the first line drug for patients with poor glycemic control on an appropriate diet [17]. However, polymorphisms of above mentioned genes need to be considered when using SUs.

Meglitinides

Meglitinides (repaglinide and nateglinide) are a new class of insulin secretagogues, which structurally unlike to sulfonylureas. They decrease blood glucose by stimulating release of insulin. Repaglinide, similar to sulfonylureas, closes the β cell KATP channel. Inhibition of the KATP channel depolarizes the cells and causes cell calcium channel open, as a result, the calcium influx induces insulin secretion [18]. Nateglinide is also a KATP channel inhibitor, but its selective enhancement of early-phase insulin secretion enables nateglinide to minimize postmeal hyperglycemia with minimal propensity for hypoglycemia [19]. A number of individual differences in the meglitinides therapeutic efficacy have been reported [6], our previous studies showed that genetic polymorphisms were one of the major contributors [20-23]. Thus, those studies will be discussed in this section.

Polymorphisms of CYP: One of the meglitinides, repaglinide is mainly metabolized by CYP3A4 and CYP2C8 [24]. CYP2C8*3 variant was related with reduced plasma concentrations of repaglinide [25]. Subjects with CYP2C8*1/*3 genotype showed lower AUC (0-infinity) and Cmax of repaglinide compared with those with CYP2C8*1/*1 genotype (P<0.05). CYP3A4 metabolizes 66% repaglinide, it is reported that CYP3A4*18 affects repaglinide pharmacokinetics[26], carriers with CYP3A4*18 have reduced clearance. Nateglinide is mainly metabolized by CYP2C9. It is reported that moderate dose adjustment based on CYP2C9 genotypes help reduce interindividual variability of anti-hyperglycemic effects of nateglinide. Carriers of CYP2C9*3/*3 genotype have a slightly higher risk of hypoglycemia compared to carriers of CYP2C9*1, particularly when taking nateglinide doses above 120 mg [27]. Therefore, CYP genetic polymorphisms play an important role in the pharmacokinetics of meglitinides.

Polymorphisms of KCNJ11: KCNJ11 plays a crucial role in insulin secretion and glucose metabolism. Although it is not the target of meglitinides, its gene polymorphism influences the action of meglitinides by influencing KATP channel sensitivity to ATP [28]. Our study found that subjects with at least one A allele of KCNJ11 rs5219 polymorphism showed lower level of fasting plasma glucose (FPG) and postprandial plasma glucose (PPG) (P<0.05), thus patients with A allele are more sensitive to repaglinide [20].

Polymorphisms of SLC30A8: SLC30A8 encodes ion channel zinc transporter protein member 8 (ZnT-8), which carries zinc from the cytoplasm into insulin secretory vesicles. It is a critical molecule during the insulin maturation and release process [29]. Nonsynonymous SNP rs13266634 (973C>T) [30] and missense mutation rs16889462 (974G>A) [31] have been found to be associated with T2DM onset and development. Our study showed that SLC30A8 rs13266634 (973C>T) and rs16889462 (974G>A) polymorphisms were associated with repaglinide therapeutic efficacy in Chinese T2DM patients [21]. T2DM patients with C allele of rs13266634 or an allele of rs16889462 have better repaglinide drug efficacy than TT or GG genotypes.

Polymorphisms of NAMPT: Nicotinamide adenine dinucleotide (NAD) is an essential coenzyme in many metabolic reactions [32]. Nicotinamide phosphoribosyl transferase (NAMPT), the rate-limiting enzyme during nicotinamide conversion into nicotinamide nucleotide (nicotinamide mononucleotide; NMN), is an NAD biosynthetic enzyme that plays precise role in the regulation of glucose metabolism by affecting the insulin secretion in β cells [33]. Thus, NAMPT may be involved in the repaglinide action. Despite contradictory results were reported about the correlation of NAMPT gene polymorphisms and T2DM susceptibility [34,35], our result showed that -948G>T and -3186C>T polymorphisms are not associated with T2DM susceptibility in Chinese population. However, -3186C>T polymorphism is associated with plasma levels of postprandial insulin secretion (PINS) and cholesterol (CHO) in Chinese T2DM patients with repaglinide monotherapy [22].We also found that frequencies of -3186C> T polymorphism in Chinese Han population were different from those in Caucasian populations, thus, different dosages of repaglinide may be used among different populations.

Polymorphisms of insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2): IGF2BP2 participates in the insulin signaling pathway and insulin secretion by targeting insulin-like growth factor 2 (IGF-2), which is an important growth and insulin signaling molecule [36]. T2DM patients with different IGF2BP2 genotypes showed various levels of insulin secretion [37]. It has also been confirmed that IGF2BP2 gene polymorphisms were related with T2DM susceptibility, especially in Asia populations [31,38]. Our previous study indicated that IGF2BP2 rs1470579 and rs4402960 polymorphisms increase T2DM risk in the Chinese population. In addition, rs1470579 and rs4402960 polymorphisms in the IGF2BP2 gene showed reduced therapeutic efficacy of repaglinide treatment [23]. Patients with C allele of rs1470579 have reduced treatment on FPG (P<0.05) and PPG (P<0.05). Patients with T allele of rs4402960 exhibited enhanced effect of repaglinide treatment of PINS (P<0.01). These results provided basis for clinical dose adjustment in the future.

3.2.6) Polymorphisms of TCF7L2: As TCF7L2 plays an important role in insulin secretion, glucose metabolism and β-cells functional regulation, mutations of TCF7L2 were reported to be associated with T2DM susceptibilities in different ethnic, including Chinese Han population [39]. We investigated whether TCF7L2 gene polymorphisms were associated with repaglinide efficacy, the results showed that rs290487 (C>T) variants enhanced repaglinide effect in Chinese patients with T2DM [20]. Subjects with TCF7L2 rs290487 TT genotype are more sensitive to repaglinide than those with CC or CT genotypes. So far, there were no other reports about the association between TCF7L2 gene variant and repaglinide therapeutic effect, thus larger sample size is needed to confirm our results. The potential mechanism underlying the enhanced repaglinide efficacy in individuals with TT genotype for TCF7L2 needs future detailed studies.

Polymorphisms of KCNQ: KCNQ1 (potassium voltage-gated channel KQT-like subfamily, member 1) encodes the pore-forming subunit of a voltage-gated K+ channel (KvLQT1). Early KCNQ1 genetic polymorphism studies focused on cardiac diseases [40,41]. Later, it is reported that KCNQ1 was also expressed in pancreatic islets and the cultured insulin-secreting INS-1 cells and may play a role in regulation of insulin secretion [42]. In Chinese Han population, KCNQ1 gene polymorphisms are associated with type 2 diabetes and impaired fasting glucose, suggesting that the KCNQ1 variants may play a role in the pathogenesis of type 2 diabetes through impaired insulin secretion of pancreatic β-cells [43]. Yu et al. [44] recently found that KCNQ1 rs2237897 were associated with repaglinide efficacy and might also be associated with rosiglitazone response, in Chinese patients with type 2 diabetes. However, how KCNQ1 polymorphisms affect outcome of these two drugs is still unknown and needs to be investigated in the future.

Metformin

Metformin’s primary effect was to reduce hepatic glucose output by increasing insulin suppression of gluconeogenesis. At the molecular level, the effects of metformin are mediated via AMP activated protein kinase (AMPK) [45]. One possible mechanism is to inhibit mitochondrial respiratory chain and thus indirectly activate AMPK by altering cellular ATP/AMP ratios [46]. The European Association for the Study of Diabetes (EASD) guidelines recommended metformin as initial pharmacotherapy for T2DM [47]. Since the publication of results of the United Kingdom Prospective Diabetes Study (UKPDS) in 1998, metformin has become the most widely prescribed oral agent for the treatment of T2DM.

Polymorphisms of organic cation transporter 1 (OCT1): Metformin is a good substrate of human OCT1 (SLC22A1) which is primarily expressed in the liver [48]. Therefore, OCT1 is important for inhibition of hepatic gluconeogenesis, which is one of the major pharmacologic effects of metformin. Previous study showed that liver OCT1 knockout mice abolished hepatic lactate production, suggested that OCT1 is crucial for metformin transporting [49]. There were some recent works on the role of SLC22A1 variation on metformin response. Shu et al. [50] investigated four non-synonymous polymorphisms of SLC22A1; they found that these polymorphisms significantly decreased metformin transport in vitro. The same group reported in another study that SLC22A1 polymorphisms were correlated with pharmacokinetics of metformin in healthy volunteers [51]. Taken together, SLC22A1 polymorphisms contribute to interindividual variability of metformin efficacy.

Polymorphisms of serine racemase (SRR): SRR synthesizes D-serine, which exist mainly in brain and some peripheral organs, such as pancreas [52,53]. Both D-serine and the neurotransmitter glutamate bind to the N-methyl D-aspartate (NMDA) receptors and trigger excitatory neurotransmission in the brain [54,55]. Both D-serine and SRR present in the pancreas. Glutamate signaling has function in peripheral tissues, including the pancreas, positively modulates secretion of both glucagon and insulin in pancreatic islets [56,57]. Thus, dysfunction of D-serine could alter glutamate signaling and affect insulin or glucagon secretion in T2DM pathogenesis. Our previous study showed that SRR polymorphism rs391300 was associated with metformin therapeutic efficacy in Chinese T2DM patients. The mutant allele carriers showed better fasting plasma glucose (FPG), postprandial plasma glucose (PPG) and plasma concentrations of CHO improvement that wild-type patients [58].

Thiazolidinediones (TZDs)

TZDs (pioglitazone, rosiglitazone) are selective PPARγ ligands that activate peroxisome proliferator-activated receptors (PPARs), which is a class of intracellular receptor. They modulate transcription of insulinsensitive genes involved in the control of glucose and lipid metabolism in lipidic, muscular tissues and liver, thus stimulate muscle glucose uptake and suppress hepatic glucose output [59].

Polymorphisms of PPARG gene: Peroxisome proliferatoractivated receptor γ (PPARγ), targets of TZDs, is encoded by PPARG. Peroxisome proliferator-activated receptor γ2 (PPARγ2), one isoform of PPAR γ [60], is a transcription factor involved in energy homeostasis, adipogenesis and insulin sensitivity. Deeb et al. [61] first reported that PPARγ2 Pro12Ala variant contributed to T2DM risk in the Japanese- Americans. Recently, a meta-analysis study based on forty-one studies in the last 10 years summarized that, Pro12Ala polymorphism in the PPARγ2 gene is associated with insulin sensitivity, the G allele carriers showed significantly lower risks of T2DM in Caucasians populations[62]. It is reported that carriers with PPARγ2 Pro12Ala genotype in the gene had a better therapeutic response to rosiglitazone than those with the Pro12Pro genotype[63]. The Pro12Ala variant of the PPARG gene is also associated with PPARγ agonist-induced fluid retention and edema [64]. However, there was no association between Pro12Ala variant and therapy efficacy of pioglitazone [65], Pro12Ala polymorphism also can’t explain the primary pioglitazone therapy failure [66], suggesting that the drug-treatment response is independent from pharmacogenetic effects of pioglitazone.

Polymorphisms of adiponectin: Adiponectin, one of the important adipocytokines secreted by adipocytes, plays a vital role in the regulation of insulin sensitivity and glucose homeostasis. We found that adiponectin polymorphisms 45T>G and 11377C>G are significantly associated with therapeutic efficacy of multiple-dose rosiglitazone treatment in Chinese patients with T2DM [67]. Namvaran et al. [68] recently demonstrated that adiponectin 45T>G was an important determinant of T2DM in the Iranian population, but adiponectin 45T>G and adiponectin receptor-2 795G>A polymorphisms were not significantly associated with the response of pioglitazone in Iranian population. Therefore, further functional studies and large scale studies are needed to explore the association between adiponectin gene and TZDs drug effect.

Polymorphisms of peroxisome proliferator activated receptor-g coactivator-1a (PGC-1α): PGC-1α, a transcriptional coactivator of PPAR γ, is involved in the glucose-lipid homeostasis [69]. An autosomal genomic scan study revealed that PGC-1α genomic region on 4p15.1 is associated with fasting serum insulin concentrations [70]. We studied 2 common polymorphisms, PGC-1α Thr394Thr and Gly482Ser, which were reported to be significantly associated with risks of T2DM in the previous investigation [71,72]. Our result suggested that PGC-1α Thr394Thr and Gly482Ser polymorphisms were associated with therapeutic efficacy of multiple-dose rosiglitazone treatment in Chinese patients with T2DM, carriers of A allele of Thr394Thr or Ser482 allele of Gly482Ser showed a poor therapeutic efficacy to rosiglitazone when compared with GG or Gly/Gly carriers [73].

Polymorphisms of uncoupling protein 2 (UCP2): UCP2 is a candidate gene of metabolic disorders; it negatively regulates glucose-stimulated insulin secretion. Tian et al. [74] found that UCP2 expression increase induced by chronic exposure of pancreatic islets to palmitate was prevented by rosiglitazone. -866 G>A variant, a prevalent polymorphism in the human UCP2 gene promoter, was found to have significant impact on its transcriptional activity and T2DM pathogenesis [75]. Our study showed that there was a strong association between rosiglitazone treatment success and UCP2-866 G>A polymorphism [76].

Polymorphisms of tumor necrosis factor-α (TNF-α): TNF-α is partly secreted by adipocyte, it causes insulin resistance through inhibiting lipogenesis, stimulating lipolysis, impairing insulin signal pathway and decreasing insulin-induced glucose uptake [77]. G-308A was one of the most common polymorphisms located in the promoter region of TNF-α gene. Some in vitro experiments demonstrated that G-308A variant increases transcriptional activation of TNF-α, because it is located in a consensus sequence of the transcription factor AP-2 [78]. Our clinical trial results showed that TNF-α G-308A polymorphism was not associated with T2DM susceptibility but with the therapeutic efficacy of rosiglitazone in T2DM patients [79]. But the frequency of TNF-α G-308A allele in Asian population was so low that we only found 4 mutant allele carriers in the study. Therefore, a larger sample size study is needed to confirm our conclusion in the future. TZDs are widely used antidiabetic drugs with proven efficacy on improve hyperglycemia, insulin sensitivity and cardio metabolic profile. However, they have several severe adverse effects. For example, troglitazone was withdrawn in 2000 due to increased risk of hepatotoxicity [80]. Rosiglitazone was also associated with an increase myocardial infarction incidence. Pioglitazone have common adverse effects of weight gain, pedal edema and bone loss [81]. Wolford et al. [82] demonstrated that differential response to troglitazone may due to sequence variation in PPARG. Thus, it is likely that genetic polymorphisms correlated with TZDs adverse effects. Future studies are needed to confirm whether gene polymorphisms can explain these drug adverse effects.

Conclusion

Numerous studies investigated the influence of pharmacogenetics to OADs. Some genetic polymorphisms were concluded to be associated with differential response to OADs. However, results of some polymorphisms were contradictory, thus these polymorphisms need to be studied in a larger and well-designed cohorts investigation. With the development of pharmacogenetics, more and more mechanisms of interindividual variability of OADs response will be unveiled. These studies will lead to personalized therapy of T2DM patients.

Acknowledgements

This work was supported by the National High-Tech R&D Program of China (863 Program) (2009AA022704), National Natural Science Foundation of China (81173129), Program for Chang Jiang Scholars and Innovative Research Team in University of Ministry of Education of China (IRT0946) and the Open Foundation of Innovative Platform in University of Hunan Province of China (10K078).

References

  1. Wild S, Roglic G, Green A, Sicree R, King H (2004) Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care 27: 1047-1053.
  2. Permutt MA, Wasson J, Cox N (2005) Genetic epidemiology of diabetes. J Clin Invest 115: 1431-1439.
  3. Yin JY, Han LF, Huang Q, Xu XJ, Zhou HH, et al. (2011) ABCC1 polymorphism Arg723Gln (2168G> A) is associated with lung cancer susceptibility in a Chinese population. Clin Exp Pharmacol Physiol 38: 632-637.
  4. Yin JY, Huang Q, Yang Y, Zhang JT, Zhong MZ, et al. (2009) Characterization and analyses of multidrug resistance-associated protein 1 (MRP1/ABCC1) polymorphisms in Chinese population. Pharmacogenet Genomics 19: 206-216.
  5. Glamoclija U, Jevric-Cauševic A (2010) Genetic polymorphisms in diabetes: influence on therapy with oral antidiabetics. Acta Pharm 60: 387-406.
  6. Kirchheiner J, Roots I, Goldammer M, Rosenkranz B, Brockmöller J (2005) Effect of genetic polymorphisms in cytochrome p450 (CYP) 2C9 and CYP2C8 on the pharmacokinetics of oral antidiabetic drugs: clinical relevance. Clin Pharmacokinet 44: 1209-1225.
  7. Holstein A, Beil W (2009) Oral antidiabetic drug metabolism: pharmacogenomics and drug interactions. Expert Opin Drug Metab Toxicol 5: 225-241.
  8. Holstein A, Plaschke A, Ptak M, Egberts EH, El-Din J, et al. (2005) Association between CYP2C9 slow metabolizer genotypes and severe hypoglycaemia on medication with sulphonylurea hypoglycaemic agents. Br J Clin Pharmacol 60: 103-106.
  9. Holstein A, Hahn M, Patzer O, Seeringer A, Kovacs P, et al. (2011) Impact of clinical factors and CYP2C9 variants for the risk of severe sulfonylurea-induced hypoglycemia. Eur J Clin Pharmacol 67: 471-476.
  10. Gloyn AL, Hashim Y, Ashcroft SJ, Ashfield R, Wiltshire S, et al. (2001) Association studies of variants in promoter and coding regions of beta-cell ATP-sensitive K-channel genes SUR1 and Kir6.2 with Type 2 diabetes mellitus (UKPDS 53). Diabet Med 18: 206-212.
  11. Florez JC, Burtt N, de Bakker PI, Almgren P, Tuomi T, et al. (2004) Haplotype structure and genotype-phenotype correlations of the sulfonylurea receptor and the islet ATP-sensitive potassium channel gene region. Diabetes 53: 1360-1368.
  12. Nielsen EM, Hansen L, Carstensen B, Echwald SM, Drivsholm T, et al. (2003) The E23K variant of Kir6.2 associates with impaired post-OGTT serum insulin response and increased risk of type 2 diabetes. Diabetes 52: 573-577.
  13. Sesti G, Laratta E, Cardellini M, Andreozzi F, Del Guerra S, et al. (2006) The E23K variant of KCNJ11 encoding the pancreatic beta-cell adenosine 5'-triphosphate-sensitive potassium channel subunit Kir6.2 is associated with an increased risk of secondary failure to sulfonylurea in patients with type 2 diabetes. J Clin Endocrinol Metab 91: 2334-2339.
  14. Pearson ER (2009) Translating TCF7L2: from gene to function. Diabetologia 52: 1227-1230.
  15. Pearson ER, Donnelly LA, Kimber C, Whitley A, Doney AS, et al. (2007) Variation in TCF7L2 influences therapeutic response to sulfonylureas: a GoDARTs study. Diabetes 56: 2178-2182.
  16. Rafiq M, Flanagan SE, Patch AM, Shields BM, Ellard S, et al. (2008) Effective treatment with oral sulfonylureas in patients with diabetes due to sulfonylurea receptor 1 (SUR1) mutations. Diabetes Care 31: 204-209.
  17. Pearson ER, Liddell WG, Shepherd M, Corrall RJ, Hattersley AT (2000) Sensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1alpha gene mutations: evidence for pharmacogenetics in diabetes. Diabet Med 17: 543-545.
  18. Holstein A, Egberts EH (2003) Risk of hypoglycaemia with oral antidiabetic agents in patients with Type 2 diabetes. Exp Clin Endocrinol Diabetes 111: 405-414.
  19. Hu S, Boettcher BR, Dunning BE (2003) The mechanisms underlying the unique pharmacodynamics of nateglinide. Diabetologia 46 Suppl 1: M37-43.
  20. Yu M, Xu XJ, Yin JY, Wu J, Chen X, et al. (2010) KCNJ11 Lys23Glu and TCF7L2 rs290487(C/T) polymorphisms affect therapeutic efficacy of repaglinide in Chinese patients with type 2 diabetes. Clin Pharmacol Ther 87: 330-335.
  21. Huang Q, Yin JY, Dai XP, Wu J, Chen X, et al. (2010) Association analysis of SLC30A8 rs13266634 and rs16889462 polymorphisms with type 2 diabetes mellitus and repaglinide response in Chinese patients. Eur J Clin Pharmacol 66: 1207-1215.
  22. Sheng FF, Dai XP, Qu J, Lei GH, Lu HB, et al. (2011) NAMPT -3186C/T polymorphism affects repaglinide response in Chinese patients with Type 2 diabetes mellitus. Clin Exp Pharmacol Physiol 38: 550-554.
  23. Huang Q, Yin JY, Dai XP, Pei Q, Dong M, et al. (2010) IGF2BP2 variations influence repaglinide response and risk of type 2 diabetes in Chinese population. Acta Pharmacol Sin 31: 709-717.
  24. Bidstrup TB, Bjørnsdottir I, Sidelmann UG, Thomsen MS, Hansen KT (2003) CYP2C8 and CYP3A4 are the principal enzymes involved in the human in vitro biotransformation of the insulin secretagogue repaglinide. Br J Clin Pharmacol 56: 305-314.
  25. Niemi M, Leathart JB, Neuvonen M, Backman JT, Daly AK, et al. (2003) Polymorphism in CYP2C8 is associated with reduced plasma concentrations of repaglinide. Clin Pharmacol Ther 74: 380-387.
  26. Ruzilawati AB, Gan SH (2010) CYP3A4 genetic polymorphism influences repaglinide's pharmacokinetics. Pharmacology 85: 357-364.
  27. Kirchheiner J, Meineke I, Müller G, Bauer S, Rohde W, et al. (2004) Influence of CYP2C9 and CYP2D6 polymorphisms on the pharmacokinetics of nateglinide in genotyped healthy volunteers. Clin Pharmacokinet 43: 267-278.
  28. Hattersley AT, Ashcroft FM (2005) Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes 54: 2503-2513.
  29. Chimienti F, Devergnas S, Favier A, Seve M (2004) Identification and cloning of a beta-cell-specific zinc transporter, ZnT-8, localized into insulin secretory granules. Diabetes 53: 2330-2337.
  30. Zeggini E, Scott LJ, Saxena R, Voight BF, Marchini JL, et al. (2008) Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet 40: 638-645.
  31. Wu Y, Li H, Loos RJ, Yu Z, Ye X, et al. (2008) Common variants in CDKAL1, CDKN2A/B, IGF2BP2, SLC30A8, and HHEX/IDE genes are associated with type 2 diabetes and impaired fasting glucose in a Chinese Han population. Diabetes 57: 2834-2842.
  32. Lin SJ, Guarente L (2003) Nicotinamide adenine dinucleotide, a metabolic regulator of transcription, longevity and disease. Curr Opin Cell Biol 15: 241-246.
  33. Revollo JR, Körner A, Mills KF, Satoh A, Wang T, et al. (2007) Nampt/PBEF/Visfatin regulates insulin secretion in beta cells as a systemic NAD biosynthetic enzyme. Cell Metab 6: 363-375.
  34. Zhang YY, Gottardo L, Thompson R, Powers C, Nolan D, et al. (2006) A visfatin promoter polymorphism is associated with low-grade inflammation and type 2 diabetes. Obesity (Silver Spring) 14: 2119-2126.
  35. Paschou P, Kukuvitis A, Yavropoulou MP, Dritsoula A, Giapoutzidis V, et al. (2010) Genetic variation in the visfatin (PBEF1/NAMPT) gene and type 2 diabetes in the Greek population. Cytokine 51: 25-27.
  36. Christiansen J, Kolte AM, Hansen TO, Nielsen FC (2009) IGF2 mRNA-binding protein 2: biological function and putative role in type 2 diabetes. J Mol Endocrinol 43: 187-195.
  37. Groenewoud MJ, Dekker JM, Fritsche A, Reiling E, Nijpels G, et al. (2008) Variants of CDKAL1 and IGF2BP2 affect first-phase insulin secretion during hyperglycaemic clamps. Diabetologia 51: 1659-1663.
  38. Takeuchi F, Serizawa M, Yamamoto K, Fujisawa T, Nakashima E, et al. (2009) Confirmation of multiple risk Loci and genetic impacts by a genome-wide association study of type 2 diabetes in the Japanese population. Diabetes 58: 1690-1699.
  39. Chang YC, Chang TJ, Jiang YD, Kuo SS, Lee KC, et al. (2007) Association study of the genetic polymorphisms of the transcription factor 7-like 2 (TCF7L2) gene and type 2 diabetes in the Chinese population. Diabetes 56: 2631-2637.
  40. Wang Q, Curran ME, Splawski I, Burn TC, Millholland JM, et al. (1996) Positional cloning of a novel potassium channel gene: KVLQT1 mutations cause cardiac arrhythmias. Nat Genet 12: 17-23.
  41. Neyroud N, Tesson F, Denjoy I, Leibovici M, Donger C, et al. (1997) A novel mutation in the potassium channel gene KVLQT1 causes the Jervell and Lange-Nielsen cardioauditory syndrome. Nat Genet 15: 186-189.
  42. Ullrich S, Su J, Ranta F, Wittekindt OH, Ris F, et al. (2005) Effects of I(Ks) channel inhibitors in insulin-secreting INS-1 cells. Pflugers Arch 451: 428-436.
  43. Qi Q, Li H, Loos RJ, Liu C, Wu Y, et al. (2009) Common variants in KCNQ1 are associated with type 2 diabetes and impaired fasting glucose in a Chinese Han population. Hum Mol Genet 18: 3508-3515.
  44. Yu W, Hu C, Zhang R, Wang C, Qin W, et al. (2011) Effects of KCNQ1 polymorphisms on the therapeutic efficacy of oral antidiabetic drugs in Chinese patients with type 2 diabetes. Clin Pharmacol Ther 89: 437-442.
  45. Zhou G, Myers R, Li Y, Chen Y, Shen X, et al. (2001) Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108: 1167-1174.
  46. Owen MR, Doran E, Halestrap AP (2000) Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J 348 Pt 3: 607-614.
  47. Campbell RK (2009) Type 2 diabetes: where we are today: an overview of disease burden, current treatments, and treatment strategies. J Am Pharm Assoc (2003) 49 Suppl 1: S3-9.
  48. Kimura N, Masuda S, Tanihara Y, Ueo H, Okuda M, et al. (2005) Metformin is a superior substrate for renal organic cation transporter OCT2 rather than hepatic OCT1. Drug Metab Pharmacokinet 20: 379-386.
  49. Wang DS, Kusuhara H, Kato Y, Jonker JW, Schinkel AH, et al. (2003) Involvement of organic cation transporter 1 in the lactic acidosis caused by metformin. Mol Pharmacol 63: 844-848.
  50. Shu Y, Sheardown SA, Brown C, Owen RP, Zhang S, et al. (2007) Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J Clin Invest 117: 1422-1431.
  51. Shu Y, Brown C, Castro RA, Shi RJ, Lin ET, et al. (2008) Effect of genetic variation in the organic cation transporter 1, OCT1, on metformin pharmacokinetics. Clin Pharmacol Ther 83: 273-280.
  52. Wolosker H, Blackshaw S, Snyder SH (1999) Serine racemase: a glial enzyme synthesizing D-serine to regulate glutamate-N-methyl-D-aspartate neurotransmission. Proc Natl Acad Sci U S A 96: 13409-13414.
  53. Wolosker H, Sheth KN, Takahashi M, Mothet JP, Brady RO Jr, et al. (1999) Purification of serine racemase: biosynthesis of the neuromodulator D-serine. Proc Natl Acad Sci U S A 96: 721-725.
  54. Mothet JP, Parent AT, Wolosker H, Brady RO Jr, Linden DJ, et al. (2000) D-serine is an endogenous ligand for the glycine site of the N-methyl-D-aspartate receptor. Proc Natl Acad Sci U S A 97: 4926-4931.
  55. Wolosker H, Dumin E, Balan L, Foltyn VN (2008) D-amino acids in the brain: D-serine in neurotransmission and neurodegeneration. FEBS J 275: 3514-3526.
  56. Gonoi T, Mizuno N, Inagaki N, Kuromi H, Seino Y, et al. (1994) Functional neuronal ionotropic glutamate receptors are expressed in the non-neuronal cell line MIN6. J Biol Chem 269: 16989-16992.
  57. Inagaki N, Kuromi H, Gonoi T, Okamoto Y, Ishida H, et al. (1995) Expression and role of ionotropic glutamate receptors in pancreatic islet cells. FASEB J 9: 686-691.
  58. Dong M, Gong ZC, Dai XP, Lei GH, Lu HB, et al. (2011) Serine racemase rs391300 G/A polymorphism influences the therapeutic efficacy of metformin in Chinese patients with diabetes mellitus type 2. Clin Exp Pharmacol Physiol 38: 824-829.
  59. Mascitelli L, Pezzetta F (2005) Thiazolidinediones. N Engl J Med 352: 205-207.
  60. Beamer BA, Negri C, Yen CJ, Gavrilova O, Rumberger JM, et al. (1997) Chromosomal localization and partial genomic structure of the human peroxisome proliferator activated receptor-gamma (hPPAR gamma) gene. Biochem Biophys Res Commun 233: 756-759.
  61. Deeb SS, Fajas L, Nemoto M, Pihlajamäki J, Mykkänen L, et al. (1998) A Pro12Ala substitution in PPARgamma2 associated with decreased receptor activity, lower body mass index and improved insulin sensitivity. Nat Genet 20: 284-287.
  62. Huguenin GV, Rosa G (2010) The Ala allele in the PPAR-gamma2 gene is associated with reduced risk of type 2 diabetes mellitus in Caucasians and improved insulin sensitivity in overweight subjects. Br J Nutr 104: 488-497.
  63. Kang ES, Park SY, Kim HJ, Kim CS, Ahn CW, et al. (2005) Effects of Pro12Ala polymorphism of peroxisome proliferator-activated receptor gamma2 gene on rosiglitazone response in type 2 diabetes. Clin Pharmacol Ther 78: 202-208.
  64. Hansen L, Ekstrøm CT, Tabanera Y Palacios R, Anant M, Wassermann K, et al. (2006) The Pro12Ala variant of the PPARG gene is a risk factor for peroxisome proliferator-activated receptor-gamma/alpha agonist-induced edema in type 2 diabetic patients. J Clin Endocrinol Metab 91: 3446-3450.
  65. Blüher M, Lübben G, Paschke R (2003) Analysis of the relationship between the Pro12Ala variant in the PPAR-gamma2 gene and the response rate to therapy with pioglitazone in patients with type 2 diabetes. Diabetes Care 26: 825-831.
  66. Mathur SK, Rathore R, Chandra S, Sharma BB, Sharma L, et al. (2009) Primary pioglitazone failure in Asian Indian diabetics is not related to common Pro12Ala polymorph of PPAR-gamma gene. Indian J Physiol Pharmacol 53: 175-180.
  67. Sun H, Gong ZC, Yin JY, Liu HL, Liu YZ, et al. (2008) The association of adiponectin allele 45T/G and -11377C/G polymorphisms with Type 2 diabetes and rosiglitazone response in Chinese patients. Br J Clin Pharmacol 65: 917-926.
  68. Namvaran F, Rahimi-Moghaddam P, Azarpira N, Dabbaghmanesh MH (2011) Polymorphism of adiponectin (45T/G) and adiponectin receptor-2 (795G/A) in an Iranian population: relation with insulin resistance and response to treatment with pioglitazone in patients with type 2 diabetes mellitus. Mol Biol Rep .
  69. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, et al. (2001) Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1. Nature 413: 131-138.
  70. Pratley RE, Thompson DB, Prochazka M, Baier L, Mott D, et al. (1998) An autosomal genomic scan for loci linked to prediabetic phenotypes in Pima Indians. J Clin Invest 101: 1757-1764.
  71. Vimaleswaran KS, Radha V, Ghosh S, Majumder PP, Deepa R, et al. (2005) Peroxisome proliferator-activated receptor-gamma co-activator-1alpha (PGC-1alpha) gene polymorphisms and their relationship to Type 2 diabetes in Asian Indians. Diabet Med 22: 1516-1521.
  72. Al-Khalili L, Forsgren M, Kannisto K, Zierath JR, Lönnqvist F, et al. (2005) Enhanced insulin-stimulated glycogen synthesis in response to insulin, metformin or rosiglitazone is associated with increased mRNA expression of GLUT4 and peroxisomal proliferator activator receptor gamma co-activator 1. Diabetologia 48: 1173-1179.
  73. Zhang KH, Huang Q, Dai XP, Yin JY, Zhang W, et al. (2010) Effects of the peroxisome proliferator activated receptor-? coactivator-1a (PGC-1a) Thr394Thr and Gly482Ser polymorphisms on rosiglitazone response in Chinese patients with type 2 diabetes mellitus. J Clin Pharmacol 50: 1022-1030.
  74. Tian JY, Li G, Gu YY, Zhang HL, Zhou WZ, et al. (2006) Role and mechanism of rosiglitazone on the impairment of insulin secretion induced by free fatty acids on isolated rat islets. Chin Med J (Engl) 119: 574-580.
  75. Bulotta A, Ludovico O, Coco A, Di Paola R, Quattrone A, et al. (2005) The common -866G/A polymorphism in the promoter region of the UCP-2 gene is associated with reduced risk of type 2 diabetes in Caucasians from Italy. J Clin Endocrinol Metab 90: 1176-1180.
  76. Yang M, Huang Q, Wu J, Yin JY, Sun H, et al. (2009) Effects of UCP2 -866 G/A and ADRB3 Trp64Arg on rosiglitazone response in Chinese patients with Type 2 diabetes. Br J Clin Pharmacol 68: 14-22.
  77. Borst SE (2004) The role of TNF-alpha in insulin resistance. Endocrine 23: 177-182.
  78. Pihlajamäki J, Ylinen M, Karhapää P, Vauhkonen I, Laakso M (2003) The effect of the -308A allele of the TNF-alpha gene on insulin action is dependent on obesity. Obes Res 11: 912-917.
  79. Liu HL, Lin YG, Wu J, Sun H, Gong ZC, et al. (2008) Impact of genetic polymorphisms of leptin and TNF-alpha on rosiglitazone response in Chinese patients with type 2 diabetes. Eur J Clin Pharmacol 64: 663-671.
  80. Kohlroser J, Mathai J, Reichheld J, Banner BF, Bonkovsky HL (2000) Hepatotoxicity due to troglitazone: report of two cases and review of adverse events reported to the United States Food and Drug Administration. Am J Gastroenterol 95: 272-276.
  81. Rizos CV, Elisaf MS, Mikhailidis DP, Liberopoulos EN (2009) How safe is the use of thiazolidinediones in clinical practice? Expert Opin Drug Saf 8: 15-32.
  82. Wolford JK, Yeatts KA, Dhanjal SK, Black MH, Xiang AH, et al. (2005) Sequence variation in PPARG may underlie differential response to troglitazone. Diabetes 54: 3319-3325.
Citation: Yin J, Guo Y, Huang Q, Sun H, Zhou H, et al. (2012) Pharmacogenetics of Oral Antidiabetic Drugs: Potential Clinical Application. Endocrinol Metabol Syndrome S5:003.

Copyright: © 2012 Yin J, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top