Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Review

1.

Introduction

2.

Solute carrier transporters

3.

The ATP-binding cassette transporter

4.

Conclusion

5.

Expert opinion

Intestinal transporters: enhanced absorption through P-glycoprotein-related drug interactions Parvin Zakeri-Milani & Hadi Valizadeh† †

Tabriz University of Medical Sciences, Drug Applied Research Center, Department of Pharmaceutics, Tabriz, Iran

Introduction: There are many factors that can affect the absorption process of orally administered drugs. Intestinal transporters play an important role in drug absorption. These transporters are divided into two major classes: the solute carriers and the ATP-binding cassette (ABC) transporters. P-glycoprotein (P-gp), belonging to the ABC transporter superfamily, flushes out the substrate drugs from a cell, thus regulating the intestinal absorption of drugs. Areas covered: This review gives a brief overview of uptake and efflux transporters localized in the intestine. However, because P-gp has been identified as an important underlying mechanism of drug interactions in humans, the review is strongly focused on summarizing the currently available data on the impact of P-gp for absorption of drugs. Expert opinion: The concomitant use of P-gp substrates and inhibitors (preferably in a single nanocarrier formulation) could be an effective and safe way to improve the bioavailability of drugs. It seems the study of P-gp and modulating its activity may be an interesting therapeutic goal to be considered in future research. Keywords: drug interaction, inhibitor, intestinal absorption, P-glycoprotein, transporter Expert Opin. Drug Metab. Toxicol. (2014) 10(6):859-871

1.

Introduction

Although oral drug delivery systems have many advantages over systems intended for other routes of drug administration, many drugs cannot be administered orally because of poor and/or highly variable uptake in the systemic circulation. Therefore, nowadays huge amount of research is directed toward modifying physicochemical properties of drugs to increase their gastrointestinal absorption [1]. In summary, there are three categories of factors that affect the rate and extent of oral absorption of drugs. The first category is physicochemical properties of a drug, including solubility, lipophilicity, intestinal permeability, pKa, particle size and so on. Physiological factors, such as gastrointestinal pH, gastric emptying, small intestinal transit time and absorption mechanism, fall into the second category, and finally, the third category comprises dosage form factors, such as solution, capsule, tablet, suspension and so on [2]. Among the physiological factors that impact the intestinal absorption, membrane drug transporter proteins are of great importance in drug disposition in the body. Intestinal transporters are membrane proteins that regulate the transport of molecules into and out of enterocytes. There are numerous classes of transporters, each with different substrate specificity, affinity and capacity, as well as specific tissue and cellular expression patterns. This review aims to represent the role of some important intestinal transporters probably involved in intestinal 10.1517/17425255.2014.905543 © 2014 Informa UK, Ltd. ISSN 1742-5255, e-ISSN 1744-7607 All rights reserved: reproduction in whole or in part not permitted

859

P. Zakeri-Milani & H. Valizadeh

. .

.

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

.

.

Membrane drug transporter proteins are of great importance in drug disposition in the body. Two major transporter superfamilies, the ATP-binding cassette and solute carrier family, are present in the intestinal tissues. Recently, fourth-generation P-glycoprotein (P-gp) inhibitors with less toxicity and more potential were introduced. In the light of numerous evidences indicating the effects of P-gp inhibitors on the pharmacokinetics of substrate drugs, clinicians should be aware of the hazards and advantages of concomitant administration of these two. Because of their reported P-gp inhibition activities, excipient selection is an important factor to consider in rational formulation design for P-gp substrates.

This box summarizes the key points contained in the article.

bioavailability of oral drugs with focus on P-glycoprotein (P-gp) drug efflux transporter localized at the intestinal barrier. The relevance of possible interactions of drugs and excipients with P-gp and their effect on intestinal absorption of pharmaceutical compounds are presented.

2.

Organic anion transporters According to pH-partition theory, only unionized form of drugs can traverse the intestinal epithelium by passive diffusion. Therefore, transporter systems exist for ionic agents, which could mediate their intestinal absorption. Involvement of specific anion transporters in the intestinal absorption of anionic drugs has been suggested. These transporters are categorized into two main classes: OATs and organic anion transporting polypeptides (OATPs), each having several structurally related isoforms. OATs have a broad range of clinically important substrates such as b lactam antibiotics, NSAIDs, antiviral drugs and so on. However, due to the distribution pattern of this transporter family, which limits its expression to kidney and liver, and, to a lesser extent, to brain, muscle, eye and placenta, the role of the OAT family in the intestinal absorption of substrate drugs seems to be negligible. In contrast, there are evidences showing that several OATP isoforms are expressed in human small intestine. These genes were previously classified within the SLC21A, but currently, based on their putative phylogenetic relationships and the chronology of identification, they are classified within the OATP/SLCO superfamily [5]. Among them, OATP-B (2B1), OATP-D (3A1), OATP-E (4A1) and OATP1A2 isoforms were identified to be expressed in human intestine [6,7]. Several pharmaceutical compounds such as methotrexate, fexofenadine, digoxin, pravastatin and NSAIDs are substrates of OATP isoforms [8-10]. It was shown that grapefruit juice (GJ) acts as an inhibitor of intestinal OATP2B1. In a study by Tapaninen et al., 61 and 81% reduction in AUC and Cmax of aliskiren (an OATP2B1 substrate) were observed with concomitant administration of GJ, respectively [11]. The same reduced oral plasma exposure was shown for fexofenadine in the presence of grapefruit, orange and apple juices [12]. This effect is likely mediated by inhibition of intestinal absorption via OATP1A2. However, a recent study by Imanaga et al. reports that in some conditions, OATP2B1 may also mediate the intestinal uptake of fexofenadine [13]. The similar effects of grapefruit and/or orange juice were also reported for other substrates of OATP1A2 such as celiprolol [14], atenolol [15], talinolol [16] and ciprofloxacin [17]. 2.2

Article highlights.

Solute carrier transporters

According to the Human Genum Organization Gene Nomenclature Committee, two major transporter superfamilies, the ATP-binding cassette (ABC) and solute carrier (SLC) family, are present in the intestinal tissues. SLC transporter genes consist of 52 distinct subfamilies, including proton-dependent oligopeptide transporters (POT, SLC15A), organic anion transporters (OAT, SLC family 21A [SLC21A]), organic cation transporters (OCT, SLC22A), nucleoside transporters (CNT, SLC28A; ENT, SLC29A) and the monocarboxylate transporters (MCT, SLC16A) (Table 1).

POT family Nowadays, a wide variety of peptidomimetic drugs is produced on commercial scale and used as therapeutic agents for the treatment of various diseases, including hypertension, AIDS and cancer. An uptake system for di- and tripeptides is present at brush border membrane of intestinal enterocytes, which is also capable of transporting the structurally related drugs. This subfamily, also known as SLC15A, includes peptide transporters 1 and 2, PepT1 (SLC15A1) and PepT2 (SLC15A2); peptide/histidine transporters 1 and 2, PHT1 (SLC15A4) and PHT2 (SLC15A3). However, PepT2 has not been shown to be expressed in GI tract [3]. Among these, PepT1 has captured the most attention in mucosal drug delivery. Its density increases from the duodenum to the ileum and is most abundant at the villus tip cells [4]. 2.1

860

Organic cation transporters OCTs are required to enhance the intestinal uptake and absorption of a significant number of cationic drugs, including b blockers, antihistamines, endogenous bioactive amine (e.g., epinephrine, choline, dopamine and guanidine), skeletal muscle relaxants and so on. However, several anionic and uncharged compounds are also substrates of these transporters. For example, transport of anionic prostaglandins E2 and F2 is mediated by human OCT1 and OCT2 [18]. Although the other member of this family, OCT3, shows overlap in substrate and inhibitor specificity with OCT1-2 isoforms, their affinity and maximum transport rate are distinctly different [19,20]. Moreover, three more novel OCTs were identified, 2.3

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

Intestinal transporters

Table 1. Major SLC transporters expressed in human small intestine.

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Gene name

Protein

Mechanism

Cellular localization

SLC15A1 (SLC15 family)

PEPT1

H+/peptide symporter

A

SLC22A1 (SLC22 family) SLC22A4 (SLC22 family) SLC22A5 (SLC22 family)

OCT1

OC uniporter

B

OCTN1

A

A

A

SLCO1A2 (SLCO family)

OATP1A2

H+ or OC antiporter OC antiporter Na+ symporter (carnitine) OA antiporter

SLCO2B1 (SLCO family)

OATP2B1

OA antiporter

OCTN2

Examples of drug substrates

Cefadroxil, ampicillin, amoxicillin, bestatin, cefaclor, emocaprilat, cefixime, enalapril, temocapril, midodrine, valganciclovir, valacyclovir Quinidine, acyclovir, ganciclovir, metformin, quinine, cimetidine, zidovudine Ergothioneine, mepyramine, quinidine, gabapentin, verapamil Verapamil, mepyramine, emetine, quinidine, valproate, cephaloridine

B

Enalapril, fexofenadine, indomethacin, levofloxacin, ouabain, rocuronium, temocaprilat, rosuvastatin, pitavastatin, methotrexate, imatinib, saquinavir Pravastatin, benzylpenicillin, atorvastatin, pitavastatin, fluvastatin, bosentan, glibenclamide, rosuvastatin

A: Apical; B: Basolateral; OAT: Organic anion transporters; OCT: Organic cation transporters; SLC: Solute carriers.

namely, OCTN1-3. The members of this subfamily have different abilities to interact with organic cationic drugs and carnitine. Human OCTN1 work as H+/organic cation antiporter, whereas OCTN2 is a Na+/carnitine cotransporter with a high affinity to carnitine. OCTN2 maintains the carnitine homeostasis. Carnitine is essential for entry of long-chain fatty acids into mitochondria, which turns fat into energy. It also transports the generated toxic and waste compounds out of mitochondria to prevent their accumulation. Both OCTN1 and OCTN2 proteins are expressed in human small intestinal enterocytes. Human OCTN2 can also operate as polyspecific, Na+-independent OCT, which transports cations in both directions across the plasma membrane [21]. Nucleoside transporters (SLC28A, CNT; SLC29A, ENT)

2.4

Two distinct families of transporter proteins mediate the intestinal absorption of nucleoside analogues: the high-affinity concentrative CNT, SLC28 and the low-affinity equilibrative CNTs (ENT, SLC29). The human SLC28 family located in the brush border membrane consists of three Na+-dependent isoforms, SLC28A1 (CNT1), SLC28A2 (CNT2) and SLC28A3 (CNT3). On the other hand, in the basolateral membrane of absorptive epithelia, an Na+-independent family of CNTs is located, which contains four members, ENT1 (SLC29A1), ENT2 (SLC29A2), ENT3 (SLC29A3) and ENT4 (SLC29A4). This family was distinguished from the ENTs by their facilitated diffusion transport mechanism [19]. Nucleosides are glycosylamines consisting of a nucleobase bound to a ribose or deoxyribose sugar via a b-glycosidic linkage. In addition to their biological importance, their importance to cellular physiology and function is fundamental. In medicine, several nucleoside analogues are used as antiviral or anticancer agents. Zidovudin, lamivudine, cytarabine,

gemcitabine, ribavirin and 5-flourouracil are examples of drugs for which interactions with CNTs have been extensively documented [22]. A novel member of the ENT family (SLC29) has been identified. Unlike the other ENT members, the novel transporter, plasma membrane monoamine transporter (PMAT), specifically transports monoamines, such as serotonin, dopamine and norepinephrine. However, its substrate and inhibitor specificity extensively overlaps with those of OCTs. PMAT is also known to play a role in drug transport at the intestine [18,20]. Monocarboxylate transporters (SLC16A: MCT) The members of this family are proton-dependent transporter proteins responsible for symport of monocarboxylates. To date, 14 members (MCT1-MCT14) of this family with distinct transport properties and tissue distribution have been identified [23,24]. However, only the first four isoforms (MCT1-4) have been demonstrated to facilitate proton-linked transport of metabolically important monocarboxylates such as lactate, pyruvate and ketone bodies [25,26]. A wide range of endogenous and exogenous compounds, including butyrate, acetate, propionate, g-hydroxybutyric acid [25], pravastatin, simvastatin, atorvastatin, XP13512, carindacillin, some b lactam antibiotics, penicillins, NSAIDs, valproic acid and nateglinide, are also substrates for MCTs [27-32]. Two isoforms in the MCT family, MCT8 and MCT10, exhibit proton-independent transport of substrates. MCT10 (SLC16A10; TAT1) transports aromatic amino acids, whereas MCT8 functions as thyroid hormones, T3 and T4 transporter. However, the role of the other MCT family members remains unknown [33,34]. Previous studies have demonstrated the expression and membrane localization of MCTs along the length of the human GI tract [35,36]. According to the results of those experiments, MCT1, 4 and 5 are the 2.5

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

861

P. Zakeri-Milani & H. Valizadeh

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Table 2. ATP-binding cassette transporters. Name/symbol

No. of members

Subfamily Subfamily Subfamily Subfamily Subfamily Subfamily Subfamily

14 11 13 4 1 3 5

A (ABC1) B (MDR/TAP) C (CFTR/MRP) D (ALD) E (OABP) F (GCN20) G (white)

ABC: ATP-binding cassette; MRP: Multidrug resistance associated proteins.

MRPX) are specific genes expressed along the GI tract that determine xenobiotic absorption. The Breast Cancer Resistance Protein (BCRP, ABCG2) is also expressed in small intestine. This transporter was first cloned from a doxorubicin-resistant MCF7 breast cancer cell line and is responsible for several human malignancies. It was also shown to be involved in cancer cells resistance to many chemotherapeutic agents [46]. Here we provide a more thorough discussion on ABCB1 as an efflux transporter. 3.1

P-glycoprotein (ABCB1) P-gp structure, distribution and function

3.1.1

predominant isoforms expressed in intestine with maximal expression in distal colonic regions. It was also shown that there are differences in preferential localization of these transporters in such a way that MCT1 is confined to apical membranes, whereas MCT4 and 5 are restricted to basolateral membranes of colonocytes. In contrast to previous data suggesting that MCT3 expression is restricted to retinal epithelium [37,38], nowadays there are data supporting its expression in human GI tract at a very low level. Expressions of MCT2 and 6 were not observed in the human intestine [35]. 3.

The ATP-binding cassette transporter

The ABC genes represent the largest family of transmembrane proteins. This family is one of the five distinct families of membrane protein characterized by Saier et al. [39]. The four others are multidrug and toxic compound extrusion transporters, major facilitator superfamily transporters, resistance nodulation division transporters and small multidrug resistance transporters [40]. ABC proteins bind ATP and use the energy to drive transport of a wide variety of substrates across extra- and intracellular membranes, including metabolic products, lipids and sterols, inorganic anions, metal ions, peptides, amino acids, sugars and drugs [41,42]. It is present in all prokaryotes, as well as plants, fungi, yeast and animals. The human genome contains 51 known human ABC transporters arranged in seven distinct subfamilies of proteins [43-45]. A list of all known human ABC genes subfamilies is displayed in Table 2 [41,42]. The basic structure that defines the members of this protein family is the combination of highly conserved ATP-binding and transmembrane domains (TMDs). In mammals, the functionally active ABC proteins consist of four distinct domains, two TMDs, also called membrane-spanning domain or integral membrane domain, and two nucleotidebinding domains (NBDs or ABC domain). These four domains may be present within one polypeptide chain (full transporters) or within two separate proteins (half transporters). Among ABC transporters, the ABCB and ABCC subfamilies contain the most important transporters influencing intestinal absorption in human. For instance, P-gp (ABCB1, P-gp) and multidrug resistance-associated proteins (ABCC, 862

P-glycoprotein was originally discovered in 1976 by Juliano and Ling as an efflux pump. They selected Chinese hamster ovary cells for colchicine resistance, which showed pleiotropic cross-resistance to a wide range of amphiphilic drugs. Through studies they found a cell surface glycoprotein that appeared unique to mutant cells displaying altered drug permeability, which they named permeability glycoprotein or P-glycoprotein [47]. This 170 kDa protein comprises 1276 -- 1280 amino acids with a tandemly duplicated structure. Each half of the molecule contains a NBD and a membrane-bound domain (TMD). Membrane-bound domains include six transmembrane segments. Figure 1 shows the schematic structure of P-gp. The gene responsible for encoding P-gp (commonly known as MDR1) belongs to the ABCB family of ABC transporters [48]. This gene is expressed in membrane of many cells, including cells in the kidney, liver, lungs, adrenal gland, colon, brain, placental syncytial trophoblasts and intestine. Some mononuclear cells in peripheral blood such as neutral killer cells and T lymphocytes and also human hematopoietic stem cells express P-gp as well. Additionally, a low-level of P-gp expression was detected in prostate, skeletal muscle, heart, spleen, stomach, skin and ovary [49]. P-gp detoxifies cells by removing hundreds of chemically unrelated toxins and metabolites from the cells. But this gate keeper protein has been demonstrated to possess broader substrate specificity. It preferentially transports hydrophobic, amphipathic molecules ranging in size from 100 to 4000 Da. However, some neutral compounds such as cyclosporine A and digoxin, negatively charged molecules such as atorvastine and fexofenadine, and some hydrophilic drugs such as methotrexate are also substrates of P-gp. This substrate promiscuity is a distinctive feature of P-gp describing the importance of polyspecific drug binding for logical drug design. However, the CYP3A subfamily, the most abundant cytochrome P450 enzymes expressed in the intestine, may be functionally linked to P-gp [50,51]. It is possible that P-gp may influence first-pass metabolism in a complex cooperative manner. They have overlapping substrate specificity of inhibitors and inducers. It appears that P-gp prevents the saturation of CYP3A4 in the enterocytes by limiting the total drug transport across the membrane. This could lead to increased duration of exposure of the drug to the enzyme, providing greater extent of

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

Intestinal transporters

Glycolysation TMD1

TMD2 Extracellular Cell membrane Intracellular

P P P

NH2

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Nucleotide binding domain

Phosphorylation Nucleotide binding domain

COOH

Figure1. Structure of P-gp efflux pump. P-gp: P-glycoprotein; TMDs: Transmembrane domains.

metabolism. Moreover, the generated metabolites are substrates for P-gp and are actively transported out of the enterocytes by P-gp. Therefore, the metabolites do not compete with the metabolism of parent drug [52]. P-gp mechanism of action A conceptual model proposed for P-gp--drug interaction is the hydrophobic vacuum cleaner model. Based on this model, drugs partition into the inner leaflet of the membrane and are then flipped to the outer leaflet in a nonspecific manner [40]. From the molecular point of view, this transport process involves a number of key steps comprising substrate binding, binding site reorientation, energy supplying for transport, coupling substrate binding and energy supplying, substrate dissociation, and resetting the membrane transport pump [53]. However, the unifying mechanism of these steps remains unclear. On the other hand, there are difficulties in attaining complete structural information of human P-gp. Because human and mouse P-gp share nearly 87% protein sequence identity, the reported X-ray structure of mouse P-gp could also help to understand its conformational changes during transport process in human. For instance, previous studies based on crystal structure and crosslinking analysis have suggested that membrane P-gp is in the closed conformation in the presence or absence of ATP. Additionally, the closed conformation has a high affinity for drug substrates. Indeed, it is improbable that under normal physiological conditions P-gp adopt open conformation because the ATP concentration inside the cell (3 -- 5 mM) is much more than the P-gp affinity for ATP (km » 0.5 mM) [54]. It seems the ‘ATP-driven switch’ mechanism, in which nucleotide-driven interaction of the nucleotide-binding domains leads to resetting of the membrane-bound domains and reduces drug affinity, may be reflected in the inward- and outward-facing structure of P-gp [48]. The interior of P-gp ‘binding pocket’ is lined with a variety of amino acids, including hydrophobic amino acids and those containing aromatic compounds. This 3.1.2

variation of residues inside the pocket explains the ability of mammalian P-gp to accommodate a wide range of substrates. Understanding P-gp structure and knowing where substrates bind may help scientists design more effective drugs. That means only redesigning of existing drugs and modifying portions of the drugs (instead of designing new drugs) could make them work better. However, predicting whether a ligand will be substrate or inhibitor remains challenging. Bearing in mind the potential for co-administration of pharmaceutical compounds of both kinds in clinical practice, it is of great importance to elucidate the possibility of P-gp-mediated drug--drug interaction. Given that the majority of drugs are developed for oral delivery, the following sections, therefore, will focus to a large degree on the role of P-gp in intestinal drug absorption. In human intestine, P-gp is highly expressed on the brush-border membrane of superficial columnar epithelial cells of the ileum and colon, and its expression gradually decreases proximally into the jejunum, duodenum, and stomach [55-57]. Generally, P-gp inhibition occurs by three mechanisms: i) blocking drug-binding site either competitively, noncompetitively or allosterically; ii) interfering with ATP hydrolysis; and iii) altering integrity of cell membrane lipids [58,59]. For example, cyclosporin A and verapamil are competitive inhibitors of P-gp, whereas cis-(Z)-flupentixol, a thioxanthene derivative, inhibits the P-gp-mediated drug transport through an allosteric mechanism. That means it prevents substrate translocation and dissociation, leading to stable but reversible P-gp-substrate complex [60]. For anthranilic acid derivative, tariquidar (XR9576), an allosteric effect on substrate recognition or ATP hydrolysis was demonstrated [61]. Inhibition of ATP hydrolysis was also reported for vanadate. In this case, a stable transition complex of P-gp/vanadate/ ADP is formed. However, XR9576 binds to the TMDs of P-gp. Finally, P-gp inhibitor surfactants are considered to act on secondary and tertiary structure, resulting in altering integrity of membrane lipids [58]. However, for many others, the inhibitory mechanisms are yet to be fully understood.

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

863

P. Zakeri-Milani & H. Valizadeh

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Table 3. List of some P-gp inhibitors belonging to three known generations. First generation

Second generation

Third generation

Verapamil Reserpine Progesterone Tamoxifen Felodipine Nifedipine Erythromycin Flufenazine Diltiazem Chlorpromazine Quinidine Cyclosporin A

Dexverapamil Gallopamil PSC 833 (Valspodar) VX-710 (Biricodar) MS-209 GF 120918 (Elacridar) Reversin 121 Reversin 125

Tariquidar (XR9576) KR30031 Cyclopropyldibenzosuberane Zosuquidar (LY335979) Laniquidar (R101933) Substituted diarylimidazole ONT-093

P-gp inhibitors and their pharmacokinetic advantages

3.1.3

Based on the specificity and affinity, there are three main generations of P-gp inhibitors. The first-generation inhibitors are pharmacologically active substances with clinical use for other indications, which have been shown to inhibit P-gp as well. Verapamil, a calcium channel blocker, is a typical example of this class [62]. Using first-generation P-gp inhibitors could cause pharmacological side effects and toxicity. Moreover, designing and developing such combination pharmaceutical products requires long time. Therefore, the second-generation modulators were presented, which are more specific with less side effect inhibitors, such as dexverapamil or dexniguldipine. However, complicated unfavorable drug-- drug interactions as a result of inhibition of two or more ABC transporters by second-generation inhibitors limit their clinical use. A third generation of P-gp inhibitors comprised compounds such as tariquidar, with high affinity to P-gp at nanomolar concentrations. The potency of this generation appeared to be about 10-fold more than the first- and second-generation inhibitors. Recently, fourth-generation P-gp inhibitors with less toxicity and more potential at concentrations that can modulate P-gp compared with previous generations were introduced. Using natural products and their derivatives and the design of peptidomimetics and dual-activity ligands emerged as the fourth generation [63,64]. Table 3 shows some examples of P-gp efflux pumps inhibitors belonging to three known generations. Various studies on the regulation of P-gp by its inhibitors have been reported. For example, in an attempt to improve the oral bioavailability of paclitaxel, Woo et al. have reported that apical-to-basolateral permeation of paclitaxel in Caco-2 monolayers was increased in the presence of KR-30031, a verapamil analogue with fewer cardiovascular effects. The ability of KR-30031 to reduce this efflux transport is equal to that of verapamil, a well-known P-gp inhibitor [65]. 864

In a study by Novak et al. digoxin intestinal absorption was assessed when co-administered with acetaminophen. They reported > 200% increase in digoxin concentration in portal vein compared with digoxin alone. It was demonstrated that apical-to-basolateral transport of acetaminophen was not affected by verapamil, a typical P-gp inhibitor [66]. On the other hand, acetaminophen accumulation was the same in P-gp knock-down and wild-type HepG2 cells [66]. These findings suggested that acetaminophen was not a P-gp substrate and there was no possibility of competitive P-gp inhibition. An in situ single-pass intestinal perfusion study in rats indicated phenytoin to be a P-gp substrate [67]. To complete the study, phenytoin was administered orally to male rats in the presence and absence of verapamil. The obtained results showed an elevated mean AUC and mean maximum plasma concentration of drug when co-administered with classical P-gp inhibitor, verapamil [67]. In a similar study using singlepass intestinal perfusion technique in rats, the elevated effective intestinal permeability of cyclosporine was reported in the presence of clarithromycin and erythromycin as efflux pump inhibitors [68,69]. In another study involving the same inhibitors, the effective intestinal permeability of digoxin as a P-gp substrate was explored in rats [70,71]. Again the macrolides enhanced intestinal transport of digoxin significantly as a function of P-gp efflux transporter inhibition. In another study, applicability of rhamnolipids as absorption enhancers in Caco-2 cells was investigated. Rhamnolipids were demonstrated to inhibit P-gp activity and reduce efflux ratio (basolateral-to-apical/apical-to-basolateral) of rhodamine 123, a P-gp substrate [72]. Herbal medicines are more and more often used in recent years to enhance well-being and treat disease, including anxiety, arthritis, depression, high blood pressure, insomnia, hormonal imbalances, migraines and so on. Although so much is still unknown and speculative, nowadays several studies are centered on the potential interaction of plant-derived products with concomitantly used drugs. For instance, Oga et al. have shown that digoxin absorptive transport in Caco-2 cells was significantly enhanced in concurrent administration with Vernonia amygdalina, Carica papaya and Vernonia amygdalina, phytomedicines commonly used during malaria therapy [73]. Another well-known example is GJ. Wide consumption of GJ is attributed to its effects in reducing atherosclerotic plaque formation and inhibiting breast cancer cell proliferation and mammary cell tumorigenesis. In 1989, researchers found that GJ can significantly increase oral bioavailability of drugs. This was achieved from an interaction study of felodipine and alcohol in which GJ was used as a flavor to mask the taste of the ethanol [74]. GJ was also reported to be a potent inhibitor of P-gp-mediated colchicine [75], talinolol [76], and digoxin [77] transport in in vitro systems. In another study, the effects of furanocoumarin derivatives in GJ on the uptake of vinblastine by Caco-2 cells were examined. All GJ constituents were able to decrease the P-gp-mediated vinblastine efflux at concentrations ranging from 0.1 to 20 µM [77,78].

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Intestinal transporters

Cyclosporine is also a well-known P-gp substrate. It is also a substrate of CYP3A4. However, based on the study conducted by Lown et al. P-gp may be a more important determinant of cyclosporine absorption than enteric CYP3A [79]. On the other hand, there is considerable evidence indicating that GJ has minimal effect on P-gp in vivo [77,80-83]. Because GJ phytochemicals can also alter the activity of enzymes (esterases, sulfotransferases) and transporters other than P-gp (OATPs) in the body, therefore when the levels of a P-gp substrate drug are increased with GJ administration, it is difficult to explain the reason. For clopidogrel, an antiplatelet agent with poor aqueous solubility, which is a P-gp efflux pump substrate, the natural compound naringin showed more than threefold absorption enhancement in everted gut sac model [84]. In that study, quinidine was also proved to increase clopidogrel absorption by P-gp inhibition. It should be noted, however, that long-term use of GJ may cause P-gp induction, leading to reduction in therapeutic plasma levels of certain drugs. The probable reason is the interaction of GJ components with aryl hydrocarbon receptor that upregulate the P-gp expression [85]. It was demonstrated that the activity of P-gp in Caco-2 cells could also be affected by the extracts of bitter melon, soybean, dokudami and welsh onion, bitter melon being the most potent inhibitor [86]. 1-Monopalmitin, a simple monoglyceride with a great amount of fatty acid chain, was found to be the major inhibitor of P-gp in bitter melon [86,87]. This study was followed by Barta et al., who analyzed the effects of two common monoglyceride components of various lipid excipients, 1-monoolein and 1-monostearin, on the activity and expression of P-gp in Caco-2 cells. The results suggested that these two monoglycerides are inhibitors of P-gp [1]. Herbal products mistletoe, Natto K2, Agaricus and green tea also showed inhibitory effect on P-gp in vitro. These products enhanced digoxin transport in differentiated and polarized Caco-2 cells [88]. St John’s wort (Hypericum perforatum) is one of the most commonly used herbal antidepressants. It was recognized as a potent P-gp inducer, and according the available human data it was able to decrease the blood concentrations of amitriptyline, cyclosporine, digoxin, fexofenadine, indinavir, methadone, midazolam, nevirapine, phenprocoumon, simvastatin, tacrolimus, theophylline and warfarin. Therefore, co-administration of drugs with St. John’s wort and their possible interaction would be a major safety concern [89,90]. A study by Fortuna and colleagues demonstrated the possible active efflux mediated by P-gp of a series of dibenz (b,f) azepine-5-carboxamide derivatives. They found that after addition of verapamil the mucosal-to-serosal permeability of oxcarbazepine, R-licarbazepine, BIA 2-024 and carbamazepine-10,11-epoxide, lowered, whereas serosal-to-mucosal permeability increased, suggesting the involvement of P-gp on the transport of those compounds. Carbamazepine itself was not shown to be P-gp substrate. This study was performed

using freshly excised mouse jejunal segments mounted in Ussing chambers [91]. Pharmacoresistance of antiepileptic drugs in the treatment of epilepsy and psychiatric disorders and their transport by P-gp were also investigated by Zhang et al. using polarized cell lines MDCKII and LLC transfected with the human MDR1 gene [92]. Many factors can alter P-gp-based drug interactions. One of these factors is genetic differences of P-gp. Polymorphism at exon 26 (C3435) has been shown to influence the level of intestinal P-gp and consequently the amount of substrate drug absorption. It was also suggested that the effect of genetic variants might be substrate specific [93]. However, more research is needed to confirm this finding. Because P-gp kinetic is saturable, the relative contribution of intestinal P-gp to overall drug absorption is important only when a very small oral dose is given (e.g., digoxin, cyclosporine A, talinolol), or the dissolution and diffusion rates of the drug are very slow [94-97]. Typically for poor soluble drugs, solubilizing agents or excipients are included in the experiment medium. This leads to better solubility and concentration gradient, which could promote drug absorption. On the other hand, nano-sized formulations providing larger surface area for better dissolution and permeation across biological membranes are considered as promising tools for drug delivery. Recently, micro/nano-sized self-emulsifying systems were recognized as effective approach for improvement of drug pharmacokinetic behavior. These systems are isotropic mixtures of oil, surfactant, co-surfactant and drug capable of forming fine o/w nanoemulsion when introduced into gastrointestinal medium. The drug is delivered in the dissolved state inside the emulsion droplets, which can be easily absorbed. On the other hand, some of these lipids and surfactants have established P-gp modulation activity. Hence, selfemulsifying systems provide a good opportunity for enhanced oral bioavailability of the substrate molecules by P-gp modulation as well. D-a-Tocopheryl polyethylene glycol 1000 succinate (TPGS or vitamin E TPGS), a novel nonionic surfactant, has been extensively used in drug delivery systems, including self-emulsifying systems [98,99]. There are several studies reporting its P-gp inhibition and absorption enhancement effects [100-102]. This effect was found to appear below TPGS critical micellization concentration of 0.20% (w/w) [103]. The micellar property for TPGS is provided by hydrophilic head of PEG 1000, which shows the most P-gp inhibition effect. The ability of P-gp inhibition was also seen in other nonionic surfactants such as Tween 80, Pluronic E8100, Pluronic E6100 and Cremophor EL. However, TPGS was the most effective inhibitor [98]. Recently, the effects of Tween excipients on the activity and expression of P-gp were investigated in our laboratory. The results indicated that Tween 20 at concentration of 0.01% (w/v) and Tween 40 at concentration of 0.05% (w/v) were able to block P-gp efflux pump significantly [104]. In another study by Yu et al., Brij35, another pharmaceutical excipient, was introduced as a P-gp

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

865

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

P. Zakeri-Milani & H. Valizadeh

inhibitor and potential permeation enhancer of bis(12)hupyridone [105]. This effect was seen at concentrations lower than its critical micelle concentration. It seems hydrophiliclipophilic balance (HLB values) of excipients could play a role in their multidrug-resistance-reversing effects. This issue was examined for a series of surfactant excipients in both Caco-2 cell line and the everted gut sacs of rat intestine [106]. The optimal transport enhancement effect on the epirubicin uptake (as a P-gp substrate drug) was characteristic of surfactants with intermediate HLB values ranging from 10 to 17. Furthermore, the effects of sodium deoxycholate and sodium caprate on the transport of epirubicin were examined using the same protocol [107]. The study suggested that these two absorption enhancers could be used as P-gp modulators in drug formulations. For tacrolimus self-microemulsifying drug delivery system, developed by Wang et al., TPGS and Cremophor EL40 were chosen as P-gp inhibitors. This immunosuppressant drug exhibited sevenfold and eightfold increase in drug absorption compared with drug simple solution administered to the rats [108]. TPGS was also used in novel multifunctional TPGS/PLGA nanoparticles [109]. The novel nanoparticles improved the uptake of the loaded drug (SN-38) by novel mechanisms, including clathrin-mediated endocytosis of unbroken nanoparticles and at the same time escaping from the recognition of P-gp. The enhancing effects of Pluronic F68 and Labrasol on the intestinal absorption and pharmacokinetics of rifampicin were also investigated by an in situ single-pass perfusion method [110]. The results of a study conducted by Du¨nnhaupt and colleagues on Caco-2 cells approved that S-protected thiolated chitosan is promising excipient for drug delivery systems providing improved permeation-enhancing and P-gp inhibition effects [111]. Phospholipids (e.g., phosphatidylcholine) are extensively used as excipients for poorly soluble drugs formulations. According to Simon et al., they were proven to be able to significantly inhibit P-gp in vitro [112]. However, this capability needs to be further elucidated in vivo. Irinotecan, a potent anticancer drug, has no oral formulation available so far. The reason is its high P-gp induced efflux, which results in very low oral bioavailability. Although there have been some studies addressing the issue, most recently its SMEDDS formulation was developed with excipients having P-gp modulation activity and examined further by in vitro, ex vivo and in vivo methods [113]. The optimized formulation was found to be beneficial in enhancing the oral accessibility of P-gp substrate drug, irinotecan. Moreover, the inhibitory effects of polyethylene glycols for the intestinal P-gp function were assessed in previous studies [114,115]. The findings revealed that glycols with different molecular weights and their derivatives are useful excipients to inhibit the function of P-gp in the intestine. Finally, as a new insight into the world of pharmaceutical excipients, acrylic copolymers (Eudragits) have also been discovered to have potential P-gp inhibition effects when tested on Caco-2 cells [116]. However, further work is required regarding a better 866

understanding of the role of Eudragits in drug absorption in intestine. 4.

Conclusion

In the light of numerous evidences indicating the effects of P-gp inhibitors on the pharmacokinetics of substrate drugs, especially their intestinal absorption, which was reviewed in this paper, clinicians should be aware of the potential for these interactions and patients need to be educated about the hazards and advantages of concomitant administration of these two. Pharmacists must also take responsibility for monitoring drug interactions and notifying the physician and patient about potential problems. For the purpose of extrapolating the in vitro results to the usual clinical setting, substrate and inhibitor exposure manner must be consistent with the real amounts that are administered by the patients. 5.

Expert opinion

SLC and ABC transporters expressed in the intestine are increasingly being recognized as significant determinants of drug absorption and drug--drug interaction. There are many examples in literature indicating the effect of the impact of transport proteins on the pharmacokinetics of established drugs. Therefore, it seems that clarification of the relative contribution of the target transporter to the overall intestinal membrane transport of a particular drug is of great importance. Although many studies have been made on the structure and function of transporter proteins, much remains undetermined regarding their molecular mechanisms, regulation, and structure--function relationships. On the other hand, there are also extensive data supporting the modulation of drug bioavailability through P-gp regulation. This regulation could occur by co-administered drugs or by components in food groups. Using P-gp substrates, prodrugs could be considered as a new strategy to bypass the efflux process. However, most of the available data have derived from in vitro systems, studying P-gp inhibition in isolation. Although these studies are valuable, they may not indicate the clinically important improvements in in vivo bioavailability of substrate drugs. On the other hand, P-gp expression and activity is dependent on individual’s genetic makeup, age, diseases state, and so on. Therefore, to obtain a valid conclusions and better understanding of the role of P-gp in drug interactions, more work is required. Moreover, for the purpose of extrapolating in vitro results to the human situation in vivo, the drug substrate and also inhibitor concentrations have to be selected carefully. If the P-gp inhibition occurs at doses larger than what is used normally, in this case intolerable side effects by the P-gp inhibitor can occur. We should also consider the nonspecific action of P-gp inhibitors on P-gp and their nonselective distribution to nontarget organs. This may result in complication in their clinical use. Therefore, continued development of more specific inhibiting agents

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Intestinal transporters

may be of great interest in future works. More studies on the exact mechanism of the suppressing effect of P-gp modulators are also needed. The probable mechanisms are alteration of plasma membrane fluidity, ATPase activity, changing surface P-gp level and intracellular ATP level. A detailed study of any conformational changes in inhibition process will reveal important data on the molecular events responsible for this communication. This kind of knowledge will benefit the rational design of P-gp inhibitors with higher efficacy and lower toxicity. Nanocarriers (such as liposomes, niosomes, polymeric nanoparticles, dendrimers and polymer--drug conjugates) are promising drug delivery systems capable of P-gp inhibition. Although some research articles have been published about nanomedicines’ P-gp inhibition effect, many challenges remain. Future works should focus on the biological mechanisms by which these nanomedicines overcome P-gp. It seems Bibliography Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

Barta CA, Sachs-Barrable K, Feng F, Wasan KM. Effects of monoglycerides on P-glycoprotein: modulation of the activity and expression in Caco-2 cell monolayers. Mol Pharm 2008;5(5):863-75

2.

Song NN, Zhang SY, Liu CX. Overview of factors affecting oral drug absorption. Asian J Drug Metab Pharmacokinet 2004;4(3):167-76

3.

Steffansen B, Nielsen CU, Brodin B, et al. Intestinal solute carriers: an overview of trends and strategies for improving oral drug absorption. Eur J Pharm Sci 2004;21(1):3-16 This article provides excellent background information on the subject.

..

4.

Lee VH. Membrane transporters. Eur J Pharm Sci 2000;11(Suppl 2):S41-50

5.

Hagenbuch B, Meier PJ. Organic anion transporting polypeptides of the OATP/ SLC21 family: phylogenetic classification as OATP/SLCO superfamily, new nomenclature and molecular/functional properties. Pflu¨gers Arch 2004;447(5):653-65

6.

Kobayashi D, Nozawa T, Imai K, et al. Involvement of human organic anion transporting polypeptide OATP-B (SLC21A9) in pH-dependent transport across intestinal apical membrane. J Pharmacol Exp Ther 2003;306(2):703-8

7.

performing in vivo studies is crucial to the development of more efficacious nanoparticulate drug delivery systems with the purpose of P-gp inhibition. In addition, because of the lack of enough high-quality three-dimensional structural information about P-gp, designing molecules that circumvent P-gp efflux is still hampered. Therefore, only a small number of structure-based prediction models have been developed in recent years.

Declaration of interest The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Grandvuinet AS, Vestergaard HT, Rapin N, Steffansen B. Intestinal transporters for endogenic and pharmaceutical organic anions: the challenges of deriving in-vitro kinetic parameters for the prediction of clinically relevant drug-drug interactions. J Pharm Pharmacol 2012;64(11):1523-48

8.

Sai Y, Tsuji A. Transporter-mediated drug delivery: recent progress and experimental approaches. Drug Discov Today 2004;9(16):712-20

9.

Koepsell H. Polyspecific organic cation transporters: their functions and interactions with drugs. Trends Pharmacol Sci 2004;25(7):375-81

10.

Hagenbuch B, Meier PJ. The superfamily of organic anion transporting polypeptides. Biochim Biophys Acta 2003;1609(1):1-18

11.

Tapaninen T, Neuvonen PJ, Niemi M. Grapefruit juice greatly reduces the plasma concentrations of the OATP2B1 and CYP3A4 substrate aliskiren. Clin Pharmacol Ther 2010;88(3):339-42

12.

13.

Dresser GK, Kim RB, Bailey DG. Effect of grapefruit juice volume on the reduction of fexofenadine bioavailability: possible role of organic anion transporting polypeptides. Clin Pharmacol Ther 2005;77(3):170-7 Imanaga J, Kotegawa T, Imai H, et al. The effects of the SLCO2B1 c.1457C > T polymorphism and apple juice on the pharmacokinetics

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

of fexofenadine and midazolam in humans. Pharmacogenet Genomics 2011;21(2):84-93 14.

Lilja JJ, Juntti-Patinen L, Neuvonen PJ. Orange juice substantially reduces the bioavailability of the beta-adrenergic-blocking agent celiprolol. Clin Pharmacol Ther 2004;75(3):184-90

15.

Lilja JJ, Raaska K, Neuvonen PJ. Effects of orange juice on the pharmacokinetics of atenolol. Eur J Clin Pharmacol 2005;61(5-6):337-40

16.

Schwarz UI, Seemann D, Oertel R, et al. Grapefruit juice ingestion significantly reduces talinolol bioavailability. Clin Pharmacol Ther 2005;77(4):291-301

17.

Neuhofel AL, Wilton JH, Victory JM, et al. Lack of bioequivalence of ciprofloxacin when administered with calcium-fortified orange juice: a new twist on an old interaction. J Clin Pharmacol 2002;42(4):461-6

18.

Engel K, Wang J. Interaction of organic cations with a newly identified plasma membrane monoamine transporter. Mol Pharmacol 2005;68(5):1397-407

19.

Pastor-Anglada M, Cano-Soldado P, Molina-Arcas M, et al. Cell entry and export of nucleoside analogues. Virus Res 2005;107(2):151-64

20.

Engel K, Zhou M, Wang J. Identification and characterization of a novel monoamine transporter in the human brain. J Biol Chem 2004;279(48):50042-9

867

P. Zakeri-Milani & H. Valizadeh

21.

Koepsell H. The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 2013;34(2-3):413-35

22.

Young JD, Yao SY, Baldwin JM, et al. The human concentrative and equilibrative nucleoside transporter families, SLC28 and SLC29. Mol Aspects Med 2013;34(2-3):529-47

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

23.

24.

25.

Halestrap AP, Meredith D. The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflu¨gers Arch 2004;447(5):619-28 Halestrap AP. The monocarboxylate transporter family–Structure and functional characterization. IUBMB Life 2012;64(1):1-9 Lam WK, Felmlee MA, Morris ME. Monocarboxylate transporter-mediated transport of gamma-hydroxybutyric acid in human intestinal Caco-2 cells. Drug Metab Dispos 2010;38(3):441-7

32.

33.

34.

35.

36.

26.

Halestrap AP. The SLC16 gene family structure, role and regulation in health and disease. Mol Aspects Med 2013;34(2-3):337-49

27.

Morris ME, Felmlee MA. Overview of the proton-coupled MCT (SLC16A) family of transporters: characterization, function and role in the transport of the drug of abuse gamma-hydroxybutyric acid. AAPS J 2008;10(2):311-21

37.

Li YH, Ito K, Tsuda Y, et al. Mechanism of intestinal absorption of an orally active beta-lactam prodrug: uptake and transport of carindacillin in Caco-2 cells. J Pharmacol Exp Ther 1999;290(3):958-64

38.

28.

29.

Emoto A, Ushigome F, Koyabu N, et al. H(+)-linked transport of salicylic acid, an NSAID, in the human trophoblast cell line BeWo. Am J Physiol Cell Physiol 2002;282(5):C1064-75

30.

Utoguchi N, Audus KL. Carrier-mediated transport of valproic acid in BeWo cells, a human trophoblast cell line. Int J Pharm 2000;195(1-2):115-24

31.

868

Wu X, Whitfield LR, Stewart BH. Atorvastatin transport in the Caco-2 cell model: contributions of P-glycoprotein and the proton-monocarboxylic acid co-transporter. Pharm Res 2000;17(2):209-15

Okamura A, Emoto A, Koyabu N, et al. Transport and uptake of nateglinide in Caco-2 cells and its inhibitory effect on human monocarboxylate transporter MCT1. Br J Pharmacol 2002;137(3):391-9 Kim DK, Kanai Y, Chairoungdua A, et al. Expression cloning of a Na+-independent aromatic amino acid transporter with structural similarity to H+/monocarboxylate transporters. J Biol Chem 2001;276(20):17221-8 Enerson BE, Drewes LR. Molecular features, regulation, and function of monocarboxylate transporters: implications for drug delivery. J Pharm Sci 2003;92(8):1531-44 Gill RK, Saksena S, Alrefai WA, et al. Expression and membrane localization of MCT isoforms along the length of the human intestine. Am J Physiol Cell Physiol 2005;289(4):C846-52 Price NT, Jackson VN, Halestrap AP. Cloning and sequencing of four new mammalian monocarboxylate transporter (MCT) homologues confirms the existence of a transporter family with an ancient past. Biochem J 1998;329(Pt 2):321-8

43.

Tarling EJ, de Aguiar Vallim TQ, Edwards PA. Role of ABC transporters in lipid transport and human disease. Trends Endocrinol Metab 2013;24(7):342-50

44.

Glavinas H, Krajcsi P, Cserepes J, Sarkadi B. The role of ABC transporters in drug resistance, metabolism and toxicity. Curr Drug Deliv 2004;1(1):27-42

45.

Sharom FJ. ABC multidrug transporters: structure, function and role in chemoresistance. Pharmacogenomics 2008;9(1):105-27

46.

Sukowati CH, Rosso N, Pascut D, et al. Gene and functional up-regulation of the BCRP/ABCG2 transporter in hepatocellular carcinoma. BMC Gastroenterol 2012;12:160

47.

Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim Biophys Acta 1976;455(1):152-62 First article to identify P-gp as efflux pump.

.

48.

Philp NJ, Yoon H, Grollman EF. Monocarboxylate transporter MCT1 is located in the apical membrane and MCT3 in the basal membrane of rat RPE. Am J Physiol 1998;274(6 Pt 2):R1824-8

Ward AB, Szewczyk P, Grimard V, et al. Structures of P-glycoprotein reveal its conformational flexibility and an epitope on the nucleotide-binding domain. Proc Natl Acad Sci USA 2013;110(33):13386-91

49.

Krishna R, Yu L. editors. Biopharmaceutics application in drug development. Springer, NY, USA; 2008

50.

Kato M, Chiba K, Hisaka A, et al. The intestinal first-pass metabolism of substrates of CYP3A4 and P-glycoprotein-quantitative analysis based on information from the literature. Drug Metab Pharmacokinet 2003;18(6):365-72

51.

Marchetti S, Mazzanti R, Beijnen JH, Schellens JH. Concise review: clinical relevance of drug drug and herb drug interactions mediated by the ABC transporter ABCB1 (MDR1, P-glycoprotein). Oncologist 2007;12(8):927-41

52.

Washington N, Washington C, Wilson C. Physiological pharmaceutics: barriers to drug absorption. 2nd edition. Taylor and Francis, Inc., NY, USA; 2001

Saier MH Jr, Tran CV, Barabote RD. TCDB: the transporter classification database for membrane transport protein analyses and information. Nucleic Acids Res 2006;34(Database issue):D181-6

40.

Wong K, Ma J, Rothnie A, et al. Towards understanding promiscuity in multidrug efflux pumps. Trends Biochem Sci 2014;39(1):8-16 Very new work that summarizes structural and computational studies toward full elucidation of P-gp recognition and transport.

41.

Dean M, Hamon Y, Chimini G. The human ATP-binding cassette (ABC) transporter superfamily. J Lipid Res 2001;42(7):1007-17

Yoon H, Fanelli A, Grollman EF, Philp NJ. Identification of a unique monocarboxylate transporter (MCT3) in retinal pigment epithelium. Biochem Biophys Res Commun 1997;234(1):90-4

39.

.

42.

Vasiliou V, Vasiliou K, Nebert DW. Human ATP-binding cassette (ABC) transporter family. Hum Genomics 2009;3(3):281-90

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

Intestinal transporters

53.

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

54.

55.

56.

57.

Pollock NL, McDevitt CA, Collins R, et al. Improving the stability and function of purified ABCB1 and ABCA4: the influence of membrane lipids. Biochim Biophys Acta 2014;1838(1):134-47 Loo TW, Bartlett MC, Clarke DM. Human P-glycoprotein is active when the two halves are clamped together in the closed conformation. Biochem Biophys Res Commun 2010;395(3):436-40 Ho GT, Moodie FM, Satsangi J. Multidrug resistance 1 gene (P-glycoprotein 170): an important determinant in gastrointestinal disease? Gut 2003;52(5):759-66 MacLean C, Moenning U, Reichel A, Fricker G. Closing the gaps: a full scan of the intestinal expression of P-glycoprotein, breast cancer resistance protein, and multidrug resistanceassociated protein 2 in male and female rats. Drug metab Dispos 2008;36(7):1249-54 MacLean C, Moenning U, Reichel A, Fricker G. Regional absorption of fexofenadine in rat intestine. Eur J Pharm Sci 2010;41(5):670-4

58.

Amin ML. P-glycoprotein Inhibition for Optimal Drug Delivery. Drug Target Insights 2013;7:27-34

59.

Varma MV, Ashokraj Y, Dey CS, Panchagnula R. P-glycoprotein inhibitors and their screening: a perspective from bioavailability enhancement. Pharmacol Res 2003;48(4):347-59

60.

61.

62.

..

Maki N, Hafkemeyer P, Dey S. Allosteric modulation of human P-glycoprotein. Inhibition of transport by preventing substrate translocation and dissociation. J Biol Chem 2003;278(20):18132-9

63.

64.

65.

66.

67.

68.

69.

70.

74.

Bailey DG, Spence JD, Edgar B, et al. Ethanol enhances the hemodynamic effects of felodipine. Clin Invest Med 1989;12(6):357-62

75.

Dahan A, Amidon GL. Grapefruit juice and its constituents augment colchicine intestinal absorption: potential hazardous interaction and the role of p-glycoprotein. Pharm Res 2009;26(4):883-92

76.

Woo JS, Lee CH, Shim CK, Hwang SJ. Enhanced oral bioavailability of paclitaxel by coadministration of the P-glycoprotein inhibitor KR30031. Pharm Res 2003;20(1):24-30

Spahn-Langguth H, Langguth P. Grapefruit juice enhances intestinal absorption of the P-glycoprotein substrate talinolol. Eur J Pharm Sci 2001;12(4):361-7

77.

Novak A, Carpini GD, Ruiz ML, et al. Acetaminophen inhibits intestinal P-glycoprotein transport activity. J Pharm Sci 2013;102(10):3830-7

Hanley MJ, Cancalon P, Widmer WW, Greenblatt DJ. The effect of grapefruit juice on drug disposition. Exp Opin Drug Metab Toxicol 2011;7(3):267-86

78.

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(6):1369-77

79.

Lown KS, Mayo RR, Leichtman AB, et al. Role of intestinal P-glycoprotein (mdr1) in interpatient variation in the oral bioavailability of cyclosporine. Clin Pharmacol Ther 1997;62(3):248-60

80.

Zakeri-Milani P, Damani S, Islambulchilar Z, et al. Effect of Erythromycine on the intestinal transport of cyclosporine. JBUMS 2009;11(2):7-15

Becquemont L, Verstuyft C, Kerb R, et al. Effect of grapefruit juice on digoxin pharmacokinetics in humans. Clin Pharmacol Ther 2001;70(4):311-16

81.

Zakeri-Milani P, Valizadeh H, Islambulchilar Z, et al. Effect of erythromycin and clarithromycin on the intestinal transport of digoxin. Pharm Sci 2009;15(1):75-82

Parker RB, Yates CR, Soberman JE, Laizure SC. Effects of grapefruit juice on intestinal P-glycoprotein: evaluation using digoxin in humans. Pharmacotherapy 2003;23(8):979-87

82.

Yasui N, Kondo T, Furukori H, et al. Effects of repeated ingestion of grapefruit juice on the single and multiple oral-dose pharmacokinetics and pharmacodynamics of alprazolam. Psychopharmacology (Berl) 2000;150(2):185-90

83.

Shi J, Montay G, Leroy B, Bhargava VO. Effects of itraconazole or grapefruit juice on the pharmacokinetics of telithromycin. Pharmacotherapy 2005;25(1):42-51

Li H, Yan Z, Ning W, et al. Using rhodamine 123 accumulation in CD8 cells as a surrogate indicator to study the P-glycoprotein modulating effect of cepharanthine hydrochloride in vivo. J Biomed Biotechnol 2011;2011:281651 Palmeira A, Sousa E, Vasconcelos MH, Pinto MM. Three decades of P-gp inhibitors: skimming through several generations and scaffolds. Curr Med Chem 2012;19(13):1946-2025

Neerati P, Ganji D, Bedada SK. Study on in situ and in vivo absorption kinetics of phenytoin by modulating P-glycoprotein with verapamil in rats. Eur J Pharm Sci 2011;44(1-2):27-31 Zakeri-Milani P, Valizadeh H, Islambulchilar Z, et al. Investigation of the intestinal permeability of ciclosporin using the in situ technique in rats and the relevance of P-glycoprotein. Arzneimittelforschung 2008;58(4):188-92

Modok S, Heyward C, Callaghan R. P-glycoprotein retains function when reconstituted into a sphingolipid- and cholesterol-rich environment. J Lipid Res 2004;45(10):1910-18

71.

Valizadeh H, Mehtari M, Zakeri-Milani P. Evidence for enhanced intestinal absorption of digoxin by P-glycoprotein inhibitors. Trop J Pharm Res 2012;11(6):939-45

Constantinides PP, Wasan KM. Lipid formulation strategies for enhancing intestinal transport and absorption of P-glycoprotein (P-gp) substrate drugs: in vitro/in vivo case studies. J Pharm Sci 2007;96(2):235-48 An important study providing the usefulness of lipid-based formulations for P-gp inhibition.

72.

Jiang L, Long X, Meng Q. Rhamnolipids enhance epithelial permeability in Caco-2 monolayers. Int J Pharm 2013;446(1-2):130-5

84.

73.

Oga EF, Sekine S, Shitara Y, Horie T. P-glycoprotein mediated efflux in Caco-2 cell monolayers: the influence of herbals on digoxin transport. J Ethnopharmacol 2012;144(3):612-17

Lassoued MA, Sfar S, Bouraoui A, Khemiss F. Absorption enhancement studies of clopidogrel hydrogen sulphate in rat everted gut sacs. J Pharm Pharmacol 2012;64(4):541-52

85.

Lim GE, Li T, Buttar HS. Interactions of grapefruit juice and cardiovascular

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

869

P. Zakeri-Milani & H. Valizadeh

rat and human intestinal epithelia. J Pharm Exp Ther 2001;296(2):584-91

medications: a potential risk of toxicity. Exp Clin Cardiol 2003;8(2):99-107 86.

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

87.

Konishi T, Satsu H, Hatsugai Y, et al. Inhibitory effect of a bitter melon extract on the P-glycoprotein activity in intestinal Caco-2 cells. Br J Pharmacol 2004;143(3):379-87 Konishi T, Satsu H, Hatsugai Y, et al. A bitter melon extract inhibits the P-glycoprotein activity in intestinal Caco-2 cells: monoglyceride as an active compound. Biofactors 2004;22(1-4):71-4

97.

Ueda CT, Lemaire M, Gsell G, et al. Apparent dose-dependent oral absorption of cyclosporin A in rats. Biopharm Drug Dispos 1984;5(2):141-51

98.

Guo Y, Luo J, Tan S, et al. The applications of Vitamin E TPGS in drug delivery. Eur J Pharm Sci 2013;49(2):175-86

99.

Wempe MF, Wright C, Little JL, et al. Inhibiting efflux with novel non-ionic surfactants: rational design based on vitamin E TPGS. Int J Pharm 2009;370(1-2):93-102

88.

Engdal S, Nilsen OG. Inhibition of P-glycoprotein in Caco-2 cells: effects of herbal remedies frequently used by cancer patients. Xenobiotica 2008;38(6):559-73

89.

Hennessy M, Kelleher D, Spiers JP, et al. St Johns wort increases expression of P-glycoprotein: implications for drug interactions. Br J Clin Pharmacol 2002;53(1):75-82

100. Collnot EM, Baldes C, Schaefer UF, et al. Vitamin E TPGS P-glycoprotein inhibition mechanism: influence on conformational flexibility, intracellular ATP levels, and role of time and site of access. Mol Pharm 2010;7(3):642-51

Zhou S, Chan E, Pan SQ, et al. Pharmacokinetic interactions of drugs with St John’s wort. J Psychopharmacol 2004;18(2):262-76

101. Constantinides PP, Han J, Davis SS. Advances in the use of tocols as drug delivery vehicles. Pharm Res 2006;23(2):243-55

Fortuna A, Alves G, Falcao A, Soares-daSilva P. Evaluation of the permeability and P-glycoprotein efflux of carbamazepine and several derivatives across mouse small intestine by the Ussing chamber technique. Epilepsia 2012;53(3):529-38

102. Parsa A, Saadati R, Abbasian Z, et al. Enhanced permeability of Etoposide across everted sacs of rat small intestine by vitamin E-TPGS. Iran J Pharm Res 2013;12(Suppl):37-46

90.

91.

92.

93.

94.

95.

96.

870

Zhang C, Zuo Z, Kwan P, Baum L. In vitro transport profile of carbamazepine, oxcarbazepine, eslicarbazepine acetate, and their active metabolites by human P-glycoprotein. Epilepsia 2011;52(10):1894-904 Karlsson L, Green H, Zackrisson AL, et al. ABCB1 gene polymorphisms are associated with fatal intoxications involving venlafaxine but not citalopram. Int J Legal Med 2013;127(3):579-86 Lin JH, Yamazaki M. Role of P-glycoprotein in pharmacokinetics: clinical implications. Clin Pharmacokinet 2003;42(1):59-98 Wetterich U, Spahn-Langguth H, Mutschler E, et al. Evidence for intestinal secretion as an additional clearance pathway of talinolol enantiomers: concentration- and dose-dependent absorption in vitro and in vivo. Pharm Res 1996;13(4):514-22 Stephens RH, O’Neill CA, Warhurst A, et al. Kinetic profiling of P-glycoprotein-mediated drug efflux in

103. Dintaman JM, Silverman JA. Inhibition of P-glycoprotein by D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS). Pharm Res 1999;16(10):1550-6 104. Hodaee D, Baradaran B, Valizadeh H, et al. The effect of Tween excipients on expression and activity of P-glycoprotein in Caco-2 cells. Pharmind 2014. [Epub ahead of print] 105. Yu H, Hu YQ, Ip FC, et al. Intestinal transport of bis(12)-hupyridone in Caco-2 cells and its improved permeability by the surfactant Brij-35. Biopharm Drug Dispos 2011;32(3):140-50 106. Lo YL. Relationships between the hydrophilic-lipophilic balance values of pharmaceutical excipients and their multidrug resistance modulating effect in Caco-2 cells and rat intestines. J Control Release 2003;90(1):37-48 107. Lo YL, Huang JD. Effects of sodium deoxycholate and sodium caprate on the transport of epirubicin in human intestinal epithelial Caco-2 cell layers and everted gut sacs of rats. Biochem Pharmacol 2000;59(6):665-72

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

108. Wang Y, Sun J, Zhang T, et al. Enhanced oral bioavailability of tacrolimus in rats by self-microemulsifying drug delivery systems. Drug Dev Ind Pharm 2011;37(10):1225-30 109. Wang Y, Guo M, Lu Y, et al. Alpha-tocopheryl polyethylene glycol succinate-emulsified poly(lactic-coglycolic acid) nanoparticles for reversal of multidrug resistance in vitro. Nanotechnology 2012;23(49):495103 110. Ma L, Wei Y, Zhou Y, et al. Effects of Pluronic F68 and Labrasol on the intestinal absorption and pharmacokinetics of rifampicin in rats. Arch Pharm Res 2011;34(11):1939-43 111. Dunnhaupt S, Barthelmes J, Rahmat D, et al. S-protected thiolated chitosan for oral delivery of hydrophilic macromolecules: evaluation of permeation enhancing and efflux pump inhibitory properties. Mol Pharm 2012;9(5):1331-41 112. Simon S, Schubert R. Inhibitory effect of phospholipids on P-glycoprotein: cellular studies in Caco-2, MDCKII mdr1 and MDCKII wildtype cells and P-gp ATPase activity measurements. Biochim Biophys Acta 2012;1821(9):1211-23 113. Negi LM, Tariq M, Talegaonkar S. Nano scale self-emulsifying oil based carrier system for improved oral bioavailability of camptothecin derivative by P-Glycoprotein modulation. Colloids Surf B Biointerfaces 2013;111C:346-53 . The article describes the usefulness of self-emulsifying nanocarriers as P-gp modulators. 114. Shen Q, Lin Y, Handa T, et al. Modulation of intestinal P-glycoprotein function by polyethylene glycols and their derivatives by in vitro transport and in situ absorption studies. Int J Pharm 2006;313(1-2):49-56 115. Hugger ED, Audus KL, Borchardt RT. Effects of poly(ethylene glycol) on efflux transporter activity in Caco-2 cell monolayers. J Pharm Sci 2002;91(9):1980-90 116. Mohammadzadeh R, Baradaran B, Valizadeh H, et al. Reduced ABCB1 expression and activity in the presence of acrylic copolymers. Advanced Pharmaceutical Bulletin 2014;4(3):219-24 .. Very new work reporting the P-gp inhibitory effect of Eudragits.

Intestinal transporters

Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Washington University Library on 06/23/14 For personal use only.

Affiliation

Parvin Zakeri-Milani1 & Hadi Valizadeh†2 † Author for correspondence 1 Tabriz University of Medical Sciences, Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz, Iran 2 Professor of Pharmaceutics, Tabriz University of Medical Sciences, Drug Applied Research Center and Faculty of Pharmacy, Department of Pharmaceutics, Tabriz, Iran Tel: +98 411 339 2649; Fax: +98 411 334 4798; E-mail: [email protected]

Expert Opin. Drug Metab. Toxicol. (2014) 10(6)

871

Intestinal transporters: enhanced absorption through P-glycoprotein-related drug interactions.

There are many factors that can affect the absorption process of orally administered drugs. Intestinal transporters play an important role in drug abs...
374KB Sizes 0 Downloads 5 Views