Next Article in Journal
Investigation of the Formation Process of Two Piracetam Cocrystals during Grinding
Previous Article in Journal
Mucoadhesive Gels Designed for the Controlled Release of Chlorhexidine in the Oral Cavity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transporter-Mediated Drug–Drug Interactions with Oral Antidiabetic Drugs

Institute of Experimental and Clinical Pharmacology and Toxicology, Clinical Pharmacology and Clinical Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
Pharmaceutics 2011, 3(4), 680-705; https://doi.org/10.3390/pharmaceutics3040680
Submission received: 11 August 2011 / Revised: 29 August 2011 / Accepted: 8 October 2011 / Published: 12 October 2011

Abstract

: Uptake transporters (e.g., members of the SLC superfamily of solute carriers) and export proteins (e.g., members of the ABC transporter superfamily) are important determinants for the pharmacokinetics of drugs. Alterations of drug transport due to concomitantly administered drugs that interfere with drug transport may alter the kinetics of drug substrates. In vitro and in vivo studies indicate that many drugs used for the treatment of metabolic disorders and cardiovascular diseases (e.g., oral antidiabetic drugs, statins) are substrates for uptake transporters and export proteins expressed in the intestine, the liver and the kidney. Since most patients with type 2 diabetes receive more than one drug, transporter-mediated drug-drug interactions are important molecular mechanisms leading to alterations in oral antidiabetic drug pharmacokinetics with the risk of adverse drug reactions. This review focuses on uptake transporters of the SLCO/SLC21 (OATP) and SLC22 (OCT/OAT) family of solute carriers and export pumps of the ABC (ATP-binding cassette) transporter superfamily (especially P-glycoprotein) as well as the export proteins of the SLC47 (MATE) family and their role for transporter-mediated drug-drug interactions with oral antidiabetic drugs.

1. Introduction

Drug effects result from the interplay of multiple processes that influence drug absorption, metabolism and excretion as well as drug response. Although, in past decades, most studies have focused on the importance of drug metabolizing enzymes (e.g., cytochrome P450 monooxygenases), in recent years it became evident that, in addition, transport proteins located in distinct membrane domains are important for drug effects. Furthermore, since all metabolizing enzymes are located intracellularly, the uptake of drugs from the extracellular space across the plasma membrane into the cell is a prerequisite for subsequent metabolism. Generally, transport proteins can be subdivided into two major groups: uptake transporters, mediating the transport of substances and drugs from the outside into cells and export proteins responsible for the translocation of substances or drugs and drug metabolites out of cells.

Uptake transporters mostly belong to the superfamily of solute carriers (SLCs [1]). Today, the SLC superfamily is comprised of 378 different transport proteins grouped into 51 different transporter families. For detailed information on the SLC superfamily see [2]. Important transporter families within the SLC transporter superfamily are the SLC21/SLCO family, the SLC22 family and the SLC10 family. Organic anion transporting polypeptides (OATPs) are members of the SLC21/SLCO family [37] whereas organic cation transporters (OCTs) and organic anion transporters (OATs) are members of the SLC22 family [8]. This review describes oral antidiabetic drugs and transporter-mediated drug-drug interactions and we especially focus on the uptake transporters OATP1B1 (gene symbol: SLCO1B1), OATP1B3 (SLCO1B3) and OATP2B1 (SLCO2B1) of the SLC21/SLCO family, the SLC22 family members OCT1 (SLC22A1), OCT2 (SLC22A2) and OCT3 (SLC22A3) and the sodium-dependent bile salt transporter NTCP belonging to the SLC10 family. In human hepatocytes (Figure 1), the OATP family members OATP1B1 and OATP1B3 are predominantly, if not exclusively, expressed [911]. OATP2B1, the third highly expressed hepatic OATP, is also expressed in enterocytes (Figure 1) of the intestinum [12] and in several other tissues such as the heart [13]. The SLC22 family members OCT1 and OCT3 [1416] as well as the SLC10 family member NTCP are also expressed in hepatocytes [17]. In addition, the presence of OCT2 and OCT3 in renal epithelial cells has been demonstrated (Figure 1; [18]).

Substrates for OATP1B1, OATP1B3 and OATP2B1 include several endogenously synthesized organic anions such as bile salts and hormone metabolites as well as widely prescribed drugs like HMG-CoA-reductase inhibitors (statins, e.g., atorvastatin [1921]) and antibiotics (e.g., benzylpenicillin [22]). Physiological substrates of OCTs include hormones (e.g., corticosterone [23]) and neurotransmitters (e.g., epinephrine [23]) whereas OAT3 transports cAMP [24] and cGMP [25]. Drugs transported by OCTs include antineoplastic agents (cisplatin [26,27]) as well as the antidiabetic drug metformin [28]. Interestingly, the sodium-dependent bile salt transporter NTCP also transports drugs as it has been demonstrated for the HMG-CoA-reductase inhibitor rosuvastatin [29].

Efflux transporters mainly belong to the ATP-binding cassette (ABC) transporter family [3033]. These export pumps use energy derived from ATP hydrolysis to mediate substrate transport, often against a concentration gradient. Today, the human ABC superfamily consists of 49 different ABC transporters grouped into 6 different ABC families [ABCA–ABCG (for detailed information see [34]). In the focus of the review are the ABCB family member P-glycoprotein (P-gp, ABCB1, [35]), the bile salt export pump BSEP (ABCB11) and the ABCG family member BCRP (ABCG2). P-gp and BCRP are expressed in enterocytes, hepatocytes and renal epithelial cells (Figure 1; [3638]), whereas the expression of BSEP is restricted to hepatocytes (Figure 1; [39]).

P-gp is the best characterized human drug transporter with a very broad substrate spectrum transporting a variety of different drugs [4042]. The ABCG family member BCRP, transports several endogenous compounds and is further capable of transporting a variety of different drugs including statins, calcium channel blockers and antivirals [43].

The MATE transporter family (multidrug and toxin extrusion; gene symbol SLC47) consists of two members, MATE1 (SLC47A1) and MATE2 [MATE2-K (SLC47A2)], which are localized to apical membrane domains. Whereas human MATE1 is strongly expressed in liver and kidney (Figure 1) and to a lesser extent in several other tissues including skeletal muscle and testis [44,45], MATE2 is almost exclusively expressed in the kidney and localized in the luminal membrane of proximal tubular epithelial cells. In contrast to the ATP-driven ABC transporters, MATE proteins are electroneutral transporters using an oppositely directed proton gradient as driving force (Figure 1). MATE1 and MATE2 have similar substrate and inhibitor specificities which overlap with those of OCTs. Endogenous substrates include organic cations (e.g., creatinine, guanidine) as well as clinically used drugs such as the antimalarial drug quinine [45,46], the antineoplastic agent cisplatin [27] and the antidiabetic drug metformin [46]. Detailed information on tissue distribution and substrate spectrum of the respective uptake and export transporters are summarized in the mentioned reviews [8,14,33,47].

After administration and passage through the intestine, oral antidiabetic drugs have to be taken up from the portal venous blood via the basolateral membrane into hepatocytes before they are metabolized, cause drug effects via intrahepatic mechanisms or are transported back into the systemic circulation for extrahepatic effects. So far, little is known on the involvement of oral antidiabetic drugs into intestinal transporter-mediated drug-drug interactions [48]. The characteristics of some widely used oral antidiabetic drugs together with their mode of action, metabolizing enzymes and transport proteins relevant for their membrane translocation or drug-drug interactions are summarized in Table 1. Since patients with type 2 diabetes are commonly treated with more than one drug (in most cases one or more oral antidiabetic drug and additionally a statin and an antihypertensive drug), it is essential to understand the molecular mechanisms underlying drug-drug interactions, which might cause changes in the effect or the pharmacokinetics of these drugs. Aside from metabolizing enzymes [4952] it is now well established that also modification of transport function is involved in these drug-drug interactions. Therefore, the role of transport proteins for drug-drug interactions with frequently used oral antidiabetic drugs presented in table 1 are in the focus of this review.

2. Oral Antidiabetic Drugs and OATPs

OATP1B1, OATP1B3 and OATP2B1 are localized in the basolateral membrane transporting substances and drugs from the portal venous blood into hepatocytes [4,47]. OATP2B1 is further expressed in enterocytes and localized there to the luminal membrane mediating the uptake of substances and drugs from the intestinal lumen into the body [12]. Several drugs have been identified as substrates for these OATPs including antibiotics and HMG-CoA-reductase inhibitors (statins) [5,10]. Repaglinide was one of the first oral antidiabetic drugs shown to interact with hepatic OATPs. More indirect evidence for the involvement of OATP1B1 in repaglinide pharmacokinetics has been published by Niemi and colleagues [52]. In this study they investigated the in vivo effect of cyclosporine, a known inhibitor of CYP3A4 and OATP1B1 on the pharmacokinetics and pharmacodynamics of repaglinide. They found that cyclosporine raised the plasma concentrations of concomitantly administered repaglinide probably by inhibiting its OATP1B1-mediated hepatic uptake and the subsequent metabolism by CYP3A4. In the same year the authors published a second in vivo study [63] demonstrating that a polymorphic variant of the OATP1B1 protein is a major determinant of the interindividual variability in the pharmacokinetics of repaglinide. Interestingly, polymorphic variants of the OATP1B1 protein seem not to have a significant effect on nateglinide, the second frequently used meglitinide derivative [66] and did not influence the pharmacokinetics of rosiglitazone or pioglitazone [90]. Based on these observations, Bachmakov et al. investigated the effect of repaglinide and rosiglitazone on OATP1B1-, OATP1B3- and OATP2B1-mediated transport in vitro [64]. Using stably transfected HEK cells recombinantly overexpressing these OATP family members and BSP as prototypic substrate, they found that both rosiglitazone and repaglinide inhibited the uptake mediated by these three transporters (Figure 2). Both oral antidiabetic drugs showed a potent uptake inhibition with IC50 values around 10 μM. In the same study they also used pravastatin as substrate for OATP1B1 and OATP1B3 and they demonstrated that repaglinide at a concentration of 10 μM significantly inhibited OATP1B1-mediated pravastatin uptake, whereas the pravastatin uptake by OATP1B3 was only slightly reduced and significantly inhibited only at a repaglinide concentration of 100 μM (Figure 3). An interesting observation was made analyzing the effect of rosiglitazone on OATP1B1- and OATP1B3-mediated pravastatin uptake. Whereas repaglinide at a concentration of 10 μM inhibited the uptake, rosiglitazone at the same concentration stimulated pravastatin uptake by OATP1B1 and OATP1B3 and at the same time shows inhibition only at the highest tested concentration of 100 μM (Figure 3). This demonstrated that not only uptake inhibition but also stimulation of drug uptake could be a result of drug-drug interactions. Recently, this stimulatory effect has been analyzed in detail using pravastatin as substrate and non-steroidal anti-inflammatory drugs as interacting substances [91]. In this study the authors showed that ibuprofen stimulated OATP1B1- and OATP1B3-mediated pravastatin uptake likely by an allosteric mechanism without being transported. In the case of rosiglitazone one can assume that at low concentrations the uptake of pravastatin can be stimulated by acting as allosteric modulator and that only at high substrate concentrations a competitive inhibition can be detected. However, the clinical relevance of these findings remains to be clarified.

Glibenclamide is a frequently prescribed insulin secretagogue stimulating the insulin secretion from pancreatic β-cells. It is extensively metabolized by CYP2C9, CYP2C19 and CYP3A4 [53,55] and seems to be a substrate for OATP2B1 [54]. Using HEK cells overexpressing OATP2B1 Satoh et al. have shown that grapefruit and citrus juice inhibited OATP2B1-mediated glibenclamide uptake [54]. Since OATP2B1 is also localized in the apical membrane of enterocytes this may result in a reduced intestinal absorption of orally administered glibenclamide and of other drugs that are substrates of OATP2B1. Interestingly, this could not be confirmed in an in vivo study analyzing the effect of clarithromycin and grapefruit juice on the pharmacokinetics of glibenclamide [92]. In this study 12 subjects ingested 250 mg clarithromycin or placebo with 200 mL grapefruit juice three times daily. On day three, they ingested in addition 0.875 mg glibenclamide with sugar water or grapefruit juice and the concentrations of glibenclamide and clarithromycin in plasma, glucose in blood and excretion of the metabolite hydroxyglibenclamide in urine were measured. These analyses demonstrated that clarithromycin increased plasma concentrations of glibenclamide, maybe by inhibiting OATP2B1-mediated uptake and CYP3A4 metabolism in the intestine, but no effect of grapefruit juice on glibenclamide pharmacokinetics could be detected. The results of this study were confirmed in a study by Niemi et al. demonstrating that clarithromycin also increases the plasma concentrations and effects of simultaneously administered repaglinide [49].

An interaction of atorvastatin with glibenclamide has been demonstrated using MDCK cells stably expressing OATP2B1 [13]. In this study the authors investigated the expression of OATP2B1 in human heart samples and analyzed OATP2B1-mediated atorvastatin and glibenclamide transport and transport inhibition by simultaneously administered drugs. They demonstrated that OATP2B1-mediated glibenclamide transport was inhibited not only by atorvastatin but also by simvastatin, cerivastatin and estrone-3-sulfate (E3S), whereas OATP2B1-mediated E3S uptake was inhibited by gemfibrozil. A potential hazardous interaction between gemfibrozil and repaglinide has been described in 2003 by Niemi and coworkers [50]. It was shown that this interaction persists for at least 12 h after administration of gemfibrozil [93] suggesting that not only the inhibition of uptake transporters but also the inhibition of the drug metabolizing enzyme CYP2C8 might by important for this drug-drug interaction. Another study investigated the effects of atorvastatin on repaglinide pharmacokinetics in relation to SLCO1B1 polymorphism and it could be demonstrated that atorvastatin raises repaglinide plasma concentration, probably by inhibiting OATP1B1 [94]. Other OATP-related drug-drug interactions with oral antidiabetic drugs are summarized in Table 2. Taken together, these analyses revealed that OATPs are important molecular targets of transporter-mediated drug-drug interactions since their substrate spectrum includes a variety of frequently prescribed drugs often administered concomitantly with oral antidiabetic drugs.

3. Oral Antidiabetic Drugs and OCTs

OCTs are members of the SLC22 family expressed in several tissues including intestine, liver and kidney (Figure 1; [8]). Since a large number of clinically used drugs are administered orally, of which approximately 40% are cations or weak bases at physiological pH [119], transport proteins for cations are important determinants of drug pharmacokinetics. Several antidiabetic drugs have been identified as substrates or inhibitors of OCT proteins. These include metformin which is a substrate for OCT1 (Km value = 1 470 μM [82]), OCT2 (Km value = 990 μM [82]) and OCT3 (Km value = 2 260 μM [15]), whereas phenformin [120], repaglinide [121] and rosiglitazone [64] all interact with OCT1. Furthermore, genetic variations in the SLC22A1 gene encoding OCT1 were associated with altered pharmacokinetics and pharmacodynamics of metformin [122,123], which might pose a higher risk for side effects of metformin, especially lactic acidosis ([86], for review see [124]). Several OCT-mediated drug-drug interactions have been described using either metformin as a substrate for OCTs or other oral antidiabetic drugs inhibiting OCT-mediated transport. In vivo it has been shown that genetic variations in the SLC22A2 gene encoding human OCT2 are associated with alterations in metformin pharmacokinetics and that in the presence of simultaneously administered cimetidine the renal clearance of metformin was decreased [108,109]. This uptake inhibition could be confirmed in vitro using HEK and MDCK cells overexpressing OCT1 [28] and OCT2 [110,111]. For OCT1, the IC50 value for cimetidine-induced inhibition of metformin uptake was 150 μM [28], for OCT2 a Ki value of 147 μM could be detected for the same drug combination [111]. Other drugs inhibiting OCT-mediated metformin uptake are the BCR-ABL inhibitors imatinib and nilotinib [112]. Imatinib inhibited the uptake with IC50 values of 1.5 μM, 5.8 μM and 4.4 μM for OCT1, OCT2 and OCT3, respectively whereas nilotinib inhibited the same transporters with IC50 values of 2.9 μM (OCT1), >30 μM (OCT2) and 0.3 μM (OCT3). Furthermore, in the same study the inhibition of OCT-mediated metformin uptake by the EGFR (epidermal growth factor receptor) inhibitor gefitinib was studied demonstrating an inhibition of OCT1, OCT2 and OCT3 with IC50 values of 1.1 μM, 24.4 μM and 5.5 μM, respectively.

Since the uptake of drugs from blood into the renal tubular cells is a key determinant for renal secretion, inhibition of OCT2-mediated drug transport may influence systemic plasma concentration. For beta-blockers this uptake inhibition has been demonstrated in vitro using MDCK cells recombinantly overexpressing OCT2. In this study, Bachmakov and coworkers showed that OCT2-mediated metformin uptake was significantly inhibited by the beta-blockers bisoprolol, carvedilol, metoprolol and propranolol with IC50 values of 2.4 μM, 2.3 μM, 50.2 μM and 8.3 μM, respectively [106].

Interestingly, the uptake and the effect of one oral antidiabetic drug can also be inhibited by a second antidiabetic drug. This has been demonstrated in vitro analyzing OCT1- and OCT2-mediated metformin uptake and uptake inhibition by the DPP-4 inhibitor sitagliptin [87]. In this study the authors showed that sitagliptin inhibited OCT1- and OCT2-mediated metformin uptake with IC50 values of 34.9 μM and 40.8 μM, respectively. Furthermore, the inhibition of metformin-induced activation of AMPK (5′ adenosine monophosphate-activated protein kinase) signaling was investigated demonstrating that treatment with sitagliptin in MDCK-OCT1 and HepG2 cells resulted in a reduced level of phosphorylated AMPK with Ki values of 38.8 μM and 43.4 μM, respectively.

Summarizing these data (in combination with data presented in Table 2) one can assume that, in addition to OATP-mediated interactions, the inhibition of OCT proteins may be an important determinant of oral antidiabetic drug pharmacokinetics.

4. Oral Antidiabetic Drugs and Export Proteins

Most export proteins that mediate the export of drugs and drug metabolites out of cells belong to the superfamily of ABC transporters. P-glycoprotein (P-gp) is the best characterized efflux transporter localized in the apical membrane of enterocytes, the bile canalicular membrane and proximal tubule cells of the renal epithelium (Figure 1) mediating the export of a variety of different drugs belonging to various drug classes [35]. In diabetes therapy, several in vivo and in vitro studies have shown that P-gp is also involved in drug-drug interactions. It has been demonstrated in vivo that the macrolide clarithromycin increased the Cmax and the AUC (area under the plasma concentration time curve) of coadministered glibenclamide [92] and thus could lead to hypoglycaemia due to elevated plasma concentrations of glibenclamide [60]. Further P-gp-mediated drug-drug interactions could be observed in several in vitro studies. Using P-gp overexpressing cells it could be demonstrated that glibenclamide inhibited P-gp-mediated efflux of the model substrate rhodamine 123 [57]. Inhibition of glibenclamide export resulting in increased intracellular accumulation was also detected in the presence of colchicine [56]. Another study conducted by Hemauer and coworkers showed that rosiglitazone and metformin were transported by P-gp using placental inside-out oriented brush border membrane vesicles. Additionally, it was reported that inhibition of P-gp by verapamil markedly decreased the transport of rosiglitazone and metformin [72].

A second well-studied export pump involved in the transport of oral antidiabetic drugs is the breast cancer resistance protein BCRP (gene symbol ABCG2). This transporter is localized in the apical membrane of enterocytes and renal epithelial cells as well as in the canalicular membrane of hepatocytes (Figure 1). Several in vitro studies have indicated that oral antidiabetic drugs are substrates for BCRP and that coadministration of other drugs may inhibit this BCRP-mediated transport. Like for P-gp, Hemauer et al. provided evidence that BCRP participates in the transport of metformin. They could show that the BCRP inhibitor KO143 lead to a decreased metformin transport [72]. In addition, several studies using cells stably expressing BCRP revealed that glibenclamide export is inhibited by prototypic inhibitors, such as the antibiotic novobiocin or fumitremorgin C [57,97]. Pollex et al. further investigated the functional consequences of a frequent genetic variation in the ABCG2 gene encoding human BCRP. They found that the variation BCRPp.Q141K (ABCG2c.412C>A) showed a higher Michaelis Menten constant (Km value) and a higher maximal transport rate (Vmax value) for BCRP-mediated glibenclamide transport as compared to the transport mediated by the wild-type BCRP protein [97].

Interesting and recently characterized transporters responsible for the export of substances and drugs out of cells are members of the MATE family (multidrug and toxin extrusion protein; gene symbol SLC47). As known so far, this family consists of the two members MATE1 [125] and MATE2 (also designated as MATE2-K [45]). MATE1 is expressed in several tissues including liver and kidney, whereas expression of MATE2 seems to be restricted to renal epithelial cells (Figure 1; [44]). Unlike ABC transporters, these export proteins use an inward-directed proton gradient for exporting substrates out of cells. Several endogenous substances and drugs have been identified as substrates of these transport proteins [126] with metformin as the best studied oral antidiabetic drug transported by MATE1 and MATE2 [46]. The importance of MATE1 for the pharmacokinetics of metformin has been investigated in Mate1 (−/−) knockout mice [127]. After a single intravenous administration of metformin (5 mg/kg), a two-fold increase in the AUC of metformin in Mate1 (−/−) mice as compared to wild-type mice was detected. Furthermore, the renal clearance of metformin was only 18% of that measured in Mate1 (+/+) mice clearly demonstrating the essential role of MATE1 in the systemic clearance of metformin.

Inhibition studies using single-transfected HEK cells recombinantly expressing MATE1 or MATE2 showed that some tyrosine kinase inhibitors (TKIs) are potent inhibitors of MATE-mediated transport [112]. In detail, imatinib, nilotinib, gefitinib and erlotinib inhibited MATE1- and MATE2-mediated metformin transport with unbound Cmax/IC50 values higher than 0.1, suggesting that this transporter-mediated drug-drug interaction with TKIs may also be relevant in vivo and therefore affect the disposition, efficacy and toxicity of metformin and possibly of other drugs that are substrates for these export proteins. A further analysis of drug interaction studies has been performed by zu Schwabedissen and coworkers [105]. They demonstrated a significant inhibition of MATE1-mediated metformin transport by using single-transfected HeLa cells expressing human MATE1 and a panel of 24 different drugs. Most potent inhibitors (all tested at a concentration of 25 μM) were cimetidine with 21.08 ± 0.81%, trimethoprim with 25.35 ± 0.49% and ritonavir with 25.75 ± 9.91% residual metformin transport. Furthermore, the IC50 values of MATE1-mediated metformin transport were determined for ritonavir (15.4 ± 2.5 μM), ranitidine (18.9 ± 7.3 μM), rapamycin (3.27 ± 0.46 μM) and mitoxantrone (4.4 ± 1.3 μM). All of these IC50 values are below the reported peak plasma concentrations, suggesting that these interactions may also play a role in vivo [105]. Another interaction of MATE1-mediated metformin uptake has been analyzed in HEK cells stably expressing MATE1 and the frequently prescribed antibiotic trimethoprim as well as the antimalarial drug chloroquine. The experiments revealed a strong inhibition for both substances with an IC50 value of 6.2 μM calculated for trimethoprim and a Ki value of 2.8 μM for chloroquine [107].

Some studies also investigated the effect of drugs on OCT- and MATE-mediated transport using double-transfected cells recombinantly expressing an uptake transporter of the OCT family together with MATE1 or MATE2 [128,129]. In order to examine the effect of cimetidine on transcellular metformin transport, an OCT2-MATE1 double-transfected MDCK cell line was used [111]. These experiments demonstrated that the transcellular metformin transport was moderately inhibited by 1 μM cimetidine and almost completely inhibited by 1 mM cimetidine. Interestingly, the intracellular accumulation of metformin was inhibited only by 1 mM cimetidine but was increased by the low concentration of 1 μM cimetidine under the same experimental conditions. These results suggest that low cimetidine concentrations had no inhibitory effect on OCT2-mediated uptake whereas MATE1 was inhibited by intracellular cimetidine resulting in an increased intracellular metformin concentration. Taken together, these studies demonstrated that beside the inhibition of uptake transporters also the inhibition of export proteins may be important and clinically relevant for drug-drug interactions with oral antidiabetic drugs. Furthermore, experimental approaches using double-transfected cell lines simultaneously expressing an uptake transporter and an export protein indicated a crucial interaction of uptake transporters and export pumps.

Figure 1. Uptake and export transporters involved in the intestinal absorption (enterocyte) and the hepatic (hepatocyte) and renal (renal epithelium) excretion of oral antidiabetic drugs. Uptake transporters (red): OATP = members of the organic anion transporting polypeptide family; OCT = members of the organic cation transporters; export transporters (blue): MRP2 = multidrug resistance protein 2; BSEP = bile salt export pump; BCRP = breast cancer resistance protein; P-gp = P-glycoprotein; MATE = members of the multidrug and toxin extrusion protein family.
Figure 1. Uptake and export transporters involved in the intestinal absorption (enterocyte) and the hepatic (hepatocyte) and renal (renal epithelium) excretion of oral antidiabetic drugs. Uptake transporters (red): OATP = members of the organic anion transporting polypeptide family; OCT = members of the organic cation transporters; export transporters (blue): MRP2 = multidrug resistance protein 2; BSEP = bile salt export pump; BCRP = breast cancer resistance protein; P-gp = P-glycoprotein; MATE = members of the multidrug and toxin extrusion protein family.
Pharmaceutics 03 00680f1 1024
Figure 2. Inhibition of OATP1B1- (top), OATP1B3- (middle) and OATP2B1-mediated (bottom) BSP uptake by the oral antidiabetic drugs rosiglitazone (left) and repaglinide (right). Data are shown as the percentage of BSP uptake (0.05 μM BSP for OATP1B1- and 1 μM BSP for OATP1B3- and OATP2B1-mediated transport; modified from [64]) in the absence of the respective drug.
Figure 2. Inhibition of OATP1B1- (top), OATP1B3- (middle) and OATP2B1-mediated (bottom) BSP uptake by the oral antidiabetic drugs rosiglitazone (left) and repaglinide (right). Data are shown as the percentage of BSP uptake (0.05 μM BSP for OATP1B1- and 1 μM BSP for OATP1B3- and OATP2B1-mediated transport; modified from [64]) in the absence of the respective drug.
Pharmaceutics 03 00680f2 1024
Figure 3. Inhibition of OATP1B1- (top) and OATP1B3-mediated (bottom) pravastatin uptake (50 μM) by the oral antidiabetic drugs repaglinide (Repa) and rosiglitazone (Rosi). Data are shown as the percentage of OATP1B1- or OATP1B3-mediated pravastatin uptake in the absence of the respective drug (without). *P < 0.05; **P < 0.01 vs. control. (modified from [64]).
Figure 3. Inhibition of OATP1B1- (top) and OATP1B3-mediated (bottom) pravastatin uptake (50 μM) by the oral antidiabetic drugs repaglinide (Repa) and rosiglitazone (Rosi). Data are shown as the percentage of OATP1B1- or OATP1B3-mediated pravastatin uptake in the absence of the respective drug (without). *P < 0.05; **P < 0.01 vs. control. (modified from [64]).
Pharmaceutics 03 00680f3 1024
Table 1. Characteristics of selected oral antidiabetic drugs.
Table 1. Characteristics of selected oral antidiabetic drugs.
Oral antidiabetic drugsCharacteristicsActionElimination#MetabolismTransporters+References

GlibenclamideSulfonylurea derivatives, insulin secretagoguesStimulation of insulin secretion from pancreatic β-cells by binding to ATP-dependent potassium channels
Side effects: weight gain, hypoglycaemia
Renal (50%)a
Biliary (50%)a
CYP2C9
CYP2C19
CYP3A4
OATP2B1
P-gp
BCRP
[53,54]
[55,56]
[55,57]
GlimepirideRenal (60%)b
Biliary (40%)b
CYP2C9-[58,59]
[60,61]*

RepaglinideMeglitinide derivatives, insulin secretagoguesStimulation of insulin secretion from pancreatic ß-cells by binding to ATP-dependent potassium channels Side effects: mild hypoglycaemiac, dRenal (<8%)c
Biliary (90%)c
CYP2C8
CYP3A4
OATP1B1
OATP1B3
[62,63]
[62,64]
NateglinideRenal (83%)d
Biliary (10%)d
CYP2C9
CYP3A4
-[65,66]
[65]
[67]*

RosiglitazoneTiazolidine-diones, insulin sensitizersPeroxisome proliferator-activated receptor γ agonists (PPAR) Side effects: fluid retention, increased incidence of heart failure and fracture risk; hepatotoxicity (troglitazone)Renal (65%)
Biliary (30%)
[68]
CYP2C8
CYP2C9
OATP1B1
OATP1B3
BCRP
P-gp
[64,70]
[58,64]
[71]
[72]
PioglitazoneRenal (45%)e
Biliary (55%)e
CYP2C8
CYP3A4
OATP1B1
OATP1B3
[73,74]
[58,74]
TroglitazoneRenal (3%)
Biliary (85%)
[69]
CYP3A4
CYP2C8
OATP1B1
OATP1B3
BCRP
P-gp
BSEP
[58,74]
[58,75]
[76]
[76]
[77]
[7881]*

MetforminBiguanideActivation of AMP-activated protein kinase (AMPK) and suppression of glucagon-stimulated glucose production, increase in glucose uptake in muscle and hepatic cells Side Effect: lactic acidosisBiliary (100%)f-OCT1-3
MATE1-2
[15,82]
[46,58]
[8386]*

SitagliptinDipeptidyl peptidase-4 inhibitor (DDP-4)Prolongation of Glucagon-like peptide 1 (GLP-1) action by inhibition of DPP-4 Side effects: nausea, hypoglycaemiagRenal (87%)g
Biliary (13%)g
-OCT1-2
OAT3
P-gp
[87]
[88]
[88]
[89]*

*References refer to general characteristics of the respective oral antidiabetic drug(s);+Oral antidiabetic drug is substrate and/or inhibitor of the respective transporter;#according to the Summary of Product Characteristics—SPC (Fachinformation) if not stated otherwise;aGlibenclamide (Euglucon) product information, Sanofi-Aventis GmbH, Frankfurt a. M., Germany, 2008bGlimepiride (Amaryl) product information, Sanofi-Aventis GmbH, Frankfurt a. M., Germany, 2006cRepaglinide (NovoNorm)product information, Novo Nordisk Pharma AG, Küsnacht, Switzerland, 2010;dNateglinide (Starlix) product information, Novartis Pharma GmbH, Nürnberg, Germany, 2009;ePioglitazone (Actos) product information, Takeda Pharma, Aachen, Germany, 2010;fMetformin (Glucophage) product information, Merck Pharma GmbH, Darmstadt, Germany, 2006;gSitagliptin (Januvia) product information, MSD Shark & Dohme GmbH, Haar, Germany, 2010.

Table 2. Selected human solute carriers (SLC) and ATP-binding cassette (ABC) transporters involved in interactions with oral antidiabetic drugs.
Table 2. Selected human solute carriers (SLC) and ATP-binding cassette (ABC) transporters involved in interactions with oral antidiabetic drugs.
Oral antidiabetic drugInhibitor/victim compoundIn vivo/in vitroEffectCYPTransportersReference(s)
Glibenclamide5-CFDAin vitroInhibition of 5-CFDA efflux-MRP1-3[57]
8-FcAin vitroInhibition of 8-FcA uptake (IC50 = 1.1 and 2.7 μM)-OATP1B1, OATP1B3[95]
Atorvastatinin vitroInhibition of atorvastatin uptake-OATP2B1[13]
Atorvastatinin vitroInhibition of glibenclamide uptake-OATP2B1[13]
Calceinin vitroInhibition of calcein efflux-MRP1[96]
Cerivastatinin vitroInhibition of glibenclamide uptake-OATP2B1[13]
Clarithromycinin vivoCmax and AUC of glibenclamide ↑CYP3A4P-gp[92]
Colchicinein vitroIncreased intracellular accumulation of colchicine-P-gp[56]
Estrone-3-sulfatein vitroInhibition of E3S uptake-OATP2B1[13]
Estrone-3-sulfatein vitroInhibition of glibenclamide uptake-OATP2B1[13]
Fumitremorgin Cin vitroIncreased intracellular accumulation of glibenclamide-BCRP[97]
GCDCin vitroInhibition of GCDC transport (IC50 = 7.6 μM)-BSEP[77]
Ginkgo leaf extractin vitroInhibition of glibenclamide uptake (IC50 = 15.4 μg/mL)-OATP2B1[98]
Glycocholatein vitroInhibition of glycocholate transport (IC50 = 18.8 μM)-BSEP[77]
Grapefruit juicein vitroInhibition of glibenclamide uptake-OATP2B1[54]
Grapefruit juicein vivoNo effect on glibenclamide pharmacokinetics--[92]
Green tea extractin vitroInhibition of glibenclamide uptake (IC50 = 24.6 μg/mL)-OATP2B1[98]
Indomethacinin vitroDecreased glibenclamide uptake in IOVs-MRP1[72]
Indomethacinin vitroIncreased intracellular accumulation of glibenclamide-MRP3[57]
KO143in vitroDecreased glibenclamide uptake in IOVs-BCRP[72]
Nicardipineex vivo aIncrease in mean fetal-to-maternal concentration ratio-BCRP[99]
Novobiocinin vitroIncreased intracellular accumulation of glibenclamide-BCRP[57,100]
Orange juicein vitroInhibition of glibenclamide uptake-OATP2B1[54]
Pitavastatinin vitroInhibition of pitavastatin uptake-OATP1B1[101]
Rhodamine 123in vitroInhibition of rhodamine 123 efflux-P-gp[57]
Rifampinin vivoAUC of glibenclamide ↑ 2.3-fold-OATP1B1[102]
Simvastatinin vitroInhibition of glibenclamide uptake-OATP2B1[13]
Taurocholatein vitroInhibition of taurocholate uptake-NTCP[103]
Taurocholatein vitroIncreased intracellular accumulation of taurocholate-BSEP[103]
Taurocholatein vitroInhibition of taurocholate transport (Ki = 27.5 μM)-BSEP[104]
Taurocholatein vitroInhibition of taurocholate transport (IC50 = 14.7 μM)-BSEP[77]
TCDCin vitroInhibition of TCDC transport (IC50 = 15.8 μM)-BSEP[77]
Verapamilin vitroDecreased glibenclamide uptake in IOVs-P-gp[72]
GlimepirideGemfibrozilin vivoAUC of Glimepiride ↑ 23%CYP2C9-[59]
MetforminAmprenavirin vitroInhibition of metformin uptake-OCT2, MATE1[105]
Bisoprololin vitroInhibition of metformin uptake (IC50 = 2.4 μM)-OCT2[106]
Carvidololin vitroInhibition of metformin uptake (IC50 = 2.3 μM)-OCT2[106]
Chloroquinein vitroInhibition of metformin uptake (Ki = 2.8 μM)-MATE1[107]
Cimetidinein vivoReduction of renal clearance-OCT2[108,109]
Cimetidinein vitroInhibition of metformin uptake (IC50 = 11 μM)-OCT2[110]
Cimetidinein vitroInhibition of metformin uptake (Ki = 147 μM)-OCT2[111]
Cimetidinein vitroInhibition of metformin uptake (IC50 = 158 μM)-OCT1[28]
Cimetidinein vitroInhibition of metformin uptake-OCT2, MATE1[105]
Cimetidinein vitroInhibition of metformin uptake (Ki = 1.1 μM)-MATE1[111]
Dipyridamolein vitroInhibition of metformin uptake-OCT2, MATE1[105]
Erlotinibin vitroInhibition of metformin uptake
IC50 (μM) = 0.4 (OCT1), 5.2 (OCT2), 4.2 (OCT3)-OCT1-3
IC50 (μM) = 7.9 (MATE1), 3.5 (MATE2)-MATE1-2[112]
Fenfluraminein vitroInhibition of metformin uptake-OCT2[113]
Gefitinibin vitroInhibition of metformin uptake
IC50 (μM) = 1.1 (OCT1), 24.4 (OCT2), 5.5 (OCT3)-OCT1-3
IC50 (μM) = 1.8 (MATE1), 0.2 (MATE2)-MATE1-2[112]
Imatinibin vitroInhibition of metformin uptake
IC50 (μM) = 1.5 (OCT1), 5.8 (OCT2), 4.4 (OCT3)-OCT1-3
IC50 (μM) = 0.05 (MATE1), 0.5 (MATE2)-MATE1-2[112]
Ketoconazolein vitroInhibition of metformin uptake-OCT2, MATE1[105]
KO143in vitroDecreased metformin uptake in IOVs-BCRP[72]
Metoprololin vitroInhibition of metformin uptake (IC50 = 50.2 μM)-OCT2[106]
Mexiletinein vitroInhibition of metformin uptake-OCT2[113]
MPP+in vitroInhibition of MPP+ uptake (IC50 = 3.4 mM)-OCT1[15]
MPP+in vitroInhibiton of MPP+ uptake (IC50 = 397 μM)-OCT2[110]
MPP+in vitroInhibition of MPP+ uptake (IC50 = 2.9 mM)-OCT3[15]
Nilotinibin vitroInhibition of metformin uptake
IC50 (μM) = 2.9 (OCT1), >30 (OCT2), 0.3 (OCT3)-OCT1-3
IC50 (μM) = 3.4 (MATE1), 1.8 (MATE2)-MATE1-2[112]
Probenicidein vitroInhibition of metformin uptake-OCT2, MATE1[105]
Propranololin vitroInhibition of metformin uptake (IC50 = 8.3 μM)-OCT2[106]
Pyrimethaminein vitroInhibition of metformin uptake-OCT2, MATE1[105]
Quinidinein vitroInhibition of metformin uptake (IC50 = 55 μM)-OCT1[114]
Ranitidinein vitroInhibition of metformin uptake-OCT2, MATE1[105]
Rapamycinin vitroInhibition of metformin uptake-MATE1[105]
Sitagliptinin vitroInhibition of metformin uptake (Ki = 34.9 μM, 40.8 μM)-OCT1, OCT2[87]
Trimethoprimin vitroInhibition of metformin uptake-OCT2, MATE1[105]
Verapamilin vitroDecreased metformin uptake in IOVs-P-gp[72]
PioglitazoneEstrone-3-sulfatein vitroInhibition of E3S uptake-OATP1B1, OATP1B3[74]
RepaglinideAtorvastatinin vivoAUC of Repaglinide ↑ 1.2-fold-OATP1B1[94]
Clarithromycinin vivoAUC of Repaglinide ↑ 40%CYP3A4OATP1B1[49]
Cyclosporin Ain vivoAUC of Repaglinide ↑ 2.4-foldCYP3A4OATP1B1[52]
FMTXIn vitroInhibition of FMTX uptake (IC50 = 1.1 μM, 4.8 μM)-OATP1B1, OATP1B3[115]
Gemfibrozilin vivoAUC of Repaglinide ↑ 8.1-foldCYP2C8OATP1B1[50,93]
Pravastatinin vitroInhibition of pravastatin uptake-OATP1B1, OATP1B3[64]
Rifampinin vivoAUC of Repaglinide ↓60%CYP3A4-[116]
Telithromycinin vivoAUC of Repaglinide ↑ 77%CYP3A4OATP1B1[51]
RosiglitazoneCalceinin vitroInhibition of calcein efflux-P-gp[76]
Estrone-3-sulfatein vitroInhibition of E3S uptake-OATP1B1[74]
Metforminin vitroInhibition of rosiglitazone uptake in IOVs (IC50 = 0.6 μM)-P-gp[72]
PhAin vitroInhibition of PhA efflux-BCRP[76]
PhAin vitroInhibition of PhA efflux (IC50 = 25 μM)-BCRP[71]
Pravastatinin vitroStimulationb and inhibition of pravastatin uptake-OATP1B1, OATP1B3[64]
Verapamilin vitroDecreased rosiglitazone uptake in IOVs-P-gp[72]
SitagliptinCimetidinein vitroInhibition of cimetidine uptake (IC50 = 160 μM)-OAT3[88]
Cimetidinein vitroInhibition of sitagliptin uptake (IC50 = 79 μM)-OAT3[88]
Cyclosporin Ain vitroInhibition of sitagliptin uptake (IC50 = 1 μM)-P-gp[88]
Fenofibric acidin vitroInhibition of sitagliptin uptake (IC50 = 2.2 μM)-OAT3[88]
Furosemidein vitroInhibition of sitagliptin uptake (IC50 = 1.7 μM)-OAT3[88]
Ibuprofenin vitroInhibition of sitagliptin uptake (IC50 = 3.7 μM)-OAT3[88]
Indapamidein vitroInhibition of sitagliptin uptake (IC50 = 11.2 μM)-OAT3[88]
MPP+in vitroInhibition of MPP+ uptake (Ki = 34.4 μM, 44.7 μM)-OCT1, OCT2[87]
Probenicidin vitroInhibition of sitagliptin uptake (IC50 = 5.6 μM)-OAT3[88]
Quinaprilin vitroInhibition of sitagliptin uptake (IC50 = 6.2 μM)-OAT3[88]
Salycylatein vitroInhibition of salycylate uptake-OAT3[87]
TroglitazoneCalceinin vitroInhibition of calcein efflux-P-gp[76]
Estradiol-17ß-glucoronidein vitroInhibition of E-17ß-G uptake (Ki = 1 μM)-OATP1B1[117]
Estradiol-17ß-glucoronidein vitroInhibition of E-17ß-G uptake (IC50= 1.2 μM, 15.7 μM)-OATP1B1, OATP1B3[75]
Estrone-3-sulfatein vitroInhibition of E3S uptake-OATP1B1[74]
GCDCin vitroInhibition of GCDC transport (IC50 = 24.4 μM)-BSEP[77]
Glycocholatein vitroInhibition of glycocholate transport (IC50 = 16 μM)-BSEP[77]
PhAin vitroInhibition of PhA efflux-BCRP[76]
Taurocholatein vitroInhibition of taurocholate transport (IC50 = 9.5 μM)-BSEP[77]
Taurocholatein vitroInhibition of taurocholate uptake-NTCP[118]
Taurocholatein vitroDecreased biliary excretion of taurocholate-BSEP[118]
TCDCin vitroInhibition of TCDC transport (IC50 = 27.6 μM)-BSEP[77]
Troglitazone SulfateEstrone-3-sulfatein vitroInhibition of E3S uptake-OATP1B1, OATP1B3[74]

aDual perfusion system of isolated human placental lobules;bStimulation at low rosiglitazone concentration (10 μM); Abbreviations: 5-CFDA, 5-carboxy fluorescein diacetate; 8-FcA, 8-fluorescein-cAMP; AUC, area under the concentration time curve; BCRP, breast cancer resistance protein; BSEP, bile salt export pump; cmax, maximum peak concentration in plasma; E3S, estrone-3-sulfate; E17βG, estradiol-17β-glucoronide; FMTX, fluorescein-methotrexate; GC, glycocholate; GCDC, glycochenodeoxycholate; IOV, inside-out placental brush border membrane vesicles; KO143, BCRP-selective inhibitor (Pyrazino[1′,2′:1,6]pyrido[3,4-b]indole-3-propanoicacid, 1,2,3,4,6,7,12,12a-octahydro-9-methoxy-6-(2-methylpropyl)-1,4-dioxo-,1,1-dimethylethyl ester, (3S,6S,12aS)-); MATE, multidrug and toxin extrusion protein; MPP+, 1-methyl-4-phenylpyridinium; MRP, multidrug resistance protein; NTCP, sodium-taurocholate cotransporting polypeptide; OATP, organic anion transporting polypeptide; OCT, organic cation transporter; PhA, pheophorbide A; P-gp, P-glycoprotein; TCDC, taurochenodeoxycholate.

Reference

  1. Hediger, M.A.; Romero, M.F.; Peng, J.B.; Rolfs, A.; Takanaga, H.; Bruford, E.A. The ABCs of Solute Carriers: Physiological, Pathological and Therapeutic Implications of Human Membrane Transport Proteins. Pflugers Arch. 2004, 447, 465–468. [Google Scholar]
  2. SLC. Available online: www.bioparadigms.org/SLC (accessed on 11 July 2011).
  3. Hagenbuch, B.; Meier, P.J. Organic Anion Transporting Polypeptides of the OATP/SLC21 Family: Phylogenetic Classification as OATP/ SLCO Superfamily, New Nomenclature and Molecular/Functional Properties. Pflugers Arch. 2004, 447, 653–665. [Google Scholar]
  4. König, J. Uptake Transporters of the Human OATP Family: Molecular Characteristics, Substrates, Their Role in Drug-Drug Interactions, and Functional Consequences of Polymorphisms. In Handbook of Experimental Pharmacology; Springer Verlag: Berlin, Germany, 2011; pp. 1–28. [Google Scholar]
  5. Seithel, A.; Glaeser, H.; Fromm, M.F.; König, J. The Functional Consequences of Genetic Variations in Transporter Genes Encoding Human Organic Anion-Transporting Polypeptide Family Members. Expert Opin. Drug Metab. Toxicol. 2008, 4, 51–64. [Google Scholar]
  6. Kindla, J.; Fromm, M.F.; König, J. In Vitro Evidence for the Role of OATP and OCT Uptake Transporters in Drug-Drug Interactions. Expert Opin. Drug Metab. Toxicol. 2009, 5, 489–500. [Google Scholar]
  7. Fahrmayr, C.; Fromm, M.F.; König, J. Hepatic OATP and OCT Uptake Transporters: Their Role for Drug-Drug Interactions and Pharmacogenetic Aspects. Drug Metab. Rev. 2010, 42, 380–401. [Google Scholar]
  8. Koepsell, H.; Endou, H. The SLC22 Drug Transporter Family. Pflugers Arch. 2004, 447, 666–676. [Google Scholar]
  9. Hsiang, B.; Zhu, Y.; Wang, Z.; Wu, Y.; Sasseville, V.; Yang, W.P.; Kirchgessner, T.G. A Novel Human Hepatic Organic Anion Transporting Polypeptide (OATP2). Identification of a Liver-Specific Human Organic Anion Transporting Polypeptide and Identification of Rat and Human Hydroxymethylglutaryl-CoA Reductase Inhibitor Transporters. J. Biol. Chem. 1999, 274, 37161–37168. [Google Scholar]
  10. König, J.; Seithel, A.; Gradhand, U.; Fromm, M.F. Pharmacogenomics of Human OATP Transporters. Naunyn-Schmied. Arch. Pharmacol. 2006, 372, 432–443. [Google Scholar]
  11. König, J.; Cui, Y.; Nies, A.T.; Keppler, D. A Novel Human Organic Anion Transporting Polypeptide Localized to the Basolateral Hepatocyte Membrane. Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 278, G156–G164. [Google Scholar]
  12. Kobayashi, D.; Nozawa, T.; Imai, K.; Nezu, J.; Tsuji, A.; Tamai, I. Involvement of Human Organic Anion Transporting Polypeptide OATP-B (SLC21A9) in pH-Dependent Transport across Intestinal Apical Membrane. J. Pharmacol. Exp. Ther. 2003, 306, 703–708. [Google Scholar]
  13. Grube, M.; Köck, K.; Oswald, S.; Draber, K.; Meissner, K.; Eckel, L.; Bohm, M.; Felix, S.B.; Vogelgesang, S.; Jedlitschky, G.; Siegmund, W.; Warzok, R.; Kroemer, H.K. Organic Anion Transporting Polypeptide 2B1 Is a High-Affinity Transporter for Atorvastatin and Is Expressed in the Human Heart. Clin. Pharmacol. Ther. 2006, 80, 607–620. [Google Scholar]
  14. Koepsell, H.; Lips, K.; Volk, C. Polyspecific Organic Cation Transporters: Structure, Function, Physiological Roles, and Biopharmaceutical Implications. Pharm. Res. 2007, 24, 1227–1251. [Google Scholar]
  15. Nies, A.T.; Koepsell, H.; Winter, S.; Burk, O.; Klein, K.; Kerb, R.; Zanger, U.M.; Keppler, D.; Schwab, M.; Schaeffeler, E. Expression of Organic Cation Transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) Is Affected by Genetic Factors and Cholestasis in Human Liver. Hepatology 2009, 50, 1227–1240. [Google Scholar]
  16. Gorboulev, V.; Ulzheimer, J.C.; Akhoundova, A.; Ulzheimer-Teuber, I.; Karbach, U.; Quester, S.; Baumann, C.; Lang, F.; Busch, A.E.; Koepsell, H. Cloning and Characterization of Two Human Polyspecific Organic Cation Transporters. DNA Cell Biol. 1997, 16, 871–881. [Google Scholar]
  17. Hagenbuch, B.; Meier, P.J. Molecular Cloning, Chromosomal Localization, and Functional Characterization of a Human Liver Na+/Bile Acid Cotransporter. J. Clin. Invest. 1994, 93, 1326–1331. [Google Scholar]
  18. Jonker, J.W.; Schinkel, A.H. Pharmacological and Physiological Functions of the Polyspecific Organic Cation Transporters: OCT1, 2, and 3 (SLC22A1-3). J. Pharmacol. Exp. Ther. 2004, 308, 2–9. [Google Scholar]
  19. Niessen, J.; Jedlitschky, G.; Grube, M.; Bien, S.; Schwertz, H.; Ohtsuki, S.; Kawakami, H.; Kamiie, J.; Oswald, S.; Starke, K.; Strobel, U.; Siegmund, W.; Rosskopf, D.; Greinacher, A.; Terasaki, T.; Kroemer, H.K. Human Platelets Express Organic Anion-Transporting Peptide 2B1, an Uptake Transporter for Atorvastatin. Drug Metab. Dispos. 2009, 37, 1129–1137. [Google Scholar]
  20. Kameyama, Y.; Yamashita, K.; Kobayashi, K.; Hosokawa, M.; Chiba, K. Functional Characterization of SLCO1B1 (OATP-C) Variants, SLCO1B1*5, SLCO1B1*15 and SLCO1B1*15+C1007G, by Using Transient Expression Systems of HeLa and HEK293 Cells. Pharmacogenet. Genomics 2005, 15, 513–522. [Google Scholar]
  21. Schwarz, U.I.; Meyer zu Schwabedissen, H.E.; Tirona, R.G.; Suzuki, A.; Leake, B.F.; Mokrab, Y.; Mizuguchi, K.; Ho, R.H.; Kim, R.B. Identification of Novel Functional Organic Anion-Transporting Polypeptide 1B3 Polymorphisms and Assessment of Substrate Specificity. Pharmacogenet. Genomics 2011, 21, 103–114. [Google Scholar]
  22. Tamai, I.; Nezu, J.; Uchino, H.; Sai, Y.; Oku, A.; Shimane, M.; Tsuji, A. Molecular Identification and Characterization of Novel Members of the Human Organic Anion Transporter (OATP) Family. Biochem. Biophys. Res. Commun. 2000, 273, 251–260. [Google Scholar]
  23. Koepsell, H.; Schmitt, B.M.; Gorboulev, V. Organic Cation Transporters. Rev. Physiol. Biochem. Pharmacol. 2003, 150, 36–90. [Google Scholar]
  24. Cha, S.H.; Sekine, T.; Fukushima, J.I.; Kanai, Y.; Kobayashi, Y.; Goya, T.; Endou, H. Identification and Characterization of Human Organic Anion Transporter 3 Expressing Predominantly in the Kidney. Mol. Pharmacol. 2001, 59, 1277–1286. [Google Scholar]
  25. Cropp, C.D.; Komori, T.; Shima, J.E.; Urban, T.J.; Yee, S.W.; More, S.S.; Giacomini, K.M. Organic Anion Transporter 2 (SLC22A7) Is a Facilitative Transporter of cGMP. Mol. Pharmacol. 2008, 73, 1151–1158. [Google Scholar]
  26. Ciarimboli, G.; Ludwig, T.; Lang, D.; Pavenstadt, H.; Koepsell, H.; Piechota, H.J.; Haier, J.; Jaehde, U.; Zisowsky, J.; Schlatter, E. Cisplatin Nephrotoxicity Is Critically Mediated Via the Human Organic Cation Transporter 2. Am. J. Pathol. 2005, 167, 1477–1484. [Google Scholar]
  27. Yonezawa, A.; Masuda, S.; Yokoo, S.; Katsura, T.; Inui, K. Cisplatin and Oxaliplatin, but Not Carboplatin and Nedaplatin, Are Substrates for Human Organic Cation Transporters (SLC22A1-3 and Multidrug and Toxin Extrusion Family). J. Pharmacol. Exp. Ther. 2006, 319, 879–886. [Google Scholar]
  28. Umehara, K.I.; Iwatsubo, T.; Noguchi, K.; Kamimura, H. Comparison of the Kinetic Characteristics of Inhibitory Effects Exerted by Biguanides and H2-Blockers on Human and Rat Organic Cation Transporter-Mediated Transport: Insight into the Development of Drug Candidates. Xenobiotica 2007, 37, 618–634. [Google Scholar]
  29. Ho, R.H.; Tirona, R.G.; Leake, B.F.; Glaeser, H.; Lee, W.; Lemke, C.J.; Wang, Y.; Kim, R.B. Drug and Bile Acid Transporters in Rosuvastatin Hepatic Uptake: Function, Expression, and Pharmacogenetics. Gastroenterology 2006, 130, 1793–1806. [Google Scholar]
  30. Borst, P.; Evers, R.; Kool, M.; Wijnholds, J. A Family of Drug Transporters: The Multidrug Resistance-Associated Proteins. J. Natl. Cancer Inst. 2000, 92, 1295–1302. [Google Scholar]
  31. Deeley, R.G.; Westlake, C.; Cole, S.P. Transmembrane Transport of Endo- and Xenobiotics by Mammalian ATP-Binding Cassette Multidrug Resistance Proteins. Physiol. Rev. 2006, 86, 849–899. [Google Scholar]
  32. Keppler, D.; Kamisako, T.; Leier, I.; Cui, Y.; Nies, A.T.; Tsujii, H.; König, J. Localization, Substrate Specificity, and Drug Resistance Conferred by Conjugate Export Pumps of the MRP Family. Adv. Enzyme Regul. 2000, 40, 339–349. [Google Scholar]
  33. Dean, M.; Rzhetsky, A.; Allikmets, R. The Human ATP-Binding Cassette (ABC) Transporter Superfamily. Genome Res. 2001, 11, 1156–1166. [Google Scholar]
  34. ABC1 (subfamily A). Availabe online: www.nutrigene.4t.com/humanABC.Htm. (accessed on 11 July 2011).
  35. Fromm, M.F. Importance of P-Glycoprotein for Drug Disposition in Humans. Eur. J. Clin. Invest 2003, 33, 6–9. [Google Scholar]
  36. Cascorbi, I. P-Glycoprotein: Tissue Distribution, Substrates, and Functional Consequences of Genetic Variations. In Handbook of Experimental Pharmacology; Springer Verlag: Berlin, Germany, 2011; pp. 261–283. [Google Scholar]
  37. Maliepaard, M.; Scheffer, G.L.; Faneyte, I.F.; van Gastelen, M.A.; Pijnenborg, A.C.; Schinkel, A.H.; van De Vijver, M.J.; Scheper, R.J.; Schellens, J.H. Subcellular Localization and Distribution of the Breast Cancer Resistance Protein Transporter in Normal Human Tissues. Cancer Res. 2001, 61, 3458–3464. [Google Scholar]
  38. Huls, M.; Brown, C.D.; Windass, A.S.; Sayer, R.; van den Heuvel, J.J.; Heemskerk, S.; Russel, F.G.; Masereeuw, R. The Breast Cancer Resistance Protein Transporter ABCG2 Is Expressed in the Human Kidney Proximal Tubule Apical Membrane. Kidney Int. 2008, 73, 220–225. [Google Scholar]
  39. Stieger, B.; Meier, Y.; Meier, P.J. The Bile Salt Export Pump. Pflugers Arch. 2007, 453, 611–620. [Google Scholar]
  40. Hoffmann, U.; Kroemer, H.K. The ABC Transporters MDR1 and MRP2: Multiple Functions in Disposition of Xenobiotics and Drug Resistance. Drug Metab. Rev. 2004, 36, 669–701. [Google Scholar]
  41. Kivistö, K.T.; Niemi, M.; Fromm, M.F. Functional Interaction of Intestinal CYP3A4 and P-Glycoprotein. Fundam. Clin. Pharmacol. 2004, 18, 621–626. [Google Scholar]
  42. Fromm, M.F. P-Glycoprotein: A Defense Mechanism Limiting Oral Bioavailability and CNS Accumulation of Drugs. Int. J. Clin. Pharmacol. Ther. 2000, 38, 69–74. [Google Scholar]
  43. Schwabedissen, H.E.; Kroemer, H.K. In Vitro and in Vivo Evidence for the Importance of Breast Cancer Resistance Protein Transporters (BCRP/MXR/ABCP/ABCG2). In Handbook of Experimental Pharmacology; Springer Verlag: Berlin, Germany, 2011; pp. 325–371. [Google Scholar]
  44. Otsuka, M.; Matsumoto, T.; Morimoto, R.; Arioka, S.; Omote, H.; Moriyama, Y. A Human Transporter Protein That Mediates the Final Excretion Step for Toxic Organic Cations. Proc. Natl. Acad. Sci. USA 2005, 102, 17923–17928. [Google Scholar]
  45. Masuda, S.; Terada, T.; Yonezawa, A.; Tanihara, Y.; Kishimoto, K.; Katsura, T.; Ogawa, O.; Inui, K. Identification and Functional Characterization of a New Human Kidney-Specific H+/Organic Cation Antiporter, Kidney-Specific Multidrug and Toxin Extrusion 2. J. Am. Soc. Nephrol. 2006, 17, 2127–2135. [Google Scholar]
  46. Tanihara, Y.; Masuda, S.; Sato, T.; Katsura, T.; Ogawa, O.; Inui, K. Substrate Specificity of MATE1 and MATE2-K, Human Multidrug and Toxin Extrusions/H+-Organic Cation Antiporters. Biochem. Pharmacol. 2007, 74, 359–371. [Google Scholar]
  47. Hagenbuch, B.; Meier, P.J. The Superfamily of Organic Anion Transporting Polypeptides. Biochim. Biophys. Acta 2003, 1609, 1–18. [Google Scholar]
  48. Tamai, I. Oral Drug Delivery Utilizing Intestinal OATP Transporters. Adv. Drug Deliv. Rev. 2011. In Press. [Google Scholar]
  49. Niemi, M. The Cytochrome P4503A4 Inhibitor Clarithromycin Increases the Plasma Concentrations and Effects of Repaglinide. Clin. Pharmacol. Ther. 2001, 70, 58–65. [Google Scholar]
  50. Niemi, M.; Backman, J.T.; Neuvonen, M.; Neuvonen, P.J. Effects of Gemfibrozil, Itraconazole, and Their Combination on the Pharmacokinetics and Pharmacodynamics of Repaglinide: Potentially Hazardous Interaction between Gemfibrozil and Repaglinide. Diabetologia 2003, 46, 347–351. [Google Scholar]
  51. Kajosaari, L.; Niemi, M.; Backman, J.; Neuvonen, P. Telithromycin, but Not Montelukast, Increases the Plasma Concentrations and Effects of the Cytochrome P450 3A4 and 2C8 Substrate Repaglinide. Clin. Pharmacol. Ther. 2006, 79, 231–242. [Google Scholar]
  52. Kajosaari, L.; Niemi, M.; Neuvonen, M.; Laitila, J.; Neuvonen, P.; Backman, J. Cyclosporine Markedly Raises the Plasma Concentrations of Repaglinide. Clin. Pharmacol. Ther. 2005, 78, 388–399. [Google Scholar]
  53. Zhou, L.; Naraharisetti, S.B.; Liu, L.; Wang, H.; Lin, Y.S.; Isoherranen, N.; Unadkat, J.D.; Hebert, M.F.; Mao, Q. Contributions of Human Cytochrome P450 Enzymes to Glyburide Metabolism. Biopharm. Drug Dispos. 2010, 31, 228–242. [Google Scholar]
  54. Satoh, H. Citrus Juices Inhibit the Function of Human Organic Anion-Transporting Polypeptide OATP-B. Drug Metab. Dispos. 2005, 33, 518–523. [Google Scholar]
  55. Naritomi, Y.; Terashita, S.; Kagayama, A. Identification and Relative Contributions of Human Cytochrome P450 Isoforms Involved in the Metabolism of Glibenclamide and Lansoprazole: Evaluation of an Approach Based on the in Vitro Substrate Disappearance Rate. Xenobiotica 2004, 34, 415–427. [Google Scholar]
  56. Golstein, P.E.; Boom, A.; van Geffel, J.; Jacobs, P.; Masereel, B.; Beauwens, R. P-Glycoprotein Inhibition by Glibenclamide and Related Compounds. Pflugers Arch. 1999, 437, 652–660. [Google Scholar]
  57. Gedeon, C.; Behravan, J.; Koren, G.; Piquette-Miller, M. Transport of Glyburide by Placental ABC Transporters: Implications in Fetal Drug Exposure. Placenta 2006, 27, 1096–1102. [Google Scholar]
  58. Kirchheiner, J.; Roots, I.; Goldammer, M.; Rosenkranz, B.; Brockmöller, J. Effect of Genetic Polymorphisms in Cytochrome P450 (CYP) 2C9 and CYP2C8 on the Pharmacokinetics of Oral Antidiabetic Drugs: Clinical Relevance. Clin. Pharmacokinet. 2005, 44, 1209–1225. [Google Scholar]
  59. Niemi, M.; Neuvonen, P.J.; Kivistö, K.T. Effect of Gemfibrozil on the Pharmacokinetics and Pharmacodynamics of Glimepiride. Clin. Pharmacol. Ther. 2001, 70, 439–445. [Google Scholar]
  60. Holstein, A.; Egberts, E.H. Risk of Hypoglycaemia with Oral Antidiabetic Agents in Patients with Type 2 Diabetes. Exp. Clin. Endocrinol. Diabetes 2003, 111, 405–414. [Google Scholar]
  61. Proks, P.; Reimann, F.; Green, N.; Gribble, F.; Ashcroft, F. Sulfonylurea Stimulation of Insulin Secretion. Diabetes 2002, 51, S368–S376. [Google Scholar]
  62. Bidstrup, T.B.; Bjornsdottir, I.; Sidelmann, U.G.; Thomsen, M.S.; Hansen, K.T. CYP2C8 and CYP3A4 Are the Principal Enzymes Involved in the Human in Vitro Biotransformation of the Insulin Secretagogue Repaglinide. Br. J. Clin. Pharmacol. 2003, 56, 305–314. [Google Scholar]
  63. Niemi, M.; Backman, J.T.; Kajosaari, L.I.; Leathart, J.B.; Neuvonen, M.; Daly, A.K.; Eichelbaum, M.; Kivistö, K.T.; Neuvonen, P.J. Polymorphic Organic Anion Transporting Polypeptide 1B1 Is a Major Determinant of Repaglinide Pharmacokinetics. Clin. Pharmacol. Ther. 2005, 77, 468–478. [Google Scholar]
  64. Bachmakov, I.; Glaeser, H.; Fromm, M.F.; König, J. Interaction of Oral Antidiabetic Drugs with Hepatic Uptake Transporters: Focus on Organic Anion Transporting Polypeptides and Organic Cation Transporter 1. Diabetes 2008, 57, 1463–1469. [Google Scholar]
  65. Niemi, M.; Backman, J.T.; Juntti-Patinen, L.; Neuvonen, M.; Neuvonen, P.J. Coadministration of Gemfibrozil and Itraconazole Has Only a Minor Effect on the Pharmacokinetics of the CYP2C9 and CYP3A4 Substrate Nateglinide. Br. J. Clin. Pharmacol. 2005, 60, 208–217. [Google Scholar]
  66. Kalliokoski, A.; Backman, J.T.; Neuvonen, P.J.; Niemi, M. Effects of the SLCO1B1*1B Haplotype on the Pharmacokinetics and Pharmacodynamics of Repaglinide and Nateglinide. Pharmacogenet. Genomics 2008, 18, 937–942. [Google Scholar]
  67. Scheen, A.J. Drug-Drug and Food-Drug Pharmacokinetic Interactions with New Insulinotropic Agents Repaglinide and Nateglinide. Clin. Pharmacokinet. 2007, 46, 93–108. [Google Scholar]
  68. Cox, P.J.; Ryan, D.A.; Hollis, F.J.; Harris, A.M.; Miller, A.K.; Vousden, M.; Cowley, H. Absorption, Disposition, and Metabolism of Rosiglitazone, a Potent Thiazolidinedione Insulin Sensitizer, in Humans. Drug Metab. Dispos. 2000, 28, 772–780. [Google Scholar]
  69. Loi, C.M.; Young, M.; Randinitis, E.; Vassos, A.; Koup, J.R. Clinical Pharmacokinetics of Troglitazone. Clin. Pharmacokinet. 1999, 37, 91–104. [Google Scholar]
  70. Kirchheiner, J.; Thomas, S.; Bauer, S.; Tomalikscharte, D.; Hering, U.; Doroshyenko, O.; Jetter, A.; Stehle, S.; Tsahuridu, M.; Meineke, I. Pharmacokinetics and Pharmacodynamics of Rosiglitazone in Relation to CYP2C8 Genotype. Clin. Pharmacol. Ther. 2006, 80, 657–667. [Google Scholar]
  71. Solbach, T.F.; Paulus, B.; Weyand, M.; Eschenhagen, T.; Zolk, O.; Fromm, M.F. ATP-Binding Cassette Transporters in Human Heart Failure. Naunyn-Schmied. Arch. Pharmacol. 2008, 377, 231–243. [Google Scholar]
  72. Hemauer, S.J.; Patrikeeva, S.L.; Nanovskaya, T.N.; Hankins, G.D.V.; Ahmed, M.S. Role of Human Placental Apical Membrane Transporters in the Efflux of Glyburide, Rosiglitazone and Metformin. Am. J. Obstet. Gynecol. 2010, 202. [Google Scholar]
  73. Tornio, A.; Niemi, M.; Neuvonen, P.J.; Backman, J.T. Trimethoprim and the CYP2C8*3 Allele Have Opposite Effects on the Pharmacokinetics of Pioglitazone. Drug Metab. Dispos. 2008, 36, 73–80. [Google Scholar]
  74. Nozawa, T.; Sugiura, S.; Nakajima, M.; Goto, A.; Yokoi, T.; Nezu, J.; Tsuji, A.; Tamai, I. Involvement of Organic Anion Transporting Polypeptides in the Transport of Troglitazone Sulfate: Implications for Understanding Troglitazone Hepatotoxicity. Drug Metab. Dispos. 2004, 32, 291–294. [Google Scholar]
  75. Gui, C.; Miao, Y.; Thompson, L.; Wahlgren, B.; Mock, M.; Stieger, B.; Hagenbuch, B. Effect of Pregnane X Receptor Ligands on Transport Mediated by Human OATP1B1 and OATP1B3. Eur. J. Pharmacol. 2008, 584, 57–65. [Google Scholar]
  76. Weiss, J.; Sauer, A.; Herzog, M.; Böger, R.H.; Haefeli, W.E.; Benndorf, R.A. Interaction of Thiazolidinediones (Glitazones) with the ATP-Binding Cassette Transporters P-Glycoprotein and Breast Cancer Resistance Protein. Pharmacology 2009, 84, 264–270. [Google Scholar]
  77. Kis, E.; Ioja, E.; Nagy, T.; Szente, L.; Heredi-Szabo, K.; Krajcsi, P. Effect of Membrane Cholesterol on BSEP/Bsep Activity: Species Specificity Studies for Substrates and Inhibitors. Drug Metab. Dispos. 2009, 37, 1878–1886. [Google Scholar]
  78. Martens, F.M.; Visseren, F.L.; Lemay, J.; de Koning, E.J.; Rabelink, T.J. Metabolic and Additional Vascular Effects of Thiazolidinediones. Drugs 2002, 62, 1463–1480. [Google Scholar]
  79. Giannini, S.; Serio, M.; Galli, A. Pleiotropic Effects of Thiazolidinediones: Taking a Look Beyond Antidiabetic Activity. J. Endocrinol. Invest. 2004, 27, 982–991. [Google Scholar]
  80. Kalaitzidis, R.G.; Sarafidis, P.A.; Bakris, G.L. Effects of Thiazolidinediones Beyond Glycaemic Control. Curr. Pharm. Des. 2009, 15, 529–536. [Google Scholar]
  81. Masubuchi, Y. Metabolic and Non-Metabolic Factors Determining Troglitazone Hepatotoxicity: A Review. Drug Metab. Pharmacokinet. 2006, 21, 347–356. [Google Scholar]
  82. Kimura, N.; Masuda, S.; Tanihara, Y.; Ueo, H.; Okuda, M.; Katsura, T.; Inui, K. Metformin Is a Superior Substrate for Renal Organic Cation Transporter OCT2 Rather Than Hepatic OCT1. Drug Metab. Pharmacokinet. 2005, 20, 379–386. [Google Scholar]
  83. Dunn, C.J.; Peters, D.H. Metformin. A Review of Its Pharmacological Properties and Therapeutic Use in Non-Insulin-Dependent Diabetes Mellitus. Drugs 1995, 49, 721–749. [Google Scholar]
  84. Abbud, W.; Habinowski, S.; Zhang, J.Z.; Kendrew, J.; Elkairi, F.S.; Kemp, B.E.; Witters, L.A.; Ismail-Beigi, F. Stimulation of AMP-Activated Protein Kinase (AMPK) Is Associated with Enhancement of Glut1-Mediated Glucose Transport. Arch. Biochem. Biophys. 2000, 380, 347–352. [Google Scholar]
  85. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; Musi, N.; Hirshman, M.F.; Goodyear, L.J.; Moller, D.E. Role of AMP-Activated Protein Kinase in Mechanism of Metformin Action. J. Clin. Invest. 2001, 108, 1167–1174. [Google Scholar]
  86. Kruse, J.A. Metformin-Associated Lactic Acidosis. J. Emerg. Med. 2001, 20, 267–272. [Google Scholar]
  87. Choi, M.K.; Jin, Q.R.; Ahn, S.H.; Bae, M.A.; Song, I.S. Sitagliptin Attenuates Metformin-Mediated AMPK Phosphorylation through Inhibition of Organic Cation Transporters. Xenobiotica 2010, 40, 817–825. [Google Scholar]
  88. Chu, X.Y.; Bleasby, K.; Yabut, J.; Cai, X.; Chan, G.H.; Hafey, M.J.; Xu, S.; Bergman, A.J.; Braun, M.P.; Dean, D.C.; Evers, R. Transport of the Dipeptidyl Peptidase-4 Inhibitor Sitagliptin by Human Organic Anion Transporter 3, Organic Anion Transporting Polypeptide 4C1, and Multidrug Resistance P-Glycoprotein. J. Pharmacol. Exp. Ther. 2007, 321, 673–683. [Google Scholar]
  89. Scheen, A.J. Pharmacokinetics of Dipeptidylpeptidase-4 Inhibitors. Diabetes Obes. Metab. 2010, 12, 648–658. [Google Scholar]
  90. Kalliokoski, A.; Neuvonen, M.; Neuvonen, P.J.; Niemi, M. No Significant Effect of SLCO1B1 Polymorphism on the Pharmacokinetics of Rosiglitazone and Pioglitazone. Br. J. Clin. Pharmacol. 2008, 65, 78–86. [Google Scholar]
  91. Kindla, J.; Müller, F.; Mieth, M.; Fromm, M.F.; König, J. Influence of Non-Steroidal Anti-Inflammatory Drugs on Organic Anion Transporting Polypeptide (OATP) 1B1- and OATP1B3-Mediated Drug Transport. Drug Metab. Dispos. 2011, 39, 1047–1053. [Google Scholar]
  92. Lilja, J.J.; Niemi, M.; Fredrikson, H.; Neuvonen, P.J. Effects of Clarithromycin and Grapefruit Juice on the Pharmacokinetics of Glibenclamide. Br. J. Clin. Pharmacol. 2007, 63, 732–740. [Google Scholar]
  93. Tornio, A.; Niemi, M.; Neuvonen, M.; Laitila, J.; Kalliokoski, A.; Neuvonen, P.J.; Backman, J.T. The Effect of Gemfibrozil on Repaglinide Pharmacokinetics Persists for at Least 12 h after the Dose: Evidence for Mechanism-Based Inhibition of CYP2C8 in Vivo. Clin. Pharmacol. Ther. 2008, 84, 403–411. [Google Scholar]
  94. Kalliokoski, A.; Backman, J.T.; Kurkinen, K.J.; Neuvonen, P.J.; Niemi, M. Effects of Gemfibrozil and Atorvastatin on the Pharmacokinetics of Repaglinide in Relation to SLCO1B1 Polymorphism. Clin. Pharmacol. Ther. 2008, 84, 488–496. [Google Scholar]
  95. Bednarczyk, D. Fluorescence-Based Assays for the Assessment of Drug Interaction with the Human Transporters OATP1B1 and OATP1B3. Anal. Biochem. 2010, 405, 50–58. [Google Scholar]
  96. Payen, L.; Delugin, L.; Courtois, A.; Trinquart, Y.; Guillouzo, A.; Fardel, O. The Sulphonylurea Glibenclamide Inhibits Multidrug Resistance Protein (MRP1) Activity in Human Lung Cancer Cells. Br. J. Pharmacol. 2001, 132, 778–784. [Google Scholar]
  97. Pollex, E.K.; Anger, G.; Hutson, J.; Koren, G.; Piquette-Miller, M. Breast Cancer Resistance Protein (BCRP)-Mediated Glyburide Transport: Effect of the C421A/Q141K BCRP Single-Nucleotide Polymorphism. Drug Metab. Dispos. 2010, 38, 740–744. [Google Scholar]
  98. Fuchikami, H. Effects of Herbal Extracts on the Function of Human Organic Anion-Transporting Polypeptide OATP-B. Drug Metab. Dispos. 2006, 34, 577–582. [Google Scholar]
  99. Pollex, E.; Lubetsky, A.; Koren, G. The Role of Placental Breast Cancer Resistance Protein in the Efflux of Glyburide across the Human Placenta. Placenta 2008, 29, 743–747. [Google Scholar]
  100. Gedeon, C.; Anger, G.; Piquette-Miller, M.; Koren, G. Breast Cancer Resistance Protein: Mediating the Trans-Placental Transfer of Glyburide across the Human Placenta. Placenta 2008, 29, 39–43. [Google Scholar]
  101. Hirano, M.; Maeda, K.; Shitara, Y.; Sugiyama, Y. Drug-Drug Interaction between Pitavastatin and Various Drugs Via OATP1B1. Drug Metab. Dispos. 2006, 34, 1229–1236. [Google Scholar]
  102. Zheng, H.X.; Huang, Y.; Frassetto, L.A.; Benet, L.Z. Elucidating Rifampin's Inducing and Inhibiting Effects on Glyburide Pharmacokinetics and Blood Glucose in Healthy Volunteers: Unmasking the Differential Effects of Enzyme Induction and Transporter Inhibition for a Drug and Its Primary Metabolite. Clin. Pharmacol. Ther. 2008, 85, 78–85. [Google Scholar]
  103. Mita, S. Inhibition of Bile Acid Transport across Na+/Taurocholate Cotransporting Polypeptide (SLC10A1) and Bile Salt Export Pump (ABCB11)-Coexpressing LLC-PK1 Cells by Cholestasis-Inducing Drugs. Drug Metab. Dispos. 2006, 34, 1575–1581. [Google Scholar]
  104. Byrne, J.A.; Strautnieks, S.S.; Mieli-Vergani, G.; Higgins, C.F.; Linton, K.J.; Thompson, R.J. The Human Bile Salt Export Pump: Characterization of Substrate Specificity and Identification of Inhibitors. Gastroenterology 2002, 123, 1649–1658. [Google Scholar]
  105. Meyer zu Schwabedissen, H.E.; Verstuyft, C.; Kroemer, H.K.; Becquemont, L.; Kim, R.B. Human Multidrug and Toxin Extrusion 1 (MATE1/SLC47A1) Transporter: Functional Characterization, Interaction with OCT2 (SLC22A2), and Single Nucleotide Polymorphisms. Am. J. Physiol. Renal. Physiol. 2010, 298, F997–F1005. [Google Scholar]
  106. Bachmakov, I.; Glaeser, H.; Endress, B.; Mörl, F.; König, J.; Fromm, M.F. Interaction of Beta-Blockers with the Renal Uptake Transporter OCT2. Diabetes Obes. Metab. 2009, 11, 1080–1083. [Google Scholar]
  107. Müller, F.; König, J.; Glaeser, H.; Schmidt, I.; Zolk, O.; Fromm, M.F.; Maas, R. Molecular Mechanism of Renal Tubular Secretion of the Antimalarial Drug Chloroquine. Antimicrob. Agents Chemother. 2011, 55, 3091–3098. [Google Scholar]
  108. Somogyi, A.; Stockley, C.; Keal, J.; Rolan, P.; Bochner, F. Reduction of Metformin Renal Tubular Secretion by Cimetidine in Man. Br. J. Clin. Pharmacol. 1987, 23, 545–551. [Google Scholar]
  109. Wang, Z.J.; Yin, O.Q.; Tomlinson, B.; Chow, M.S. OCT2 Polymorphisms and in-Vivo Renal Functional Consequence: Studies with Metformin and Cimetidine. Pharmacogenet. Genomics 2008, 18, 637–645. [Google Scholar]
  110. Zolk, O.; Solbach, T.F.; König, J.; Fromm, M.F. Functional Characterization of the Human Organic Cation Transporter 2 Variant p.270Ala>Ser. Drug Metab. Dispos. 2009, 37, 1312–1318. [Google Scholar]
  111. Tsuda, M.; Terada, T.; Ueba, M.; Sato, T.; Masuda, S.; Katsura, T.; Inui, K. Involvement of Human Multidrug and Toxin Extrusion 1 in the Drug Interaction between Cimetidine and Metformin in Renal Epithelial Cells. J. Pharmacol. Exp. Ther. 2009, 329, 185–191. [Google Scholar]
  112. Minematsu, T.; Giacomini, K.M. Interactions of Tyrosine Kinase Inhibitors with Organic Cation Transporters and Multidrug and Toxic Compound Extrusion Proteins. Mol. Cancer Ther. 2011, 10, 531–539. [Google Scholar]
  113. Zolk, O.; Solbach, T.F.; König, J.; Fromm, M.F. Structural Determinants of Inhibitor Interaction with the Human Organic Cation Transporter OCT2 (SLC22A2). Naunyn-Schmied. Arch. Pharmacol. 2008, 379, 337–348. [Google Scholar]
  114. Umehara, K.I.; Iwatsubo, T.; Noguchi, K.; Usui, T.; Kamimura, H. Effect of Cationic Drugs on the Transporting Activity of Human and Rat OCT/Oct 1-3 in Vitro and Implications for Drug-Drug Interactions. Xenobiotica 2008, 38, 1203–1218. [Google Scholar]
  115. Gui, C.; Obaidat, A.; Chaguturu, R.; Hagenbuch, B. Development of a Cell-Based High-Throughput Assay to Screen for Inhibitors of Organic Anion Transporting Polypeptides 1B1 and 1B3. Curr. Chem. Genomics 2010, 4, 1–8. [Google Scholar]
  116. Niemi, M.; Backman, J.T.; Neuvonen, M.; Neuvonen, P.J.; Kivistö, K.T. Rifampin Decreases the Plasma Concentrations and Effects of Repaglinide. Clin. Pharmacol. Ther. 2000, 68, 495–500. [Google Scholar]
  117. Gui, C.; Wahlgren, B.; Lushington, G.H.; Hagenbuch, B. Identification, Ki Determination and CoMFA Analysis of Nuclear Receptor Ligands as Competitive Inhibitors of OATP1B1-Mediated Estradiol-17beta-Glucuronide Transport. Pharmacol. Res. 2009, 60, 50–56. [Google Scholar]
  118. Marion, T.L.; Leslie, E.M.; Brouwer, K.L. Use of Sandwich-Cultured Hepatocytes to Evaluate Impaired Bile Acid Transport as a Mechanism of Drug-Induced Hepatotoxicity. Mol. Pharm. 2007, 4, 911–918. [Google Scholar]
  119. Neuhoff, S.; Ungell, A.L.; Zamora, I.; Artursson, P. Ph-Dependent Bidirectional Transport of Weakly Basic Drugs across Caco-2 Monolayers: Implications for Drug-Drug Interactions. Pharm. Res. 2003, 20, 1141–1148. [Google Scholar]
  120. Dresser, M.J.; Xiao, G.; Leabman, M.K.; Gray, A.T.; Giacomini, K.M. Interactions of N-Tetraalkylammonium Compounds and Biguanides with a Human Renal Organic Cation Transporter (hOCT2). Pharm. Res. 2002, 19, 1244–1247. [Google Scholar]
  121. Ahlin, G.; Karlsson, J.; Pedersen, J.M.; Gustavsson, L.; Larsson, R.; Matsson, P.; Norinder, U.; Bergstrom, C.A.; Artursson, P. Structural Requirements for Drug Inhibition of the Liver Specific Human Organic Cation Transport Protein 1. J. Med. Chem. 2008, 51, 5932–5942. [Google Scholar]
  122. Shu, Y.; Brown, C.; Castro, R.A.; Shi, R.J.; Lin, E.T.; Owen, R.P.; Sheardown, S.A.; Yue, L.; Burchard, E.G.; Brett, C.M.; Giacomini, K.M. Effect of Genetic Variation in the Organic Cation Transporter 1, OCT1, on Metformin Pharmacokinetics. Clin. Pharmacol. Ther. 2008, 83, 273–280. [Google Scholar]
  123. Shu, Y.; Sheardown, S.A.; Brown, C.; Owen, R.P.; Zhang, S.; Castro, R.A.; Ianculescu, A.G.; Yue, L.; Lo, J.C.; Burchard, E.G.; Brett, C.M.; Giacomini, K.M. Effect of Genetic Variation in the Organic Cation Transporter 1 (OCT1) on Metformin Action. J. Clin. Invest. 2007, 117, 1422–1431. [Google Scholar]
  124. Zolk, O. Disposition of Metformin: Variability Due to Polymorphisms of Organic Cation Transporters. Ann. Med. 2011. in press. [Google Scholar]
  125. Omote, H.; Hiasa, M.; Matsumoto, T.; Otsuka, M.; Moriyama, Y. The MATE Proteins as Fundamental Transporters of Metabolic and Xenobiotic Organic Cations. Trends Pharmacol. Sci. 2006, 27, 587–593. [Google Scholar]
  126. Nies, A.T.; Koepsell, H.; Damme, K.; Schwab, M. Organic Cation Transporters (OCTs, MATEs), in Vitro and in Vivo Evidence for the Importance in Drug Therapy. In Handbook of Experimental Pharmacology; Springer Verlag: Berlin, Germany, 2011; pp. 105–167. [Google Scholar]
  127. Tsuda, M.; Terada, T.; Mizuno, T.; Katsura, T.; Shimakura, J.; Inui, K. Targeted Disruption of the Multidrug and Toxin Extrusion 1 (Mate1) Gene in Mice Reduces Renal Secretion of Metformin. Mol. Pharmacol. 2009, 75, 1280–1286. [Google Scholar]
  128. Sato, T.; Masuda, S.; Yonezawa, A.; Tanihara, Y.; Katsura, T.; Inui, K. Transcellular Transport of Organic Cations in Double-Transfected MDCK Cells Expressing Human Organic Cation Transporters hOCT1/hMATE1 and hOCT2/hMATE1. Biochem. Pharmacol. 2008, 76, 894–903. [Google Scholar]
  129. König, J.; Zolk, O.; Singer, K.; Hoffmann, C.; Fromm, M.F. Double-Transfected MDCK Cells Expressing Human OCT1/MATE1 or OCT2/MATE1: Determinants of Uptake and Transcellular Translocation of Organic Cations. Br. J. Pharmacol. 2011, 163, 546–555. [Google Scholar]

Share and Cite

MDPI and ACS Style

Klatt, S.; Fromm, M.F.; König, J. Transporter-Mediated Drug–Drug Interactions with Oral Antidiabetic Drugs. Pharmaceutics 2011, 3, 680-705. https://doi.org/10.3390/pharmaceutics3040680

AMA Style

Klatt S, Fromm MF, König J. Transporter-Mediated Drug–Drug Interactions with Oral Antidiabetic Drugs. Pharmaceutics. 2011; 3(4):680-705. https://doi.org/10.3390/pharmaceutics3040680

Chicago/Turabian Style

Klatt, Sabine, Martin F. Fromm, and Jörg König. 2011. "Transporter-Mediated Drug–Drug Interactions with Oral Antidiabetic Drugs" Pharmaceutics 3, no. 4: 680-705. https://doi.org/10.3390/pharmaceutics3040680

Article Metrics

Back to TopTop