Abstract
ATP-binding cassette (ABC) transporters, P-glycoprotein (P-gp, ABCB1) and ABCG2, are membrane proteins that couple the energy derived from ATP hydrolysis to efflux many chemically diverse compounds across the plasma membrane, thereby playing a critical and important physiological role in protecting cells from xenobiotics. These transporters are also implicated in the development of multidrug resistance (MDR) in cancer cells that have been treated with chemotherapeutics. One approach to blocking the efflux capability of an ABC transporter in a cell or tissue is inhibiting the activity of the transporters with a modulator. Since ABC transporter modulators can be used in combination with chemotherapeutics to increase the effective intracellular concentration of anticancer drugs, the possible impact of modulators of ABC drug transporters is of great clinical interest. Another possible clinical use of modulators that has recently attracted attention is their ability to increase oral bioavailability or increase tissue penetration of drugs transported by the transporters. Several preclinical and clinical studies have been performed to evaluate the feasibility and the safety of this approach. The primary focus of this review is to discuss progress made in recent years in the identification and applicability of compounds that may serve as ABC transporter modulators and the possible role of these compounds in altering the pharmacokinetics and pharmacodynamics of therapeutic drugs used in the clinic.
Keywords: ABC transporters, ABCG2, blood-brain barrier, chemotherapy, modulators, multidrug resistance, oral bioavailability, P-glycoprotein
Multidrug resistance and ABC drug transporters
Chemotherapy is usually the most effective treatment for cancer patients with advanced/metastatic tumors or hematological malignancies. However, cancer cells often develop simultaneous resistance to many functionally and structurally unrelated anti-cancer drugs, a phenomenon known as multidrug resistance (MDR), which is a major problem in the treatment of cancer (1). Cancer cells with the MDR phenotype may have either inherent resistance to anti-cancer drugs or resistance acquired after cycles of chemotherapy. Several mechanisms of anti-cancer drug resistance, such as reduced cellular drug uptake, increased or decreased expression of metabolic enzymes, mutation of the target, alterations of the apoptotic pathway, changes in cellular repair mechanisms and increased expression/activity of drug efflux pumps have been characterized (2).
One of the most important MDR mechanisms is an increased efflux rate of the anti-cancer drug from cancer cells by the members of the superfamily of ATP-binding cassette (ABC) transporters, which is one of the largest protein families in living organisms (3, 4). There are 48 genes in the human genome that encode ABC transporters, which are divided into seven subfamilies (ABCA-ABCG) based on the amino acid sequence identity of ATP-binding domains (4). ATP-binding domains, also known as nucleotide binding domains (NBDs), and trans-membrane domains (TMDs), each with six alpha helices, are the essential units of an ABC transporter. The NBD has the highly conserved Walker A, B, and signature C motifs and are well conserved in all the organisms. On the other hand, the TMD confers transport substrate specificity, and is not well conserved (5, 6). The members of this family play a crucial role in physiology, and mutations in ABC transporters result in several human diseases including progressive familial intrahepatic cholestasis (ABCB11), Dubin-Johnson syndrome (ABCC2), cystic fibrosis (ABCC7), and adrenoleukodystrophy (ABCD1) (3, 4). So far, at least 15 transporters have been shown to export anti-cancer drugs in vitro (7). Among them, P-glycoprotein (P-gp; MDR1, ABCB1), multidrug resistance-associated protein 1 (MRP1, ABCC1), and ABCG2 (breast cancer resistance protein; BCRP, mitoxantrone resistance protein; MXR) are considered major players in the development of MDR in cancer cells.
P-gp, discovered in 1976, is one of the best characterized ABC transporters (8). It is composed of two homologous halves, each containing a NBD and a TMD, and transports exogenous and endogenous amphipathic substrates out of cells using energy from ATP (9). It is localized at the apical surface of the cells and is highly expressed in capillary endothelial cells of the blood-brain barrier, placental trophoblasts, the testes, intestines, the liver, kidneys and the adrenal gland (3). These tissues function as barriers, suggesting the physiological role of P-gp is to protect the body from xenobiotics and toxins. P-gp pumps out many structurally unrelated anti-cancer drugs, such as vinca alkaloids (vinblastine, vincristine, vindesine, vinorelbine), anthracyclines (doxorubicin, daunorubicin) and taxanes (paclitaxel, docetaxel), suggesting the flexible nature of the substrate binding site of P-gp (10, 11). P-gp is highly expressed in leukemia, breast, ovarian, colon, kidney, adrenocortical, and hepatocellular cancers and its overexpression is inversely correlated with poor clinical prognosis (12–14).
ABCG2 is a half transporter which contains one TMD and one NBD, and is therefore thought to homodimerize or heterodimerize to form the functional unit (15–17). Interestingly, similar to the MDR family of transporters in yeast, the location of the TMD and NBD is reversed in ABCG2 compared to P-gp (18). Similar to P-gp, ABCG2 is localized to the apical membrane in epithelial cells and normally expressed in organs such as the placenta, brain, liver, prostate, and intestine (16). ABCG2 is also detected in hematopoietic and other stem cells, suggesting that it may play an important role in the protective function of pluripotent stem cells (19). Overexpression of ABCG2 renders cancer cells resistant to many anti-cancer drugs including mitoxantrone, topotecan and methotrexate and it is associated with poor response to chemotherapy in leukemia and breast cancer patients (20, 21).
MRP1 (ABCC1) was the first member of the MRP family to be identified (in 1992) and has been linked to the development of MDR (22). The structure of MRP1 is similar to that of P-gp, except five additional transmembrane helices are present at the amino-terminal end of the transporter. It is highly expressed in the adrenal gland, bladder, choroid plexus, colon, in erythrocytes, bone marrow, the kidneys, lungs, placenta, spleen, stomach, testes, in helper T cells and in muscle cells (23). MRP1 transports some substrates conjugated with glucuronide, sulfate or glutathione, vinca alkaloids, anthracyclines, methotrexate and also leukotriene C4, which is an endogenous substrate for the transporter (24, 25). The localization of MRP1 is different from that of P-gp, as it is expressed in the basolateral membrane in polarized epithelial cells and transports substrates in to the bloodstream (26). Overexpression of MRP1 has also been shown in lung, breast, prostate, and ovarian cancer, gastrointestinal carcinoma, melanoma, and leukemia (27). While some studies have reported MRP1 expression levels to be of prognostic significance (28, 29), others have found no correlation between clinical outcome and its expression (30, 31). A comprehensive role of MRP1 in clinical drug resistance is still debatable; therefore the present review will mainly focus on two major ABC drug transporters, P-gp and ABCG2.
Approaches to improving chemotherapy
A combination of two or multiple drugs is often used in chemotherapy, as each drug inhibits a specific target and the combination therefore could maximize the killing effect on cancer cells, additively and synergistically (32). The combination of drugs targets several cellular pathways simultaneously, which not only augments the tumoricidal effect of anti-cancer drugs, but also lessens the occurrence of drug resistance, thereby providing an optimal therapeutic outcome (2).
In addition to conventional anti-cancer drugs, many novel drugs have been developed based on an understanding of molecular mechanisms, which revealed a number of new potential targets for drugs. These novel agents such as the monoclonal antibody cetuximab, which targets the extracellular domain of EGFR by blocking ligand binding, and the small-molecule inhibitor erlotinib, that blocks EGFR intracellular tyrosine kinase activity, have demonstrated clinical advantages over the established drug regimen (33). Several newer approaches such as cancer vaccines, antisense oligonucleotides, and small-interfering RNAs (siRNAs) are also promising approaches for targeting cancer cells and deserve further clinical investigation (33). These approaches show a great promise for improving the efficiency of chemotherapy, but are still not sufficient to alter the outcome for many cancer patients.
ABC drug transporters: Targets for improving chemotherapy
MDR mediated by ABC transporters in cancer cells can be overcome by methods such as specific inhibition of transporters at the cell surface level, blocking the signaling pathways that control amplification and overexpression of these transporters, and targeting the transcription factors that regulate the expression of the pumps (34). Historically, research on the reversal of ABC transporter-mediated MDR has been directed towards inhibiting the activity of the transporter at the cell surface by drugs known as modulators or inhibitors and due to the scope of this review, we will focus on this strategy as a possible way to improve chemotherapy and/or oral bioavailability. A majority of modulator drugs used for this purpose are basically transport substrates of ABC drug transporters that inhibit efflux of anti-cancer agents in both in vitro and in vivo systems.
Almost three decades ago, Tsuruo et al. discovered that verapamil (a calcium channel blocker used as a coronary vasodilator) enhanced the cytotoxicity of vincristine and vinblastine in a P-gp-expressing vincristine-resistant cell line (35). Slater et al. later reported that cyclosporine A (CsA), an immunosuppressant drug, completely reversed primary resistance to vincristine and daunorubicin in a drug-resistant cell line of human T cell acute lymphatic leukemia (36). Additional studies showed that both verapamil and CsA reversed the MDR phenotype by interacting directly at the drug-substrate site on P-gp (37–39). Several other ‘off the shelf’ drugs which were already in clinical use such as nicardipine and nifedipine were shown to reverse Pgp-mediated MDR (reviewed in (34)) but many of these agents were toxic, as higher concentrations of these drugs were required for inhibiting the transporters. These drugs have been described as the first generation modulators of P-gp. In 1991, an analog of CsA PSC833 (valspodar) was synthesized which was not only 10-fold more potent than CsA in inhibiting Pgp activity but also showed no immunosuppressive side effect (40). Discovery of this molecule marked the beginning of the second generation of ABC transporter modulators. Many other modulators were developed based on the quantitative structural activity relationship approach. A number of clinical studies were carried out using these modulators. These trials demonstrated some advantage over the previous agents, but patients still suffered from side effects associated with the therapy (41–44). Since then, third generation modulators such as tariquidar (XR9576) (45), elacridar (GF120918) (46), zosuquidar (LY335979) (47) and dofequidar (48) have been developed which have shown improved selectivity and inhibitory activity towards ABC transporters. Martin et al. reported that tariquidar, unlike verapamil and CsA, was not a transport substrate of P-gp, and that it inhibits P-gp function by binding at a site distinct from the vinblastine and paclitaxel site (49). Tariquidar potentiated the cytotoxicity of several drugs including doxorubicin, paclitaxel, etoposide, and vincristine at very low concentrations (25–80 nM). Co-administration of tariquidar potentiated the antitumor activity of doxorubicin without a significant increase in toxicity in mice (50). Two Phase III trials of tariquidar in combination with paclitaxel/carboplatin, or vinorelbine were initiated in patients with non-small-cell lung cancer (NSCLC) but these studies were terminated later owing to chemotherapy-related toxicity in the tariquidar arm (51). Recent results of another phase I study with vinorelbine have indicated shown that tariquidar is a potent P-gp antagonist, without significant side effects and much less pharmacokinetic interaction than previous P-gp antagonists (52). This and other ongoing clinical studies at the National Cancer Institute (NIH, Bethesda, USA) with tariquidar suggest that there is still optimism that therapeutic modulation of MDR mediated by ABC transporters may be beneficial to patients.
The leukotriene LTD4 receptor antagonist MK571 and a fungal toxin, fumitremorgin C (FTC) were identified as specific modulators of MRP1 and ABCG2, respectively (53, 54). Since then MK571 and FTC have been used as standard modulators for inhibiting MRP1- and ABCG2-mediated transport, respectively, in laboratory assays. However, unlike P-gp, clinically useful modulators of MRP1 and ABCG2 have not been extensively investigated in clinical trials. Some modulators of P-gp such as biricodar (VX-710) dofequidar (MS-209), elacridar (GF120918) or CBT-1 have also been shown to inhibit MRP-1 and/or ABCG2-mediated drug transport/resistance in in vitro and in clinical studies (48, 55–61). Although the effect of these modulators on each transporter is not well established in clinical trials, these drugs might be useful against cancer cells which express a combination of transporters linked to MDR. The efficacy and safety of these modulators in clinical studies is still under evaluation.
Newer approaches have been developed in the laboratory to overcome ABC transporter-mediated MDR, including a monoclonal antibody that binds to human P-gp and inhibits drug transport (62), siRNA technology that specifically decreases the expression of ABCB1 (63), and regulation of transcription factors that inhibit P-gp activation (64). Although these approaches might help to overcome transporter-mediated MDR, clinical studies using these alternative approaches are still lacking to establish their efficacy in cancer patients.
Modulators of ABC drug transporters and oral bioavailability of chemotherapeutics
Although modulators of ABC drug transporters were initially envisioned as drugs that can be used to overcome ABC transporter-mediated MDR in cancer cells or tumor tissue, these drugs are recapturing attention for another possible use: increasing oral bioavailability of drugs which were earlier ruled out as oral chemotherapeutic agents due to poor bioavailability (65). Pgp and ABCG2 have now been confirmed to play significant physiological roles including preventing the uptake of many drugs and food components from the gut into the body. Both P-gp and ABCG2 are expressed in the brush-border membranes of enterocytes in the intestine, which results in the excretion of their drug-substrates into the lumen, resulting in a potentially limiting net absorption (Figure 1). The association of drug transporter levels in the intestine and oral drug bioavailability was initially demonstrated by Spareboom et al., who showed that P-gp in the intestine limits the uptake of orally administered paclitaxel (66). Later Greiner et al. also showed that the plasma concentration of digoxin, another P-gp substrate, was inversely correlated with intestinal P-gp expression in patients (67). These observations led researchers to evaluate if modulators of ABC transporters could possibly be used to temporarily inhibit the efflux activities of the transporters, thereby increasing the oral bioavailability and plasma concentrations of some poorly absorbed drugs. Several preclinical in vitro and in vivo studies have since then reported the use of specific modulators of ABC drug transporters to increase the oral bioavailability or efficacy of drugs such as docetaxel, vinblastine, etoposide, digoxin, indinavir, saquinavir, tacrolimus, nelfinavir, talinolol, topotecan, methotrexate, irinotecan, SN-38, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and erlotinib, which are either P-gp or ABCG2 substrates (reviewed in (65)). This is consistent with biochemical studies demonstrating that there is significant overlap in substrate specificity of Pgp, ABCG2 and MRP1 (11). Recently, we showed that inhibition of intestinal abcg2 in mice by curcumin results in increased oral bioavailability of its substrate, sulfalasalazine, which suggested that curcumin, a non-toxic natural product modulator of P-gp and ABCG2, may be used to enhance drug exposure (68). A very recent study reported a newly discovered modulator of Pgp, HM30181 increased oral bioavailability of paclitaxel in rats and inhibition of growth of tumor xenografts in mice which was better than intravenous paclitaxel (69). These results identify HM30181, as a highly selective and potent inhibitor of MDR1, which in combination with paclitaxel may provide an orally effective anti-tumor regimen. Table 1 summarizes recent studies which reported the use of P-gp or ABCG2 modulators to increase the oral bioavailability or plasma concentrations of drugs. These studies suggest a pharmacological advantage of using chemotherapeutic drugs in combination with modulators of ABC drug transporters.
Figure 1.
Schematic showing the localization of P-gp and ABCG2 at the apical surface in the (a) intestinal enterocytes and b) brain capillary endothelial cells. The drug substrates of the transporters are pumped inside the lumen of the intestine and brain capillaries by these transporters, resulting in reduced oral bioavailability and decreased brain accumulation (see text for details).
Table 1.
Preclinical/clinical studies demonstrating the use of modulators of ABC drug transporters to improve oral bioavailability
Transporter | Drug | Inhibitor | Reference |
---|---|---|---|
P-gp | Paclitaxel | HM30181 | (69) |
P-gp | Paclitaxel | Valspodar (PSC833) | (93) |
P-gp | Docetaxel | Ritonavir | (84) |
P-gp | Talinolol | Verapamil | (94) |
P-gp | Paclitaxel, Digoxin, Fexofenadine | Biochanin | (95) |
P-gp | Paclitaxel | Elacridar (GF120918) | (96) |
P-gp | Paclitaxel | CyclosporinA | (97) |
P-gp | Docetaxel | Ritonavir | (98) |
P-gp | Paclitaxel | Cyclosporin A | (99) |
P-gp | Digoxin | Quinidine | (100) |
P-gp | Docetaxel | Cyclosporin A | (101) |
P-gp | Digoxin | Talinolol | (102) |
P-gp | Paclitaxel | Elacridar (GF120918) | (103) |
P-gp | Paclitaxel | HM30181 | (69) |
P-gp, ABCG2 | Topotecan | Elacridar (GF120918) | (82, 104) |
P-gp, ABCG2 | Etoposide | Elacridar (GF120918) | (105) |
P-gp, ABCG2 | Topotecan | Elacridar (GF120918) | (106) |
ABCG2 | Sulfasalazine | Curcumin | (68) |
ABCG2 | Methotrexate | Pantoprazole | (107) |
ABCG2 | Irinotecan | Gefitinib | (108) |
ABCG2 | Methotrexate | Omeprazole/lansoprazole; | (109) |
Although using modulators in combination with other drugs is an appealing strategy, it does have the risk of increased toxicities associated with the combination therapy because both P-gp and ABCG2 are also expressed in epithelial cells of the colon, adrenal cortex, kidney, liver and bile canalicular membranes and gall bladder. These organs influence the excretion of drugs. Moreover, as P-gp and ABCG2 also protect vital organs such as the brain, the testes, and the fetus, against toxins that enter the body, inhibition of the activity of these transporters may lead to toxicity associated with these organs. In addition, some modulators also inhibit metabolic enzymes such as cytochrome P450-3A4, which may significantly delay drug clearance from the body (70). Considering these factors, the use of modulators in a combination regimen for improving oral bioavailability is an approach which should be carefully monitored for minimal toxicity associated with improved therapeutic outcome.
Modulators of ABC drug transporters increase brain accumulation of chemotherapeutics
P-gp and ABCG2 expression in the brain has been found in numerous species, including humans, primates, rats, mice, and pigs (71). These transporters are principally expressed at the luminal membrane of the endothelial cells in brain capillaries (blood-brain barrier; BBB) and pump substrates back into the circulation, which is a critical determinant for the efficacy/toxicity of chemotherapeutics for the brain (Figure 1). The importance of P-gp in the brain was first demonstrated by Schinkel et al., who showed that brain uptake of drugs was higher in P-gp knockout mice, suggesting that Pgp protects the brain from toxic effects under physiological conditions (72). Later, several other studies reported that P-gp drug-substrate levels were significantly higher in the brains of mdr1a knockout mice than wild-type mice (73–76). Brain accumulation of topotecan, a camptothecin analog and a substrate of both P-gp and ABCG2 that is used for treating ovarian, cervical and small cell lung cancer, was also found to be 3.7-fold higher in mdr1a/b/Abcg2-knockout mice compared to wild-type mice (77). Recently, Lagas et al. also showed P-gp and ABCG2 knockout mice have drastically increased dasatinib brain concentrations, both after oral and i.p. administration (78). Collectively, these studies provide strong evidence for the notion that inhibition of ABC transporters at the BBB can also be used to enhance the efficacy of drugs which are targeted for brain disorders and do not penetrate the BBB by virtue of being substrates of the transporters. This has been demonstrated in several in vivo and in vitro studies in which modulators of ABC transporters such as elacridar, pantoprazole, valspodar and zosuquidar were used to increase accessibility of several clinically important drugs into the brain (78–80). Table 2 summarizes the list of modulators that have been evaluated for the purpose of increasing brain accumulation of clinically important drugs. Taken together, modulators of ABC drug transporters can possibly be used to augment brain accumulation of drugs which cannot cross the BBB due to the action of transporters, thereby increasing their therapeutic potential.
Table 2.
Preclinical/clinical studies demonstrating use of modulators of ABC drug transporters to improve brain accumulation of drugs.
Transporter | Drug | Inhibitor | Reference |
---|---|---|---|
P-gp | Vincristine | Quercetin | (113) |
P-gp | Colchicine, Vinblastine | Valspodar (PSC833), Elacridar (GF120918) | (114) |
P-gp | Colchicine, Vinblastine | Verapamil, Valspodar (PSC-833) | (115, 116) |
P-gp | Colchicine | Valspodar (PSC 833) | (117) |
P-gp | Paclitaxel | Cyclosporin A, Valspodar (PSC833), Elacridar (GF120918) | (118) |
P-gp | Paclitaxel | Zosuquidar (LY335979) | (119) |
P-gp | Paclitaxel | PSC833 (Valspodar) | (120) |
P-gp, ABCG2 | Dasatinib | Elacridar (GF120918), Zosuquidar (LY335979) (only for P-gp) | (121) |
P-gp, ABCG2 | Dasatinib | Elacridar (GF120918) | (78) |
P-gp | Imatinib | Valsopodar (PSC 833), Zosuquidar (LY335979) | (80, 122) |
P-gp, ABCG2 | Imatinib | Elacridar (GF120918), Pantoprazole | (79) |
ABCG2 | Imatinib | Elacridar (GF120918) | (80, 122) |
ABCG2 | Mitoxantrone | Elacridar (GF120918) | (123) |
ABCG2 | Mitoxantrone | Elacridar (GF120918) | (124) |
Modulators as adjuvants in clinical studies
As stated above, inhibition of ABC drug transporters by modulators may be used to achieve two independent clinical objectives: (1) reversal of MDR in drug resistant tumors during chemotherapy (2) increasing a drug’s exposure in body {tissues} for purposes such as increasing oral bioavailability or enhanced brain accumulation of chemotherapeutics. Inhibiting ABC transporters as a way to reverse drug resistance in tumors has been reported several times, but most studies failed to show a dramatic improvement in clinical outcome. We have earlier reviewed the recent progress made in clinical studies by third generation ABC transporter modulators (34). Here, we will describe recent clinical studies reporting promising results. In a Phase I/II clinical trial, Zosuquidar (LY335979) in combination with CHOP (cyclophosphamide, hydroxydaunorubicin, oncovin (vincristine), and prednisone) was shown to have little effect on the pharmacokinetics of anti-cancer drugs in patients with non-Hodgkin’s lymphoma, which suggests that this combination may be pursued further in phase III studies (81). Another phase I dose escalation study showed that a combination of oral topotecan and tariquidar was well tolerated in patients and warranted a further phase II evaluation (82). Saeki et al. also reported that a dual inhibitor of Pgp and MRP1, dofequidar (MS-209) in combination with cyclophosphamide, doxorubicin, and fluorouracil (CAF) displayed a significantly increased efficacy in breast cancer patients who did not receive prior therapy. Although the patient numbers in these analyses were small, the results are important within these clinically significant patient populations (83). In addition, studies from Oostendorp et al. also suggest that co-administration of ritonavir with docetaxel could also be evaluated for its efficacy in patients with solid tumors (84). Although more positive outcomes are expected now because of the increasing knowledge about modulators with drug transporters, studies such as those reported by Lhomme et al. compared the safety and efficacy of paclitaxel administered with or without PSC833, in patients with advanced ovarian or primary peritoneal cancer, showed that the addition of PSC833 to paclitaxel did not lead to an arrest of the disease progression or overall survival and the combination was more toxic compared with paclitaxel in untreated patients with advanced ovarian or primary peritoneal cancer (85).
The apparent lack of response to modulators to reverse MDR in clinical studies can be attributed to multiple factors. One of the most challenging issues in using modulators with chemotherapeutic agents is alteration of the pharmacokinetics of the chemotherapeutic agent, which has a toxic effect. In addition, inhibition of ABC drug transporters adds to non-specific dose limiting toxic effects and it may also result in drug-drug interaction related side effects. As a result, many of these studies had to be discontinued because of toxicities in patients (51). Genetic variability (SNPs) in ABC transporters (discussed below) and the variability in the expression levels of transporters among individuals selected for the clinical studies may also be one of the factors responsible for the poor outcome of the use of modulators. Another important aspect is patient selection in clinical studies. As development of MDR is a multifactorial phenomenon, reversal of MDR may have been studied in patients in whom drug resistance was not due to the overexpression of ABC drug transporters. Considering this, patients who show higher levels of Pgp or ABCG2 in tumor tissue should be the ones selected to evaluate the modulators in clinical studies. Moreover, the presence of other ABC transporters in tumors can complicate the use of specific modulators of ABC transporters. Taking the above factors into consideration, the use of modulators to inhibit ABC drug transporter mediated-MDR in patients is a strategy that is still a viable approach for treating drug resistant cancer. However, it is critical to find the right inhibitor for the right transporter with the right dosing, which would allow minimal pharmacokinetic interaction of the chemotherapeutic drug and maximal specific inhibition of the transporters in the target tumor.
Single-nucleotide polymorphisms in ABC drug transporters and its role in modulator efficiency
ABC drug transporters directly influence drug efficacy and toxicity and the expression of these transporters determines the degree of resistance of cancer cells to chemotherapy. Several single nucleotide polymorphisms (SNPs) in the coding region of both P-gp and ABCG2 have been reported that influence their substrate specificity and interaction with the transporters [reviewed in (86, 87)]. Such genetic variability in transporters often explains the inter-individual variability in interactions with substrate or modulators and drug disposition’, ultimately resulting in differences in clinical endpoints including toxicity and response to the modulators. Kimchi-Sarfaty et al. reported that a synonymous mutation (C3435T) in a particular MDR1 haplotype causes a change in the conformation of substrate/modulator binding sites (88). Another example of a common non-synonymous SNP (421C>A) in ABCG2 has been linked to increased oral bioavailability of topotecan in patients, decreased survival of prostate cancer patients and increased exposure to atorvastatin and rosuvastatin, statins commonly used in the treatment of high cholesterol (89–91). In addition, SNPs in metabolic enzymes such as cytochrome P450s (CYPs) are often found to be associated with altered pharmacokinetics of transporter substrate drugs (92). This is especially important when the metabolism of the drug/modulator by the CYPs may be the rate-limiting step in drug clearance. Since the efficacy and toxicity of drugs is ultimately determined by plasma pharmacokinetic parameters, studies investigating polymorphisms in ABC drug transporters as they relate to drug administration are becoming increasingly important in the clinical setting. In conclusion, polymorphisms in ABC drug transporters in various ethnic populations may significantly affect the pharmacokinetics of substrates or modulators, suggesting that they may play a more important role than expected in the efficiency of therapeutic treatment of cancer patients.
Conclusions and therapeutic perspective
In the last decade, a huge amount of effort has been invested in the field of ABC drug transporters to identify, develop, and clinically evaluate a variety of agents known to antagonize the function of these transporters as a means of overcoming tumor resistance. The application of this approach—using modulators as adjuvants in chemotherapy especially for drug-resistant tumors is still debatable due to very little success in the clinical studies that have been performed. The major reasons for the failure of this strategy could be explained in retrospect by multiple factors and variable components that are involved in the development of drug resistance in patients. It still seems that it will take some time to develop an ideal modulator that can be used to overcome drug resistance that develops as a result of cancer treatment or one that can be used in combination with other treatments because of complex substrate recognition factors and the physiological relevance of ABC drug transporters. Moreover, these transporters are only one part of a multi-component system that works to develop the drug resistance phenotype in a cancer cell. Physiologically, transporters play a vital role in protecting the cells from xenobiotics. Altering the function of any of these transporters may lead to a severe physiological imbalance resulting in high levels of toxicity. Therefore it is essential to evaluate the following factors critically to expect a successful outcome from the use of modulators of ABC drug transporters in the clinic: (a) Toxicity is one of the significant issues reported in several clinical studies, as most of these modulators showed non-specific toxic effects which are not considered acceptable during chemotherapy and prevents their safe usage during treatment; (b) Overlapping substrate specificity and redundancy in the expression of ABC transporters for the same substrate makes it very difficult to target one modulator for a specific drug transporter. Therefore, an ideal modulator should be able to specifically recognize and inhibit the ABC drug transporter responsible for drug resistance in a specific tumor; (c) Patient selection for clinical studies is a factor which seems to have been completely ignored in previous clinical studies and may well be a major reason for the failure of those trials. Patients whose tumors express high levels of transporters will obviously receive the most benefit from modulators. Therefore, drug-resistance reversal trials should ideally be performed in individuals with tumors that initially are chemosensitive but develop drug resistance following initial therapy, which is marked by an increase in the expression of ABC drug transporters; (d) In addition, SNPs in an ABC drug transporter are also linked to sensitivity to drugs. Therefore it is imperative to determine polymorphisms in these transporters in patients before making decisions about combination chemotherapy; (e) Improved in vivo imaging studies with probes specific for ABC transporters and high throughput genotype analysis can provide a quantitative assessment of the functioning of the transporters and would help in identifying patients for whom modulators of ABC drug transporter may be used as adjuvants to improve the clinical outcome of chemotherapy. It would also be important to evaluate whether the use of modulators of ABC drug transporters would prevent the selection of resistant cells with characteristics of cancer stem cells with increased expression of ABC transporters such as P-gp or ABCG2; (f) It may be worthwhile to test whether short-term treatment with modulators to block P-gp or ABCG2 just prior to chemotherapy would be effective. This type of short-term treatment could minimize not only the toxic side effects associated with the use of modulators but also help to lower the required doses of chemotherapeutic drugs.
Acknowledgments
We thank George Leiman for editorial assistance in preparation of the manuscript.
Grant Support: This work was supported by the Intramural Research Program of the NIH, National Cancer Institute, Center for Cancer Research.
Footnotes
Declaration of Interest: All authors disclose that they do not have any affiliation with any organization with a financial interest, direct or indirect, in the subject matter or materials discussed in the manuscript that may affect the conduct or reporting of the work submitted under the heading.
References
- 1.Gottesman MM, Ling V. The molecular basis of multidrug resistance in cancer: the early years of P-glycoprotein research. FEBS Lett. 2006;580:998–1009. doi: 10.1016/j.febslet.2005.12.060. [DOI] [PubMed] [Google Scholar]
- 2.O’Connor R. A review of mechanisms of circumvention and modulation of chemotherapeutic drug resistance. Curr Cancer Drug Targets. 2009;9:273–80. doi: 10.2174/156800909788166583. [DOI] [PubMed] [Google Scholar]
- 3.Gottesman MM, Ambudkar SV. Overview: ABC transporters and human disease. J Bioenerg Biomembr. 2001;33:453–8. doi: 10.1023/a:1012866803188. [DOI] [PubMed] [Google Scholar]
- 4.Dean M, Annilo T. Evolution of the ATP-binding cassette (ABC) transporter superfamily in vertebrates. Annu Rev Genomics Hum Genet. 2005;6:123–42. doi: 10.1146/annurev.genom.6.080604.162122. [DOI] [PubMed] [Google Scholar]
- 5.Ambudkar SV, Dey S, Hrycyna CA, Ramachandra M, Pastan I, Gottesman MM. Biochemical, cellular, and pharmacological aspects of the multidrug transporter. Annu Rev Pharmacol Toxicol. 1999;39:361–98. doi: 10.1146/annurev.pharmtox.39.1.361. [DOI] [PubMed] [Google Scholar]
- 6.Sauna ZE, Smith MM, Muller M, Kerr KM, Ambudkar SV. The mechanism of action of multidrug-resistance-linked P-glycoprotein. J Bioenerg Biomembr. 2001;33:481–91. doi: 10.1023/a:1012875105006. [DOI] [PubMed] [Google Scholar]
- 7.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. doi: 10.1016/s0169-409x(02)00169-2. [DOI] [PubMed] [Google Scholar]
- 8.Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochimica et Biophysica Acta (BBA) - Biomembranes. 1976;455:152–62. doi: 10.1016/0005-2736(76)90160-7. [DOI] [PubMed] [Google Scholar]
- 9.Sauna ZE, Kim IW, Ambudkar SV. Genomics and the mechanism of P-glycoprotein (ABCB1) J Bioenerg Biomembr. 2007;39:481–7. doi: 10.1007/s10863-007-9115-9. [DOI] [PubMed] [Google Scholar]
- 10.Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58. doi: 10.1038/nrc706. [DOI] [PubMed] [Google Scholar]
- 11.Sauna ZE, Ambudkar SV. Evolution and function of the multidrug resistance-linked ABC transporters in bacteria and cancer cells. In: Ponte-Sucre A, editor. ABC transporters in microorganisms. Caister Academic Press; UK: 2009. pp. 35–50. [Google Scholar]
- 12.Callaghan R, Crowley E, Potter S, Kerr ID. P-glycoprotein: So Many Ways to Turn It On. J Clin Pharmacol. 2008;48:365–78. doi: 10.1177/0091270007311568. [DOI] [PubMed] [Google Scholar]
- 13.Penson RT, Oliva E, Skates SJ, Glyptis T, Fuller AF, Jr, Goodman A, et al. Expression of multidrug resistance-1 protein inversely correlates with paclitaxel response and survival in ovarian cancer patients: a study in serial samples. Gynecol Oncol. 2004;93:98–106. doi: 10.1016/j.ygyno.2003.11.053. [DOI] [PubMed] [Google Scholar]
- 14.Chan HS, Haddad G, Thorner PS, DeBoer G, Lin YP, Ondrusek N, et al. P-glycoprotein expression as a predictor of the outcome of therapy for neuroblastoma. N Engl J Med. 1991;325:1608–14. doi: 10.1056/NEJM199112053252304. [DOI] [PubMed] [Google Scholar]
- 15.Allikmets R, Schriml LM, Hutchinson A, Romano-Spica V, Dean M. A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res. 1998;58:5337–9. [PubMed] [Google Scholar]
- 16.Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, et al. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A. 1998;95:15665–70. doi: 10.1073/pnas.95.26.15665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Miyake K, Mickley L, Litman T, Zhan Z, Robey R, Cristensen B, et al. Molecular cloning of cDNAs which are highly overexpressed in mitoxantrone-resistant cells: demonstration of homology to ABC transport genes. Cancer Res. 1999;59:8–13. [PubMed] [Google Scholar]
- 18.Bauer BE, Wolfger H, Kuchler K. Inventory and function of yeast ABC proteins: about sex, stress, pleiotropic drug and heavy metal resistance. Biochim Biophys Acta-Biomembr. 1999;1461:217–36. doi: 10.1016/s0005-2736(99)00160-1. [DOI] [PubMed] [Google Scholar]
- 19.Scharenberg CW, Harkey MA, Torok-Storb B. The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood. 2002;99:507–12. doi: 10.1182/blood.v99.2.507. [DOI] [PubMed] [Google Scholar]
- 20.Noguchi K, Katayama K, Mitsuhashi J, Sugimoto Y. Functions of the breast cancer resistance protein (BCRP/ABCG2) in chemotherapy. Adv Drug Deliv Rev. 2009;61:26–33. doi: 10.1016/j.addr.2008.07.003. [DOI] [PubMed] [Google Scholar]
- 21.Robey RW, Polgar O, Deeken J, To KW, Bates SE. ABCG2: determining its relevance in clinical drug resistance. Cancer Metastasis Rev. 2007;26:39–57. doi: 10.1007/s10555-007-9042-6. [DOI] [PubMed] [Google Scholar]
- 22.Cole SP, Bhardwaj G, Gerlach JH, Mackie JE, Grant CE, Almquist KC, et al. Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science. 1992;258:1650–4. doi: 10.1126/science.1360704. [DOI] [PubMed] [Google Scholar]
- 23.Eckford PD, Sharom FJ. ABC efflux pump-based resistance to chemotherapy drugs. Chem Rev. 2009;109:2989–3011. doi: 10.1021/cr9000226. [DOI] [PubMed] [Google Scholar]
- 24.Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 2000;92:1295–302. doi: 10.1093/jnci/92.16.1295. [DOI] [PubMed] [Google Scholar]
- 25.Ishikawa T, Kuo MT, Furuta K, Suzuki M. The human multidrug resistance-associated protein (MRP) gene family: from biological function to drug molecular design. Clin Chem Lab Med. 2000;38:893–7. doi: 10.1515/CCLM.2000.130. [DOI] [PubMed] [Google Scholar]
- 26.Evers R, Zaman GJR, Van Deemter L, Jansen H, Calafat J, Oomen LCJM, et al. Basolateral localization and export activity of the human multidrug resistance - associated protein in polarized pig kidney cells. J Clin Invest. 1996;97:1211–8. doi: 10.1172/JCI118535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hipfner DR, Deeley RG, Cole SP. Structural, mechanistic and clinical aspects of MRP1. Biochim Biophys Acta. 1999;1461:359–76. doi: 10.1016/s0005-2736(99)00168-6. [DOI] [PubMed] [Google Scholar]
- 28.Stewart AJ, Canitrot Y, Baracchini E, Dean NM, Deeley RG, Cole SP. Reduction of expression of the multidrug resistance protein (MRP) in human tumor cells by antisense phosphorothioate oligonucleotides. Biochem Pharmacol. 1996;51:461–9. doi: 10.1016/0006-2952(95)02220-1. [DOI] [PubMed] [Google Scholar]
- 29.Bordow SB, Haber M, Madafiglio J, Cheung B, Marshall GM, Norris MD. Expression of the multidrug resistance-associated protein (MRP) gene correlates with amplification and overexpression of the N-myc oncogene in childhood neuroblastoma. Cancer Res. 1994;54:5036–40. [PubMed] [Google Scholar]
- 30.Nooter K, Brutel de la Riviere G, Look MP, van Wingerden KE, Henzen-Logmans SC, Scheper RJ, et al. The prognostic significance of expression of the multidrug resistance-associated protein (MRP) in primary breast cancer. Br J Cancer. 1997;76:486–93. doi: 10.1038/bjc.1997.414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Arts HJ, Katsaros D, de Vries EG, Massobrio M, Genta F, Danese S, et al. Drug resistance-associated markers P-glycoprotein, multidrug resistance-associated protein 1, multidrug resistance-associated protein 2, and lung resistance protein as prognostic factors in ovarian carcinoma. Clin Cancer Res. 1999;5:2798–805. [PubMed] [Google Scholar]
- 32.Goldberg RM, Sargent DJ, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK, et al. A randomized controlled trial of fluorouracil plus leucovorin, irinotecan, and oxaliplatin combinations in patients with previously untreated metastatic colorectal cancer. J Clin Oncol. 2004;22:23–30. doi: 10.1200/JCO.2004.09.046. [DOI] [PubMed] [Google Scholar]
- 33.Ma WW, Adjei AA. Novel agents on the horizon for cancer therapy. CA Cancer J Clin. 2009;59:111–37. doi: 10.3322/caac.20003. [DOI] [PubMed] [Google Scholar]
- 34.Shukla S, Wu CP, Ambudkar SV. Development of inhibitors of ATP-binding cassette drug transporters: present status and challenges. Expert Opin Drug Metab Toxicol. 2008;4:205–23. doi: 10.1517/17425255.4.2.205. [DOI] [PubMed] [Google Scholar]
- 35.Tsuruo T, Iida H, Tsukagoshi S, Sakurai Y. Overcoming of vincristine resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastine by verapamil. Cancer Res. 1981;41:1967–72. [PubMed] [Google Scholar]
- 36.Slater LM, Sweet P, Stupecky M, Gupta S. Cyclosporin A reverses vincristine and daunorubicin resistance in acute lymphatic leukemia in vitro. J Clin Invest. 1986;77:1405–8. doi: 10.1172/JCI112450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cornwell MM, Pastan I, Gottesman MM. Certain calcium channel blockers bind specifically to multidrug-resistant human KB carcinoma membrane vesicles and inhibit drug binding to P-glycoprotein. J Biol Chem. 1987;262:2166–70. [PubMed] [Google Scholar]
- 38.Foxwell BM, Mackie A, Ling V, Ryffel B. Identification of the multidrug resistance-related P-glycoprotein as a cyclosporine binding protein. Mol Pharmacol. 1989;36:543–6. [PubMed] [Google Scholar]
- 39.Goldberg H, Ling V, Wong PY, Skorecki K. Reduced cyclosporin accumulation in multidrug-resistant cells. Biochem Biophys Res Commun. 1988;152:552–8. doi: 10.1016/s0006-291x(88)80073-1. [DOI] [PubMed] [Google Scholar]
- 40.Twentyman PR, Bleehen NM. Resistance modification by PSC-833, a novel non-immunosuppressive cyclosporin [corrected] Eur J Cancer. 1991;27:1639–42. doi: 10.1016/0277-5379(91)90435-g. [DOI] [PubMed] [Google Scholar]
- 41.Benson AB, 3rd, Trump DL, Koeller JM, Egorin MI, Olman EA, Witte RS, et al. Phase I study of vinblastine and verapamil given by concurrent iv infusion. Cancer Treat Rep. 1985;69:795–9. [PubMed] [Google Scholar]
- 42.Verweij J, Herweijer H, Oosterom R, van der Burg ME, Planting AS, Seynaeve C, et al. A phase II study of epidoxorubicin in colorectal cancer and the use of cyclosporin-A in an attempt to reverse multidrug resistance. Br J Cancer. 1991;64:361–4. doi: 10.1038/bjc.1991.307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bradshaw DM, Arceci RJ. Clinical relevance of transmembrane drug efflux as a mechanism of multidrug resistance. J Clin Oncol. 1998;16:3674–90. doi: 10.1200/JCO.1998.16.11.3674. [DOI] [PubMed] [Google Scholar]
- 44.Friedenberg WR, Rue M, Blood EA, Dalton WS, Shustik C, Larson RA, 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;106:830–8. doi: 10.1002/cncr.21666. [DOI] [PubMed] [Google Scholar]
- 45.Roe M, Folkes A, Ashworth P, Brumwell J, Chima L, Hunjan S, et al. Reversal of P-glycoprotein mediated multidrug resistance by novel anthranilamide derivatives. Bioorg Med Chem Lett. 1999;9:595–600. doi: 10.1016/s0960-894x(99)00030-x. [DOI] [PubMed] [Google Scholar]
- 46.Hyafil F, Vergely C, Du Vignaud P, Grand-Perret T. In vitro and in vivo reversal of multidrug resistance by GF120918, an acridonecarboxamide derivative. Cancer Res. 1993;53:4595–602. [PubMed] [Google Scholar]
- 47.Dantzig AH, Shepard RL, Cao J, Law KL, Ehlhardt WJ, Baughman TM, et al. Reversal of P-glycoprotein-mediated multidrug resistance by a potent cyclopropyldibenzosuberane modulator, LY335979. Cancer Res. 1996;56:4171–9. [PubMed] [Google Scholar]
- 48.Naito M, Matsuba Y, Sato S, Hirata H, Tsuruo T. MS-209, a quinoline-type reversal agent, potentiates antitumor efficacy of docetaxel in multidrug-resistant solid tumor xenograft models. Clin Cancer Res. 2002;8:582–8. [PubMed] [Google Scholar]
- 49.Martin C, Berridge G, Mistry P, Higgins C, Charlton P, Callaghan R. The molecular interaction of the high affinity reversal agent XR9576 with P-glycoprotein. Br J Pharmacol. 1999;128:403–11. doi: 10.1038/sj.bjp.0702807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mistry P, Stewart AJ, Dangerfield W, Okiji S, Liddle C, Bootle D, et al. In vitro and in vivo reversal of P-glycoprotein-mediated multidrug resistance by a novel potent modulator, XR9576. Cancer Res. 2001;61:749–58. [PubMed] [Google Scholar]
- 51.Fox E, Bates SE. Tariquidar (XR9576): a P-glycoprotein drug efflux pump inhibitor. Expert Rev Anticancer Ther. 2007;7:447–59. doi: 10.1586/14737140.7.4.447. [DOI] [PubMed] [Google Scholar]
- 52.Abraham J, Edgerly M, Wilson R, Chen C, Rutt A, Bakke S, et al. A phase I study of the P-glycoprotein antagonist tariquidar in combination with vinorelbine. Clin Cancer Res. 2009;15:3574–82. doi: 10.1158/1078-0432.CCR-08-0938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gekeler V, Ise W, Sanders KH, Ulrich WR, Beck J. The Leukotriene LTD4 Receptor Antagonist Mk571 Specifically Modulates MRP Associated Multidrug Resistance. Biochem Biophys Res Commun. 1995;208:345–52. doi: 10.1006/bbrc.1995.1344. [DOI] [PubMed] [Google Scholar]
- 54.Rabindran SK, Ross DD, Doyle LA, Yang W, Greenberger LM. Fumitremorgin C reverses multidrug resistance in cells transfected with the breast cancer resistance protein. Cancer Res. 2000;60:47–50. [PubMed] [Google Scholar]
- 55.Germann UA, Shlyakhter D, Mason VS, Zelle RE, Duffy JP, Galullo V, et al. Cellular and biochemical characterization of VX-710 as a chemosensitizer: reversal of P-glycoprotein-mediated multidrug resistance in vitro. Anticancer Drugs. 1997;8:125–40. doi: 10.1097/00001813-199702000-00004. [DOI] [PubMed] [Google Scholar]
- 56.Minderman H, O’Loughlin KL, Pendyala L, Baer MR. VX-710 (biricodar) increases drug retention and enhances chemosensitivity in resistant cells overexpressing P-glycoprotein, multidrug resistance protein, and breast cancer resistance protein. Clin Cancer Res. 2004;10:1826–34. doi: 10.1158/1078-0432.ccr-0914-3. [DOI] [PubMed] [Google Scholar]
- 57.Seiden MV, Swenerton KD, Matulonis U, Campos S, Rose P, Batist G, et al. A phase II study of the MDR inhibitor biricodar (INCEL, VX-710) and paclitaxel in women with advanced ovarian cancer refractory to paclitaxel therapy. Gynecol Oncol. 2002;86:302–10. doi: 10.1006/gyno.2002.6762. [DOI] [PubMed] [Google Scholar]
- 58.Toppmeyer D, Seidman AD, Pollak M, Russell C, Tkaczuk K, Verma S, et al. Safety and efficacy of the multidrug resistance inhibitor Incel (biricodar; VX-710) in combination with paclitaxel for advanced breast cancer refractory to paclitaxel. Clin Cancer Res. 2002;8:670–8. [PubMed] [Google Scholar]
- 59.Gandhi L, Harding MW, Neubauer M, Langer CJ, Moore M, Ross HJ, et al. A phase II study of the safety and efficacy of the multidrug resistance inhibitor VX-710 combined with doxorubicin and vincristine in patients with recurrent small cell lung cancer. Cancer. 2007;109:924–32. doi: 10.1002/cncr.22492. [DOI] [PubMed] [Google Scholar]
- 60.Robey RW, Shukla S, Finley EM, Oldham RK, Barnett D, Ambudkar SV, et al. Inhibition of P-glycoprotein (ABCB1)- and multidrug resistance-associated protein 1 (ABCC1)-mediated transport by the orally administered inhibitor, CBT-1((R)) Biochem Pharmacol. 2008;75:1302–12. doi: 10.1016/j.bcp.2007.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Robey RW, To KK, Polgar O, Dohse M, Fetsch P, Dean M, et al. ABCG2: a perspective. Adv Drug Deliv Rev. 2009;61:3–13. doi: 10.1016/j.addr.2008.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mechetner EB, Roninson IB. Efficient inhibition of P-glycoprotein-mediated multidrug resistance with a monoclonal antibody. Proc Natl Acad Sci U S A. 1992;89:5824–8. doi: 10.1073/pnas.89.13.5824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Widmer N, Rumpold H, Untergasser G, Fayet A, Buclin T, Decosterd LA. Resistance reversal by RNAi silencing of MDR1 in CML cells associated with increase in imatinib intracellular levels. Leukemia. 2007;21:1561–2. doi: 10.1038/sj.leu.2404671. [DOI] [PubMed] [Google Scholar]
- 64.Jin S, Gorfajn B, Faircloth G, Scotto KW. Ecteinascidin 743, a transcription-targeted chemotherapeutic that inhibits MDR1 activation. Proc Natl Acad Sci U S A. 2000;97:6775–9. doi: 10.1073/pnas.97.12.6775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Oostendorp RL, Beijnen JH, Schellens JHM. The biological and clinical role of drug transporters at the intestinal barrier. Cancer Treat Rev. 2009;35:137–47. doi: 10.1016/j.ctrv.2008.09.004. [DOI] [PubMed] [Google Scholar]
- 66.Sparreboom A, van Asperen J, Mayer U, Schinkel AH, Smit JW, Meijer DKF, 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;94:2031–5. doi: 10.1073/pnas.94.5.2031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Greiner B, Eichelbaum M, Fritz P, Kreichgauer H-P, von Richter O, Zundler J, et al. The role of intestinal P-glycoprotein in the interaction of digoxin and rifampin. J Clin Invest. 1999;104:147–53. doi: 10.1172/JCI6663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shukla S, Zaher H, Hartz A, Bauer B, Ware J, Ambudkar S. Curcumin Inhibits the Activity of ABCG2/BCRP1, a Multidrug Resistance-Linked ABC Drug Transporter in Mice. Pharm Res. 2009;26:480–7. doi: 10.1007/s11095-008-9735-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kwak J-O, Lee SH, Lee GS, Kim MS, Ahn Y-G, Lee JH, et al. Selective inhibition of MDR1 (ABCB1) by HM30181 increases oral bioavailability and therapeutic efficacy of paclitaxel. Eur J Pharmacol. 2010;627:92–8. doi: 10.1016/j.ejphar.2009.11.008. [DOI] [PubMed] [Google Scholar]
- 70.Yasuda K, Lan L-b, Sanglard D, Furuya K, Schuetz JD, Schuetz EG. Interaction of Cytochrome P450 3A Inhibitors with P-Glycoprotein. J Pharmacol Exp Ther. 2002;303:323–32. doi: 10.1124/jpet.102.037549. [DOI] [PubMed] [Google Scholar]
- 71.Loscher W, Potschka H. Drug resistance in brain diseases and the role of drug efflux transporters. Nat Rev Neurosci. 2005;6:591–602. doi: 10.1038/nrn1728. [DOI] [PubMed] [Google Scholar]
- 72.Schinkel AH, Wagenaar E, Mol CA, van Deemter L. P-glycoprotein in the blood-brain barrier of mice influences the brain penetration and pharmacological activity of many drugs. J Clin Invest. 1996;97:2517–24. doi: 10.1172/JCI118699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Schinkel AH, Smit JJM, Van Tellingen O, Beijnen JH, Wagenaar E, Van Deemter L, 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. doi: 10.1016/0092-8674(94)90212-7. [DOI] [PubMed] [Google Scholar]
- 74.Mayer U, Wagenaar E, Dorobek B, Beijnen JH, Borst P, Schinkel AH. Full blockade of intestinal P-glycoprotein and extensive inhibition of blood-brain barrier P-glycoprotein by oral treatment of mice with PSC833. J Clin Invest. 1997;100:2430–6. doi: 10.1172/JCI119784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Miyama T, Takanaga H, Matsuo H, Yamano K, Yamamoto K, Iga T, et al. P-glycoprotein-mediated transport of itraconazole across the blood- brain barrier. Antimicrob Agents Chemother. 1998;42:1738–44. doi: 10.1128/aac.42.7.1738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kawahara M, Sakata A, Miyashita T, Tamai I, Tsuji A. Physiologically based pharmacokinetics of digoxin in mdr1a knockout mice. J Pharm Sci. 1999;88:1281–7. doi: 10.1021/js9901763. [DOI] [PubMed] [Google Scholar]
- 77.de Vries NA, Zhao J, Kroon E, Buckle T, Beijnen JH, van Tellingen O. P-Glycoprotein and Breast Cancer Resistance Protein: Two Dominant Transporters Working Together in Limiting the Brain Penetration of Topotecan. Clin Cancer Res. 2007;13:6440–9. doi: 10.1158/1078-0432.CCR-07-1335. [DOI] [PubMed] [Google Scholar]
- 78.Lagas JS, van Waterschoot RAB, van Tilburg VACJ, Hillebrand MJ, Lankheet N, Rosing H, et al. Brain Accumulation of Dasatinib Is Restricted by P-Glycoprotein (ABCB1) and Breast Cancer Resistance Protein (ABCG2) and Can Be Enhanced by Elacridar Treatment. Clin Cancer Res. 2009;15:2344–51. doi: 10.1158/1078-0432.CCR-08-2253. [DOI] [PubMed] [Google Scholar]
- 79.Breedveld P, Pluim D, Cipriani G, Wielinga P, van Tellingen O, Schinkel AH, 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;65:2577–82. doi: 10.1158/0008-5472.CAN-04-2416. [DOI] [PubMed] [Google Scholar]
- 80.Bihorel S, Camenisch G, Lemaire M, Scherrmann J-M. Modulation of the Brain Distribution of Imatinib and its Metabolites in Mice by Valspodar, Zosuquidar and Elacridar. Pharm Res. 2007;24:1720–8. doi: 10.1007/s11095-007-9278-4. [DOI] [PubMed] [Google Scholar]
- 81.Morschhauser F, Zinzani PL, Burgess M, Sloots L, Bouafia F, Dumontet C. Phase I/II trial of a P-glycoprotein inhibitor, Zosuquidar.3HCl trihydrochloride (LY335979), given orally in combination with the CHOP regimen in patients with non-Hodgkin’s lymphoma. Leuk Lymphoma. 2009;48:708–15. doi: 10.1080/10428190701190169. [DOI] [PubMed] [Google Scholar]
- 82.Kuppens IELM, Witteveen EO, Jewell RC, Radema SA, Paul EM, Mangum SG, et al. A phase I, randomized, open-label, parallel-cohort, dose-finding study of elacridar (GF120918) and oral topotecan in cancer patients. Clin Cancer Res. 2007;13:3276–85. doi: 10.1158/1078-0432.CCR-06-2414. [DOI] [PubMed] [Google Scholar]
- 83.Saeki T, Nomizu T, Toi M, Ito Y, Noguchi S, Kobayashi T, et al. Dofequidar Fumarate (MS-209) in Combination With Cyclophosphamide, Doxorubicin, and Fluorouracil for Patients With Advanced or Recurrent Breast Cancer. J Clin Oncol. 2007;25:411–7. doi: 10.1200/JCO.2006.08.1646. [DOI] [PubMed] [Google Scholar]
- 84.Oostendorp RL, Huitema A, Rosing H, Jansen RS, ter Heine R, Keessen M, et al. Coadministration of Ritonavir Strongly Enhances the Apparent Oral Bioavailability of Docetaxel in Patients with Solid Tumors. Clin Cancer Res. 2009;15:4228–33. doi: 10.1158/1078-0432.CCR-08-2944. [DOI] [PubMed] [Google Scholar]
- 85.Lhomme C, Joly F, Walker JL, Lissoni AA, Nicoletto MO, Manikhas GM, et al. Phase III Study of Valspodar (PSC 833) Combined With Paclitaxel and Carboplatin Compared With Paclitaxel and Carboplatin Alone in Patients With Stage IV or Suboptimally Debulked Stage III Epithelial Ovarian Cancer or Primary Peritoneal Cancer. J Clin Oncol. 2008;26:2674–82. doi: 10.1200/JCO.2007.14.9807. [DOI] [PubMed] [Google Scholar]
- 86.Fung KL, Gottesman MM. A synonymous polymorphism in a common MDR1 (ABCB1) haplotype shapes protein function. Biochimica et Biophysica Acta (BBA) - Proteins & Proteomics. 2009;1794:860–71. doi: 10.1016/j.bbapap.2009.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sissung T, Baum C, Kirkland C, Gao R, Gardner E, Figg W. Pharmacogenetics of Membrane Transporters: An Update on Current Approaches. Mol Biotechnol. 2010;44:152–67. doi: 10.1007/s12033-009-9220-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kimchi-Sarfaty C, Oh JM, Kim I-W, Sauna ZE, Calcagno AM, Ambudkar SV, et al. A “Silent” Polymorphism in the MDR1 Gene Changes Substrate Specificity. Science. 2007;315:525–8. doi: 10.1126/science.1135308. [DOI] [PubMed] [Google Scholar]
- 89.Sparreboom A, Loos WJ, Burger H, Sissung TM, Verweij J, Figg WD, et al. Effect of ABCG2 genotype on the oral bioavailability of topotecan. Cancer Biol Ther. 2005;4:650–8. doi: 10.4161/cbt.4.6.1731. [DOI] [PubMed] [Google Scholar]
- 90.Gardner ER, Ahlers CM, Shukla S, Sissung TM, Ockers SB, Price DK, et al. Association of the ABCG2 C421A polymorphism with prostate cancer risk and survival. BJU Int. 2008;102:1694–9. doi: 10.1111/j.1464-410X.2008.07913.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Keskitalo JE, Zolk O, Fromm MF, Kurkinen KJ, Neuvonen PJ, Niemi M. ABCG2 Polymorphism Markedly Affects the Pharmacokinetics of Atorvastatin and Rosuvastatin. Clin Pharmacol Ther. 2009;86:197–203. doi: 10.1038/clpt.2009.79. [DOI] [PubMed] [Google Scholar]
- 92.Crettol S, Deglon J-J, Besson J, Croquette-Krokar M, Hammig R, Gothuey I, et al. ABCB1 and cytochrome P450 genotypes and phenotypes: Influence on methadone plasma levels and response to treatment[ast] Clin Pharmacol Ther. 2006;80:668–81. doi: 10.1016/j.clpt.2006.09.012. [DOI] [PubMed] [Google Scholar]
- 93.van Asperen J, van Tellingen O, Sparreboom A, Schinkel AH, Borst P, Nooijen WJ, et al. Enhanced oral bioavailability of paclitaxel in mice treated with the P-glycoprotein blocker SDZ PSC 833. Br J Cancer. 1997;76:1181–3. doi: 10.1038/bjc.1997.530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Spahn-Langguth H, Baktir G, Radschuweit A, Okyar A, Terhaag B, Ader P, et al. P-glycoprotein transporters and the gastrointestinal tract: Evaluation of the potential in vivo relevance of in vitro data employing talinolol as model compound. Int J Clin Pharmacol Ther. 1998;36:16–24. [PubMed] [Google Scholar]
- 95.Sean XP, David MR, Martin C, Earl D, Robert D, Jane F. Altered oral bioavailability and pharmacokinetics of P-glycoprotein substrates by coadministration of biochanin A. J Pharm Sci. 2006;95:1984–93. doi: 10.1002/jps.20664. [DOI] [PubMed] [Google Scholar]
- 96.Bardelmeijer HA, Beijnen JH, Brouwer KR, Rosing H, Nooijen WJ, Schellens JHM, et al. Increased Oral Bioavailability of Paclitaxel by GF120918 in Mice through Selective Modulation of P-glycoprotein. Clin Cancer Res. 2000;6:4416–21. [PubMed] [Google Scholar]
- 97.Van Asperen J, Van Tellingen O, Van Der Valk MA, Rozenhart M, Beijnen JH. Enhanced oral absorption and decreased elimination of paclitaxel in mice cotreated with cyclosporin A. Clin Cancer Res. 1998;4:2293–7. [PubMed] [Google Scholar]
- 98.Bardelmeijer HA, Ouwehand M, Buckle T, Huisman MT, Schellens JHM, Beijnen JH, et al. Low systemic exposure of oral docetaxel in mice resulting from extensive first-pass metabolism is boosted by ritonavir. Cancer Res. 2002;62:6158–64. [PubMed] [Google Scholar]
- 99.Meerum Terwogt JM, Malingre MM, Beijnen JH, Ten Bokkel Huinink WW, Rosing H, Koopman FJ, et al. Coadministration of oral cyclosporin A enables oral therapy with paclitaxel. Clin Cancer Res. 1999;5:3379–84. [PubMed] [Google Scholar]
- 100.Doering W. Quinidine-digoxin interaction. Pharmacokinetics, underlying mechanism and clinical implications. N Engl J Med. 1979;301:400–4. doi: 10.1056/NEJM197908233010803. [DOI] [PubMed] [Google Scholar]
- 101.Malingre MM, Richel DJ, Beijnen JH, Rosing H, Koopman FJ, Ten Bokkel Huinink WW, et al. Coadministration of cyclosporine strongly enhances the oral bioavailability of docetaxel. J Clin Oncol. 2001;19:1160–6. doi: 10.1200/JCO.2001.19.4.1160. [DOI] [PubMed] [Google Scholar]
- 102.Westphal K, Weinbrenner A, Giessmann T, Stuhr M, Franke G, Zschiesche M, et al. Oral bioavailability of digoxin is enhanced by talinolol: Evidence for involvement of intestinal P-glycoprotein. Clin Pharmacol Ther. 2000;68:6–12. doi: 10.1067/mcp.2000.107579. [DOI] [PubMed] [Google Scholar]
- 103.Malingre MM, Beijnen JH, Rosing H, Koopman FJ, Jewell RC, Paul EM, et al. Co-administration of GF120918 significantly increases the systemic exposure to oral paclitaxel in cancer patients. Br J Cancer. 2001;84:42–7. doi: 10.1054/bjoc.2000.1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kruijtzer CM, Beijnen JH, Rosing H, ten Bokkel Huinink WW, Schot M, Jewell RC, et al. Increased oral bioavailability of topotecan in combination with the breast cancer resistance protein and P-glycoprotein inhibitor GF120918. J Clin Oncol. 2002;20:2943–50. doi: 10.1200/JCO.2002.12.116. [DOI] [PubMed] [Google Scholar]
- 105.Allen JD, Van Dort SC, Buitelaar M, van Tellingen O, Schinkel AH. Mouse breast cancer resistance protein (Bcrp1/Abcg2) mediates etoposide resistance and transport, but etoposide oral availability is limited primarily by P-glycoprotein. Cancer Res. 2003;63:1339–44. [PubMed] [Google Scholar]
- 106.Kruijtzer CMF, Beijnen JH, Rosing H, ten Bokkel Huinink WW, Schot M, Jewell RC, et al. Increased Oral Bioavailability of Topotecan in Combination With the Breast Cancer Resistance Protein and P-Glycoprotein Inhibitor GF120918. J Clin Oncol. 2002;20:2943–50. doi: 10.1200/JCO.2002.12.116. [DOI] [PubMed] [Google Scholar]
- 107.Breedveld P, Zelcer N, Pluim D, Sonmezer O, Tibben MM, Beijnen JH, 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;64:5804–11. doi: 10.1158/0008-5472.CAN-03-4062. [DOI] [PubMed] [Google Scholar]
- 108.Stewart CF, Leggas M, Schuetz JD, Panetta JC, Cheshire PJ, Peterson J, et al. Gefitinib enhances the antitumor activity and oral bioavailability of irinotecan in mice. Cancer Res. 2004;64:7491–9. doi: 10.1158/0008-5472.CAN-04-0096. [DOI] [PubMed] [Google Scholar]
- 109.Joerger M, Huitema ADR, Van Den Bongard HJGD, Baas P, Schornagel JH, Schellens JHM, 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. doi: 10.1111/j.1365-2125.2005.02513.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Marchetti S, de Vries NA, Buckle T, Bolijn MJ, van Eijndhoven MAJ, Beijnen JH, et al. Effect of the ATP-binding cassette drug transporters ABCB1, ABCG2, and ABCC2 on erlotinib hydrochloride (Tarceva) disposition in in vitro and in vivo pharmacokinetic studies employing Bcrp1−/−/Mdr1a/1b−/− (triple-knockout) and wild-type mice. Mol Cancer Ther. 2008;7:2280–7. doi: 10.1158/1535-7163.MCT-07-2250. [DOI] [PubMed] [Google Scholar]
- 111.Leggas M, Panetta JC, Zhuang Y, Schuetz JD, Johnston B, Bai F, et al. Gefitinib modulates the function of multiple ATP-binding cassette transporters in vivo. Cancer Res. 2006;66:4802–7. doi: 10.1158/0008-5472.CAN-05-2915. [DOI] [PubMed] [Google Scholar]
- 112.Sparreboom A, van Asperen J, Mayer U, Schinkel AH, Smit JW, Meijer DK, 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;94:2031–5. doi: 10.1073/pnas.94.5.2031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Mitsunaga Y, Takanaga H, Matsuo H, Naito M, Tsuruo T, Ohtani H, et al. Effect of bioflavonoids on vincristine transport across blood-brain barrier. Eur J Pharmacol. 2000;395:193–201. doi: 10.1016/s0014-2999(00)00180-1. [DOI] [PubMed] [Google Scholar]
- 114.Cisternino S, Rousselle C, Dagenais C, Scherrmann JM. Screening of multidrug-resistance sensitive drugs by in situ brain perfusion in P-glycoprotein-deficient mice. Pharm Res. 2001;18:183–90. doi: 10.1023/a:1011080418027. [DOI] [PubMed] [Google Scholar]
- 115.Drion N, Lemaire M, Lefauconnier J-M. Role of P-Glycoprotein in the Blood-Brain Transport of Colchicine and Vinblastine. J Neurochem. 1996;67:1688–93. doi: 10.1046/j.1471-4159.1996.67041688.x. [DOI] [PubMed] [Google Scholar]
- 116.Drion N, Risede P, Cholet N, Chanez C, Scherrmann JM. Role of P-170 glycoprotein in colchicine brain uptake. J Neurosci Res. 1997;49:80–8. [PubMed] [Google Scholar]
- 117.Desrayaud S, Guntz P, Scherrmann JM, Lemaire M. Effect of the P-glycoprotein inhibitor, SDZ PSC 833, on the blood and brain pharmacokinetics of colchicine. Life Sci. 1997;61:153–63. doi: 10.1016/s0024-3205(97)00370-6. [DOI] [PubMed] [Google Scholar]
- 118.Kemper EM, van Zandbergen AE, Cleypool C, Mos HA, Boogerd W, Beijnen JH, et al. Increased Penetration of Paclitaxel into the Brain by Inhibition of P-Glycoprotein. Clin Cancer Res. 2003;9:2849–55. [PubMed] [Google Scholar]
- 119.Kemper EM, Cleypool C, Boogerd W, Beijnen J, Tellingen O. The influence of the P-glycoprotein inhibitor zosuquidar trihydrochloride (LY335979) on the brain penetration of paclitaxel in mice. Cancer Chemother Pharmacol. 2004;53:173–8. doi: 10.1007/s00280-003-0720-y. [DOI] [PubMed] [Google Scholar]
- 120.Fellner S, Bauer B, Miller DS, Schaffrik M, Fankhanel M, Spruss T, et al. Transport of paclitaxel (Taxol) across the blood-brain barrier in vitro and in vivo. J Clin Invest. 2002;110:1309–18. doi: 10.1172/JCI15451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Chen Y, Agarwal S, Shaik NM, Chen C, Yang Z, Elmquist WF. P-glycoprotein and breast cancer resistance protein influence brain distribution of dasatinib. J Pharmacol Exp Ther. 2009;330:956–63. doi: 10.1124/jpet.109.154781. [DOI] [PubMed] [Google Scholar]
- 122.Bihorel S, Camenisch G, Lemaire M, Scherrmann JM. Influence of breast cancer resistance protein (Abcg2) and p-glycoprotein (Abcb1a) on the transport of imatinib mesylate (Gleevec) across the mouse blood-brain barrier. J Neurochem. 2007;102:1749–57. doi: 10.1111/j.1471-4159.2007.04808.x. [DOI] [PubMed] [Google Scholar]
- 123.Lee YJ, Kusuhara H, Jonker JW, Schinkel AH, Sugiyama Y. Investigation of efflux transport of dehydroepiandrosterone sulfate and mitoxantrone at the mouse blood-brain barrier: a minor role of breast cancer resistance protein. J Pharmacol Exp Ther. 2005;312:44–52. doi: 10.1124/jpet.104.073320. [DOI] [PubMed] [Google Scholar]
- 124.Cisternino S, Mercier C, Bourasset F, Roux F, Scherrmann JM. Expression, up-regulation, and transport activity of the multidrug-resistance protein Abcg2 at the mouse blood-brain barrier. Cancer Res. 2004;64:3296–301. doi: 10.1158/0008-5472.can-03-2033. [DOI] [PubMed] [Google Scholar]