In Vitro Permeability & Drug Transporter Services

Cyprotex is a specialist provider of ADME and PK services and offer a range of in vitro permeability and uptake and efflux drug transporter assays. We offer both screening assays and full regulatory DDI and drug transporter packages according to FDA, European Medicines Agency (EMA), PMDA and ICH guidance.

In Vitro Permeability & Drug Transporters Services

Evotec, and wholly owned subsidiary Cyprotex, offer a range of in vitro  permeability and uptake and efflux drug transporter assays. We offer both screening assays and full regulatory DDI and drug transporter packages according to FDA, EMA, PMDA and ICH guidance. Data from these regulatory packages are used for IND and NDA submissions during preclinical development and clinical stage projects.

Discover More About Our In Vitro Permeability & Drug Transporter Services:

In Vitro Permeability Assays

The permeability of drugs is an important factor in oral absorption, BBB permeation and transport of drugs into tissues and across cell membranes. The permeability of a drug across a membrane is dependent on the passive permeability as well as the susceptibility of the drug to undergo active efflux or uptake by drug transporter proteins.

We offer a number of different models to study permeability including Caco-2 cells, MDCK-MDR1 cells and PAMPA as well as a panel of transporter assays which cover the key transporters recommended in the regulatory guidelines and those of emerging scientific interest. For more information on DDI studies, read our ADME guide or our DDI regulatory guidance booklet.

Drug Transporter Assays

Drug transporters exist in many tissues including, but not limited to, the intestinal epithelia, the hepatocytes and bile canaliculi, the kidney proximal tubules and the brain capillary endothelial cells.

The main transporters in these tissues are illustrated below.

Understanding whether your compound interacts with drug transporters (as a substrate or inhibitor) is an important stage in the drug development process. In vitro transporter interaction assays are used towards identifying if clinical drug-drug interaction studies are required.

The FDA guidance for industry (2020) and the EMA guideline on the investigation of drug interactions (adopted 2012) provides recommendations on the most clinically-relevant drug transporters for evaluation in the drug discovery and development process:

  • P-gp
  • BCRP
  • OATP1B1
  • OATP1B3
  • OAT1
  • OAT3
  • OCT2
  • MATE1
  • MATE2-K

In addition to the main transporters above, the EMA and the International Transporter Consortium3,4 also recommends preferably evaluating:

  • BSEP
  • OCT1

In addition to these broadly recommended transporter studies, there are a number of other potentially clinically relevant transporters which may be important for particular drug-discovery programs. These are discussed in the International Transporter Consortium (ITC) review whitepapers published in March 20105, July 20136 and November 20184. Read our review article: Studying the right transporter at the right time: an in vitro strategy for assessing drug-drug interaction risk during drug discovery and development.

We provide an extensive portfolio of drug transporter services and provides support to in vitro drug interaction studies including data required for regulatory submission.

ABC Transporter Interactions

P-glycoprotein

P-gp (P-glycoprotein) is one of the most well-recognized efflux transporters. It is expressed in many tissues, including the intestine, brain, and kidney. P-gp inhibition has been shown to be responsible for several clinical drug-drug interactions. For example, clarithromycin can inhibit the transport of the P-gp substrate digoxin, resulting in an elevation of plasma levels and a decrease in renal clearance7.

Recommendations for studying P-gp inhibition and substrate identification are outlined in the FDA Guidance – In Vitro Drug Interaction Studies – Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions Guidance for Industry (January 2020)1 and in the EMA Guideline on the Investigation of Drug Interactions (Adopted 2012)2. Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our P-glycoprotein substrate identification and our P-glycoprotein inhibition services.

BCRP

BCRP (Breast Cancer Resistance Protein) is an efflux transporter expressed in several tissues such as the gastrointestinal tract, liver, brain endothelium, mammary tissue, testis, and placenta. BCRP is known to play a role in clinical drug-drug interactions. In addition, clinically relevant genetic polymorphisms have been shown to impact on the PK (e.g., irinotecan8, rosuvastatin9, sulfasalasine10 and topotecan11) and toxicity (e.g., gefitinib-induced diarrhoea12) of marketed drugs. Recommendations for studying BCRP inhibition and substrate identification are outlined in the FDA Guidance for In Vitro Drug Interaction Studies (2020)1 and in the EMA Guideline on the Investigation of Drug Interactions (Adopted 2012)2. Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our BCRP substrate identification and our BCRP inhibition services.

BSEP

The Bile Salt Export Pump (BSEP) is a member of the ATP binding cassette family of transporters and is located on the canalicular membrane of hepatocytes. It is involved in transport of taurocholate and other cholate conjugates from hepatocytes to the bile and plays an important function in bile formation and bile flow13.

The EMA2 and ITC3 suggest preferably evaluating the potential of new chemical entities to inhibit the BSEP transporter.

Learn more about our BSEP inhibition service.

MRP

MRP2 (multidrug resistance associated protein 2; ABCC2), MRP3 (ABCC3) and MRP4 (ABCC4) are ATP binding cassette (ABC) efflux transporters which are located on the brush border membrane of enterocytes (MRP2), the canalicular membrane (MRP2) or sinusoidal membrane (MRP3, MRP4) of hepatocytes, the brush border membrane of renal proximal tubule epithelial cells (MRP2, MRP4) and at the blood-brain barrier (MRP4)4. Consequently, these efflux transporters influence the absorption, distribution, metabolism and excretion of endobiotics (e.g. bile acids), drugs/and or metabolites within the body.

Learn more about our MRP substrate identification and our MRP inhibition services.

SLC Transporter Interactions

OATP1B1

Organic Anion Transporting Polypeptide 1B1 (OATP1B1) is expressed on the sinusoidal membrane of hepatocytes where it is responsible for the uptake of several marketed drugs including some statins14. There is evidence for clinical drug-drug interactions involving some statin drugs and cyclosporine which appear to be mediated, at least in part, by inhibition of OATP1B115,16.

The FDA guidance1 and the EMA guidelines2 recommend investigating for potential OATP1B1 substrates and inhibitors due to the role of OATP1B1 in drug-drug interactions and the impact of genetic polymorphism of this transporter on therapy outcome and toxicity. According to the most recent regulatory guidance, it is only necessary to evaluate potential OATP1B1 substrates when hepatic clearance of the investigational drug is significant (e.g. hepatic elimination (hepatic or biliary clearance) is more than or equal to 25% of the total clearance). We follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our OATP1B1 substrate identification and our OATP1B1 inhibition services.

OATP1B3

Organic Anion Transporting Polypeptide 1B3 (OATP1B3) is an uptake transporter expressed on the sinusoidal membrane of hepatocytes. It has a substrate specificity that overlaps somewhat with OATP1B117. Because of the prominent expression of these transporters on the sinusoidal membrane of hepatocytes, they represent a critical mechanism for uptake of drugs into the liver.

The FDA guidance1 and the EMA guidelines2 recommend the evaluation of new chemical entities for their potential to act as substrates or inhibitors of OATP1B3 in vitro. It is only necessary to evaluate potential OATP1B3 substrates when hepatic clearance of the investigational drug is significant (e.g., hepatic elimination (hepatic or biliary clearance) is more than or equal to 25% of the total clearance). Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our OATP1B3 substrate identification and our OATP1B3 inhibition services.

OAT1

Organic Anion Transporter 1 (OAT1) is part of the SLC superfamily (SLC22A6). It is a transmembrane protein expressed predominantly at the basolateral membrane of proximal tubular cells of the kidneys and plays a central role in renal organic anion transport18. OAT1 is involved in the uptake of a wide range of relatively small and hydrophilic organic anions from plasma into the cytoplasm of the proximal tubular cells of the kidneys for subsequent exit across the apical membrane for excretion via the urine19. It has an essential role in the disposition of NSAIDs, antiviral drugs, diuretics, antitumour drugs and β-lactam antibiotics18.

The FDA guidance1 and the EMA guidelines2 recommend investigating for potential OAT1 substrates and inhibitors due to the role of OAT1 in drug-drug interactions. It is only necessary to evaluate potential OAT1 substrates when renal active secretion of the investigational drug is significant (e.g., active secretion by the kidney is more than or equal to 25% of total clearance). Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our OAT1 substrate identification and our OAT1 inhibition services.

OAT3

Organic Anion Transporter 3 (OAT3) is part of the SLC superfamily (SLC22A8). Like OAT1, it is primarily expressed at the basolateral membrane of proximal tubular cells of the kidneys, facilitating its role is in the renal transport of organic anions18.

OAT3 exhibits a broader substrate specificity than OAT1, and accepts amphipathic and hydrophilic organic anions and some organic cations18. Drugs which are renally cleared and are actively secreted by OATs may be susceptible to increases in AUC as a result of OAT3 inhibition. Examples of these clinically relevant interactions include the interaction of probenecid with acyclovir, resulting in a 32% decline in renal clearance, a 40% increase in AUC, and an 18% increase in the terminal plasma half-life of acyclovir following probenecid administration20.

The FDA guidance1 and the EMA guidelines2 recommend investigating for potential OAT3 substrates and inhibitors due to the role of OAT3 in drug-drug interactions. It is only necessary to evaluate potential OAT3 substrates when renal active secretion of the investigational drug is significant (e.g., active secretion by the kidney is more than or equal to 25% of total clearance). Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our OAT3 substrate identification and our OAT3 inhibition services.

OCT2

Organic Cation Transporter 2 (OCT2) is a member of the SLC family of transporters (SLC22A2). This transporter is expressed on the cells of the kidney proximal tubules where it is involved in the renal clearance of drug substrates21. Drug-drug interactions involving OCT2 may result in decreased renal clearance of the victim drug and a corresponding increase in exposure (e.g. dolutegravir interaction with metformin46).

The FDA guidance1 and the EMA guidelines2 recommend investigating for potential OCT2 substrates and inhibitors due to the role of OCT2 in drug-drug interactions. It is only necessary to evaluate potential OCT2 substrates when renal active secretion of the investigational drug is significant (e.g., active secretion by the kidney is more than or equal to 25% of total clearance). Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our OCT2 substrate identification and our OCT2 inhibition services.

MATE1

Multidrug and toxin extrusion 1 (MATE1; SLC47A1) transporter is a key transporter for the secretion of organic cations. MATE1 can either act as an uptake or efflux transporter, depending on an oppositely directed proton gradient as the driving force. Therefore, extracellular alkalinisation or intracellular acidification increases MATE1-mediated uptake in vitro, whereas extracellular acidification increases efflux by MATE123. Although MATE1 has been detected in several tissues, the main organ of expression appears to be the kidney. It is specifically located in the brush border (apical) membrane of the proximal and distal convoluted tubules24. Single nucleotide polymorphisms (SNPs) in the SLC47A1 gene have been implicated in altered metformin disposition in humans25.

Drug-drug interactions involving MATE1 may result in decreased renal clearance of the victim drug and a corresponding increase in exposure (e.g., cimetidine interaction with metformin22).

The FDA guidance1 and the EMA guidelines2 recommend investigating for potential MATE1 substrates and inhibitors due to the role of MATE1 in drug-drug interactions. It is only necessary to evaluate potential MATE1 substrates when renal active secretion of the investigational drug is significant (e.g., active secretion by the kidney is more than or equal to 25% of total clearance). Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our MATE1 substrate identification and our MATE1 inhibition services.

MATE2-K

Multidrug and toxin extrusion 2-K (MATE2-K, SLC47A2) transporter is a splicing variant of MATE2. MATE2-K is an H+/organic cation antiporter which is located exclusively in the kidney on the brush border membrane of proximal tubules26. It plays an important role in extruding organic cations into the kidney27. Substrates of MATE2-K include metformin, cimetidine, TEA and procainamide, and there is considerable overlap in terms of substrate specificity with the MATE1 transporter28.

Drug-drug interactions involving MATE2-K may result in decreased renal clearance of the victim drug and a corresponding increase in exposure (e.g., cimetidine interaction with metformin22).

The FDA guidance1 and the EMA guidelines2 recommend investigating for potential MATE2-K substrates and inhibitors due to the role of MATE2-K in drug-drug interactions. It is only necessary to evaluate potential MATE2-K substrates when renal active secretion of the investigational drug is significant (e.g., active secretion by the kidney is more than or equal to 25% of total clearance). Cyprotex follow the decision trees for identifying if an in vivo drug-drug interaction study is required using in vitro protocols recommended by the regulatory authorities.

Learn more about our MATE2-K substrate identification and our MATE2-K inhibition services.

OCT1

Organic Cation Transporter 1 (OCT1) is a member of the SLC superfamily (SLC22A1) and is located on the basolateral membrane of hepatocytes and enterocytes29. It mediates facilitated transport of small (hydrophilic) organic cations30. The OCTs have been implicated in several clinically relevant drug interactions4.

The EMA2 and ITC4 suggest that the investigation of new chemical entities for their ability to inhibit or be substrates of OCT1 in vitro could be considered.

Learn more about our OCT1 substrate identification and our OCT1 inhibition services.

OATP1A2

Organic Anion Transporting Polypeptide 1A2 (OATP1A2) is a member of the SLC transporter family (SLCO1A2). Highest expression of OATP1A2 mRNA is observed in the brain, but it is also present in the liver, intestine, kidneys, lung and testes. OATP1A2 has a broad range of substrates including endogenous compounds such as bile acids, steroid hormones and their conjugates and thyroid hormones as well as various drugs such as fexofenadine, ouabain and methoxtrexate. The transport does exhibit pH dependency with methotrexate increasing 7 fold at pH 5.0 compared with pH 7.431.

OATP1A2 is localized on the brush border membrane of enterocytes in the duodenum where it is thought to be involved in absorption and in the cholangiocytes of the liver where it is thought to be involved in reabsorption of xenobiotics excreted into the bile. It also plays a role in the renal transport and the blood brain barrier transport of xenobiotics. The transporter is thought to be important in breast cancer where mRNA expression in breast cancer tissue has been found to be almost 10 fold higher than in healthy tissue, and it is believed that OATP1A2 may enhance hormone dependent breast cancer proliferation by facilitating estrone 3-sulfate uptake in the cells31.

Learn more about our OATP1A2 substrate identification and our OATP1A2 inhibition services.

OATP2B1

Organic Anion Transporting Polypeptide 2B1 (OATP2B1) is a member of the SLC transporter family (SLCO2B1). As well as being localized in the sinusoidal membrane of the liver where it is involved in uptake of drugs or endogenous substances and in the intestine where it plays a role in uptake and absorption, OATP2B1 is also expressed in the brain, lung, spleen, kidney, heart, placenta and ovaries32. OATP2B1 transport appears to be pH-dependent with an increased activity at acidic pH such as that observed in the weakly acidic intestine32. The transporter is also expressed in a number of solid tumours. As the solid tumour microenvironment is acidic, it is thought OATP2B1 could also play a role in drug delivery to tumour cells33. Substrates of OATP2B1 include steroid conjugates, thyroid hormone and numerous drugs including statins32,34.

Learn more about our OATP2B1 substrate identification and our OATP2B1 inhibition services.

OAT2

Organic Anion Transporter 2 (OAT2) is a member of the SLC transporter family (SLC22A7). Expression of OAT2 is primarily expressed in the basolateral membrane of renal proximal tubule cells and is involved in active renal secretion of drugs or endogenous molecules35. OAT2 is also expressed in the liver and several other tissues in the body36. Substrates for OAT2 include acetyl salicylate, prostaglandin E2, dicarboxylates, glutamates, PAH as well as some anitvirals36. Of particular note is the transport of the endogenous substrate, cGMP, and it is thought therefore that OAT2 may be involved in modulation of intracellular signalling.

Learn more about our OAT2 substrate identification and our OAT2 inhibition services.

OAT4

OAT4 is a member of the SLC transporter family (SLC22A11). It is localized primarily in the syncytiotrophoblast cells of the placenta where it plays a role in regulating transport of hormones, drugs and toxins across the maternal-fetal barrier, and also in the apical membrane of renal proximal tubule cells where it is important for the reabsorption of organic anions, including sulfate conjugates36,37. Substrates of OAT4 include sulfated steroids, NSAIDs, anti-hypertensives, prostaglandins and uric acid36.

Learn more about our OAT4 substrate identification and our OAT4 inhibition services.

OCTN2

The organic cation / carnitine transporter 2 (OCTN2) is a member of the SLC transporter family (SLC22A5). It is expressed in the brush border membrane of the proximal renal tubule cells where it acts as a high affinity Na+/carnitine co-transporter and is responsible for the reabsorption of L-carnitine and acetyl carnitine. These endogenous molecules are important in the transport of long chain fatty acids into mitochondria and subsequent energy production by β-oxidation38. OCTN2 is also expressed in skeletal muscle, placenta, heart, pancreas, liver, lung, intestine and brain tissue39. Transport can be bidirectional, with the direction dependent on the substrate. Other known substrates of OCTN2 include verapamil, cephaloridine and oxaliplatin39. Polymorphisms in OCTN2 have been associated with the clinical progression of intestinal inflammation in conditions such as Crohn's disease39.

Learn more about our OCTN2 substrate identification and our OCTN2 inhibition services.

PEPT1

Peptide transporter 1 (PEPT1) is a member of the SLC family of transporters (SLC15A1). It is a low affinity high capacity transporter which is proton dependent. PEPT1 is localised on the brush border membrane of the intestine and to a lesser extent on renal epithelial cells. It is also known to be present in certain cancer cells. The primary role of PEPT1 is the uptake of dipeptides, tripeptides or free amino acids in the small intestine following dietary protein digestion within the gastrointestinal tract40. Certain β-lactam antibiotics and anticancer agents are also substrates for PEPT1 in the intestine. Uptake by PEPT1 in the intestine is also a popular route for prodrug design for drug delivery. Prodrugs which have amino acids as promoieties have been designed to be substrates of PEPT1 to improve oral absorption and bioavailability. This has been successful in the case of anti-virals such as valacyclovir and valganciclovir41. There is also evidence of PEPT1 playing a role in the pathogenesis of intestinal inflammation42.

Learn more about our PEPT1 substrate identification and our PEPT1 inhibition services.

PEPT2

Peptide transporter 2 (PEPT2) is a member of the SLC transporter family (SLC15A2). It is a high affinity low capacity transporter. Like PEPT1, it is involved in the proton-coupled transport of dipeptides, tripeptides, amino acids and peptide-like drugs. PEPT2 is widely distributed in the body and is expressed in the kidney, central nervous system and lung as well as several other tissues43.

Learn more about our PEPT2 substrate identification and our PEPT2 inhibition services.

NTCP

The sodium/taurocholate cotransporting polypeptide (NTCP) is a member of the SLC transporter family (SLC10A1). NTCP is present on the sinusoidal membrane of hepatocytes where it is responsible for transporting bile salts into hepatocytes as part of the enterohepatic recirculation process. This is a process whereby bile is secreted by the hepatocytes, stored in the gall bladder and secreted into the intestine, from which many of its constituents are reabsorbed and recirculate via the portal vein back to the liver for re-uptake by NTCP. Although bile salts are the main substrate for NTCP, other substrates include estrone 3-sulfate, bromosulfophthalein, dehydroepiandrosterone sulfate and thyroid hormones44. It has also been reported that NTCP contributes significantly to active hepatocyte uptake of statins45.

Learn more about our NTCP substrate identification and our NTCP inhibition services.

Preclinical species Oatp1b Interactions

Species differences in drug transporters with regard to their tissue distribution, expression levels and substrate specificity can be problematic for preclinical cross-species extrapolation of drug disposition (clearance) and DDI potential to human. The main hepatic uptake transporters expressed on the sinusoidal membrane of hepatocytes in preclinical species, which maybe rate-determining in the elimination of drugs, are rat Oatp1b2, dog Oatp1b4 and cynomolgus monkey Oatp1b1. Learn more about our Preclinical Species Oatp substrate identification and our Preclinical Species Oatp inhibition services.

References

1) FDA Guidance – In Vitro Drug Interaction Studies – Cytochrome P450 Enzyme- and Transporter-Mediated Drug Interactions Guidance for Industry (January 2020)
2) The European Medicines Agency (EMA) Guideline on the Investigation of Drug Interactions (Adopted 2012)
3) Kenna JG et al., (2018) Can bile salt export pump inhibition testing in drug discovery and development reduce liver injury risk? An International Transporter Consortium perspective. Clin Pharmacol Ther 104(5); 916-932
4) Zamek-Gliszczynski MJ et al., (2018) Transporters in drug development: 2018 ITC recommendations for transporters of emerging clinical importance. Clin Pharmacol Ther 104(5); 890-899
5) The International Transporter Consortium (2010) Membrane transporters in drug
development. Nat Rev Drug Disc 9; 215-236
6) Hillgren KM et al., (2013) Emerging transporters of clinical importance: an update from the International Transporter Consortium. Clin Pharmacol Ther 94(1); 52-63
7) Wakasugi H et al. (1998) Effect of clarithromycin on renal excretion of digoxin: Interaction with P-glycoprotein. Clin Pharmacol Ther 64; 123-128
8) Zhou Q et al. (2005) Pharmacogenetic profiling across the irinotecan pathway in Asian patients with cancer. Br J Clin Pharmacol 59; 415-424
9) Zhang W et al. (2006) Role of BCRP 421C>A polymorphism on rosuvastatin pharmacokinetics in healthy Chinese males. Clin Chim Acta 373; 99-103
10) Yamasaki Y et al. (2008) Pharmacogenetic characterization of sulfasalazine disposition based on NAT2 and ABCG2 (BCRP) gene polymorphisms in humans. Clin Pharmacol Ther 84(1); 95-103
11) Sparreboom A et al. (2005) Effect of ABCG2 genotype on the oral bioavailability of topotecan. Cancer Biol Ther 4; 650-658
12) Cusatis G et al. (2006) Pharmacogenetics of ABCG2 and adverse reactions to gefitinib. J Natl Cancer Inst 98; 1739–1742
13) Stieger B et al., (2007) The bile salt export pump. Pflügers Archiv Eur J Physiol 453; 611-620
14) Chen C et al. (2005) Differential interaction of 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors with ABCB1, ABCC2, and OATP1B1. Drug Metab Dispos 33; 537-546
15) Neuvonen PJ et al. (2006) Drug interactions with lipid-lowering drugs: Mechanisms and clinical relevance. Clin Pharmacol Ther 80; 565-581
16) Shitara Y et al, (2003) Inhibition of transporter-mediated hepatic uptake as a mechanism for drug-drug Interaction between cerivastatin and cyclosporin A. J Pharmacol Exp Ther 304; 610-616
17) Klaassen CD & Aleksunes LM (2010) Xenobiotic, bile acid, and cholesterol transporters: Function and regulation. Pharmacol Rev 62(1); 1-96
18) Nozaki Y et al., (2007) Characterization of the uptake of Organic Anion Transporter (OAT) 1 and OAT3 substrates by human kidney slices. J Pharmacol Exp Ther 321; 362-369
19) El-Sheikh AAK et al., (2008) Mechanisms of renal anionic drug transport. Eur J Pharmacol 585; 245-255
20) Laskin OL et al., (1982) Effects of probenecid on the pharmacokinetics and elimination of acyclovir in humans. Antimicrob Agents Chemother 21(5); 804-7
21) Aoki M et al., (2008) Kidney-specific expression of human organic cation transporter 2 (OCT2/SLC22A2) is regulated by DNA methylation. Am J Physiol Renal Physiol 295; F165-F170
22) Somogyi A et al. (1987) Reduction of metformin renal tubular secretion by cimetidine in man. Br J Clin Pharmacol 23; 545-551
23) Müller F and Fromm MF (2011) Transporter-mediated drug–drug interactions. Pharmacogenomics 12(7); 1017-1037
24) Meyer zu Schwabedissen HE et al, (2010) Human multidrug and toxin extrusion 1 (MATE1/SLC47A1) transporter: functional characterization, interaction with OCT2 (SLC22A2), and single nucleotide polymorphisms. Am J Physiol Renal Physiol 298; F997-F1005
25) Becker ML et al, (2010) Interaction between polymorphisms in the OCT1 and MATE1 transporter and metformin response. Pharmacogenet Genomics 20(1); 38-44
26) Masuda S et al, (2006) 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 17; 2127-2135
27) Komatsu T et al, (2011) Characterization of the human MATE2 proton-coupled polyspecific organic cation exporter. Int J Biochem Cell Biol 43(6); 913-918
28) Tanihara Y et al, (2007) Substrate specificity of MATE1 and MATE2-K, human multidrug and toxin extrusions/H+-organic cation antiporters. Biochem Pharmacol 74(2); 359-371
29) Jonker JW and Schinkel AH (2004) Pharmacological and physiological functions of the polyspecific Organic Cation Transporters: OCT1, 2, and 3 (SLC22A1-3). J Pharmacol Exp Ther 308(1); 2-9
30) Jonker JW et al, (2001) Reduced hepatic uptake and intestinal excretion of organic cations in mice with a targeted disruption of the organic cation transporter 1 (Oct1 [Slc22a1]) gene. Mol Cell Biol 21(16); 5471-5477
31) Zhou Y et al., (2015) Genetic polymorphisms and function of the organic anion-transporting polypeptide 1A2 and its clinical relevance in drug disposition. Pharmacology 95: 201-208.
32) Nakanishi T and Tamai I (2012) Genetic polymorphisms of OATP transporters and their impact on intestinal absorption and hepatic disposition of drugs. Drug Metab Pharmacokinet 27(1): 106-121.
33) Visentin M et al., (2012) Substrate and pH-specific antifolate transport mediated by organic anion-transporting polypeptide 2B1 (OATP2B1-SLCO2B1). Mol Pharmacol 81(2): 134-142
34) Roth M et al., (2012) OATPs, OATs and OCTs: the organic anion and cation transporters of the SLCO and SLC22A gene superfamilies. Br J Pharmacol 165(5):1260-1287
35) Cheng Y et al. (2012). Expression of organic anion transporter 2 in the human kidney and its potential role in the tubular secretion of guanine-containing antiviral drugs. Drug Metab Dispos 40(3): 617-624.
36) Nigam SK et al., (2015) The organic anion transporter family: a systems biology perspective. Physiol Rev 95(1): 83-123
37) Ekaratanawong S et al., (2004) Human organic anion transporter 4 is a renal apical organic anion/dicarboxylate exchanger in the proximal tubules. J Pharmacol Sci 94: 297-304.
38) Ohnishi S et al., (2008) Role of Na+/L-cartinine transporter (OCTN2) in renal handling of pivaloylcarnitiine and valproylcarnitine formed during pivalic acid-containing prodrugs and valproic acid treatment. Drug Metab Pharmacokinet 23(4): 293-303
39) Park HJ et al., (2016) Identification of OCTN2 variants and their association with phenotypes of Crohn’s disease in a Korean population. Sci. Rep. 6: 22887
40) Spanier B (2014) Transcriptional and functional regulation of the intestinal peptide transporter PEPT1. J Physiol 592: 871-879
41) Gupta D et al., (2013) Increasing oral absorption of polar neuraminidase inhibitors: a prodrug transporter approach applied to oseltamivir analogue. Mol Pharm 10(2): 512-522
42) Ingersoll SA et al., (2012) The role and pathophysiological relevance of membrane transporter PepT1 in intestinal inflammation and inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol 302(5); G484-92
43) Zhao D and Lu K (2015) Substrates of the human oligopeptide transporter hPEPT2. Biosci Trends 9(4): 207-213
44) Trauner M & Boyer JL (2003) Bile salt transporters: molecular characterisation, function and regulation. Physiol Rev 83(2): 633-671
45) Bi YA et al., (2013) Quantitative assessment of the contribution of sodium-dependent taurocholate co-transporting polypeptide (NTCP) to the hepatic uptake of rosuvastatin, pitavastatin and fluvastatin. Biopharm Drug Dispos 34(8): 452-461
46) Song IH et al., (2016) The effect of dolutegravir on the pharmacokinetics of metformin in healthy subjects. J Acquir Immune Defic Syndr 72(4): 400–407

TODO

Cyprotex eStore

Order our services online.

Visit the Cyprotex eStore