Learning Objectives

After completing this course, the reader will be able to:

  1. Identify important sources of variability in drug exposure caused by drug interactions mediated by P-glycoprotein.

  2. Describe how unwanted drug–drug interactions may lead to unexpected serious toxicity or undertreatment.

  3. Prevent these interactions by individualizing pharmacotherapy; this means selecting noninteracting drugs or adapting the dose of (the) interacting drug(s).

CME Access and take the CME test online and receive 1 AMA PRA Category 1 Credit™ at CME.TheOncologist.com

Abstract

The importance of P-glycoprotein (P-gp) in drug–drug interactions is increasingly being identified. P-gp has been reported to affect the pharmacokinetics of numerous structurally and pharmacologically diverse substrate drugs. Furthermore, genetic variability in the multidrug resistance 1 gene influences absorption and tissue distribution of drugs transported. Inhibition or induction of P-gp by coadministered drugs or food as well as herbal constituents may result in pharmacokinetic interactions leading to unexpected toxicities or undertreatment. On the other hand, modulation of P-gp expression and/or activity may be a useful strategy to improve the pharmacological profile of anticancer P-gp substrate drugs.

In recent years, the use of complementary and alternative medicine (CAM), like herbs, food, and vitamins, by cancer patients has increased significantly. CAM use substantially increases the risk for interactions with anticancer drugs, especially because of the narrow therapeutic window of these compounds. However, for most CAMs, it is unknown whether they affect metabolizing enzymes and/or drug transporter activity. Clinically relevant interactions are reported between St John's wort or grapefruit juice and anticancer as well as nonanticancer drugs. CAM–drug interactions could explain, at least in part, the large interindividual variation in efficacy and toxicity associated with drug therapy in both cancer and noncancer patients.

The study of drug–drug, food–drug, and herb–drug interactions and of genetic factors affecting pharmacokinetics and pharmacodynamics is expected to improve drug safety and will enable individualized drug therapy.

Introduction

In patients, drug–drug interactions can result in unexpected life-threatening and even lethal toxicities. Up to 10% of all admissions in general hospitals are caused by improper use of drugs and combinations of drugs, resulting in potentially severe drug–drug interactions [1, 2]. Adverse drug reactions can especially be severe when these interactions involve cytotoxic anticancer agents [3, 4]. Anticancer drugs are dosed close to the maximum-tolerated dose, and factors affecting the pharmacokinetics may therefore greatly increase the likelihood of development of life-threatening toxicities.

Thus far drug–drug interactions have been thought to result from inhibition of drug metabolism, inhibition of renal drug excretion, displacement out of the protein binding, or pharmaceutical interactions. However, interference at the level of ATP binding cassette (ABC) and other transporters is increasingly being identified as the mechanism behind clinically important drug–drug interactions. Drug–drug and herb–drug interactions at the level of ABCB1 (multidrug resistance 1 [MDR1], P-glycoprotein [P-gp]) is the subject of this paper.

Milestones, Position in ABC Transporter Family, Main Molecular Mechanism

P-gp was first identified by Juliano and Ling in 1976 as a surface glycoprotein in Chinese hamster ovary cells expressing the MDR phenotype [5]. Cloning of the encoding gene and structure analysis of the protein revealed that P-gp is a 160-kDa ATP-dependent efflux transporter, belonging to the ABC transporter superfamily [6, 7].

Tissue Distribution and Physiological Function

The anatomical localization of P-gp in various tumors (where it confers the MDR phenotype) and at the apical/luminal membrane of polarized cells in several normal human tissues with excretory (liver, kidney, adrenal gland) and barrier (intestine, blood–brain barrier, placenta, blood–testis and blood–ovarian barriers) functions [811] suggests for P-gp a physiological role in detoxification and protection of the body against toxic xenobiotics and metabolites by secreting these compounds into bile, urine, and the intestinal lumen and by preventing their accumulation in the brain, testis, and fetus (Fig. 1) [12].

Schematic representations of the main sites of localization of P-glycoprotein in the body.
Figure 1

Schematic representations of the main sites of localization of P-glycoprotein in the body.

Impact of Genetic Polymorphism in the  ABCB1 Gene on Function

Currently, at least 105 variants in the ABCB1 gene have been identified, with significant differences in their frequencies among different ethnic groups. The majority of these single nucleotide polymorphisms (SNPs) involve intronic or noncoding regions, thus not affecting the P-gp amino acid sequence. However, several variants in the ABCB1 coding regions result in amino acid change and potentially affect P-gp expression and activity. Hoffmeyer et al. [13] reported an association among a SNP in exon 26 (C3435T) of ABCB1, reduction in duodenal P-gp levels, and higher peak plasma concentrations of the P-gp substrate digoxin in healthy volunteers. Confirming and contradicting studies have subsequently been published about the influence of SNPs in ABCB1 on disposition of digoxin and also on other P-gp substrate drugs (such as fexofenadine, tacrolimus, irinotecan, SN-38, paclitaxel, and cyclosporin A) and on P-gp expression and activity (see reviews [1421]). Moreover, genetic variation in ABCB1, by potentially altering the physiologic protective role of P-gp, has recently been assessed in the etiology of several human pathophysiological conditions. An increasing number of studies have associated certain SNPs in ABCB1 with susceptibility to diseases such as pharmacoresistant epilepsy, Parkinson's disease, inflammatory bowel diseases (ulcerative colitis and Crohn's disease), colorectal cancer, and renal carcinoma [2227].

Recently the ABCB1 SNP C3435T has been associated with the efficacy of antiemetic treatment with 5-hydroxytryptamine type 3 receptor antagonists (such as granisetron, ondansetron, tropisetron) in patients with cancer [28], whereas in patients affected by depression, the same polymorphism has been linked to the development of postural hypotension induced by the antidepressant nortriptyline [29].

MDR1 gene polymorphism has also been suggested to affect the therapy outcome of patients with several malignancies. Goreva et al. [30] reported an association between C3435T and G3677T SNPs in ABCB1 and the risk for drug resistance in patients with lymphoproliferative diseases. A correlation between several commonly occurring ABCB1 SNPs and overall survival and the risk for relapse has been reported in patients affected by acute myeloid leukemia treated with etoposide, mitoxantrone, or daunorubicin (well-known P-gp substrates) [31]. Moreover, the ABCB1 SNP C3435T has been suggested as a significant predictor of treatment outcome in children affected by acute lymphoblastic leukemia, although these findings have not been confirmed in adults [32, 33]. Another study showed a greater response to preoperative chemotherapy in breast cancer patients homozygous for the C3435T genotype [34], whereas conflicting results have been reported about the impact of genetic variation in the MDR1 gene (in particular, G2677T/A) on the response to paclitaxel chemotherapy in patients with ovarian carcinoma [35].

Several factors may have contributed to the conflicting findings reported in the literature: demographic data from subjects selected for the various MDR1 SNPs and the methods used to measure P-gp expression differ from one study to another (i.e., protein detection by Western blot versus immunohistochemistry, various antibodies used, etc.). Moreover, discrepancies may be related to the route of drug administration and extent of metabolism relative to P-gp–mediated transport. For instance, cyclosporine is a P-gp but also a cytochrome P450 3A4 enzyme (CYP3A4) substrate, therefore a potential P-gp effect may be hidden by CYP3A4 activity. In this regard, environmental factors, such as diet, that affect CYP enzyme activity could also influence transporter function. Differences in dietary constituents among different populations may have contributed to the conflicting results among studies. For example, one of the possible reasons hypothesized for the reported divergent effects of MDR1 SNPs on fexofenadine disposition among whites living in the U.S. and in Germany was the difference in dietary salt intake between the two populations [14, 36, 37]. Furthermore, although well-known P-gp substrate drugs that are not metabolized to a relevant extent in humans (such as digoxin, fexofenadine, talinolol) have been used as probe drugs for P-gp function in humans, the involvement of other transporters and associated genetic variability could have influenced study results. Another possible reason for the contradictory reports associating ABCB1 variants with variation in drug response is that most of the studies have not considered haplotypes, whereas several recent studies suggested that the primary determinant of functional differences in P-gp resides not in SNP differences but in ABCB1 haplotypes [38]. Given the known interpopulation differences in drug response, it is especially important to consider variability among ethnic groups and to characterize variability in haplotype structure and linkage disequilibrium and recombination within and among ethnic populations.

Additional studies involving larger samples sizes and stratification according to haplotype are required for a complete understanding of the contribution of genetic variability in ABCB1 and related proteins to drug disposition, therapeutic response, and toxicity [21, 39]. To reduce the risk of a spurious association between MDR1 genotypes and in vivo phenotypes, demographic data of subjects selected for the various MDR1 SNPs as well as sample size and environmental factors should also be considered carefully. Moreover, standardization of assays relating to P-gp protein and mRNA detection and quantification is desirable too [14].

Main Clinically Applied Substrate Classes

P-gp presents high transport capacity and broad substrate specificity: a wide number of clinically relevant drugs with structurally different features and belonging to different classes (e.g., several anticancer drugs, some HIV protease inhibitors [HPIs], H2-receptor antagonists, antiarrhythmics—cardiac glycosides and calcium channel blockers—immunosuppressive agents, corticosteroids, antiemetic and antidiarrheal agents, analgesics, antibiotics, anthelmintics, antiepileptics, sedatives, antidepressants) can be transported by P-gp (for review, see [14, 40]); in general, they are hydrophobic and amphipathic molecules in nature, uncharged or basic, although zwitterionic and negatively charged compounds can also be transported.

Inhibitors (Competitive, Noncompetitive)

Some P-gp drug substrates are able to inhibit P-gp–mediated transport of other substrates. The discovery by Tsuruo and colleagues [41] that verapamil (weak P-gp substrate) could reverse P-gp–mediated MDR in leukemia cells was followed by the identification of several other P-gp inhibitors [42, 43] that can block P-gp activity by competition for drug-binding sites (competitive inhibitors) or by blockade of the ATP hydrolysis process (noncompetitive inhibitors). The first agents identified as P-gp inhibitors were drugs (e.g., verapamil and cyclosporin A) already used in the clinic that were themselves transported by P-gp (so-called first-generation inhibitors). Because of their low substrate selectivity and the concomitant inhibition of the drug-metabolizing CYP3A4, so-called second-generation (cyclosporin A analog PSC833) and third-generation (LY335979, VX710, GF120918, XR9576) P-gp inhibitors were developed. These and other selective P-gp inhibitors have been extensively tested preclinically and in patients to reverse MDR. Of interest is that GF120918 (elacridar), originally developed as a P-gp inhibitor, was also identified as an effective breast cancer resistance protein (BCRP; ABCG2) inhibitor [44].

Recently it has been reported that benzimidazole gastric H+, K+ -ATPase proton pump inhibitors (PPIs—omeprazole, pantoprazole, lansoprazole, and rabeprazole), which are used by up to 50% of patients with cancer, are effective inhibitors of P-gp in vitro [45], although their potency towards BCRP inhibition is even greater [46]. Drug interactions with benzimidazoles are increasingly reported [1, 2, 4750]. However, it has been noted that the 50% inhibitory concentration (IC50) values of PPIs in inhibiting P-gp observed in vitro are higher than their expected intraluminal (intestinal) and plasma concentrations obtained after oral dosing in humans, making a drug–drug interaction at these levels unlikely. Considering that PPIs have also been shown to be CYP3A4 and CYP2C19 substrates, and that they are able to inhibit BCRP activity, under certain circumstances, for instance in poor metabolizers of CYP2C19, plasma levels of omeprazole and pantoprazole would reach the range of reported IC50 values, thus making a clinical drug–drug interaction possible with coadministered substrate drugs for P-gp and/or BCRP [45].

In addition, several widely used drugs have been described to inhibit P-gp function, thus potentially leading to relevant drug–drug interactions. They include various antimicrobial agents (e.g., ceftriaxone, cefoperazone, clarithromycin, erythromycin, itraconazole, ketoconazole), Ca2+ antagonists (verapamil, diltiazem, quinidine, quinine, nifedipine, nicardipine), HPIs (ritonavir, indinavir, saquinavir, nelfinavir), and other compounds such as amiodarone, propranolol, dipyridamole, tacrolimus, hydrocortisone, progesterone, and tamoxifen, to name a few [51, 52].

Furthermore, many pure herbal constituents commonly used as complementary and alternative medicines (CAMs) by cancer patients and dietary phytochemicals have been reported to modulate P-gp expression and/or activity. Indeed, piperine [53], ginsenosides [54, 55], silymarin from milk thistle and other flavonoids [56], capsaicin [57], and resveratrol [57] were reported to inhibit P-gp activity in vitro, whereas curcumin [58, 59] and curcuminoids [60] and several catechins from green tea [6163] were shown to reduce P-gp expression and activity in vitro. Importantly, some of these herbal constituents (such as piperine, silymarin) have been observed to interact with P-gp at dietary concentrations, thus making a drug–herbal interaction in vivo more likely [53, 56]. Similarly, constituents of grapefruit and orange juice were also found to block P-gp function and certain juice–drug interactions for commonly used drugs have been described too [6470]. The modulation of P-gp as well as other transporters (i.e., organic anion transporting polypeptides [OATPs]) or drug-metabolizing enzymes (such as CYP3A4) may provide an explanation for many reported clinical herb/juice–drug interactions.

Inducers

Clinical and preclinical findings reveal that the expression of P-gp (like some of the CYP isoenzymes) is inducible. Expression levels of P-gp (as well as other transporters) and drug-metabolizing enzymes appear to be regulated by nuclear receptors like the pregnane X receptor (PXR), constitutive androstane receptor, and vitamin D binding receptor [71]. Some (active constituents of) herbs, like hyperforin from St John's wort (SJW), can activate one or more of these receptors, thereby increasing the expression of metabolizing enzymes and transporters [72, 73]. Several carotenoids and their metabolites (such as retinol and β-carotene) have been shown to activate the PXR in vitro [74]. Recent in vitro studies demonstrated that several drugs, including rifampicin, paclitaxel, and reserpine, can induce CYP3A4 and MDR1 gene expression through a similar mechanism [7577]. Other putative P-gp inducers are clotrimazole, phenobarbital, phenytoin, troglitazone, and the flavonoids kaempferol and quercetin [51]. However, thus far, only rifampin and SJW have been documented to significantly induce intestinal P-gp in humans: in duodenal biopsies performed in healthy volunteers after rifampin administration, P-gp was induced 3.5-fold [78]. Similarly, administration of SJW induced human intestinal P-gp 1.4 fold [79]. For the other inducers, only in vitro data are available, thus raising doubts whether results obtained in cell lines can be extrapolated to the human in vivo situation. Moreover, recent preclinical studies demonstrate a tissue specificity of P-gp induction with potential differences among species [80]. Therefore, although interactions between P-gp substrates and commonly used drugs/CAMs that are reported to induce P-gp expression in vitro have been described in the literature (see below), clearly, preclinical animal models that behave similarly to humans in terms of transporter and metabolism induction as well as further studies in humans are needed to evaluate their clinical relevance.

Pharmacological and Toxicological Functions

The pharmacological functions of P-gp have been extensively studied in in vitro and in vivo models: P-gp was first described as a plasma membrane protein that could cause MDR in tumor cells by actively extruding a wide range of structurally diverse compounds, thus contributing to the resistance against chemotherapy occurring in several cancers. In addition, the strategic physiological distribution of P-gp in organs that play key roles in processes of drug absorption, distribution, and elimination suggests that P-gp has a relevant impact on limiting cellular uptake of drugs out of the blood circulation into the brain, placenta, and testis and from the intestinal lumen into epithelial cells lining the gut. In addition, P-gp may also mediate excretion of drugs out of hepatocytes into the bile canalicuIi and out of renal tubules into the urine.

The effect of P-gp on the pharmacokinetics of substrate drugs has been demonstrated in vivo using mdr1a and mdr1a/1b knockout mice. Mice lacking one or both mdr genes showed significant alterations in drug absorption, distribution, and elimination [8184]: compared with wild-type mdr1a(−/−) and mdr1a/1b(−/−) mice displayed greater sensitivity to the centrally neurotoxic anthelmintic ivermectin and other known P-gp substrates like vinblastine, digoxin, and cyclosporin A. Compared with wild-type, mdr1a(−/−) and mdr1a/1b(−/−) mice also presented higher concentrations of drugs in many tissues (especially in the brain) and a slower rate of drug elimination. Other in vitro and in vivo studies documented the effect of P-gp expression on the apparent oral bioavailability of substrate drugs. Hunter et al. [85] reported the apical efflux of vinblastine across intestinal Caco-2 cell layers and the efflux was inhibited in the presence of the P-gp inhibitor verapamil and other P-gp modulators. In in vivo experiments, the apparent bioavailability of the P-gp substrate paclitaxel after oral administration was 11% in wild-type and 35% in mdr1a(−/−) mice [86]. Bardelmeijer et al. [87] reported an apparent bioavailability after oral administration of docetaxel, another P-gp substrate, of 3.6% in wild-type and 22.7% in mdr1a/1b(−/−) mice. In other studies, oral administration of the P-gp inhibitors valspodar (PSC 833) or cyclosporin A or elacridar (GF120918) followed by oral paclitaxel [8890] or by oral docetaxel [87] resulted in significantly greater apparent oral bioavailability in wild-type mice compared with those treated without the P-gp inhibitor (Fig. 2A). These findings led to important potential clinical implications: drug–drug interactions between substrates and P-gp inhibitors can modify the drug's pharmacokinetics by increasing bioavailability and organ uptake, leading to more adverse drug reactions and toxicities. Possibly, coadministration of substrates for P-gp and P-gp–inducing agents may lead to a reduction in plasma drug levels and consequently undertreatment.

Effect of cyclosporin A on the oral absorption of paclitaxel. (A): Coadministration of oral paclitaxel and cyclosporin A (CsA) in wild-type (WT) mice resulted in a significantly greater area under the curve (AUC) of paclitaxel in plasma. The effect was even greater than the AUC of paclitaxel when given to P-glycoprotein–deficient mdr1a/b knockout (KO) mice. The results indicate that P-glycoprotein effectively prevents oral uptake of paclitaxel from the gut. CsA may also have inhibited cytochrome P450 3A4 enzyme (CYP3A4) to explain the additional difference in the AUC compared with the experiment in KO mice. From Sparreboom A, van Asperen J, Mayer U et al. Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci U S A 1997;94:2031–2035, with permission. (B): Coadministration of oral paclitaxel and CsA in patients with advanced cancer resulted in a significantly greater AUC of paclitaxel in plasma. These results are the clinical proof of the concept that inhibition of P-glycoprotein (and possibly also CYP3A4 in the gut epithelium) results in a significantly greater uptake of paclitaxel from the gut leading to a significantly greater systemic exposure to paclitaxel. Reprinted from Meerum Terwogt JM, Beijnen JH, ten Bokkel Huinink WW et al. Co-administration of oral cyclosporin enables oral therapy with paclitaxel. Lancet 1998;352:285, with permission from Elsevier.
Figure 2

Effect of cyclosporin A on the oral absorption of paclitaxel. (A): Coadministration of oral paclitaxel and cyclosporin A (CsA) in wild-type (WT) mice resulted in a significantly greater area under the curve (AUC) of paclitaxel in plasma. The effect was even greater than the AUC of paclitaxel when given to P-glycoprotein–deficient mdr1a/b knockout (KO) mice. The results indicate that P-glycoprotein effectively prevents oral uptake of paclitaxel from the gut. CsA may also have inhibited cytochrome P450 3A4 enzyme (CYP3A4) to explain the additional difference in the AUC compared with the experiment in KO mice. From Sparreboom A, van Asperen J, Mayer U et al. Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci U S A 1997;94:2031–2035, with permission. (B): Coadministration of oral paclitaxel and CsA in patients with advanced cancer resulted in a significantly greater AUC of paclitaxel in plasma. These results are the clinical proof of the concept that inhibition of P-glycoprotein (and possibly also CYP3A4 in the gut epithelium) results in a significantly greater uptake of paclitaxel from the gut leading to a significantly greater systemic exposure to paclitaxel. Reprinted from Meerum Terwogt JM, Beijnen JH, ten Bokkel Huinink WW et al. Co-administration of oral cyclosporin enables oral therapy with paclitaxel. Lancet 1998;352:285, with permission from Elsevier.

Furthermore, the localization of P-gp in the placenta has been shown to play a key role in preventing fetal exposure to various potentially harmful or therapeutic compounds. Inhibition of P-gp activity in the placenta can affect the distribution and consequently the fetal toxicity and/or efficacy of P-gp substrate drugs [9194]. Drug–drug interactions should be considered very carefully in pregnant or lactating breast cancer patients who will be treated with anticancer drugs, substrates for P-gp, such as anthracyclines.

Drug–Drug Interactions

In the literature several drug–drug interactions mediated by P-gp transporters have been described (Table 1). In general, the involvement of P-gp in drug–drug interactions is difficult to prove in humans because, as a result of the overlapping substrate specificity of inhibitors and inducers between CYP3A4 and P-gp, many drug interactions may involve both CYP3A4 enzymes and P-gp [95]. Moreover, P-gp and CYP3A4 may be functionally linked, and several potential mechanisms whereby the functions of P-gp and CYP3A4 could be complementary have been proposed [96]. Furthermore, drug–drug interactions may involve additional ABC transporters as well.

Table 1

Clinically relevant drug-drug interactions involving MDR1

Table 1

Clinically relevant drug-drug interactions involving MDR1

Clinical drug–drug interactions were reported in the literature between digoxin (a good P-gp substrate) and other drugs, such as quinidine [9799], verapamil [100102], talinolol [103], clarithromycin [104], itraconazole [105], erythromycin [106], and propafenone [101, 107, 108].

Other clinically relevant drug interactions described in the literature involve the antimicrobial drug rifampicin. Rifampicin is a well-known inducer of intestinal CYP3A4. However, recent findings indicate that it can also induce P-gp expression. In a clinical study, the oral bioavailability of digoxin in eight healthy volunteers was decreased by 30% during rifampicin therapy. Intestinal biopsies obtained from the same patients before and after administration of rifampicin showed a significant increase in intestinal P-gp expression after administration of the antimicrobial drug, which correlated inversely with the oral area under the concentration–time curve (AUC) of digoxin. In addition, pretreatment with rifampicin had little effect on the renal clearance of digoxin. These results suggest that the digoxin–rifampicin interaction mainly occurs at the intestinal level and that chronic exposure to rifampicin can result in P-gp induction [78]. Similar interactions with rifampicin have been reported for talinolol [109], fexofenadine [110], and cyclosporin A [111].

Interactions mediated by P-gp that may have clinically relevant consequences have also been reported for some excipients used in pharmaceutical formulations. In in vitro experiments, polysorbate 80 was able to inhibit P-gp activity and to increase daunorubicin intracellular levels in cell cultures [112]. Polyoxyl castor oil and polysorbate 80 (substances used in drug formulations to dissolve some lipophylic and/or poorly soluble drugs, especially paclitaxel and docetaxel) were reported to increase the oral absorption of saquinavir and digoxin, respectively, through interaction with P-gp activity [113, 114].

In addition, food and dietary constituents, such as grapefruit, orange, apple, and pummelo juice, are also possible P-gp modulators. The in vivo effect of fruit juices, and in particular, grapefruit juice, on drug transport is still controversial, because some authors have predicted or reported greater whereas others have predicted or reported lower amounts of coadministered drugs reaching the systemic circulation. Indeed, several authors reported that flavonoids and furanocoumarins of grapefruit juice were able to inhibit P-gp and CYP3A4 activity, thus influencing accumulation and efflux of anticancer drugs (well-known P-gp substrates) in P-gp–overexpressing cell lines [66, 70, 115117]. In contrast, the apparent bioavailability after oral administration and the plasma concentrations of etoposide were significantly lower in subjects taking grapefruit juice [118]. Similar results were reported in healthy volunteers taking grapefruit juice and the nonmetabolized and P-gp–transported drug talinolol [119], whereas in rats, administration of grapefruit juice resulted in higher plasma concentrations and lower apparent oral clearance of talinolol [115]. The reasons for these discrepancies are still unknown: differences in the concentrations of drug-interacting compounds in the juices (such as fourano coumarins and flavonoids) have been proposed to contribute to the discrepancies in the results [120], as well as the modulation of other transporters (such as OATPs, multidrug resistance–associated proteins [MRPs]) and metabolizing enzymes by grapefruit juice constituents and other environmental factors (i.e., dietary constituents). All these findings make it difficult to predict whether a grapefruit juice–drug interaction will occur and the magnitude of such interaction. Therefore, patients should be cautious with the consumption of grapefruit juice when treated with narrow-therapeutic-index drugs (especially with drugs whose absorption has been reported to be affected by P-gp, MRPs, OATPs).

Furthermore, many other dietary food and pure herbal constituents (see above) commonly used as CAMs directly inhibit CYP and P-gp activity in vitro, and some of them (like piperine and silymarin) were shown to act as P-gp inhibitors at dietary concentrations. P-gp expression is clearly induced by the over-the-counter antidepressant herbal SJW, and clinically relevant drug–drug interactions have been reported between SJW and a wide range of drugs. Chronic administration of SJW together with cyclosporin A has been associated with a significant reduction in cyclosporin plasma levels and a higher risk for acute organ rejection in transplanted patients [121, 122]. In healthy volunteers, administration of SJW together with the HPI indinavir produced an approximately 57% lower plasma AUC of indinavir [123]. Coadministration of SJW with digoxin produced an 18% lower plasma AUC of digoxin and a 40% higher expression level of intestinal P-gp [79]. Other clinical studies confirmed that coadministration of SJW significantly reduced plasma concentrations of drugs like oral contraceptives, tacrolimus, warfarin, verapamil, fexofenadine, and some others, leading to important clinical implications, that is undertreatment and failure of therapies. Similarly, in rats and in cancer patients, the plasma concentrations of SN-38 (the active metabolite of irinotecan) were significantly lower and hematological and gastrointestinal toxicities were less when SJW was coadministered [124, 125]. Furthermore, in healthy volunteers, administration of SJW together with the protein tyrosine kinase inhibitor imatinib resulted in a significantly greater oral clearance and lower AUC, maximum concentration, and half-life of imatinib [126, 127]. Induction of CYP3A4 and enhanced P-gp expression have been suggested to be responsible for these interactions (for review, see [128135]).

However, for most CAMs, it is unknown whether they affect metabolizing enzymes and/or drug transporters, potentially leading to unwanted pharmacokinetic interactions with drug therapy. Altered expression or activity of several drug transporters and drug-metabolizing enzymes can lead to lower therapeutic efficacy or greater toxicity.

The risk for interactions is significantly high in cancer patients, considering that several anticancer drugs (such as vincristine, vinblastine, vinorelbine, irinotecan, etoposide, docetaxel, and paclitaxel) are P-gp and/or CYP3A4 substrates, as well as certain supportive care agents concomitantly and commonly used by cancer patients, such as ondansetron, fentanyl, morphine, loperamide, and domperidone [83, 136138]. Clearly, the risk for interaction is further increased by the intake of CAMs, products that are frequently used by people affected by cancer.

Possible Clinical Benefit of Drug–Drug Interactions

On the other hand, the study of drug–drug interactions with P-gp modulators is an interesting research field, because P-gp was discovered and described for its ability to confer the MDR phenotype to cancer cells. The modulation of P-gp activity was at first seen as a useful strategy for increasing the penetration and retention of anticancer drugs in resistant tumor cells, thus overcoming the intrinsic or acquired resistance against chemotherapy occurring in several cancers. Since the mid-1980s, various clinical trials with anticancer drugs in combination with P-gp modulators (calcium channel blockers—nifedipine or verapamil—or cyclosporin A) have been performed [139141]. Unfortunately, with only few exceptions [142146], these studies did not show any survival benefit for the combination of an anticancer drug plus a P-gp inhibitor [147151]. In addition, because the P-gp inhibitors used in those trials presented overlap in substrate specificity with CYP3A4 inhibitors, pharmacokinetic interactions occurred, resulting in greater toxicity. To date, some clinical trials using second- and third-generation P-gp inhibitors with the aim of reversing MDR in tumor cells have been performed and others are still ongoing [152154]. In a recent pilot phase II trial, the combination of valspodar (PSC 833, a second-generation P-gp inhibitor) plus paclitaxel (administered i.v. at a reduced dose because of the expected pharmacokinetic interaction [155]) in patients with metastatic breast cancer showed acceptable toxicity but the activity was not significantly increased compared with single-agent paclitaxel [156]. Additional trials will further explore the feasibility and efficacy of this strategy.

Modulation of P-gp activity with selective inhibitors could also be a useful strategy to increase the oral bioavailability of P-gp substrate drugs, in particular, to develop oral formulations of anticancer drugs transported by P-gp. Several preclinical animal studies (see above) and clinical trials in humans have been performed to evaluate the feasibility and the safety of this approach (coadministration of a substrate drug and a P-gp inhibitor). In a clinical study, cyclosporin A, an effective P-gp blocker, followed by oral paclitaxel (a well-known P-gp substrate) resulted in an eightfold higher systemic exposure to paclitaxel (Fig. 2B) [157]. Cyclosporin A also effectively resulted in a greater oral bioavailability of docetaxel, 91% versus 8% [158]. Elacridar, an effective inhibitor of BCRP as well as of P-gp produced a greater oral bioavailability of topotecan, 97% versus 40% [159]. Further studies in patients with advanced solid tumors confirmed that this strategy for oral treatment is at least as effective and safe as standard i.v. administration of these drugs, and clinical trials with third-generation modulators of P-gp (e.g., biricodar, zosuquidar, and laniquidar) specifically developed for MDR reversal are ongoing. The results will give insight into the possible clinical feasibility of this strategy [159163]. Indeed, an interesting clinical application of selective modulation of P-gp activity might lead to greater passage of certain drugs across the blood–brain barrier, which might profoundly extend the range of drugs available for treatment of brain disorders [164]. These include primary and metastatic tumors, microbial infections, HIV infections, mood disorders, and neurological treatment–resistant disease, for example, refractory epilepsy and schizophrenia. Furthermore, preclinical studies have shown that the brain penetration of anticancer drugs that are transported by P-gp, such as paclitaxel, docetaxel, and imatinib, can be improved by concomitant use of P-gp inhibitors, such as cyclosporin A, valspodar, elacridar, and zosuquidar [165169]. A clinical study determining the brain penetration of paclitaxel in combination with elacridar in patients with primary brain tumors is ongoing and the preliminary results are reported to be promising [170]. Similarly, clinical trials are exploring the activity of imatinib (Gleevec®; Novartis Pharmaceuticals Corporation, East Hanover, NJ) against the central nervous system (CNS) tumor glioblastoma [171] based on promising preclinical results. Taking into account that imatinib is a good P-gp and BCRP substrate drug with a limited distribution to the brain [172, 173] and that preclinical studies reported that the combination of imatinib with an effective P-gp inhibitor resulted in greater CNS accumulation [168, 174], modulation of P-gp as well as BCRP activity can be a useful strategy to improve CNS penetration of imatinib [170, 174].

However, the safety of this approach should be explored carefully as modulation of P-gp in the blood–brain barrier may lead to greater CNS accumulation of unwanted potentially toxic xenobiotics and endogenous compounds. Preclinical studies in wild-type and mdr1a/b knockout mice demonstrated that mdr1a/1b knockout mice are fertile and viable, but they are more sensitive to a range of drugs and toxins [81, 83, 84]. Moreover, absence or inhibition of P-gp activity can alter the specific pharmacodynamic activity of some P-gp substrate drugs, leading to CNS toxicity and adverse drug effects. For instance, the safe clinical use of the antidiarrheal drug loperamide may also be critically dependent on the presence of P-gp in the blood–brain barrier. Loperamide is a potent opiate, which demonstrates nearly exclusively peripheral opiate-like effects on the gastrointestinal tract and no central effects because it is a P-gp substrate. Thus, normally it cannot accumulate in the CNS. In mdr1a knockout mice, however, loperamide showed pronounced opiate-like effects and sometimes lethal effects at doses that are safe in wild-type mice [83]. In humans, coadministration of loperamide with the P-gp inhibitor quinidine produced opiate-induced respiratory depression, a clear central opiate effect that is normally not seen in humans [175].

On the same line, blocking of placental P-gp in HIV-infected pregnant women might be used to enhance HPI levels in the unborn child shortly before and during the delivery, thereby reducing the risk for HIV infection of the fetus. However, the safety of this approach needs to be studied in greater detail. Indeed, preclinical data using mdr1a/1b knockout mice demonstrated significantly greater fetal penetration of the HPIs indinavir and saquinavir, but also of other drugs and toxic compounds, indicating that placental P-gp might have a protective role for the fetus [92, 176].

Summary, Conclusions, and Perspectives

The importance of ABC transporters in drug–drug interactions is increasingly being identified. P-gp is involved in the interactions between cyclosporin A or verapamil and oral digoxin. Azole antifungals, such as fluconazole and itraconazole, interact with P-gp, explaining drug interactions with digoxin and other drugs. Benzimidazoles are transported by and inhibit P-gp. P-gp regulates oral bioavailability and tissue distribution of the immunosuppressant tacrolimus. P-gp mediates drug interactions between antiretroviral drugs and comedications. Also, genetic variability in the MDR1 gene affects absorption and tissue distribution of P-gp substrate drugs.

Furthermore, CAM use, like herbs, food, and vitamins, by patients has increased significantly in recent years. Surveys have shown that the prevalence of CAM use among cancer patients receiving conventional therapy is 54%–77%, and that about 72% of patients do not inform their treating physician. CAM use significantly increases the risk for interactions with anticancer drugs, especially because of the small therapeutic range and steep dose–toxicity curve of these drugs. Clinically relevant problems are seen with SJW and grapefruit juice. SJW significantly decreases the plasma levels of SN-38, the active metabolite of irinotecan, and increases imatinib clearance. Grapefruit juice affects the oral bioavailability of etoposide. However, it is expected that CAM–drug interactions are responsible for more of the, so far unresolved, interindividual variation and clinical problems seen in cancer and noncancer patients.

The main causes of interactions are changes in the pharmacokinetics of drugs, although interactions at the pharmacodynamic level are also possible. Many drugs are cleared by biotransformation and subsequently transported by P-gp, BCRP, or other transporters. Altered expression or activity of these proteins can lead to lower therapeutic efficacy or greater toxicity.

Increased knowledge of drug–drug, food–drug, and herb–drug interactions and of genetic factors affecting pharmacokinetics and pharmacodynamics is expected to improve drug safety and will enable drug therapy tailored to the individual patients' needs.

Disclosure of Potential Conflicts of Interest

The authors indicate no potential conflicts of interest.

References

1

Fattinger
 
K
,
Roos
 
M
,
Vergeres
 
P
et al.

Epidemiology of drug exposure and adverse drug reactions in two Swiss departments of internal medicine
.
Br J Clin Pharmacol
.
2000
;
499
:
158
167
.
2

Zoppi
 
M
,
Braunschweig
 
S
,
Kuenzi
 
UP
et al.

Incidence of lethal adverse drug reactions in the comprehensive hospital drug monitoring, a 20-year survey, 1974–1993, based on the data of Berne/St Gallen
.
Eur J Clin Pharmacol
.
2000
;
56
:
427
430
.
3

van Meerten
 
E
,
Verweij
 
J
,
Schellens
 
JH
,

Antineoplastic agents: Drug interactions of clinical significance
.
Drug Saf
.
1995
;
12
:
168
182
.
4

Beijnen
 
JH
,
Schellens
 
JHM
,

Drug interactions in oncology
.
Lancet Oncol
.
2004
;
5
:
489
496
.
5

Juliano
 
RL
,
Ling
 
V
,

A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants
.
Biochim Biophys Acta
.
1976
;
455
:
152
162
.
6

Gros
 
P
,
Croop
 
J
,
Housman
 
D
,

Mammalian multidrug resistance gene: Complete cDNA sequence indicates strong homology to bacterial transport proteins
.
Cell
.
1986
;
47
:
371
380
.
7

Higgins
 
CF
,

ABC transporters: From microorganisms to man
.
Annu Rev Cell Biol
.
1992
;
8
:
67
113
.
8

Fojo
 
AT
,
Ueda
 
K
,
Slamon
 
DJ
et al.

Expression of a multidrug-resistance gene in human tumors and tissues
.
Proc Natl Acad Sci U S A
.
1987
;
84
:
265
269
.
9

Thiebaut
 
F
,
Tsuruo
 
T
,
Hamada
 
H
et al.

Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues
.
Proc Natl Acad Sci U S A
.
1987
;
84
:
7735
7738
.
10

Arceci
 
RJ
,
Croop
 
JM
,
Horwitz
 
SB
et al.

The gene encoding multidrug resistance is induced and expressed at high levels during pregnancy in the secretory epithelium of the uterus
.
Proc Natl Acad Sci U S A
.
1988
;
85
:
4350
4354
.
11

Cordon-Cardo
 
C
,
O'Brien
 
JP
,
Casals
 
D
et al.

Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites
.
Proc Natl Acad Sci U S A
.
1989
;
86
:
695
698
.
12

Kartner
 
N
,
Riordan
 
JR
,
Ling
 
V
,

Cell surface P-glycoprotein associated with multidrug resistance in mammalian cell lines
.
Science
.
1983
;
221
:
1285
1288
.
13

Hoffmeyer
 
S
,
Burk
 
O
,
Von Richter
 
O
et al.

Functional polymorphisms of the human multidrug resistance gene: Multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo
.
Proc Natl Acad Sci U S A
.
2000
;
97
:
3473
3478
.
14

Marzolini
 
C
,
Paus
 
E
,
Buclin
 
T
et al.

Polymorphisms in human MDR1 (P-glycoprotein): Recent advances and clinical relevance
.
Clin Pharmacol Ther
.
2004
;
75
:
13
33
.
15

Mathijssen
 
RHJ
,
Marsh
 
S
,
Karlsson
 
MO
et al.

Irinotecan pathway genotype analysis to predict pharmacokinetics
.
Clin Cancer Res
.
2003
;
9
:
3246
3253
.
16

Zhou
 
Q
,
Sparreboom
 
A
,
Tan
 
EH
et al.

Pharmacogenetic profiling across the irinotecan pathway in Asian patients with cancer
.
Br J Clin Pharmacol
.
2005
;
59
:
415
424
.
17

de Jong
 
FA
,
de Jonge
 
MJ
,
Verweij
 
J
et al.

Role of pharmacogenetics in irinotecan therapy
.
Cancer Lett
.
2006
;
234
:
90
106
.
18

Yamaguchi
 
H
,
Hishinuma
 
T
,
Endo
 
N
et al.

Genetic variation in ABCB1 influences paclitaxel pharmacokinetics in Japanese patients with ovarian cancer
.
Int J Gynecol Cancer
.
2006
;
16
:
979
985
.
19

Henningsson
 
A
,
Marsh
 
S
,
Loos
 
WJ
et al.

Association of CYP2C8, CYP3A4, CYP3A5, and ABCB1 polymorphisms with the pharmacokinetics of paclitaxel
.
Clin Cancer Res
.
2005
;
11
:
8097
8104
.
20

Verstuyft
 
C
,
Schwab
 
M
,
Schaeffeler
 
E
et al.

Digoxin pharmacokinetics and MDR1 genetic polymorphisms
.
Eur J Clin Pharmacol
.
2003
;
58
:
809
812
.
21

Schwab
 
M
,
Eichelbaum
 
M
,
Fromm
 
MF
,

Genetic polymorphisms of the human MDR1 drug transporter
.
Annu Rev Pharmacol Toxicol
.
2003
;
43
:
285
307
.
22

Siddiqui
 
A
,
Kerb
 
R
,
Weale
 
ME
et al.

Association of multidrug resistance in epilepsy with a polymorphism in the drug-transporter gene ABCB1
.
N Engl J Med
.
2003
;
348
:
1442
1448
.
23

Wada
 
M
,

Single nucleotide polymorphisms in ABCC2 and ABCB1 genes and their clinical impact in physiology and drug response
.
Cancer Lett
.
2006
;
234
:
40
50
.
24

Siegsmund
 
M
,
Brinkmann
 
U
,
Schaffeler
 
E
et al.

Association of the P-glycoprotein transporter MDR1(C3435T) polymorphism with the susceptibility to renal epithelial tumors
.
J Am Soc Nephrol
.
2002
;
13
:
1847
1854
.
25

Drozdzik
 
M
,
Bialecka
 
M
,
Mysliwiec
 
K
et al.

Polymorphism in the P-glycoprotein drug transporter MDR1 gene: A possible link between environmental and genetic factors in Parkinson's disease
.
Pharmacogenetics
.
2003
;
13
:
259
263
.
26

Furuno
 
T
,
Landi
 
MT
,
Ceroni
 
M
et al.

Expression polymorphism of the blood-brain barrier component P-glycoprotein (MDR1) in relation to Parkinson's disease
.
Pharmacogenetics
.
2002
;
12
:
529
534
.
27

Schwab
 
M
,
Schaeffeler
 
E
,
Marx
 
C
et al.

Association between the C3435T MDR1 gene polymorphism and susceptibility for ulcerative colitis
.
Gastroenterology
.
2003
;
124
:
26
33
.
28

Babaoglu
 
MO
,
Bayar
 
B
,
Aynacioglu
 
AS
et al.

Association of the ABCB1 3435C>T polymorphism with antiemetic efficacy of 5-hydroxytryptamine type 3 antagonists
.
Clin Pharmacol Ther
.
2005
;
78
:
619
626
.
29

Roberts
 
RL
,
Joyce
 
PR
,
Mulder
 
RT
et al.

A common P-glycoprotein polymorphism is associated with nortriptyline-induced postural hypotension in patients treated for major depression
.
Pharmacogenomics J
.
2002
;
2
:
191
196
.
30

Goreva
 
OB
,
Grishanova
 
AY
,
Mukhin
 
OV
et al.

Possible prediction of the efficiency of chemotherapy in patients with lymphoproliferative diseases based on MDR1 gene G2677T and C3435T polymorphisms
.
Bull Exp Biol Med
.
2003
;
136
:
183
185
.
31

Illmer
 
T
,
Schuler
 
US
,
Thiede
 
C
et al.

MDR1 gene polymorphisms affect therapy outcome in acute myeloid leukemia patients
.
Cancer Res
.
2002
;
62
:
4955
4962
.
32

Jamroziak
 
K
,
Robak
 
T
,

Pharmacogenomics of MDR1/ABCB1 gene: The influence on risk and clinical outcome of haematological malignancies
.
Hematology
.
2004
;
9
:
91
105
.
33

Jamroziak
 
K
,
Balcerczak
 
E
,
Cebula
 
B
et al.

Multi-drug transporter MDR1 gene polymorphism and prognosis in adult acute lymphoblastic leukemia
.
Pharmacol Rep
.
2005
;
57
:
882
888
.
34

Kafka
 
A
,
Sauer
 
G
,
Jaeger
 
C
et al.

Polymorphism C3435T of the MDR-1 gene predicts response to preoperative chemotherapy in locally advanced breast cancer
.
Int J Oncol
.
2003
;
22
:
1117
1121
.
35

Green
 
H
,
Soderkvist
 
P
,
Rosenberg
 
P
et al.

mdr-1 single nucleotide polymorphisms in ovarian cancer tissue: G2677T/A correlates with response to paclitaxel chemotherapy
.
Clin Cancer Res
.
2006
;
12
:
854
859
.
36

Drescher
 
S
,
Schaeffeler
 
E
,
Hitzl
 
M
et al.

MDR1 gene polymorphisms and disposition of the P-glycoprotein substrate fexofenadine
.
Br J Clin Pharmacol
.
2002
;
53
:
526
534
.
37

Dresser
 
G
,
Schwartz
 
U
,
Wilkinson
 
G
et al.

Fexofenadine bio-availability modulated by dietary salt [abstract]
.
Clin Pharmacol Ther
.
2001
;
69
:
P23
.
38

Kroetz
 
DL
,
Pauli-Magnus
 
C
,
Hodges
 
LM
et al.

Sequence diversity and haplotype structure in the human ABCB1 (MDR1, multidrug resistance transporter) gene
.
Pharmacogenetics
.
2003
;
13
:
481
494
.
39

Kurata
 
Y
,
Ieiri
 
I
,
Kimura
 
M
et al.

Role of human MDR1 gene polymorphism in bioavailability and interaction of digoxin, a substrate of P-glycoprotein
.
Clin Pharmacol Ther
.
2002
;
72
:
209
219
.
40

Schinkel
 
AH
,
Jonker
 
JW
,

Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: An overview
.
Adv Drug Deliv Rev
.
2003
;
55
:
3
29
.
41

Tsuruo
 
T
,
Iida
 
H
,
Tsukagoshi
 
S
et al.

Overcoming of vincristine resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastine by verapamil
.
Cancer Res
.
1981
;
41
:
1967
1972
.
42

Sikic
 
BI
,

Pharmacological approaches to reversing multidrug resistance
.
Semin Hematol
.
1997
;
34
(
suppl 5
):
40
47
.
43

Sikic
 
BI
,

New approaches in cancer treatment
.
Ann Oncol
.
1999
;
10
(
suppl 6
):
149
153
.
44

e Bruin
 
M
,
Miyake
 
K
,
Litman
 
T
et al.

Reversal of resistance by GF120918 in cell lines expressing the ABC half-transporter, MXR
.
Cancer Lett
.
1999
;
146
:
117
126
.
45

Pauli-Magnus
 
C
,
Rekersbrink
 
S
,
Kloz
 
U
et al.

Interaction of omeprazole, lansoprazole and pantoprazole with P-glycoprotein
.
Naunyn-Schmiederbergs Arch Pharmacol
.
2001
;
364
:
551
557
.
46

Breedveld
 
P
,
Zelcer
 
N
,
Pluim
 
D
et al.

Mechanism of the pharmacokinetic interaction between methotrexate and benzimidazoles: Potential role for breast cancer resistance protein in clinical drug-drug interactions
.
Cancer Res
.
2004
;
64
:
5804
5811
.
47

Reid
 
T
,
Yuen
 
A
,
Catolico
 
M
et al.

Impact of omeprazole on the plasma clearance of methotrexate
.
Cancer Chemother Pharmacol
.
1993
;
33
:
82
84
.
48

Mannesse
 
CK
,
Derkx
 
FH
,
de Ridder
 
MA
et al.

Contribution of adverse drug reactions to hospital admission of older patients
.
Age Ageing
.
2000
;
29
:
35
39
.
49

Tröger
 
U
,
Stotzel
 
B
,
Martens-Lobenhoffe
 
J
et al.

Drug Points: Severe myalgia from an interaction between treatments with pantoprazole and methotrexate
.
BMJ
.
2002
;
324
:
1497
.
50

De Maat
 
MM
,
Ekhart
 
GC
,
Huitema
 
AD
et al.

Drug interactions between antiretroviral drugs and comedicated agents
.
Clin Pharmacokinet
.
2003
;
42
:
223
282
.
51

DuBuske
 
LM
,

The role of P-glycoprotein and organic anion-transporting polypeptides in drug interactions
.
Drug Saf
.
2005
;
28
:
789
801
.
52

Sankatsing
 
SUC
,
Beijnen
 
JH
,
Schinkel
 
AH
et al.

P glycoprotein in human immunodeficiency virus type 1 infection and therapy
.
Antimicrob Agents Chemother
.
2004
;
48
:
1073
1081
.
53

Bhardwaj
 
RK
,
Glaeser
 
H
,
Becquemont
 
L
et al.

Piperine, a major constituent of black pepper, inhibits human P-glycoprotein and CYP3A4
.
J Pharmacol Exp Ther
.
2002
;
302
:
645
650
.
54

Kim
 
S-W
,
Kwon
 
HY
,
Chi
 
D-W
et al.

Reversal of P-glycoprotein-mediated multidrug resistance by ginsenoside Rg(3)
.
Biochem Pharmacol
.
2003
;
65
:
75
82
.
55

Choi
 
C-H
,
Kang
 
G
,
Min
 
Y-D
,

Reversal of P-glycoprotein-mediated multidrug resistance by protopanaxatriol ginsenosides from Korean red ginseng
.
Planta Med
.
2003
;
69
:
235
240
.
56

Zhang
 
S
,
Morris
 
ME
,

Effects of the flavonoids biochanin A, morin, phloretin, and silymarin on P-glycoprotein-mediated transport
.
J Pharmacol Exp Ther
.
2003
;
304
:
1258
1267
.
57

Nabekura
 
T
,
Kamiyama
 
S
,
Kitagawa
 
S
,

Effects of dietary chemopreventive phytochemicals on P-glycoprotein function
.
Biochem Biophys Res Commun
.
2005
;
327
:
866
870
.
58

Romiti
 
N
,
Tongiani
 
R
,
Cervelli
 
F
et al.

Effects of curcumin on P-glycoprotein in primary cultures of rat hepatocytes
.
Life Sci
.
1998
;
62
:
2349
2358
.
59

Anuchapreeda
 
S
,
Leechanachai
 
P
,
Smith
 
MM
et al.

Modulation of P-glycoprotein expression and function by curcumin in multidrug-resistant human KB cells
.
Biochem Pharmacol
.
2002
;
64
:
573
582
.
60

Limtrakul
 
P
,
Anuchapreeda
 
S
,
Buddhasukh
 
D
,

Modulation of human multidrug-resistance MDR-1 gene by natural curcuminoids
.
BMC Cancer
.
2004
;
4
:
13
.
61

Sadzuka
 
Y
,
Sugiyama
 
T
,
Sonobe
 
T
,

Efficacies of tea components on doxorubicin induced antitumor activity and reversal of multidrug resistance
.
Toxicol Lett
.
2000
;
114
:
155
162
.
62

Jodoin
 
J
,
Demeule
 
M
,
Beliveau
 
R
,

Inhibition of the multidrug resistance P-glycoprotein activity by green tea polyphenols
.
Biochim Biophys Acta
.
2002
;
1542
:
149
159
.
63

Mei
 
Y
,
Qian
 
F
,
Wei
 
D
et al.

Reversal of cancer multidrug resistance by green tea polyphenols
.
J Pharm Pharmacol
.
2004
;
56
:
1307
1314
.
64

Xu
 
J
,
Go
 
ML
,
Lim
 
L-Y
,

Modulation of digoxin transport across Caco-2 cell monolayers by citrus fruit juices: Lime, lemon, grapefruit, and pummelo
.
Pharm Res
.
2003
;
20
:
169
176
.
65

Zhou
 
S
,
Lim
 
LY
,
Chowbay
 
B
,

Herbal modulation of P-glycoprotein
.
Drug Metab Rev
.
2004
;
36
:
57
104
.
66

Takanaga
 
H
,
Ohnishi
 
A
,
Matsuo
 
H
et al.

Inhibition of vinblastine efflux mediated by P-glycoprotein by grapefruit juice components in caco-2 cells
.
Biol Pharm Bull
.
1998
;
21
:
1062
1066
.
67

Takanaga
 
H
,
Ohnishi
 
A
,
Yamada
 
S
et al.

Polymethoxylated flavones in orange juice are inhibitors of P-glycoprotein but not cytochrome P450 3A4
.
J Pharmacol Exp Ther
.
2000
;
293
:
230
236
.
68

Ikegawa
 
T
,
Ushigome
 
F
,
Koyabu
 
N
et al.

Inhibition of P-glycoprotein by orange juice components, polymethoxyflavones in adriamycin-resistant human myelogenous leukemia (K562/ADM) cells
.
Cancer Lett
.
2000
;
160
:
21
28
.
69

Honda
 
Y
,
Ushigome
 
F
,
Koyabu
 
N
et al.

Effects of grapefruit juice and orange juice components on P-glycoprotein- and MRP2-mediated drug efflux
.
Br J Pharmacol
.
2004
;
143
:
856
864
.
70

Wang
 
EJ
,
Casciano
 
CN
,
Clement
 
RP
et al.

Inhibition of P-glycoprotein transport function by grapefruit juice psoralen
.
Pharm Res
.
2001
;
18
:
432
438
.
71

Xu
 
C
,
Li
 
CY-T
,
Kong
 
A-NT
,

Induction of phase I, II and III drug metabolism/transport by xenobiotics
.
Arch Pharm Res
.
2005
;
28
:
249
268
.
72

Moore
 
LB
,
Goodwin
 
B
,
Jones
 
SA
et al.

St. John's wort induces hepatic drug metabolism through activation of the pregnane X receptor
.
Proc Natl Acad Sci U S A
.
2000
;
97
:
7500
7502
.
73

Kliewer
 
SA
,
Goodwin
 
B
,
Willson
 
TM
,

The nuclear pregnane X receptor: A key regulator of xenobiotic metabolism
.
Endocr Rev
.
2002
;
23
:
687
702
.
74

Ruhl
 
R
,
Sczech
 
R
,
Landes
 
N
et al.

Carotenoids and their metabolites are naturally occurring activators of gene expression via the pregnane X receptor
.
Eur J Nutr
.
2004
;
43
:
336
343
.
75

Schuetz
 
EG
,
Beck
 
WT
,
Schuetz
 
JD
,

Modulators and substrates of P-glycoprotein and cytochrome P4503A coordinately up-regulate these proteins in human colon carcinoma cells
.
Mol Pharmacol
.
1996
;
49
:
311
318
.
76

Geick
 
A
,
Eichelbaum
 
M
,
Burk
 
O
,

Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin
.
J Biol Chem
.
2001
;
276
:
14581
14587
.
77

Synold
 
TW
,
Dussault
 
I
,
Forman
 
BM
,

The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux
.
Nat Med
.
2001
;
7
:
584
590
.
78

Greiner
 
B
,
Eichelbaum
 
M
,
Fritz
 
P
et al.

The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin
.
J Clin Invest
.
1999
;
104
:
147
153
.
79

Durr
 
D
,
Stieger
 
B
,
Kullak-Ublick
 
GA
et al.

St John's wort induces intestinal P-glycoprotein/MDR1 and intestinal and hepatic CYP3A4
.
Clin Pharmacol Ther
.
2000
;
68
:
598
604
.
80

Matheny
 
CJ
,
Ali
 
RY
,
Yang
 
X
et al.

Effect of prototypical inducing agents on P-glycoprotein and CYP3A expression in mouse tissues
.
Drug Metab Dispos
.
2004
;
32
:
1008
1014
.
81

Schinkel
 
AH
,
Smit
 
JJM
,
van Tellingen
 
O
et al.

Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs
.
Cell
.
1994
;
77
:
491
502
.
82

Schinkel
 
AH
,
Wagenaar
 
E
,
van Deemter
 
L
et al.

Absence of the mdr1a P-glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin and cyclosporin A
.
J Clin Invest
.
1995
;
96
:
1698
1705
.
83

Schinkel
 
AH
,
Wagenaar
 
E
,
Mol
 
CA
et al.

P-glycoprotein in the blood-brain barrier of mice influences the brain penetration and pharmacological activity of many drugs
.
J Clin Invest
.
1996
;
97
:
2517
2524
.
84

Schinkel
 
AH
,
Mayer
 
U
,
Wagenaar
 
E
et al.

Normal viability and altered pharmacokinetics in mice lacking mdr1-type (drug-transporting) P-glycoproteins
.
Proc Natl Acad Sci U S A
.
1997
;
94
:
4028
4033
.
85

Hunter
 
J
,
Jepson
 
MA
,
Tsuruo
 
T
et al.

Functional expression of P-glycoprotein in apical membranes of human intestinal Caco-2 cells. Kinetics of vinblastine secretion and interaction with modulators
.
J Biol Chem
.
1993
;
268
:
14991
14997
.
86

Sparreboom
 
A
,
van Asperen
 
J
,
Mayer
 
U
et al.

Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine
.
Proc Natl Acad Sci U S A
.
1997
;
94
:
2031
2035
.
87

Bardelmeijer
 
HA
,
Ouwehand
 
M
,
Buckle
 
T
et al.

Low systemic exposure of oral docetaxel in mice resulting from extensive first-pass metabolism is boosted by ritonavir
.
Cancer Res
.
2002
;
62
:
6158
6164
.
88

van Asperen
 
J
,
van Tellingen
 
O
,
Sparreboom
 
A
et al.

Enhanced oral bioavailability of paclitaxel in mice treated with the P-glycoprotein blocker SDZ PSC 833
.
Br J Cancer
.
1997
;
76
:
1181
1183
.
89

van Asperen
 
J
,
van Tellingen
 
O
,
van der Valk
 
MA
et al.

Enhanced oral absorption and decreased elimination of paclitaxel in mice cotreated with cyclosporin A
.
Clin Cancer Res
.
1998
;
4
:
2293
2297
.
90

Bardelmeijer
 
HA
,
Beijnen
 
JH
,
Brouwer
 
KR
et al.

Increased oral bioavailability of paclitaxel by GF120918 in mice through selective modulation of P-glycoprotein
.
Clin Cancer Res
.
2000
;
6
:
4416
4421
.
91

Lankas
 
GR
,
Wise
 
LD
,
Cartwright
 
ME
et al.

Placental P-glycoprotein deficiency enhances susceptibility to chemically induced birth defects in mice
.
Reprod Toxicol
.
1998
;
12
:
457
463
.
92

Smit
 
JW
,
Huisman
 
MT
,
van Tellingen
 
O
et al.

Absence or pharmacological blocking of placental P-glycoprotein profoundly increases fetal drug exposure
.
J Clin Invest
.
1999
;
104
:
1441
1447
.
93

Sudhakaran
 
S
,
Ghabrial
 
H
,
Nation
 
RL
et al.

Differential bidirectional transfer of indinavir in the isolated perfused human placenta
.
Antimicrob Agents Chemother
.
2005
;
49
:
1023
1028
.
94

Molsa
 
M
,
Heikkinen
 
T
,
Hakkola
 
J
et al.

Functional role of P-glycoprotein in the human blood-placental barrier
.
Clin Pharmacol Ther
.
2005
;
78
:
123
131
.
95

Wacher
 
VJ
,
Wu
 
CY
,
Benet
 
LZ
,

Overlapping substrate specificities and tissue distribution of cytochrome P450 3A and P-glycoprotein: Implications for drug delivery and activity in cancer chemotherapy
.
Mol Carcinog
.
1995
;
13
:
129
134
.
96

Watkins
 
PB
,

The barrier function of CYP3A4 and P-glycoprotein in the small bowel
.
Adv Drug Deliv Rev
.
1997
;
27
:
161
170
.
97

Bigger
 
JT
 Jr,
Leahey
 
EB
 Jr
,

Quinidine and digoxin: An important interaction
.
Drugs
.
1982
;
24
:
229
239
.
98

Leahey
 
EB
 Jr,
Reiffel
 
JA
,
Heissenbuttel
 
RH
et al.

Enhanced cardiac effect of digoxin during quinidine treatment
.
Arch Intern Med
.
1979
;
139
:
519
521
.
99

Doering
 
W
,

Quinidine-digoxin interaction: Pharmacokinetics, underlying mechanism and clinical implications
.
N Engl J Med
.
1979
;
301
:
400
404
.
100

Klein
 
HO
,
Lang
 
R
,
Weiss
 
E
et al.

The influence of verapamil on serum digoxin concentration
.
Circulation
.
1982
;
65
:
998
1003
.
101

Belz
 
GG
,
Doering
 
W
,
Munkes
 
R
et al.

Interaction between digoxin and calcium antagonists and antiarrhythmic drugs
.
Clin Pharmacol Ther
.
1983
;
33
:
410
417
.
102

Verschraagen
 
M
,
Koks
 
CHW
,
Schellens
 
JHM
et al.

P-glycoprotein system as a determinant of drug interactions: The case of digoxin-verapamil
.
Pharmacol Res
.
1999
;
40
:
301
306
.
103

Westphal
 
K
,
Weinbrenner
 
A
,
Giessmann
 
T
et al.

Oral bioavailability of digoxin is enhanced by talinolol: Evidence for involvement of intestinal P-glycoprotein
.
Clin Pharmacol Ther
.
2000
;
68
:
6
12
.
104

Wakasugi
 
H
,
Yano
 
I
,
Ito
 
T
et al.

Effect of clarithromycin on renal excretion of digoxin: Interaction with P-glycoprotein
.
Clin Pharmacol Ther
.
1998
;
64
:
123
128
.
105

Jalava
 
KM
,
Partanen
 
J
,
Neuvonen
 
PJ
,

Itraconazole decreases renal clearance of digoxin
.
Ther Drug Monit
.
1997
;
19
:
609
613
.
106

Maxwell
 
DL
,
Gilmour-White
 
SK
,
Hall
 
MR
,

Digoxin toxicity due to interaction of digoxin with erythromycin
.
BMJ
.
1989
;
298
:
572
.
107

Woodland
 
C
,
Verjee
 
Z
,
Giesbrecht
 
E
et al.

The digoxin-propafenone interaction: Characterization of a mechanism using renal tubular cell monolayers
.
J Pharmacol Exp Ther
.
1997
;
283
:
39
45
.
108

Calvo
 
MV
,
Martin-Suarez
 
A
,
Martin Luengo
 
C
et al.

Interaction between digoxin and propafenone
.
Ther Drug Monit
.
1989
;
11
:
10
15
.
109

Westphal
 
K
,
Weinbrenner
 
A
,
Zschiesche
 
M
et al.

Induction of P-glycoprotein by rifampin increases intestinal secretion of talinolol in human beings: A new type of drug/drug interaction
.
Clin Pharmacol Ther
.
2000
;
68
:
345
355
.
110

Hamman
 
MA
,
Bruce
 
MA
,
Haehner-Daniels
 
BD
et al.

The effect of rifampin administration on the disposition of fexofenadine
.
Clin Pharmacol Ther
.
2001
;
69
:
114
121
.
111

Hebert
 
MF
,
Roberts
 
JP
,
Prueksaritanont
 
T
et al.

Bioavailability of cyclosporine with concomitant rifampin administration is markedly less than predicted by hepatic enzyme induction
.
Clin Pharmacol Ther
.
1992
;
52
:
453
457
.
112

Woodcock
 
DM
,
Linsenmeyer
 
ME
,
Chojnowski
 
G
et al.

Reversal of multidrug resistance by surfactants
.
Br J Cancer
.
1992
;
66
:
62
68
.
113

Martin-Facklam
 
M
,
Burhenne
 
J
,
Ding
 
R
et al.

Dose-dependent increase of saquinavir bioavailability by the pharmaceutic aid cremophor EL
.
Br J Clin Pharmacol
.
2002
;
53
:
576
581
.
114

Tayrouz
 
Y
,
Ding
 
R
,
Burhenne
 
J
et al.

Pharmacokinetic and pharmaceutic interaction between digoxin and Cremophor RH40
.
Clin Pharmacol Ther
.
2003
;
73
:
397
405
.
115

Spahn-Langguth
 
H
,
Langguth
 
P
,

Grapefruit juice enhances intestinal absorption of the P-glycoprotein substrate talinolol
.
Eur J Pharm Sci
.
2001
;
12
:
361
367
.
116

Ohnishi
 
A
,
Matsuo
 
H
,
Yamada
 
S
et al.

Effect of furanocoumarin derivatives in grapefruit juice on the uptake of vinblastine by Caco-2 cells and on the activity of cytochrome P450 3A4
.
Br J Pharmacol
.
2000
;
130
:
1369
1377
.
117

Eagling
 
VA
,
Profit
 
L
,
Back
 
DJ
,

Inhibition of the CYP3A4-mediated metabolism and P-glycoprotein-mediated transport of the HIV-1 protease inhibitor saquinavir by grapefruit juice components
.
Br J Clin Pharmacol
.
1999
;
48
:
543
552
.
118

Reif
 
S
,
Nicolson
 
MC
,
Bisset
 
D
et al.

Effect of grapefruit juice intake on etoposide bioavailability
.
Eur J Clin Pharmacol
.
2002
;
58
:
491
494
.
119

Schwarz
 
UI
,
Seemann
 
D
,
Oertel
 
R
et al.

Grapefruit juice ingestion significantly reduces talinolol bioavailability
.
Clin Pharmacol Ther
.
2005
;
77
:
291
301
.
120

De Castro
 
WV
,
Mertens-Talcott
 
S
,
Rubner
 
A
et al.

Variation of flavonoids and furanocoumarins in grapefruit juices: A potential source of variability in grapefruit juice-drug interaction studies
.
J Agric Food Chem
.
2006
;
54
:
249
255
.
121

Ruschitzka
 
F
,
Meier
 
PJ
,
Turina
 
M
et al.

Acute heart transplant rejection due to Saint John's wort
.
Lancet
.
2000
;
355
:
548
549
.
122

Bauer
 
S
,
Stormer
 
E
,
Johne
 
A
et al.

Alterations in cyclosporin A pharmacokinetics and metabolism during treatment with St John's wort in renal transplant patients
.
Br J Clin Pharmacol
.
2003
;
55
:
203
211
.
123

Piscitelli
 
SC
,
Burstein
 
AH
,
Chaitt
 
D
et al.

Indinavir concentrations and St John's wort
.
Lancet
.
2000
;
355
:
547
548
.
124

Mathijssen
 
RHJ
,
Verweij
 
J
,
de Bruijn
 
P
et al.

Effects of St. John's wort on irinotecan metabolism
.
J Natl Cancer Inst
.
2002
;
94
:
1247
1249
.
125

Hu
 
Z
,
Yang
 
X
,
Ho
 
PC-L
et al.

St. John's wort modulates the toxicities and pharmacokinetics of CPT-11 (irinotecan) in rats
.
Pharm Res
.
2005
;
22
:
902
914
.
126

Smith
 
P
,
Bullock
 
JM
,
Booker
 
BM
et al.

The influence of St. John's wort on the pharmacokinetics and protein binding of imatinib mesylate
.
Pharmacotherapy
.
2004
;
24
:
1508
1514
.
127

Frye
 
RF
,
Fitzgerald
 
SM
,
Lagattuta
 
TF
et al.

Effect of St John's wort on imatinib mesylate pharmacokinetics
.
Clin Pharmacol Ther
.
2004
;
76
:
323
329
.
128

Zhou
 
S
,
Chan
 
E
,
Pan
 
SQ
et al.

Pharmacokinetic interactions of drugs with St John's wort
.
J Psychopharmacol
.
2004
;
18
:
262
276
.
129

Schwarz
 
UI
,
Buschel
 
B
,
Kirch
 
W
,

Unwanted pregnancy on self-medication with St John's wort despite hormonal contraception
.
Br J Clin Pharmacol
.
2003
;
55
:
112
113
.
130

Dresser
 
GK
,
Schwarz
 
UI
,
Wilkinson
 
GR
et al.

Coordinate induction of both cytochrome P4503A and MDR1 by St John's wort in healthy subjects
.
Clin Pharmacol Ther
.
2003
;
73
:
41
50
.
131

Wang
 
Z
,
Hamman
 
MA
,
Huang
 
SM
et al.

Effect of St John's wort on the pharmacokinetics of fexofenadine
.
Clin Pharmacol Ther
.
2002
;
71
:
414
420
.
132

Mai
 
I
,
Stormer
 
E
,
Bauer
 
S
et al.

Impact of St John's wort treatment on the pharmacokinetics of tacrolimus and mycophenolic acid in renal transplant patients
.
Nephrol Dial Transplant
.
2003
;
18
:
819
822
.
133

Meijerman
 
I
,
Beijnen
 
JH
,
Schellens
 
JHM
,

Herb-drug interactions in oncology: Focus on mechanisms of induction
.
The Oncologist
.
2006
;
11
:
742
752
.
134

Tannergren
 
C
,
Engman
 
H
,
Knutson
 
L
et al.

St John's wort decreases the bioavailability of R- and S-verapamil through induction of the first-pass metabolism
.
Clin Pharmacol Ther
.
2004
;
75
:
298
309
.
135

Hall
 
SD
,
Wang
 
Z
,
Huang
 
SM
et al.

The interaction between St John's wort and an oral contraceptive
.
Clin Pharmacol Ther
.
2003
;
74
:
525
535
.
136

Henthorn
 
TK
,
Liu
 
Y
,
Mahapatro
 
M
et al.

Active transport of fentanyl by the blood-brain barrier
.
J Pharmacol Exp Ther
.
1999
;
289
:
1084
1089
.
137

Kharasch
 
ED
,
Hoffer
 
C
,
Altuntas
 
TG
et al.

Quinidine as a probe for the role of P-glycoprotein in the intestinal absorption and clinical effects of fentanyl
.
J Clin Pharmacol
.
2004
;
44
:
224
233
.
138

Dagenais
 
C
,
Graff
 
CL
,
Pollack
 
GM
,

Variable modulation of opioid brain uptake by P-glycoprotein in mice
.
Biochem Pharmacol
.
2004
;
67
:
269
276
.
139

Rogan
 
AM
,
Hamilton
 
TC
,
Young
 
RC
et al.

Reversal of adriamycin resistance by verapamil in human ovarian cancer
.
Science
.
1984
;
224
:
994
996
.
140

Fisher
 
GA
,
Sikic
 
BI
,

Clinical studies with modulators of multidrug resistance
.
Hematol Oncol Clin North Am
.
1995
;
9
:
363
382
.
141

List
 
AF
,
Spier
 
C
,
Greer
 
J
et al.

Phase I/II trial of cyclosporine as a chemotherapy-resistance modifier in acute leukemia
.
J Clin Oncol
.
1993
;
11
:
1652
1660
.
142

Chan
 
HS
,
DeBoer
 
G
,
Thiessen
 
JJ
et al.

Combining cyclosporin with chemotherapy controls intraocular retinoblastoma without requiring radiation
.
Clin Cancer Res
.
1996
;
2
:
1499
1508
.
143

List
 
AF
,

Multidrug resistance: Clinical relevance in acute leukemia
.
Oncology (Williston Park)
.
1993
;
7
:
23
28
discussion 32, 35–38.
144

List
 
AF
,
Kopecky
 
KJ
,
Willman
 
CL
et al.

Benefit of cyclosporine modulation of drug resistance in patients with poor-risk acute myeloid leukemia: A Southwest Oncology Group study
.
Blood
.
2001
;
98
:
3212
3220
.
145

Belpomme
 
D
,
Gauthier
 
S
,
Pujade-Lauraine
 
E
et al.

Verapamil increases the survival of patients with anthracycline-resistant metastatic breast carcinoma
.
Ann Oncol
.
2000
;
11
:
1471
1476
.
146

Millward
 
MJ
,
Cantwell
 
BM
,
Munro
 
NC
et al.

Oral verapamil with chemotherapy for advanced non-small cell lung cancer: A randomised study
.
Br J Cancer
.
1993
;
67
:
1031
1035
.
147

Dalton
 
WS
,
Crowley
 
JJ
,
Salmon
 
SS
et al.

A phase III randomized study of oral verapamil as a chemosensitizer to reverse drug resistance in patients with refractory myeloma. A Southwest Oncology Group study
.
Cancer
.
1995
;
75
:
815
820
.
148

Milroy
 
R
,

A randomised clinical study of verapamil in addition to combination chemotherapy in small cell lung cancer. West of Scotland Lung Cancer Research Group, and the Aberdeen Oncology Group
.
Br J Cancer
.
1993
;
68
:
813
818
.
149

Wishart
 
GC
,
Bissett
 
D
,
Paul
 
J
et al.

Quinidine as a resistance modulator of epirubicin in advanced breast cancer: Mature results of a placebo-controlled randomized trial
.
J Clin Oncol
.
1994
;
12
:
1771
1777
.
150

Sonneveld
 
P
,
Suciu
 
S
,
Weijermans
 
P
et al.

Cyclosporin A combined with vincristine, doxorubicin and dexamethasone (VAD) compared with VAD alone in patients with advanced refractory multiple myeloma: An EORTC-HOVON randomized phase III study (06914)
.
Br J Haematol
.
2001
;
115
:
895
902
.
151

Solary
 
E
,
Drenou
 
B
,
Campos
 
L
et al.

Quinine as a multidrug resistance inhibitor: A phase 3 multicentric randomized study in adult de novo acute myelogenous leukemia
.
Blood
.
2003
;
102
:
1202
1210
.
152

Friedenberg
 
WR
,
Rue
 
M
,
Blood
 
EA
et al.

Phase III study of PSC-833 (valspodar) in combination with vincristine, doxorubicin, and dexamethasone (valspodar/VAD) versus VAD alone in patients with recurring or refractory multiple myeloma (E1A95): A trial of the Eastern Cooperative Oncology Group
.
Cancer
.
2006
;
106
:
830
838
.
153

Greenberg
 
PL
,
Lee
 
SJ
,
Advani
 
R
et al.

Mitoxantrone, etoposide, and cytarabine with or without valspodar in patients with relapsed or refractory acute myeloid leukemia and high-risk myelodysplastic syndrome: A phase III trial (E2995)
.
J Clin Oncol
.
2004
;
22
:
1078
1086
.
154

Fracasso
 
PM
,
Goldstein
 
LJ
,
de Alwis
 
DP
et al.

Phase I study of docetaxel in combination with the P-glycoprotein inhibitor, zosuquidar, in resistant malignancies
.
Clin Cancer Res
.
2004
;
10
:
7220
7228
.
155

Chico
 
I
,
Kang
 
MH
,
Bergan
 
R
et al.

Phase I study of infusional paclitaxel in combination with the P-glycoprotein antagonist PSC 833
.
J Clin Oncol
.
2001
;
19
:
832
842
.
156

Carlson
 
RW
,
O'Neill
 
AM
,
Goldstein
 
LJ
et al.

A pilot phase II trial of valspodar modulation of multidrug resistance to paclitaxel in the treatment of metastatic carcinoma of the breast (E1195): A trial of the Eastern Cooperative Oncology Group
.
Cancer Invest
.
2006
;
24
:
677
681
.
157

Meerum Terwogt
 
JM
,
Beijnen
 
JH
,
Malingre
 
MM
et al.

Coadministration of oral cyclosporin A enables oral therapy with paclitaxel
.
Clin Cancer Res
.
1999
;
5
:
3379
3384
.
158

Malingrè
 
MM
,
Richel
 
DJ
,
Beijnen
 
JH
et al.

Coadministration of cyclosporine strongly enhances the oral bioavailability of docetaxel
.
J Clin Oncol
.
2001
;
19
:
1160
1166
.
159

Kruijtzer
 
CMF
,
Beijnen
 
JH
,
Rosing
 
H
et al.

Increased oral bioavailability of topotecan in combination with the breast cancer resistance protein (BCRP) and P-glycoprotein inhibitor GF120918
.
J Clin Oncol
.
2002
;
20
:
2943
2950
.
160

Kruijtzer
 
CMF
,
Beijnen
 
JH
,
Schellens
 
JHM
,

Improvement of oral drug treatment by temporary inhibition of drug transporters and/or cytochrome P450 in the gastrointestinal tract and liver: An overview
.
The Oncologist
.
2002
;
7
:
516
530
.
161

Kruijtzer
 
CMF
,
Schellens
 
JHM
,
Mezger
 
J
et al.

Phase II and pharmacologic study of weekly oral paclitaxel plus cyclosporine in patients with advanced non-small-cell lung cancer
.
J Clin Oncol
.
2002
;
20
:
4508
4516
.
162

Helgason
 
HH
,
Kruijtzer
 
CM
,
Huitema
 
AD
et al.

Phase II and pharmacological study of oral paclitaxel (Paxoral) plus cyclosporin in anthracycline-pretreated metastatic breast cancer
.
Br J Cancer
.
2006
;
95
:
794
800
.
163

Kruijtzer
 
CM
,
Boot
 
H
,
Beijnen
 
JH
et al.

Weekly oral paclitaxel as first-line treatment in patients with advanced gastric cancer
.
Ann Oncol
.
2003
;
14
:
197
204
.
164

van Asperen
 
J
,
Mayer
 
U
,
van Tellingen
 
O
et al.

The functional role of P-glycoprotein in the blood-brain barrier
.
J Pharm Sci
.
1997
;
86
:
881
884
.
165

Kemper
 
EM
,
van Zandbergen
 
AE
,
Cleypool
 
C
et al.

Increased penetration of paclitaxel into the brain by inhibition of P-Glycoprotein
.
Clin Cancer Res
.
2003
;
9
:
2849
2855
.
166

Kemper
 
EM
,
Boogerd
 
W
,
Thuis
 
I
et al.

Modulation of the blood-brain barrier in oncology: Therapeutic opportunities for the treatment of brain tumours?
.
Cancer Treat Rev
.
2004
;
30
:
415
423
.
167

Kemper
 
EM
,
Verheij
 
M
,
Boogerd
 
W
et al.

Improved penetration of docetaxel into the brain by co-administration of inhibitors of P-glycoprotein
.
Eur J Cancer
.
2004
;
40
:
1269
1274
.
168

Dai
 
H
,
Marbach
 
P
,
Lemaire
 
M
et al.

Distribution of STI-571 to the brain is limited by P-glycoprotein-mediated efflux
.
J Pharmacol Exp Ther
.
2003
;
304
:
1085
1092
.
169

Fellner
 
S
,
Bauer
 
B
,
Miller
 
DS
et al.

Transport of paclitaxel (Taxol) across the blood-brain barrier in vitro and in vivo
.
J Clin Invest
.
2002
;
110
:
1309
1318
.
170

Breedveld
 
P
,
Beijnen
 
JH
,
Schellens
 
JHM
,

Use of P-glycoprotein and BCRP inhibitors to improve oral bioavailability and CNS penetration of anticancer drugs
.
Trends Pharmacol Sci
.
2006
;
27
:
17
24
.
171

Kilic
 
T
,
Alberta
 
JA
,
Zdunek
 
PR
et al.

Intracranial inhibition of platelet-derived growth factor-mediated glioblastoma cell growth by an orally active kinase inhibitor of the 2-phenylaminopyrimidine class
.
Cancer Res
.
2000
;
60
:
5143
5150
.
172

Petzer
 
AL
,
Gunsilius
 
E
,
Hayes
 
M
et al.

Low concentrations of STI571 in the cerebrospinal fluid: A case report
.
Br J Haematol
.
2002
;
117
:
623
625
.
173

Takayama
 
N
,
Sato
 
N
,
O'Brien
 
SG
et al.

Imatinib mesylate has limited activity against the central nervous system involvement of Philadelphia chromosome-positive acute lymphoblastic leukaemia due to poor penetration into cerebrospinal fluid
.
Br J Haematol
.
2002
;
119
:
106
108
.
174

Breedveld
 
P
,
Pluim
 
D
,
Cipriani
 
G
et al.

The effect of Bcrp1 (Abcg2) on the in vivo pharmacokinetics and brain penetration of imatinib mesylate (Gleevec): Implications for the use of breast cancer resistance protein and P-glycoprotein inhibitors to enable the brain penetration of imatinib in patients
.
Cancer Res
.
2005
;
65
:
2577
2582
.
175

Sadeque
 
AJ
,
Wandel
 
C
,
He
 
H
et al.

Increased drug delivery to the brain by P-glycoprotein inhibition
.
Clin Pharmacol Ther
.
2000
;
68
:
231
237
.
176

Huisman
 
MT
,
Smit
 
JW
,
Schinkel
 
AH
,

Significance of P-glycoprotein for the pharmacology and clinical use of HIV protease inhibitors
.
AIDS
.
2000
;
14
:
237
242
.
177

Morton
 
MR
,
Cooper
 
JW
,

Erythromycin-induced digoxin toxicity
.
DICP
.
1989
;
23
:
668
670
.
178

Woodland
 
C
,
Ito
 
S
,
Koren
 
G
,

A model for the prediction of digoxin-drug interactions at the renal tubular cell level
.
Ther Drug Monit
.
1998
;
20
:
134
138
.
179

Phillips
 
EJ
,
Rachlis
 
AR
,
Ito
 
S
,

Digoxin toxicity and ritonavir: A drug interaction mediated through P-glycoprotein?
.
AIDS
.
2003
;
17
:
1577
1578
.
180

Ding
 
R
,
Tayrouz
 
Y
,
Riedel
 
KD
et al.

Substantial pharmacokinetic interaction between digoxin and ritonavir in healthy volunteers
.
Clin Pharmacol Ther
.
2004
;
76
:
73
84
.
181

Malingrè
 
MM
,
Beijnen
 
JH
,
Rosing
 
H
et al.

Co-administration of GF120918 significantly increases the systemic exposure to oral paclitaxel in cancer patients
.
Br J Cancer
.
2001
;
84
:
42
47
.
182

Hsu
 
A
,
Granneman
 
GR
,
Cao
 
G
et al.

Pharmacokinetic interactions between two human immunodeficiency virus protease inhibitors, ritonavir and saquinavir
.
Clin Pharmacol Ther
.
1998
;
63
:
453
464
.
183

Kempf
 
DJ
,
Marsh
 
KC
,
Kumar
 
G
et al.

Pharmacokinetic enhancement of inhibitors of the human immunodeficiency virus protease by coadministration with ritonavir
.
Antimicrob Agents Chemother
.
1997
;
41
:
654
660
.
184

Hebert
 
MF
,
Lam
 
AY
,

Diltiazem increases tacrolimus concentrations
.
Ann Pharmacother
.
1999
;
33
:
680
682
.
185

Schwarz
 
UI
,
Gramatte
 
T
,
Krappweis
 
J
et al.

P-glycoprotein inhibitor erythromycin increases oral bioavailability of talinolol in humans
.
Int J Clin Pharmacol Ther
.
2000
;
38
:
161
167
.
186

Gupta
 
SK
,
Bakran
 
A
,
Johnson
 
RW
et al.

Cyclosporin-erythromycin interaction in renal transplant patients
.
Br J Clin Pharmacol
.
1989
;
27
:
475
481
.
187

Gupta
 
SK
,
Bakran
 
A
,
Johnson
 
RW
et al.

Erythromycin enhances the absorption of cyclosporin
.
Br J Clin Pharmacol
.
1988
;
25
:
401
402
.
188

Hebert
 
MF
,
Fisher
 
RM
,
Marsh
 
CL
et al.

Effects of rifampin on tacrolimus pharmacokinetics in healthy volunteers
.
J Clin Pharmacol
.
1999
;
39
:
91
96
.
189

Johne
 
A
,
Brockmoller
 
J
,
Bauer
 
S
et al.

Pharmacokinetic interaction of digoxin with an herbal extract from St John's wort (Hypericum perforatum)
.
Clin Pharmacol Ther
.
1999
;
66
:
338
345
.
190

Hebert
 
MF
,
Park
 
JM
,
Chen
 
YL
et al.

Effects of St John's wort (Hypericum perforatum) on tacrolimus pharmacokinetics in healthy volunteers
.
J Clin Pharmacol
.
2004
;
44
:
89
94
.
191

Joerger
 
M
,
Huitema
 
AD
,
van den Bongard
 
HJ
et al.

Determinants of the elimination of methotrexate and 7-hydroxy-methotrexate following high-dose infusional therapy to cancer patients
.
Br J Clin Pharmacol
.
2006
;
62
:
71
80
.
192

Meerum Terwogt
 
JM
,
Beijnen
 
JH
,
ten Bokkel Huinink
 
WW
et al.

Co-administration of oral cyclosporin enables oral therapy with paclitaxel
.
Lancet
.
1998
;
352
:
285
.

Author notes

Disclosure of potential conflicts of interest is found at the end of this article.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)