The effects of dietary and herbal phytochemicals on drug transporters

The effects of dietary and herbal phytochemicals on drug transporters

ADR-13061; No of Pages 18 Advanced Drug Delivery Reviews xxx (2016) xxx–xxx Contents lists available at ScienceDirect Advanced Drug Delivery Reviews...

920KB Sizes 1 Downloads 86 Views

ADR-13061; No of Pages 18 Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews journal homepage: www.elsevier.com/locate/addr

The effects of dietary and herbal phytochemicals on drug transporters☆

2Q1

Yan Li a,b, Jezrael Revalde c, James W. Paxton c,⁎

3 4 5 6

a

7

a r t i c l e

8 9 10 11 12

Article history: Received 4 April 2016 Received in revised form 10 August 2016 Accepted 5 September 2016 Available online xxxx

13 14 15 16 17 18 19 20 21 22 23 24 25 37 38 39 40 41 42

Chemical compounds studied in this article: Curcumin (PubChem CID: 969516) Chrysin (PubChem CID: 5281607) Digoxin (PubChem CID: 2724385) Metformin (PubChem CID: 4091) (−)-Epigallocatechin gallate (PubChem CID: 65064) Talinolol (PubChem CID: 68770) SN-38 (PubChem CID: 104842) Quinine (PubChem CID: 3034034) Quercetin (PubChem CID: 5280343) Morin (PubChem CID: 5281670)

O

1. 2.

3.

P

D

E

T R

Introduction . . . . . . . . . . . . . . . . . . . . . . . Absorption from gastrointestinal (GI) tract . . . . . . . . . 2.1. ABC transporters . . . . . . . . . . . . . . . . . 2.1.1. Pgp . . . . . . . . . . . . . . . . . . . 2.1.2. BCRP . . . . . . . . . . . . . . . . . . 2.1.3. Coordinated function of efflux and metabolism 2.2. SLC transporters . . . . . . . . . . . . . . . . . 2.2.1. OATP2B1 and OATP1A2 . . . . . . . . . . 2.2.2. Proton-coupled peptide transporters (PEPTs) 2.2.3. OCT family (OCT 1 and 2, SLC22A) . . . . . Hepatobilliary excretion . . . . . . . . . . . . . . . . . 3.1. SLC transporters . . . . . . . . . . . . . . . . . 3.1.1. OCT1 . . . . . . . . . . . . . . . . . .

U

49 50 51 52 53 54 55 56 57 58 59 60 61

Contents

Membrane transporter proteins (the ABC transporters and SLC transporters) play pivotal roles in drug absorption and disposition, and thus determine their efficacy and safety. Accumulating evidence suggests that the expression and activity of these transporters may be modulated by various phytochemicals (PCs) found in diets rich in plants and herbs. PC absorption and disposition are also subject to the function of membrane transporter and drug metabolizing enzymes. PC–drug interactions may involve multiple major drug transporters (and metabolizing enzymes) in the body, leading to alterations in the pharmacokinetics of substrate drugs, and thus their efficacy and toxicity. This review summarizes the reported in vitro and in vivo interactions between common dietary PCs and the major drug transporters. The oral absorption, distribution into pharmacological sanctuaries and excretion of substrate drugs and PCs are considered, along with their possible interactions with the ABC and SLC transporters which influence these processes. © 2016 Published by Elsevier B.V.

N C O

46 44 43

48 47

a b s t r a c t

R

Keywords: ABC transporter SLC transporter Pharmacokinetics Food–drug interactions Phytochemical

45

R O

i n f o

C

c

School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand School of Interdisciplinary Health Studies, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand Department of Pharmacology & Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand

E

b

F

1

. . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

. . . . . . . . . . . . .

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Drug Transporters: Molecular Mechanisms”. ⁎ Corresponding author. E-mail address: [email protected] (J.W. Paxton).

http://dx.doi.org/10.1016/j.addr.2016.09.004 0169-409X/© 2016 Published by Elsevier B.V.

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

0 0 0 0 0 0 0 0 0 0 0 0 0

26 27 28 29 30 31 32 33 34 35 36

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125

C

E

R

R

103 104

O

101 102

C

99 100

N

97 98

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . .

0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

128

2. Absorption from gastrointestinal (GI) tract Oral drug administration is regarded as the most convenient and safe route for drug delivery. In order for drugs to exert their beneficial effects (other than on the GI tract itself), they must be delivered to their target organ(s) and tissues by the systemic circulation. Adequate concentrations at the site(s) of action are only achieved by overcoming several absorption and metabolism barriers, both in the GI tract and in the liver (Fig. 1).

Foods/phytochemicals

Interaction with hepatic transporters can affect hepatic uptake and biliary excretion of drugs Interaction with renal transporters can affect drug excretion

Interaction withbloodbrain barrier transporters can affect drug distribution into brain

Interaction with intestinal transporters can affect drug absorption and bioavailability

U

95 96

. . . . . . . . . . . . . . . . . . . . . .

D

There is accumulating evidence that membrane transporter proteins play pivotal roles in drug absorption and disposition and thus determine the efficacy and safety of drugs [1–7]. Two major transporter superfamilies, namely the ATP-binding cassette (ABC) and the solute carrier (SLC) transporters, have been identified in the human genome. The human ABC transporter family contains 49 members with 7 subfamilies, including several important drug transporters, such as Pgp (ABCB1), MRP1-9 (ABCC1-6 and ABCC10-12, respectively) and BCRP (ABCG2) [2,8]. They actively efflux chemically diverse substrates, including amino acids, peptides, proteins, lipids, saccharides, inorganic ions, metals, and xenobiotics from cells. In most examples, transport of the substrates against their concentration and chemical-potential gradients is driven by the hydrolysis of ATP [9]. The SLC transporters include more than 300 influx transporters responsible for nutrient intake and drug disposition into important organs [10]. Both ABC and SLC transporters are present in all tissues and appear to play a fundamental role in defending the body against various exogenous toxins and carcinogens, as well as maintaining physiological homeostasis. The oral bioavailability and pharmacokinetic profile of each drug may be very different due to their great structural diversity. Consequently, the bioavailability and the distribution of these drugs at their site(s) of action in the tissue, are key factors that need to be established in association with their biological effects. Recently, various in vivo studies have demonstrated that drug transporters may determine drug disposition in various tissue and therefore efficacy and/or side effects. These transporters appear to be under tight transcriptional regulation by nuclear receptors, suggesting their functions are subject to dietary and environmental influences [2,5,11,12]. The expression and function of these transporters may be modulated by various phytochemicals (PCs) found in diets rich in plants and herbs [13–15]. PCs are most often associated with health benefits, with epidemiological and nutritional studies suggesting a protective role in the prevention of diabetes, cancer, cardiovascular and neurodegenerative diseases [16–25]. They include glucosinolates, organic isothiocyanates, dibenzocyclo-octadienes, sulfur-containing compounds of alliaceae, terpenoids (carotenoids, monoterpenes, and phytosterols), flavonoids and polyphenols (e.g., anthocyanins, flavones, flavan-3-ols, isoflavones, stilbenoids, catechin, epigallocatechin and ellagic acid) [16, 17]. The involvement of the ATP-binding cassette (ABC) and solute carrier (SLC) transporters in the pharmacokinetics of some drugs, and their

93 94

. . . . . . . . . . . . . . . . . . . . . .

E

86

91 92

. . . . . . . . . . . . . . . . . . . . . .

T

1. Introduction

89 90

. . . . . . . . . . . . . . . . . . . . . .

implications in potential diet/herb-drug clinical interactions, will be 126 explored in this review. 127

85

87 88

. . . . . . . . . . . . . . . . . . . . . .

P

84

. . . . . . . . . . . . . . . . . . . . . .

F

3.1.2. OATP1B1 and OATP1B3 . . . . . . . . . . . 3.1.3. Multidrug and toxin extrusion (MATE) proteins 3.2. ABC transporters . . . . . . . . . . . . . . . . . . . 3.2.1. Pgp . . . . . . . . . . . . . . . . . . . . 3.2.2. MRP2 . . . . . . . . . . . . . . . . . . . 3.2.3. BCRP . . . . . . . . . . . . . . . . . . . . 3.2.4. MRP1 and 3 . . . . . . . . . . . . . . . . . 4. Blood–brain barrier . . . . . . . . . . . . . . . . . . . . . 4.1. SLC transporters . . . . . . . . . . . . . . . . . . . 4.2. ABC transporters . . . . . . . . . . . . . . . . . . . 4.2.1. Pgp . . . . . . . . . . . . . . . . . . . . 4.2.2. BCRP . . . . . . . . . . . . . . . . . . . . 5. Renal excretion . . . . . . . . . . . . . . . . . . . . . . 5.1. SLC transporters . . . . . . . . . . . . . . . . . . . 5.1.1. OAT1 and 3 . . . . . . . . . . . . . . . . . 5.1.2. OCT2 . . . . . . . . . . . . . . . . . . . . 5.2. Multidrug and toxin extrusion (MATE) proteins . . . . 5.3. ABC transporters . . . . . . . . . . . . . . . . . . . 5.3.1. Pgp . . . . . . . . . . . . . . . . . . . . 5.3.2. BCRP and MRP2 . . . . . . . . . . . . . . . 6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . .

O

62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

R O

2

Fig. 1. Effect of phytochemicals on absorption and disposition of drugs in humans.

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

129 130 131 132 133 134 135

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

159 160 161 162

168 169 170 171

T

157 158

C

155 156

E

153 154

R

151 152

R

149 150

N C O

147 148

Pgp

Apical side H+

MRP2

ATP

PEPT1

BCRP

ATP

ATP

OATP 2B1

OCT1

H+

Na+

U

145 146

F

The human family of ABC transporters contains 49 members with 7 subfamilies, including several important xenobiotic transporters, such as Pgp (ABCB1), MRP1-9 (ABCC1-6 and ABCC10-12, respectively) and BCRP (ABCG2) [2,8]. They actively efflux chemically diverse substrates, including amino acids, peptides, proteins, lipids, saccharides, inorganic ions, metals, and xenobiotics from cells. In most examples, transport

143 144

O

166 167

142

2.1.1. Pgp Pgp, encoded by the multidrug resistance gene (ABCB1), is a 170-kDa protein with 1280 residues in a single polypeptide [35,36]. The protein can be split into two homologous halves, each half composed of one trans-membrane domain with 6 trans-membrane α-helices and one cytoplasmic nucleotide binding domain linked by a cytoplasmic 60 amino acid linker chain. It is an efflux transporter located on the plasma membrane in many tissues such as intestine, liver, kidney and brain. Pgp transports various structurally and pharmacologically diverse drugs, including vinca alkaloids (vinblastine, vinorelbine) [37], camptothecin derivates (topotecan) [38], tubulin polymerizing drugs (paclitaxel, docetaxel), anthracyclines (doxorubicin, idarubicin) [39], epipodophyllotoxins (etoposide) [40], tyrosine kinase inhibitors (e.g., erlotinib and imatinib), antibiotics (e.g., grepafloxacin) [41], anticonvulsants (e.g., dabigatran, rivaroxaban, and apixaban) [42], antihypertensives (e.g., nicardipine and talinolol) [43,44], digitalis glycosides (e.g., digoxin) [45], immunosuppressive agents (e.g., cyclosporine) [46], anticoagulants (e.g., ximelagatran) [47], steroids (e.g., cortisol, aldosterone, and dexamethasone) and HIV protease inhibitors (e.g., saquinavir, ritonavir, and nelfinavir) [48]. Typical synthesized inhibitors of Pgp include verapamil, cyclosporine A, tamoxifen, quinidine, erythromycin, elacridar (GF120918), zosuquidar (LY335979) and tariquidar (XR9576) [26,48–51]. Pgp is exclusively localized on apical (AP) side of intestine tissue and effluxes substrate drugs into the gut lumen and limits drug entry and contributes to intestinal drug excretion [52–54]. It can limit the bioavailability of substrates, although some drugs with high intrinsic permeability and solubility may circumvent the barrier function of intestinal Pgp [55–57]. Pgp concentrations vary considerably over the length of the GI tract, with ABCB1 mRNA and Pgp protein expression gradually increasing from the duodenum to the ileum and colon [58,59]. Since most marketed oral drugs are absorbed from the duodenum, this also extended the complexity of Pgp functionality. Nevertheless, the important roles of intestinal Pgp in absorption of its substrates have been confirmed in vivo by comparing the pharmacokinetics between: 1) wildtype and Abcb1a/1b (−/−) mice; and 2) human subjects with or without co-administration of Pgp inhibitors [54,60].

R O

2.1. ABC transporters

140 141

P

165

138 139

of the substrates against their concentration and chemical potential 172 gradients is driven by the hydrolysis of ATP [9]. 173

E

163 164

Aqueous solubility is recognized as a key factor for the bioaccessibility and thus bioavailability of many drugs. Once dissolved into GI fluids, a drug is usually absorbed across the GI wall, following the basic rule of thumb that “the more lipophilic, the more efficient is the absorption”, provided that it is not subject to transporter-mediated absorption. Atypical absorption kinetics of many drugs (which cannot be predicted from their physicochemical properties) can be caused by interactions with intestinal ABC and SLC transporters. Transporter-related absorption phenomena, such as limited or nonlinear intestinal permeability and absorption of drugs, may result in extensive variability in their oral bioavailability, resulting in inadequate plasma concentrations and lack of pharmacological effect on the one hand, or elevated concentrations and toxicity on the other. Many ABC and SLC transporters have been identified in the GI tract, including the organic anion transporting polypeptide 1A2 (OATP1A2, SLCO1A2) and 2B1 (OATP2B1, SLCO2B1), oligopeptide transporter PEPT1 (SLC15A1), P-glycoprotein (Pgp, ABCB1), multidrug resistance protein 2 (MRP2, ABCC2) and breast cancer resistance protein (BCRP, ABCG2) on the apical membrane, and organic cation transporter 1 (OCT1, SLC22A1) and MRP3, 4, 5 (ABCC3, 4 and 5) on the basolateral (blood) side (Fig. 2) [2,8,26–34]. Most PCs consumed in our diet, or taken as supplements or herbal medicines, are present in GI tract at relatively abundant concentrations. Many PCs have been reported to act as substrates/inhibitors/modulators of the ABC transporters, such as Pgp, BCRP and MRP2 (Table 1), which can severely limit the oral bioavailability of co-administered drugs. To date, the important SLC transporters involved in xenobiotic absorption and disposition mainly include the OATP family and proton-coupled peptide transporters (Fig. 2).

D

136 137

3

ATP

MRP 3

K+ ATP

MRP 4

ATP

ATP

MRP 5

OCT2

Na+

Basolateral side Fig. 2. Localization of important ABC and SLC transporters in human small intestine.

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209

4 t1:1 t1:2

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

Table 1 An overview of tissue distribution, PC substrates and inhibitors of Pgp, MRPs and BCRP (?, not determined, numbers inside brackets are IC50 values) Name Official Tissue location symbol

PC substrates

PC inhibitors

References

t1:4

Pgp

Quercetin, Narigenin, Biochanin A

[64–68,78,119–122]

t1:5

MRP1 ABCC1

All major tissues

ECG

t1:6

MRP2 ABCC2

Liver, kidney, intestine, brain

ECG 4-O-methyl-EGCG

t1:7

MRP3 ABCC3

Small intestine, pancreas, colon, placenta

Baicalein-7-glucuronide Resveratrol, resveratrol-3-glucuronide

Genistein, naringin, naringenin, hesperetin, acacetin, apigenin, chrysin, 6′,7′-epoxybergamottin, 6′,7′-dihydroxybergamottin, CBT-1, tetrandine, fangchinoline, quinine, curcumin, ECG, EGCG, CG, ginsenosides. Quercetin, Naringenin, Kaempferol,Apigenin, Genistein, Schisandrin A and B, Schisandrol A and B, Curcumin Myricetin (15.0 μM), Robinetin (22.2 μM) Curcumin (5 μM) ?

t1:8

MRP4 ABCC4

Kidney

?

t1:9

MRP5 ABCC5

Most tissues

?

t1:10

BCRP

Placenta, liver, the small intestine, colon, lung, kidney

Quercetin, genistein, resveratrol, resveratrol-3-glucuronide, resveratrol-3-sulfate, malvidin, petunidin, malvidin-3-galactoside, malvidin-3,5-diglucoside, cyanidin-3-galactoside, peonidin-3-glucoside and cyanidin-3-glucoside

R O

O

Quercetin, silymarin Quercetin, Silymarin Curcumin Curcumin, retusin, ayanin, genistein, naringenin, hesperetin, acacetin, apigenin, chrysin, chrysoeriol, laricitrin, myricetin, myricetin 3′,4′,5′-trimethylether, pinocembrin, quercitrin, tamarixetin, tricetin and tricetin 3′,4′,5′-trimethylether diosmetin, luteolin, galangin, kaempferide, kaempferol, erberine, celastrol, ellagic acid, limonin, oleanolic acid, propyl gallate, sinapic acid, ursolic acid, cyanidin, peonidin, cyaniding-3,5-diglucoside, malvidin, pelargonidin, delphinidin, petunidin, delphinidin-3-glucoside, cyaniding-3-rutinoside, malvidin-3-glucoside, pelargonidin-3,5-diglucoside, malvidin-3-galactoside, cannabinoids

[290–292] [236,293,294]

[147,293,294]

[293–295] [293–296] [102,117,126,297]

220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243

C

E

R

218 219

R

217

O

215 216

C

213 214

Several flavonoids (quercetin, genistein, naringenin and hesperetin), which constitute major classes of PCs in most herbs and fruits, are inhibitors of Pgp (Table 1) and may significantly influence intestinal absorption of Pgp substrates. For example, the oral bioavailability of nonmetabolizable Pgp substrate fexofenadine was increased by 55% when coadministered with the Pgp inhibitor quercetin in humans [15]. Although quercetin also inhibits human BCRP and MRP2 [61,62], fexofenadine was not a substrate of either transporter based on a recent study using a zinc finger nuclease-mediated ABCG2/ABCC2 knock-out Caco-2 cell monolayer model [63]. Other potent Pgp inhibitors were also identified from several furanocoumarins (e.g., 6′,7′-epoxybergamottin and 6′,7′-dihydroxybergamottin) and some alkaloids (e.g., quinine, CBT-1® (a plant bisbenzylisoquinoline alkaloid), tetrandine and fangchinoline) [64–68]. Increased absorption of Pgp substrates has also been reported in patients having concomitantly taken foods and drinks rich in phytochemicals. Some of these recorded cases have been life-threatening as many of these drugs, such as the anticancer agents (e.g., irinotecan), cardiovascular agents (e.g., digoxin), and immunosuppressants (e.g., cyclosporine), have a narrow therapeutic index. For example, a lethal pharmacokinetic interaction between quercetin and digoxin was observed in pigs, which was thought to be due to enhanced digoxin absorption caused by inhibition of intestinal Pgp efflux by quercetin [69]. In addition, 1 μM epigallocatechin-3-gallate (EGCG, a major component of green tea) significantly inhibited Pgp mediated transport of digoxin in human Caco-2 monolayer. Commercial green tea may have EGCG concentrations up to 1 mM [70], implying that inhibition of intestinal Pgp by such a popular beverage could affect disposition of Pgp substrates and thus may cause side effects in humans. In addition, modulation of intestinal Pgp expression by PCs may change the pharmacokinetics of some drugs. For example, chronic use of St. John's wort can significantly induce intestinal Pgp resulting in markedly decreased absorption and bioavailability of digoxin [71,72], indinavir [73] and fexofenadine [74] in humans. For some new oral

N

211 212

U

210

T

E

D

ABCG2

Liver, intestine, kidney brain

P

ABCB1

F

t1:3

anticoagulants (e.g., dabigatran, rivaroxaban and apixaban), coadministration of St. John's wort may cause decreased drug absorption and should be avoided [75]. Flavonoids, such as tangeritin, quercetin and naringenin, up-regulate ABCB1 gene expression due to their agonistic activity on the constitutive androstane receptor (CAR)- and pregnane X receptor (PXR)-mediated pathways [76,77], which leads to an increased Pgp protein and functional activity. In contrast, curcumin, derived from the spice turmeric, down-regulated ABCB1 gene expression in several human cancer cell lines over-expressing Pgp [78–80]. Interestingly oral dosed curcumin (60 mg/kg/day) for 4 consecutive days led to a down-regulation of the intestinal Pgp but an upregulation of hepatic Pgp levels, with no effect on renal Pgp in male Sprague–Dawley rats, indicating tissue-specific modulation [81]. The maximum plasma concentrations (Cmax) and area under the concentration–time curve (AUC(0–8h)) for oral celiprolol, administered on the fourth day at 30 mg/kg, were significantly raised by 1.9- and 1.6-fold, respectively. Piperine is another well-studied alkaloid (from black pepper) with evidence of Pgp inhibition [82,83]. Piperine inhibited Pgp-mediated digoxin and cyclosporine transport across Caco-2 cell monolayers and increased intestinal absorption of fexofenadine in rats [83,84]. In addition, piperine caused an increase in Pgp protein and ABCB1 mRNA levels after incubation of Caco-2 cells for 48 and 72 h [83]. As with curcumin, in vivo administration of piperine to rats for 14 days resulted in an increase in intestinal Pgp concentrations but a decrease in the liver [85]. Given the complexity of PC/food/herb–drug interactions, these knowledge gaps have to be addressed to prevent undesirable and harmful clinical consequences.

244

2.1.2. BCRP BCRP is a 72-kDa protein composed of 665 amino acids, which is encoded by the gene ABCG2. The highest expression of BCRP occurs in the placenta (the syncytiotrophoblasts), followed by brain capillaries, the small and large intestines, liver, lung, kidney, adrenal, mammary and sweat glands, and the endothelia of veins [86–88]. Consistent

271

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270

272 273 274 275 276

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342

383

2.2.1. OATP2B1 and OATP1A2 Several uptake transporters, such as the OATPs from the SLC transporter superfamily are also functionally expressed on the apical side of human intestine tissues [5,149]. The latter have recently been reported to be involved in the oral absorption of some drugs, but there are very few studies on their interactions with PCs [5]. Recently OATP1A2 and OATP2B1 have been suggested to mediate intestinal absorption of several drugs and PCs [29,150,151]. The co-localization of these uptake and efflux transporters with their counteracting transport mechanisms makes it more difficult to predict the outcome of their PC/food–drug interactions. For example, a decreased bioavailability of celiprolol (by 87%), talinolol and fexofenadine in the presence of grapefruit juice (GFJ) has been observed in humans [152–155]. As celiprolol, talinolol and fexofenadine are substrates of both Pgp and OATP and undergo very little metabolism, it was concluded that the inhibition of intestinal OATP uptake, rather than Pgp efflux, was the major interaction between PCs and GFJ. To add to this complexity, compounds such as aliskiren (a renin inhibitor used to treat hypertension) may be a substrate for CYP3A4, Pgp and OATP2B1 (Km 72 μM) [156]. Co-administration of aliskiren with GFJ (a well-known source of various potent inhibitors of the aforementioned enzyme/transporters) decreased its oral absorption and bioavailability [157], and was explained by inhibition of OATP2B1-

384 385

O

F

2.2. SLC transporters

R O

296 297

P

294 295

343 344

D

292 293

2.1.3. Coordinated function of efflux and metabolism Accumulating evidence suggests that intestinal ABC transporters may function as barriers to absorption of drugs and PCs (e.g., resveratrol, green tea catechins and flavonoids) by cooperating with the array of phase I and II metabolic enzymes [56,129–136]. The cytochrome P450-dependent mixed-function oxidases (CYPs) 1A, 2C, 2D and 3A are subfamilies responsible for the phase I metabolism of the majority of oral drugs/PC [137]. CYP3A4 and Pgp are both present in the enterocytes and the latter increases CYP3A4-mediated intestinal metabolism of their dual substrates by controlling the “rate of presentation” of drugs to the intracellular enzyme [133–136]. Both CYP3A4 and Pgp share many of the same inducers and inhibitors. In vivo evidence has demonstrated that the oral bioavailability of these dual substrates (e.g., paclitaxel, docetaxel, cyclosporine) may be increased by concomitant administration of common PC inhibitors (e.g., quercetin and naringenin) [138–140]. The phase II conjugation reactions most often yield a more polar and water-soluble metabolite. The most common conjugation reaction for drugs/PCs usually involves the formation of a βglucuronide which is catalysed by a family of enzymes known as the uridine diphosphoglucuronosyl transferases (UGTs). Other less common conjugations with a sulfo moiety (SO − 3 ) or glutathione may also occur, catalysed by various sulfotransferases (SULTs) and glutathione-S-transferases (GSTs), respectively. Recently it has been suggested that intestinal ABC transporters may function as barriers to absorption of drugs (e.g., raloxifene, a selective estrogen receptor modulator; ezetimibe, a cholesterol absorption inhibitor) and PCs (e.g., resveratrol, green tea catechins and flavonoids) by cooperating with phase II metabolic enzymes [56,129–132,141, 142]. For example, the oral bioavailability of raloxifene, chrysin and resveratrol is severely limited by extensive intestinal phase II metabolism and active efflux of the Phase II metabolites back into the GI tract for faecal elimination [141,143–146]. BCRP and MRP2 play pivotal roles in effluxing the phase II conjugates into intestinal lumen [147,132]. Similarly, ezetimibe glucuronide is a high-affinity substrate of MRP2, while ezetimibe is transported by Pgp and MRP2. Up-regulation of intestinal Pgp, MRP2, and UGT1A1 by rifampin was associated with markedly decreased AUC for ezetimibe and its glucuronide and increased intestinal clearance, leading to nearly abolished sterol-lowering effects [148].

E

290 291

T

288 289

C

286 287

E

284 285

R

283

R

281 282

N C O

279 280

with its tissue localization, mounting evidence suggests that BCRP functions as an efflux pump limiting fetal exposure, brain penetration, intestinal absorption of substrate xenobiotics and facilitating their mammary, biliary and kidney secretion [88–100]. Drug substrates for which BCRP has the highest affinity include sulfasalazine, mitoxantrone, topotecan, daunorubicin, flavopiridol, prazosin, cladribine and rosuvastatin [26,101–104]. BCRP transports substrate drugs more efficiently at low pH, independent of the dissociation status of the substrate [105]. Several potent BCRP inhibitors that have been identified include the Pgp antagonist GF120918 (IC50, 50 nM) [106,107], a natural product fumitremorgin C (FTC) and its analogue Ko134 (both IC50 b 1 μM) [94,108] and several kinase inhibitors, such as gefitinib (IC50, 300 nM) and imatinib (IC50, 170 nM) [109,110]. The expression of ABCG2 transcripts is greatest in the duodenum and subsequently decreases in the direction of the colon [111]. To the best of our knowledge, there is no published data of the distribution of BCRP protein expression along the human GI tract. The ABCG2 mRNA level in human jejunum is higher than that of Pgp [27,112], indicating that BCRP plays an important role in limiting the intestinal absorption of its substrates. The oral bioavailability of topotecan was higher in patients heterozygous for the c.421CNA allele of ABCG2 gene than those with wild type alleles [113]. Compared to wild type mice, knocking out mouse Abcg2 led to significant increases (~ 5–111-fold) in the oral absorption and bioavailability of substrates, including sulfasalazine, daidzein, genistein, rosuvastatin and topotecan [89,96,114,115]. Although species differences in this transporter might limit the use of these knockout models in predicting the contribution of BCRP to drug bioavailability in humans, a more recent study using humanized BCRP (hBCRP) mice confirmed the important role of BCRP in oral absorption and bioavailability of typical drugs and PC substrates mentioned above [116]. Several flavonoids derived from fruit and herbs, such as quercetin, kaempferol, resveratrol and diverse anthocyanins and anthocyanidins (commonly found in grapes and berries), malvidin, petunidin, malvidin-3-galactoside, malvidin-3,5-diglucoside, cyanidin-3-galactoside, peonidin-3-glucoside and cyanidin-3-glucoside have also been identified as BCRP substrates [105,117,118]. There is accumulating evidence, mainly from in vitro studies, that many dietary and herbal PCs are potent Pgp and BCRP inhibitors. These include curcumin, retusin, ayanin, genistein, biochanin A, quercetin, morin, phloretin, silymarin (a mixture of silibinins, isosilyin A and B, silychristin A and B, and silydianin), chrysin, hesperetin, naringenin, epicatechin gallate (ECG), catechin gallate (CG) and EGCG (Table 1) [64,117,119–124]. It has been suggested that such inhibitory PCs could be used to reverse the ABC transporter-based constraints on the GI absorption of other substrate PCs/drugs and that this may represent a useful strategy for improving their bioavailability. For example, curcumin increased the bioavailability of a well-defined BCRP substrate sulfasalazine 3.2-fold at the therapeutic dose in healthy subjects [125]. Co-administration of curcumin also increased the intestinal absorption and bioavailability of sulfasalazine in wildtype but not in Abcg2(−/−) mice [13,125], suggesting that the effect was Bcrp-mediated. Two flavonoids, chrysin (IC50, 0.20 μM) and benzoflavone, potent BCRP/Bcrp inhibitors [126], have no significant effect on oral topotecan pharmacokinetics in rats or Abcb1a/1b (−/−) mice [127]. However, the oral absorption and apparent bioavailability of topotecan increased in the presence of GF120918, a dual BCRP/Pgp inhibitor, in humans and in Pgp-deficient mice [89,128]. To add to the complexity of these interactions, chrysin increased the Cmax and bioavailability of oral nitrofurantoin (a specific BCRP/Bcrp1 substrate) in rats by more than 1.7-fold [14]. The likely explanation of this increase in bioavailability, is Bcrp1 inhibition by chrysin rather than metabolism, as the contribution of CYP1A-mediated metabolism of nitrofurantoin was reported to be minor. From these examples, it is apparent that inhibition of intestinal BCRP might be substrate/inhibitor-dependent and also speciesdependent, thus warranting further evaluation in humans.

U

277 278

5

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382

386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406

6

407 408

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

t2:1 t2:2 t2:3

Table 2 An overview of tissue distribution, PC substrates and modulators of OATPs, OATs, OCTs and MATEs (?, not determined; * substrate-dependent stimulator, numbers are Km values for substrates or IC50 values for inhibitors)

414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429

t2:4

Name

t2:5 t2:6

ECG, EGCG OATP1A2 SLCO1A2 Brain (BBB), (OATPA) Kidney, Cholangiocytes SLCO1B1 Liver ? OATP1B1 (OATP2; OATPC)

PC inhibitors

OATP1B3 (OATP8; LST-2)

SLCO1B3

Liver

ECG, EGCG

t2:11

OATP2B1 (OATPB)

SLCO2B1

Liver, brain (BBB), intestine, placenta, heart

?

t2:12

OAT1

SLC22A6

Kidney

t2:13

OAT3

SLC22A8

t2:14 t2:15

OCT1 OCT2

SLC22A1 SLC22A2

Kidney, brain (BBB and choroid plexus cells) Liver, intestine Kidney, intestine

t2:16

MATE1

SLC47A1

Liver, kidney

Quinine

t2:17

MATE2-K

SLC47A2

Kidney

Quinine

O

t2:9 t2:10

PC stimulator

References

Naringin, hesperidin, ECG, EGCG, apigenin, kaempferol, quercetin

?

[150,151,252,253,298]

Biochanin A, genistein, ursolic acid, oleanolic acid, 8-trans-p-coumaroyloxy-α-terpineol, quercetin 3-Ο-α-L-arabinopyranosyl (1→2) α-L-rhamnopyranoside, silymarin, silybin A silybin B, silychristin Quercetin 3-Ο-α-L-arabinopyranosyl (1→2) α-L-rhamnopyranoside, silymarin, silybin A silybin B, silychristin

Rutin

[150,191,204,215,299,300]

E

C

PC substrates

R

Tissue location

R

t2:7 t2:8

Official symbol

T

E

D

430 431

2.2.2. Proton-coupled peptide transporters (PEPTs) The oligopeptide transporter PEPT1 (SLC15A1) is mainly expressed at the apical membranes of intestinal epithelial [168] and is committed to intestinal absorption of various drugs, including β-lactam antibiotics (e.g., amino-penicillin and cephalosporins, such as cefaclor, cephalexin, cefadroxil, cefixime, ceftibuten, and cefdinir), antivirals (e.g., L-valylacyclovir, a prodrug of acyclovir), naturally occurring peptide JBP485, anti-bacterial agent alafosfalin, angiotensin converting enzyme inhibitors (e.g., captopril and enalapril), renin inhibitors, thrombin inhibitors, antitumor drugs (e.g., betastin, 5-aminolevulinic acid), anti-epileptic agent NP-647, anti-hypotensive midodrine, metabotropic glutamate Glu2/3 receptor agonist LY544344, and a dipeptidyl derivative of LDopa, L-Phe-Dopa [6,60]. A recent study showed resveratrol increased

O

413

R O

411 412

P

409 410

reported to strongly inhibit OATP2B1 and OATP1A2 [150]. Similarly, apigenin, kaempferol, and quercetin have been reported to efficiently inhibit OATP1A2 and OATP2B1 transport activity [151]. However, it has been suggested that the contribution of OATP1A2 to intestinal drug absorption may be very minor due to its extremely low/negligible expression in human intestinal tissues [162–165]. Consequently, dietary supplements containing large amounts of abovementioned PCs may have the potential to reduce the absorption/bioavailability of OATP2B1 drug substrates, such as imatinib [166], talinolol [167], levofloxacin, fexofenadine, statins and methotrexate [29]. Thus, modulation of intestinal OATP transport by PCs may be a novel mechanism for food/ herb–drug interactions in humans. Other PC substrates and modulators of the OATPs are listed in Table 2.

F

432 433

mediated intestinal uptake of aliskiren. This perhaps implies that intracellular binding sites of Pgp and CYP3A4 are less accessible to a co-inhibitor(s) compared with those of OATP2B1, probably resulting in “first contact first service” phenomenon. Similarly, GFJ inhibited intestinal absorption of another co-substrate, montelukast and decreased its systematic exposure, which may lead to poor antiasthmatic effects in the clinic [158]. OATP2B1 also transports the toxic metabolite of irinotecan, SN-38. Such transport is saturable and pH-dependent and is decreased in the presence of baicalin (a flavonoid) or fruit juices, such as apple juice and GFJ [159]. In vivo gastrointestinal toxicity of SN-38 is a major issue with irinotecan-based chemotherapy. In mice, SN-38-induced GI toxicity was prevented by co-ingestion with apple juice. Thus, OATP2B1 may contribute to the uptake of SN-38 by intestinal tissues, triggering gastrointestinal toxicity. Apart from the conventional inhibition of bacterial β-glucuronidase in the gut with antibiotics, inhibition of OATP2B1 may be a novel strategy to prevent gastrointestinal toxicity without disturbance of the normal intestinal microflora. Inhibition of OATP2B1 by apple juice or a mixture of phloridzin, phloretin, hesperidin, and quercetin at the concentrations present in apple juice appeared to be “irreversible” (i.e., “non-competitive”) and last for a relatively long period [160], suggesting that it may be helpful for the prophylaxis of late-onset diarrhea caused by irinotecan therapy. In addition, OATP2B1-mediated uptake of estrone-3-sulfate can be inhibited by the extracts of bilberry, echinacea, green tea, banaba, grape seed, ginkgo, and soybean in HEK293 cells stably expressing human OATP2B1 [161]. Green tea catechins, ECG and EGCG, have been

U

N

C

Kaempferol-3-glucoside, isorhamnetin-3-glucoside, catechin, EC, ECG, EGCG, kaempferol-3-rutinoside, quercetin-3-rhamnoside, quercetin-3-rutinoside, 6′,7′-dihydroxybergamottin, tangeretin, naringin, naringenin, nobiletin, silymarin, silybin A silybin B, silychristin Ellagic acid, morin (300 nM), luteolin Fisetin, (470 nM), silybin, fisetin, quercetin quercetin-3′-O-sulfate, quercetin-3′-O-sulfate, genistein-4′-O-sulfate genistein-4′-O-sulfate Daidzein-7-O-glucuronide, Morin, daidzein-7-O-glucuronide, genistein-7-O-glucuronide, genistein-7-O-glucuronide, glycitein-7-O-glucuronide, glycitein-7-O-glucuronide, quercetin-3′-O-glucuronide quercetin-3′-O-glucuronide Quinine Quinine, EGCG (14 μM) Quinine Quinine

Quinine (1.9 μM) EGCG Quinine (6.4 μM) EGCG

EGCG*, quercetin [150,199,202,215,300] 3-Ο-α-L-arabinopyranosyl (1→2) α-L-rhamnopyranoside* ? [28,161,254,300–304]

?

[272,276,280,305,306]

?

[279,280,307–309]

? Quercetin, rutin, naringenin, naringin, genistein, genistin, and xanthohumol ?

[173,184] [121,173,185]

?

[70,222]

[70,222]

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524

3.1. SLC transporters

532

OCT1, OAT2, OATP1B1 and OATP1B3 are located in the sinusoid membrane of the liver (Fig. 3), extracting xenobiotics from blood and have become recognized as important determinants of hepatic uptake and clearance of a wide variety of drugs [188].

533

O

F

531

527 528 529 530

534 535 536

3.1.1. OCT1 Human OCT1 is mainly expressed in the liver and responsible for the hepatic uptake of many cationic drugs. For example, OCT1 genotype is a determinant of hepatic uptake of metformin and thus its therapeutic effects [180,189]. OCT1 is a high-capacity transporter for sumatriptan, an anti-migraine triptan, with a Km of 55 μM and Vmax of 978 pmol/mg protein/min [190]. Hepatic uptake was not affected by the Met420del polymorphism, but was strongly reduced by Arg61Cys and Gly401Ser, and completely abolished by Gly465Arg and Cys88Arg, resulting in a doubling of plasma concentrations in humans with two deficient OCT1 alleles compared with those with fully active OCT1. Interestingly, several drugs (e.g., atropine, prazosin) are potent inhibitors of OCT1 but apparently, not substrates [4]. Similarly, EGCG and green tea inhibited OCT1 with an IC50 value for green tea of 1.4% (v/v, containing 14 μM EGCG) [70]. The unbound EGCG peak concentration is estimated to be at 5–10 μM in the portal vein after drinking green tea. Thus, EGCG might inhibit hepatic uptake of an OCT1 substrate (e.g., metformin, sumatriptan) and increase its plasma concentration in patients drinking green tea or taking herbal supplements containing a high dose of EGCG.

537 538

3.1.2. OATP1B1 and OATP1B3 OATP1B1 (LST1/OATPC/OATP-2/SLCO1B3) is a liver-specific transporter that mediates the uptake into the hepatocytes of a broad range of drugs, including statins (e.g., pravastatin [191], fluvastatin [192]), antibiotics (e.g., rifampicin [193], beta lactam antibiotics [194]), angiotension II receptor blockers (e.g., olmesartan [195], valsartan [196]), HIV protease inhibitors [197], fexofenadine [198], methotrexate [199] and SN-38 [200], as well as several endogenous substrates, such as bile salts, conjugated steroids, eicosanoids, thyroid hormones and coproporphyrins I and III [201,28,165,202–204]. Accumulating evidence suggests that genetic variants affecting OATP1B1 activity are important determinants of the hepatic clearance of its substrates [205–207]. For example, in subjects containing one non-synonymous mutations (c.521TN C), both the AUC of pravastatin and renal excretion were increased nearly two-fold compared to wild-type [208], suggesting that this single polymorphism may cause compromised OATP1B1 activity and thus diminished hepatic clearance of pravastatin. Similarly, SN-38 plasma exposure was significantly increased in 521TNC allele carriers treated with irinotecan [209–211]. Severe toxicities associated with increased SN-38 exposure have also been reported in a patient with 388AN G allele [212]. Similar to OATP1B1, OATP1B3 (LST2/OATP8/SLCO1B3) is predominantly expressed in human liver [199]. It shares with OATP1B1 many common substrates, such as statins (e.g., pravastatin, rosuvastatin), angiotension II receptor blockers (e.g., olmesartan, valsartan), rifampicin, bosentan, hormone conjugates, bile salts and thyroid hormones. Evidence has indicated that several drugs including paclitaxel, docetaxel, imatinib and telmisartan are substrates of OATP1B3, but not OATP1B1 [213,214].

556 557

R O

477 478

3. Hepatobilliary excretion

P

475 476

525 526

D

473 474

2.2.3. OCT family (OCT 1 and 2, SLC22A) Members of the OCT family (SLC22A1-3) are expressed in the human GI tract and play important roles in the disposition of organic cations. Initially OCT1 was detected on the basolateral (BL) side of human small intestinal epithelial cells by using a rat Oct1 antibody [171]. However, a recent study using a validated anti-human OCT1 antibody showed that it is exclusively expressed on the apical side of human/mouse small intestine and Caco-2 cell monolayer [172]. A short hairpin RNA-mediated SLC22A1 knockdown in Caco-2 cells decreased apical uptake of pentamidine (a specific OCT1 substrate) by ∼50%, but did not alter basolateral uptake. Similarly, chemical inhibition of OCT1/Oct1decreased apical but not basolateral uptake of pentamidine into human/mouse small intestine. Typical substrates of OCT1 include cationic drugs, such as metformin, cimetidine, imatinib and pentamidine, and non-drugs such as dopamine, prostaglandin E2, Nmethylquinine and 1-methyl-4-phenylpyridinium (MPP+) [173–178]. However, OCT1/Oct1 appears to have very limited effects on intestinal absorption of metformin based on pharmacogenetics studies in humans and in Slc22a1(−/−) mice [179,180]. The plasma membrane monoamine transporter (PMAT; SLC29) is abundantly expressed on the apical side of human intestinal tissues and has been suggested to be responsible for the intestinal absorption of metformin [181]. Based on an in vitro knock-down study, OCT1 and PMAT appeared to contribute to approximately 20% and 40%, respectively, of the apical uptake of metformin in a Caco-2 cell monolayer model [182]. In addition, type 2 diabetic patients with two reduced functioning OCT1 alleles who were treated with OCT1 inhibitors, were over four times more likely to develop GI intolerance [183]. Taken together, we postulate that intestinal OCT1 may mainly act as an apical efflux pump for metformin in vivo. Reduced intestinal OCT1 activities may increase metformin accumulation in intestine tissues, leading to GI intolerance. Green tea has recently been reported to inhibit OCT1-mediated metformin transport with IC50 values of 1.4% (v/v, containing 14 μM EGCG) [70]. A comparison of the transport activity in the presence of either 10% (v/v, containing 100 μM EGCG) green tea or 100 μM EGCG shows a higher inhibitory efficacy for green tea than for 100 μM EGCG, suggesting other components in green tea may act synergistically on OCT1. The estimated gut EGCG concentration is about 1 mM after drinking green tea and is sufficient to inhibit intestinal OCT1. This might increase risk of GI intolerance to metformin especially in patients with reduced functioning OCT1 alleles but further investigations are required. OCT2 is reported to be expressed on the basolateral side of human small intestinal epithelial cells, facilitating the permeation of organic cations across the cell membrane [171]. Green tea inhibited OCT2-mediated metformin transport with IC50 values of 7.0% (v/v, containing 70 μM EGCG). Quinine is a PC substrate for both OCT1 and 2, and may act as a competitive inhibitor of OCT1 or 2 [173,184,185]. In Caco-2 cell monolayers, basolateral uptake rate of cimetidine, a substrate of OCT2, was enhanced 145–295% when co-treated with flavonoids, such as quercetin, rutin, naringenin, naringin, genistein, genistin, and xanthohumol [121]. One possible mechanism involves the interaction of some flavonoids with a wide range of protein kinases, including protein kinase C (PKC), serine/threonine kinases, cAMP-dependent protein kinase and tyrosine

kinases [186]. Kinase Yes1 inhibition in vivo diminished OCT2 activity, significantly mitigating oxaliplatin-induced acute sensory neuropathy [187]. We hypothesize that some flavonoids may be kinase activators and thus induce OCT2 phosphorylation leading to increased OCT2 function. The exact mechanisms of this “stimulated uptake” by specific flavonoids remain unknown and further studies are required.

E

472

T

471

C

469 470

E

467 468

R

466

R

464 465

N C O

462 463

intestinal permeability of bestatin via upregulation of PEPT1 in Caco-2 cells [169]. In addition, the Cmax and AUC of bestatin were significantly increased by 2.0- and 1.6-fold, respectively, in rats pretreated with resveratrol for two weeks compared with control. An inwardly directed proton gradient, which is maintained by intestinal sodium/proton ATPases, is utilized as the driving force for PEPT1-mediated uptake. Interestingly, selected flavonoids (quercetin, genistein, naringin, diosmin, acacetin, and chrysin) have been reported to stimulate apical Na+/H+-exchange and a concomitant increase in the driving force for PEPT1, leading to enhanced absorption of cefixime in Caco-2 cells [170]. This may be a possible new mechanism for drug–PC intestinal absorption interactions.

U

460 461

7

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555

558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584

8

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

Hepatocyte Pgp OATP2B1

ATP

ATP

OCT1 OATP1B1

BCRP

Bile

MRP2

MATE1

ATP

OATP1B3

F

MRP3

Hepatocyte

Blood

R O

O

Blood

Fig. 3. Localization of uptake and efflux transporter in hepatocytes.

585 586

t3:1 t3:2 t3:3

Table 3 Common phytochemical substrates and inhibitors of Pgp, MRPs, BCRP and OATPs (I, inhibitor; S, substrate; ×, not a substrate or inhibitor; ?, not determined. The numbers are either IC50 values or Ki values (with *) in μM)

595 596

D

E

T

593 594

C

591 592

E

589 590

t3:4

Phytochemicals

Pgp

BCRP

MRP1

t3:5

Apigenin

?

I (0.055)

I

t3:6 t3:7

Biochanin A Curcumin

I I

I I (1.04)

t3:8 t3:9 t3:10

Cyanidin Daidzein EGCG

I I I (99.0)

I S, I ?

t3:11 t3:12 t3:13 t3:14 t3:15

ECG Genistein Hesperetin Hesperidin Kaempferol

? I I I I

t3:16

Myricetin

I

t3:17

Narigenin

t3:18

R

587 588

implications of altered pharmacokinetics by quercetin remain unknown and may warrant further investigation. In addition to these mainly inhibitory interactions, there is also evidence that the flavonoid, rutin, may stimulate OATP1B1mediated [3 H]dehydroepiandrosterone sulfate (DHEAS) uptake [215]. In addition, EGCG from green tea and another flavonoid quercetin 3-Ο-α- L -arabinopyranosyl (1 → 2) α- L -rhamnopyranoside showed substrate-dependent stimulation of uptake by OATP1B3 [150,215]. For example, OATP1B3-mediated uptake of Fluo-3 was noncompetitively inhibited by EGCG; whereas uptake of estrone-3sulfate was strongly stimulated by EGCG at low substrate concentrations. A similar stimulation effect has been reported for OATP1B3-mediated pravastatin uptake into isogenic cells by

P

597

There is also evidence that some PCs, such as ursolic acid, oleanolic acid, and 8-trans-p-coumaroyloxy-α-terpineol (derived from Rollinia emarginata Schlecht, Annonaceae; a plant with an antiprotozoal properties) and some flavonoids (quercetin, apigenin, kaempferol, biochanin A and genistein) are potent inhibitors of OATP1B1 [215–217]. Quercetin, apigenin, kaempferol and hyperforin also inhibited OATP1B3 activity [213,217] (Table 3). Quercetin concentrations in the portal vein were estimated to be up to 110 μM after ingesting food or herbal supplements, which is about 10-fold higher than Ki for OATP1B1/1B3 [217]. In healthy male subjects, co-administration of quercetin (500 mg orally, once daily for 14 days) increased the pravastatin AUC0–10 h and the peak plasma drug concentration, prolonged its elimination half-life and decreased its apparent clearance [218]. At this stage, the clinical

MRP4

MRP5

OATP1A2

OATP 1B1

OATP 1B3

OATP 2B1

References

I

?

?

?

? ?

? ?

? I

I (20.8*) ? ?

[61,126,151,215,217,291]

? I (5) ? ? S

? ? ?

? ? ?

? ? ?

? ? S, I (18.8)

I (0.6*) I I (3.8) ? ? I

I

I I (15) ? I S

I (32.4*) ? ?

? ? I

[117] [61,116,291] [70,119,150,313]

? S, I I I I (0.086)

S I ? ? ?

S S I ? I

? ? ? ? ?

? ? ? ? I

? ? ? ? ?

[150,313] [61,116,215] [314] [314] [126,217] [314]

I (15.0) S

?

?

?

?

[235]

S, I

I (13.6) I

? I ? ? I (16.0*) ?

I ? I (67.6) I (1.92) I (21.3)

I (1.55) I

S, I (10.4) ? ? ? I (25.2) ?

?

?

I

?

I (49.2)

[61,217,295]

Naringin

I

I

?

?

?

?

?

I

I (81.6) I

I (4.63)

[61,167,295,314]

t3:19 t3:20 t3:21

Phloretin Phloridzin Quercetin

I I S, I

I ? S,I (0.21)

? S I

? S S, I

? ? ?

? ? I

? ? I

Resveratrol Silymarin

I I

S,I I

I I

? ?

S ?

I I

× I

I (1.31) I (23.2) I (9.47) (8.7*) S I (0.3)

[310,314] [314,315] [61,126,151,295]

t3:22 t3:23

? ? I (22.0) ? ?

O

C

N

R

MRP3

U

MRP2

? ? I (8.8*) S I (1.3)

? I (33.7) ? ? S, I (12.0) S ? ? ? I (24.4*) ? I (101.1) I (197.5) ? ? I (7.8*) S I (2.2)

[290,310,311] [13,78,236,242,296,312]

[105,147,295,316,317] [120,295,300,318]

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

598 599 600 601 602 603 604 605 606 607 608 609 610

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

3.2. ABC transporters

638 639

The major ABC transporters, Pgp, MRP2 and BCRP are located in the canalicular membrane of the liver, effluxing compounds into the bile [5, 225]. Evidence has indicated that the inhibition or impairment of these hepatocyte transporters can increase the concentrations of their drug/ PC substrates in the bloodstream, and/or decrease their biliary excretion, leading to a more prolonged stay in the body.

640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671

T

633 634

C

631 632

E

629 630

3.2.1. Pgp Hepatic Pgp is mainly responsible for biliary excretion of lipophilic and cationic drugs. It was reported that the biliary excretion clearance of digoxin is reduced in mdr1a/1b knock-out mice compared with wildtype [226]. The biliary excretion of camptothecin was decreased in rats treated with Pgp inhibitors such as cyclosporin A, quercetin, naringin and naringenin, compared with controls [227]. On the other hand, regulation of hepatic Pgp by PCs may result in changes in hepatic clearance of some Pgp substrates. Oral dosed curcumin (60 mg/kg/day) for 4 consecutive days led to upregulation of hepatic Pgp levels but downregulation of the intestinal Pgp in male Sprague–Dawley rat with no effect on renal Pgp [81]. This increased absorption and bioavailability of celiprolol, a Pgp substrate with minor metabolism. Pgp may not be a rate-limiting factor for biliary excretion of celiprolol. On the other hand, male Wistar rats intragastrically administered with piperine at the equivalent daily human consumption dose of 112 μg/kg for 14 days exhibited induced intestinal Pgp levels concomitant with a downregulation of hepatic Pgp. However, the clinical importance of dietary regulation of liver Pgp remains unclear.

R

627 628

R

625 626

N C O

623 624

U

621 622

F

637

619 620

3.2.2. MRP2 MRP2 transport various drug substrates, including methotrexate, vinblastine, irinotecan and its toxic metabolite SN-38, pravastatin, ceftriaxone and ampicillin [60], fosinopril (an angiotensin-converting enzyme inhibitor) [228], fexofenadine [229], lopinavir [230] and oxaliplatin [231], as well as glucuronide and sulfate metabolites of drugs or PCs [232]. For example, the biliary excretion of SN-38 glucuronide, was significantly lower in Mrp2-deficient Eisai hyperbilirubinemic (EHBR) rats when compared with normal rats [233]. Since the major

672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694

3.2.3. BCRP BCRP plays a pivotal role in the biliary excretion of its drug substrates and the sulfate metabolites of PCs and drugs (i.e., the nonMrp2-mediated component). For example, the biliary excretion of nitrofurantoin, fluoroquinolones and pitavastatin was significantly lower in Abcg2-deficient mice compared to wild-type animals [99, 100,241]. Hepatobiliary excretion of unchanged topotecan was also substantially decreased in Pgp-deficient mice pretreated with GF120918, leading to higher systemic exposure [89]. We and other groups have demonstrated that curcumin is a very potent inhibitor for human BCRP with IC50 values of 20–2000 nM [13,126, 242]. However, although curcumin has a very low oral bioavailability as mentioned previously [237], there remains the possibility that sufficiently high concentrations are achieved in the liver to potentially inhibit hepatic BCRP. For example, the liver to plasma exposure ratio for curcumin was reported to be more than 40:1 in mice [243]. Such potential food–drug interactions with BCRP may decrease hepatobiliary excretion of its drug substrates while simultaneously increasing intestinal absorption (or decreasing intestinal secretion). This could potentially have serious consequences for substrate drugs, such as the anticancer agents (e.g., imatinib and topotecan) which have a narrow therapeutic index.

695

3.2.4. MRP1 and 3 In contrast to Pgp, BCRP and MRP2, MRP1 and 3 are located at the sinusoid membrane and are involved in transporting substrates back into the bloodstream, but are expressed at low levels under normal conditions [244]. Evidence has indicated that Mrp3-null mice have enhanced GI tract toxicity after diclofenac treatment [245]. A major reactive diclofenac metabolite, diclofenac acyl glucuronide (DCF-AG) that covalently binds to biologic targets, may contribute to these adverse GI reactions. Mrp3 plays an important role in DCF-AG disposition and it also transports baicalein-7-glucuronide, a phase II metabolite of baicalein, which is present in many herbal supplements. Thus coadministration of baicalein with diclofenac may compromise MRP3/ Mrp3-mediated efflux of DCF-AG. However, the clinical relevance of this putative PC-drug interaction remains to be revealed.

717

4. Blood–brain barrier

731

D

635 636

3.1.3. Multidrug and toxin extrusion (MATE) proteins Multidrug and toxin extrusion (MATE; SLC47A) proteins contain two members, MATE1 and MATE2-K. Hepatic MATE1 is a proton antiporter expressed in the liver canalicular membrane mediating the excretion of organic cations and zwitterions into bile [220,221]. Its substrates include the vitamin thiamine, cimetidine, metformin, guanidine, procainamide, the antiviral agents (e.g., acyclovir and ganciclovir) and the antibiotics (e.g., cephalexin and cephradine) as well as an endogenous substrate creatinine. Quinine is an MATE1 substrate and a potent inhibitor of MATE1 (IC50 value of 1.9 μM) [222]. Coadministration of quinine with the antiviral agent ritonavir (a MATE1 inhibitor) markedly increased systemic exposure and elimination halflife of quinine in healthy subjects [223]. Since quinine is a well-known CYP3A4 substrate and ritonavir is a potent CYP3A4 inhibitor, inhibition of both CYP3A4 and MATE1 would explain such dramatic changes in quinine pharmacokinetics. A recent study has reported green tea (IC50, 4.9%) and EGCG inhibit MATE1-mediated transport of metformin in vitro [70]. The clinical implication of this discovery remains unclear. In vivo in Mate1(−/−) mice, the hepatic concentration of metformin was markedly higher than in Mate1(+/+) mice, leading to metformin-induced lactic acidosis [224].

617 618

O

616

R O

615

pathway for irinotecan elimination is by biliary excretion of glucuronide conjugates in humans and rodents, the pharmacokinetics of irinotecan may be susceptible to pathological conditions where MRP2 may be deficient, such as in cholestatic liver disease or in Dubin–Johnson Syndrome, or after MRP2 inhibition/induction [2,234]. Two PC inhibitors of MRP2, curcumin (IC50 5 μM) and myricetin (IC50 15 μM), are common constituents in herb supplements [235,236]. However the bioavailability of curcumin is very low with maximum plasma concentrations of 120 nM after 8 g orally per day in pancreatic cancer patients for 4 weeks [237]. Consequently, curcumin may have very limited effects on inhibition of hepatic MRP2. However, the chronic administration of PCs may result in the up-regulation of MRP2 via activation of the pregnane X receptor (PXR) or the aryl hydrocarbon receptor (AhR) in hepatocytes [81,85,238], leading to increased hepatobiliary excretion of drugs and thus a lower bioavailability. Genetic variations of MRP2 are an important predisposing factor for herbal-induced toxic liver injuries [239], suggesting compromised MRP2 activity may lead to decreased hepatobiliary excretion of PCs or their metabolites from the liver. The biliary excretion of glucuronide and sulfate conjugates of silymarin flavonolignans was reduced by approximately 96 and 78%, respectively, in Mrp2-deficient Wistar rats compared to wild type [240]. However, the effects of PC glucuronide and sulfate conjugates on liver MRP2 activity remain unknown in humans.

P

613 614

rosiglitazone (a thiazolidinedione antidiabetic drug), resulting in a 400% increase in pravastatin uptake compared to pravastatin alone [219]. Given the complexity of such food/drug interactions and the increased popularity of green tea and other herbal supplements, caution is advised when consumed with pharmacotherapy.

E

611 612

9

696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716

718 719 720 721 722 723 724 725 726 727 728 729 730

Endothelial cells that line the microvasculature of the central 732 nervous system (CNS) constitute the blood–brain barrier (BBB), 733

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

10

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

Pgp

OATP1A2

BCRP

Blood

MRP4

ATP

ATP

ATP

O

F

Endothelial

Tight junction

R O

OAT3

Brain

Fig. 4. Localization of ABC and SLC transporters in the human BBB.

742

751 752

Generally a drug must be highly lipid-soluble, or subject to uptake transport processes, to cross the BBB, and then gain access to the brain. OATP1A2 (SLCO1A2; SLC21A3) is highly expressed in the apical (blood) side of the human BBB [251–254] and may facilitate its PC substrates, such as ECG and EGCG, to penetrate into the brain [150]. OATP1A2 transports analgesic opioids (e.g., [D-penicillamine2,5]encephalin, deltorphin II) [252], antimigraine triptans (e.g., sumatriptan), levofloxacin and methotrexate [255]. Some PCs, including ECG, EGCG, apigenin, kaempferol, and quercetin are also potent inhibitors of OATP1A2 [150, 151], and thus their influence on brain exposure to OATP1A2 substrates requires more studies.

753

4.2. ABC transporters

754

The ABC transporters may represent a major rate-limiting factor in PC distribution or access to the brain. ABCB1, ABCG2, ABCC1, ABCC3, ABCC4 and ABCC5 genes have been detected in the BBB using qRT-PCR [256]. According to a recent quantitative proteomics study, the two most abundant ABC transporters in the human BBB are BCRP and Pgp [257]. Both Pgp and BCRP have been located at the apical membrane of brain capillary endothelial cells, along with MRP1 (Fig. 4) [256,258].

755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770

C

E

R

R

O

749 750

C

747 748

N

745 746

U

743 744

4.2.2. BCRP Interestingly, the uptake of typical BCRP substrates, mitoxantrone and dehydroepiandrosterone sulfate, into the brain did not vary significantly between wild type and Bcrp knockout mice [266]. In contrast in the same knockout mouse model, significant increases in the brain concentrations of topotecan (3-fold), genistein (9-fold), daidzein (6-fold) and coumestrol (4-fold) have been reported [114,116]. Brain concentrations of topotecan were moderately, but statistically significantly increased by 1.7-fold after 60 min in Abcg2 knockout mice compared to hBCRP animals [116]. BCRP expression levels in the BBB were recently determined to be around 2-fold greater in humans than in mice [257], suggesting perhaps a more limited brain penetration of these BCRP substrates in humans. Manipulation of BBB BCRP may alter brain drug/PC exposure. In a rat hemisphere perfusion study, the brain accumulation of quercetin was dramatically increased by pretreatment with GF120918 (a Pgp and BCRP inhibitor), but not with PSC833 (a Pgp inhibitor) [267]. A study using the ex vivo rat BBB model indicated that curcumin at 50 nM inhibited BCRP activity and significantly increased the penetration of sulfasalazine across the BBB [13].To put this into perspective, curcumin plasma concentrations peaked at 120 nM in pancreatic cancer patients after long-term large oral dosing [237]. Thus clinical studies on PC–drug interaction at BBB may warrant further investigation.

789 790

P

4.1. SLC transporters

771

D

741

737 738

of Glycyrrhiza glabra (licorice)) into the cerebrum increased significantly by co-perfusion with Pgp or Mrp1 inhibitors [263]. Camptothecin (a substrate for Pgp and BCRP) passage across the rat BBB was increased by a Pgp inhibitor cyclosporine but not several potent PC inhibitors of Pgp and BCRP including quercetin, naringin and naringenin [227], possibly due to the poor bioavailability of these PCs. In addition, dietary constituents and nutraceuticals may regulate the ABC transporters by activating PXR, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) or aryl hydrocarbon receptor (AhR) at the BBB [250]. For example, rats dosed with sulforaphane (an Nrf2 ligand derived from broccoli) exhibit increased protein expression of Pgp, MRP2, and BCRP at BBB and reduced delivery to the brain of a Pgp substrate verapamil [264]. Hyperforin is a constituent of St. John's wort and it is also a human PXR but not mice PXR ligand. It increased Pgp expression and transport activity in brain capillaries isolated from transgenic mice expressing human PXR (hPXR), but had no effects on BBB Pgp in wild type mice [265]. These examples highlight the fact that we are still a long way from being able to exploit our knowledge of Pgp in the clinic.

T

739 740

selectively excluding circulating drugs and toxic agents from entering the neural parenchyma but allowing essential nutrients, hormones and some drugs into the brain [246–250]. This selectivity is attributed to specialized features unique to the BBB, including expression of tight intercellular junctions that markedly limit paracellular permeability, plus a unique expression of ABC and SLC transporters that determine the transcellular permeability of nutrients, drugs and PCs (Fig. 4).

735 736

E

734

4.2.1. Pgp There is evidence that Pgp efficiently restricts the entry of a wide variety of drugs into the brain [259–261]. It also limited CNS exposure to several PCs. For example, the brain level of cryptotanshinone (a major triterpenoid of the traditional Chinese herbal medicine, Danshen) was 15- and 2-fold higher in Abcb1a(−/−) and Abcc1(−/−) mice, respectively, compared to wild-type mice, suggesting Pgp and Mrp1 may play significant roles in the restriction of cryptotanshinone penetration across the BBB [262]. Likewise in a bilateral in situ brain perfusion model, the uptake of glabridin (a flavonoid and major active constituent

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788

791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

Brush border membrane

Basolateral membrane

MRP2

ATP

MATE1, MATE2-K

OAT1 H+

Pgp

OCT2

ATP

OAT3 BCRP

816 817

OCT2, OAT1 and 3 and OATP4C1 are expressed on the basolateral side of the proximal tubule cells (PTCs) of the kidney; whereas OAT4, Pgp, MRP2, MRP4 and BCRP are on the brush border membranes, and all are involved in transporting xenobiotics and endogenous compounds out of the blood into the urine [5,268–271] (Fig. 5).

818

5.1. SLC transporters

819

5.1.1. OAT1 and 3 OAT1 and 3 are both expressed on the basolateral side of renal proximal tubule cells and take up drugs from the blood into the proximal tubule cells [272]. OAT1 can affect many systemic biological pathways including the tricarboxylic acid (TCA) cycle, tryptophan and other amino acids, fatty acids, prostaglandins, cyclic nucleotides, odorants, polyamines, and vitamins [273]. OAT3 appears to be essential for the handling of phase I and phase II metabolites, dietary flavonoids and antioxidants [274]. OAT1 and 3 share some common drug substrates, including ACE inhibitors (e.g., captopril and quinapril), diuretics (e.g., bumetanide and furosemide), ceperosporins (e.g., ceftibuten and ceftizoxime), NSAIDs (e.g., indomethacin, salicylate, ketoprofen), adefovir, methotrexate, cimetidine and ranitidine [275]. Benzylpenicillin and farmotidine appeared to be only transported by OAT3. A recent study showed that morin and luteolin are potent OAT1 inhibitors, with IC50 values of b0.3 and 0.47 μM, respectively [276]. As the in vivo Cmax values of morin and luteolin are 10 and 30-fold higher than their IC50 values for OAT1 inhibition, the potent inhibitory effect of morin and luteolin on OAT1-mediated drug transport may also occur in vivo and lead to altered renal clearance and pharmacokinetics. The renal clearance of methotrexate was reduced by 42% in the presence of morin in the rat [277]. In the clinic, decreased methotrexate clearance due to OAT inhibition may be fatal as exemplified by the potentially life-threatening methotrexate/NSAID interaction [278]. Morin also inhibited OAT3-mediated kidney accumulation of imipenem, protecting against imipenem-induced nephrotoxicity in rabbits [279]. A more recent study also suggested that epicatechin and EGCG are effective inhibitors of mouse Oat3 [274]. Many PCs are excreted as conjugated metabolites by the kidney, and consequently, there exists the possibility of competitive inhibition of these transport processes with drugs whose major route of elimination is via the kidney. For example, the sulfated conjugates of flavonoids,

830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850

C

E

R

R

828 829

N C O

826 827

U

824 825

883

5.2. Multidrug and toxin extrusion (MATE) proteins

899

Both MATE1 and MATE2-K are localized in the apical membrane of human kidney proximal tubules functioning as the final excretion step of drugs/toxins into urine. MATE2-K shares with MATE1 some common substrates such as cimetidine, metformin, guanidine and procainamide, while the zwitterionic cephalexin and cephradine were revealed to be substrates of MATE1, but not of MATE2-K [221]. While the IC50 of green tea was 4.9% (v/v) for inhibition of MATE1-mediated metformin transport, the IC50 value for MATE2-K-mediated metformin transport could not be calculated [70]. In vitro 100 μM EGCG significantly inhibited MATE1- and MATE2-K-mediated metformin transport. Such an unbound EGCG concentration may not be achieved after oral ingestion of green tea or herbal supplements.

900 901

R O

D

813

E

5. Renal excretion

T

812

822 823

5.1.2. OCT2 In human kidney proximal tubules, OCT1 was not detected but OCT2 is abundantly expressed in all three segments and plays a pivotal role of basolateral uptake of quinine, cimetidine, ranitidine, metformin and platinum drugs [285]. A very recent study showed that jatrorrhizine and epiberberine (alkaloids found in Chinese herbal medicines) are potent inhibitors of OCT2 with IC50 values of 2.3 μM and 3.1 μM, respectively [286]. After a single oral dose in rats, Cmax of jatrorrhizine and epiberberine were 5- and 8.5-fold higher than their IC50 values for OCT2, respectively. This suggests that these alkaloids may cause in vivo inhibition of renal excretion of OCT2 substrates such as metformin. Increased metformin concentration may cause life threatening risks of lactic acidosis especially in dehydrated patients or in patients with diabetic nephropathy [287]. Thus, it may be advisable that these patients avoid herbal medicines containing jatrorrhizine and epiberberine alkaloids.

P

Fig. 5. Schematic diagram of the drug transporters in the human kidney proximal tubules.

820 821

851 852

O

ATP

Proximal tubule epithelium

814 815

such as quercetin-3′-O-sulfate and genistein-4′-O-sulfate, were efficiently taken up by OAT1 in kidney [280]. On the other hand, glucuronide conjugates including daidzein-7-O-glucuronide, genistein-7-Oglucuronide, glycitein-7-O-glucuronide and quercetin-3′-O-glucuronide appeared to be preferential substrates for kidney OAT3 (Table 2). In addition, quercetin-3′-O-sulfate effectively reduced the uptake and cytotoxicity of adefovir in OAT1-overexpressing HEK293 cells [280]. These flavonoid conjugates inhibit OAT1 and OAT3 activity at physiologically relevant concentrations [280]. Since uptake by OAT1 and 3 is a rate-limiting step in the renal secretion process of many drugs, interaction of flavonoids and their conjugates with OATs may be an important mechanism for food/herb–drug interactions via inhibition of renal uptake. In addition, the therapeutic efficacy and the toxicity of many drugs may depend on the expression of OATs. The regulation of OAT transport activity in response to various stimuli can occur at several levels such as transcription, translation, and post-translational modification [281]. Post-translational regulation is usually a quick response (minutes to hours) to variable intake of drugs, fluids, or meals, as well as metabolic activity. It modifies target proteins through biochemical reactions, including glycosylation, phosphorylation, ubiquitination, sulfation, methylation, acetylation, and hydroxylation. Okadaic acid is a potent protein phosphatase inhibitor derived from sea sponge. It downregulates mOat-mediated transport of para-aminohippuric acid by enhancing phosphorylation of mOat [282]. Similarly, enhanced hOAT1 ubiquitination may lead to increased hOAT1 degradation in the proteasome, decreased hOAT1 expression at the cell surface, and inhibited hOAT1 transport activity [283]. EGCG appears to act as a proteasome inhibitor [284]. It may therefore be of interest to investigate whether EGCG can interfere with the ubiquitination of OATs. This will help better understand OAT-mediated PC–drug interactions and may warrant further investigation.

F

OAT4

11

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882

884 885 886 887 888 889 890 891 892 893 894 895 896 897 898

902 903 904 905 906 907 908 909 910 911

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933

951

6. Conclusions

952

Due to their suggested health benefits, there is increased interest in the community on PC use as supplements or as components in herbal remedies. PCs are processed by the same absorption, distribution, metabolism and excretion (ADME) mechanisms as those used by drugs [289], and thus the potential for PC–drug interactions is apparent. However, it is very difficult to predict the possibility of clinically significant PC–drug interactions because of the lack of information on the pharmacokinetics and pharmacodynamics of most PCs. Nevertheless, the greatest PC concentrations occur in the intestines and there is accumulating clinical evidence that modulation of intestinal transporters by co-administered PCs may change oral drug absorption and bioavailability. This research has confirmed that the majority of PCs appear to be multi-targeting inhibitors/modulators of both the ABC and SLC transport proteins. Most research on the interaction of the PCs with the transporters has been undertaken in vitro and has shown that both stimulation/induction and inhibition of substrate transport may result, and that the outcome is often dependent on the specific PC investigated, its concentration, exposure time, cell line and substrate used. Thus to predict clinical outcomes from in vitro data, various factors should be considered including: the available concentration of the PC in the intestines; drug/PC regimen; the different transporter kinetics (e.g., high affinity and high capacity for ABC transporters but low affinity

948 949

953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973

C

E

946 947

R

944 945

R

942 943

O

940 941

C

938 939

N

936 937

U

934 935

References

1008

T

950

5.3.2. BCRP and MRP2 BCRP is expressed on the apical membrane of the proximal tubule cells and is involved in the tubular secretion of drug/PC substrates, such as ciprofloxacin and grepafloxacin. The kidney/plasma ratios were increased 3.6- and 1.5-fold for ciprofloxacin and grepafloxacin, respectively, in Bcrp (−/−) mice compared with wild type mice [100]. Various flavonoids are extensively metabolized by the Phase II enzymes and generate their conjugated metabolites, which are taken up from the blood by OAT1 and 3. These hydrophilic conjugates are then believed to be pumped out of the kidney proximal tubular epithelium into the urine by BCRP and OAT4, and to a lesser extent by MRP2. This coupled active secretion of conjugated metabolites of PCs may ultimately accelerate elimination of PCs from the systemic circulation in humans. However, the interaction of flavonoids and their conjugates with kidney ABC transporters may alter the pharmacokinetics of their drug substrates. Given the substrate diversity of MRP2, BCRP, and OATs, it would appear cautionary to monitor plasma concentrations for drugs with a narrow therapeutic index in patients who are also consuming large amounts of PCs, perhaps as herbal medications or health supplements.

F

915 916

O

5.3.1. Pgp Pgp is expressed on the brush border membranes and is responsible for the renal excretion of its substrates such as digoxin. It is well known that drug interactions that diminish digoxin excretion by Pgp inhibitors (e.g., quinidine, verapamil, cyclosprin A), require digoxin dose adjustment to prevent any toxicity. The mechanisms of such possiblyfatal drug–drug interactions (DDIs) have been elucidated using renal tubular LLC-PK1 cells overexpressing Pgp [288]. Using the same model, quercetin, naringenin, genistein, and xanthohumol reduced Pgp-mediated transport of cimetidine [121], implying these PCs may diminish Pgp-mediated cimetidine secretion. Kidney Pgp is also subject to regulation by various PC constituents in food, herbal remedies and nutraceuticals. Rats dosed with 128 μg/kg of capsaicin showed downregulated Pgp levels in the liver and kidney, but no change in intestinal Pgp level [85]. Similarly, a study showed higher systemic exposure for per-oral digoxin in male Wistar rats co-administered with a single oral dose of capsaicin (10 or 30 mg/kg). Systemic clearance of the drug was also lower than in control group, possibly due to decreased renal secretion of digoxin by Pgp.

974 975

R O

913 914

and high capacity for PEPT1 and PMAT); the different driving forces (e.g., ATP for ABC transporters; protons for PEPT1 and PMAT); and their specific localization and activity. The PC–drug interaction data with intestinal transporters have provided a unique perspective to better understand the complexities of intestinal drug absorption. Thus, the modulation of the expression and function of intestinal transporters by PCs should always be considered in context with other factors influencing drug absorption, such as the particular drug/PC formulations, the intestinal solubility of the drugs/PCs, intestinal transit time, regional differences of luminal pH and interactions with intestinal metabolizing enzymes. In contrast to the gut, there is considerably less information on whether various PCs, or perhaps more importantly their metabolites, can modulate the activity/expression of drug transporters in the excretory organs, such as the liver and kidney. The modulation of liver or kidney transporters may influence a drug's systemic clearance and change its residence time in the body. However, it is often extremely difficult to ascertain from in vivo results whether the transporters are solely responsible for the change in pharmacokinetics, as many PCs (e.g., flavonoids) are also substrates/modulators of various CYPs and UGTs which are also involved in the elimination of many drugs. Transporters can be up- or down-regulated by activation of specific nuclear receptors or transcription factors, but information is sparse on the modulatory effects of the various PCs on the tissue-specific expression of drug transporters. An additional complexity is also often present with in vivo studies of supplements or herbal medicines which often contain a variety of different PCs which may have additive/synergistic or even antagonistic effects. Similarly, there is a lack of data of PC–transporter interactions at the BBB and other organ barriers, which may modulate the penetration of drugs into these pharmacological sanctuaries. Further in vitro and in vivo studies, both in animal models and humans, are necessary to answer these questions. Certainly, caution is particularly advised when taking supplements or herbal remedies concurrently with drugs which have a narrow therapeutic range.

P

5.3. ABC transporters

D

912

E

12

[1] L. Zhang, C.M. Brett, K.M. Giacomini, Role of organic cation transporters in drug absorption and elimination, Annu. Rev. Pharmacol. Toxicol. 38 (1998) 431–460. [2] P. Borst, R.O. Elferink, Mammalian ABC transporters in health and disease, Annu. Rev. Biochem. 71 (2002) 537–592. [3] A.H. Schinkel, J.W. Jonker, Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview, Adv. Drug Deliv. Rev. 55 (2003) 3–29. [4] H. Koepsell, K. Lips, C. Volk, Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications, Pharm. Res. 24 (2007) 1227–1251. [5] K.M. Giacomini, S.M. Huang, D.J. Tweedie, L.Z. Benet, K.L. Brouwer, X. Chu, A. Dahlin, R. Evers, V. Fischer, K.M. Hillgren, K.A. Hoffmaster, T. Ishikawa, D. Keppler, R.B. Kim, C.A. Lee, M. Niemi, J.W. Polli, Y. Sugiyama, P.W. Swaan, J.A. Ware, S.H. Wright, S.W. Yee, M.J. Zamek-Gliszczynski, L. Zhang, Membrane transporters in drug development, Nat. Rev. Drug Discov. 9 (2010) 215–236. [6] T. Nakanishi, I. Tamai, Interaction of drug or food with drug transporters in intestine and liver, Curr. Drug Metab. 16 (2015) 753–764. [7] M.A. Hediger, M.F. Romero, J.B. Peng, A. Rolfs, H. Takanaga, E.A. Bruford, The ABCs of solute carriers: physiological, pathological and therapeutic implications of human membrane transport proteins, Pflugers Arch. 447 (2004) 465–468. [8] M. Takano, R. Yumoto, T. Murakami, Expression and function of efflux drug transporters in the intestine, Pharmacol. Ther. 109 (2006) 137–161. [9] C.F. Higgins, ABC transporters: from microorganisms to man, Annu. Rev. Cell Biol. 8 (1992) 67–113. [10] L. He, K. Vasiliou, D.W. Nebert, Analysis and update of the human solute carrier (SLC) gene superfamily, Hum. Genomics 3 (2009) 195–206. [11] A. Lancon, N. Hanet, B. Jannin, D. Delmas, J.M. Heydel, G. Lizard, M.C. Chagnon, Y. Artur, N. Latruffe, Resveratrol in human hepatoma HepG2 cells: metabolism and inducibility of detoxifying enzymes, Drug Metab. Dispos. 35 (2007) 699–703. [12] N. Petri, C. Tannergren, B. Holst, F.A. Mellon, Y. Bao, G.W. Plumb, J. Bacon, K.A. O'Leary, P.A. Kroon, L. Knutson, P. Forsell, T. Eriksson, H. Lennernas, G. Williamson, Absorption/metabolism of sulforaphane and quercetin, and regulation of phase II enzymes, in human jejunum in vivo, Drug Metab. Dispos. 31 (2003) 805–813. [13] S. Shukla, H. Zaher, A. Hartz, B. Bauer, J.A. Ware, S.V. Ambudkar, Curcumin inhibits the activity of ABCG2/BCRP1, a multidrug resistance-linked ABC drug transporter in mice, Pharm. Res. 26 (2009) 480–487. [14] X. Wang, M.E. Morris, Effects of the flavonoid chrysin on nitrofurantoin pharmacokinetics in rats: potential involvement of ABCG2, Drug Metab. Dispos. 35 (2007) 268–274.

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007

1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[47] U.G. Eriksson, H. Dorani, J. Karlsson, H. Fritsch, K.J. Hoffmann, L. Olsson, T.C. Sarich, U. Wall, K.M. Schutzer, Influence of erythromycin on the pharmacokinetics of ximelagatran may involve inhibition of P-glycoprotein-mediated excretion, Drug Metab. Dispos. 34 (2006) 775–782. [48] C. Washington, G. Duran, M. Man, B. Sikic, T. Blaschke, Interaction of anti-HIV protease inhibitors with the multidrug transporter P-glycoprotein (P-gp) in human cultured cells, J. Acquir. Immune Defic. Syndr. 19 (1998) 203–209. [49] U. Schwarz, T. Gramatte, J. Krappweis, R. Oertel, W. Kirch, P-glycoprotein inhibitor erythromycin increases oral bioavailability of talinolol in humans, Int. J. Clin. Pharmacol. Ther. 38 (2000) 161–167. [50] E. Fox, S.E. Bates, Tariquidar (XR9576): a P-glycoprotein drug efflux pump inhibitor, Expert. Rev. Anticancer. Ther. 7 (2007) 447–459. [51] C.A. McDevitt, R. Callaghan, How can we best use structural information on Pglycoprotein to design inhibitors? Pharmacol. Ther. 113 (2007) 429–441. [52] I. Cascorbi, P-glycoprotein: tissue distribution, substrates, and functional consequences of genetic variations, Handb. Exp. Pharmacol. (2011) 261–283. [53] M. Huls, F.G. Russel, R. Masereeuw, The role of ATP binding cassette transporters in tissue defense and organ regeneration, J. Pharmacol. Exp. Ther. 328 (2009) 3–9. [54] J.H. Lin, Drug–drug interaction mediated by inhibition and induction of Pglycoprotein, Adv. Drug Deliv. Rev. 55 (2003) 53–81. [55] H. Toyobuku, I. Tamai, K. Ueno, A. Tsuji, Limited influence of P-glycoprotein on small-intestinal absorption of cilostazol, a high absorptive permeability drug, J. Pharm. Sci. 92 (2003) 2249–2259. [56] Y. Li, J.P. Fawcett, H. Zhang, I.G. Tucker, Transport and metabolism of MitoQ10, a mitochondria-targeted antioxidant, in Caco-2 cell monolayers, J. Pharm. Pharmacol. 59 (2007) 503–511. [57] T. Murakami, M. Takano, Intestinal efflux transporters and drug absorption, Expert Opin. Drug Metab. Toxicol. 4 (2008) 923–939. [58] C. Zimmermann, H. Gutmann, P. Hruz, J.-P. Gutzwiller, C. Beglinger, J. Drewe, Mapping of multidrug resistance gene 1 and multidrug resistance-associated protein isoform 1 to 5 mRNA expression along the human intestinal tract, Drug Metab. Dispos. 33 (2005) 219–224. [59] G. Englund, F. Rorsman, A. Ronnblom, U. Karlbom, L. Lazorova, J. Grasjo, A. Kindmark, P. Artursson, Regional levels of drug transporters along the human intestinal tract: co-expression of ABC and SLC transporters and comparison with Caco-2 cells, Eur. J. Pharm. Sci. 29 (2006) 269–277. [60] M. Estudante, J.G. Morais, G. Soveral, L.Z. Benet, Intestinal drug transporters: an overview, Adv. Drug Deliv. Rev. 65 (2013) 1340–1356. [61] Y. Imai, S. Tsukahara, S. Asada, Y. Sugimoto, Phytoestrogens/flavonoids reverse breast cancer resistance protein/ABCG2-mediated multidrug resistance, Cancer Res. 64 (2004) 4346–4352. [62] H. Kidron, G. Wissel, N. Manevski, M. Hakli, R.A. Ketola, M. Finel, M. Yliperttula, H. Xhaard, A. Urtti, Impact of probe compound in MRP2 vesicular transport assays, Eur. J. Pharm. Sci. (2012). [63] K.E. Sampson, A. Brinker, J. Pratt, N. Venkatraman, Y. Xiao, J. Blasberg, T. Steiner, M. Bourner, D.C. Thompson, Zinc finger nuclease-mediated gene knockout results in loss of transport activity for P-glycoprotein, BCRP, and MRP2 in Caco-2 cells, Drug Metab. Dispos. 43 (2015) 199–207. [64] W.V. de Castro, S. Mertens-Talcott, H. Derendorf, V. Butterweck, Grapefruit juice–drug interactions: grapefruit juice and its components inhibit P-glycoprotein (ABCB1) mediated transport of talinolol in Caco-2 cells, J. Pharm. Sci. 96 (2007) 2808–2817. [65] S.U. Choi, S.H. Park, K.H. Kim, E.J. Choi, S. Kim, W.K. Park, Y.H. Zhang, H.S. Kim, N.P. Jung, C.O. Lee, The bisbenzylisoquinoline alkaloids, tetrandine and fangchinoline, enhance the cytotoxicity of multidrug resistance-related drugs via modulation of P-glycoprotein, Anti-Cancer Drugs 9 (1998) 255–261. [66] S. Orlowski, D. Valente, M. Garrigos, E. Ezan, Bromocriptine modulates Pglycoprotein function, Biochem. Biophys. Res. Commun. 244 (1998) 481–488. [67] R.W. Robey, S. Shukla, E.M. Finley, R.K. Oldham, D. Barnett, S.V. Ambudkar, T. Fojo, S.E. Bates, Inhibition of P-glycoprotein (ABCB1)- and multidrug resistanceassociated protein 1 (ABCC1)-mediated transport by the orally administered inhibitor, CBT-1((R)), Biochem. Pharmacol. 75 (2008) 1302–1312. [68] P. Hsiao, T. Bui, R.J. Ho, J.D. Unadkat, In vitro-to-in vivo prediction of Pglycoprotein-based drug interactions at the human and rodent blood–brain barrier, Drug Metab. Dispos. 36 (2008) 481–484. [69] Y.H. Wang, P.D. Chao, S.L. Hsiu, K.C. Wen, Y.C. Hou, Lethal quercetin–digoxin interaction in pigs, Life Sci. 74 (2004) 1191–1197. [70] J. Knop, S. Misaka, K. Singer, E. Hoier, F. Muller, H. Glaeser, J. Konig, M.F. Fromm, Inhibitory effects of green tea and (−)-epigallocatechin gallate on transport by OATP1B1, OATP1B3, OCT1, OCT2, MATE1, MATE2-K and P-glycoprotein, PLoS ONE 10 (2015), e0139370. [71] D. Durr, B. Stieger, G.A. Kullak-Ublick, K.M. Rentsch, H.C. Steinert, P.J. Meier, K. Fattinger, St John's Wort induces intestinal P-glycoprotein/MDR1 and intestinal and hepatic CYP3A4, Clin. Pharmacol. Ther. 68 (2000) 598–604. [72] M.D. Perloff, L.L. von Moltke, E. Stormer, R.I. Shader, D.J. Greenblatt, Saint John's wort: an in vitro analysis of P-glycoprotein induction due to extended exposure, Br. J. Pharmacol. 134 (2001) 1601–1608. [73] S.C. Piscitelli, A.H. Burstein, D. Chaitt, R.M. Alfaro, J. Falloon, Indinavir concentrations and St John's wort, Lancet 355 (2000) 547–548. [74] G.K. Dresser, U.I. Schwarz, G.R. Wilkinson, R.B. Kim, Coordinate induction of both cytochrome P4503A and MDR1 by St John's wort in healthy subjects, Clin. Pharmacol. Ther. 73 (2003) 41–50. [75] T. Hellwig, M. Gulseth, Pharmacokinetic and pharmacodynamic drug interactions with new oral anticoagulants: what do they mean for patients with atrial fibrillation? Ann. Pharmacother. 47 (2013) 1478–1487. [76] H. Satsu, Y. Hiura, K. Mochizuki, M. Hamada, M. Shimizu, Activation of pregnane X receptor and induction of MDR1 by dietary phytochemicals, J. Agric. Food Chem. 56 (2008) 5366–5373. [77] Y. Li, Q. Wang, X. Yao, Induction of CYP3A4 and MDR1 gene expression by baicalin, baicalein, chlorogenic acid, and ginsenoside Rf through constitutive androstane receptor- and pregnane X receptor-mediated pathways, Eur. J. Pharmacol. 640 (2010) 46–54.

E

T

[15] K.A. Kim, P.W. Park, J.Y. Park, Short-term effect of quercetin on the pharmacokinetics of fexofenadine, a substrate of P-glycoprotein, in healthy volunteers, Eur. J. Clin. Pharmacol. 65 (2009) 609–614. [16] M.J. Balunas, A.D. Kinghorn, Drug discovery from medicinal plants, Life Sci. 78 (2005) 431–441. [17] J.C. Espin, M.T. Garcia-Conesa, F.A. Tomas-Barberan, Nutraceuticals: facts and fiction, Phytochemistry 68 (2007) 2986–3008. [18] S.J. Duthie, Berry phytochemicals, genomic stability and cancer: evidence for chemoprotection at several stages in the carcinogenic process, Mol. Nutr. Food Res. 51 (2007) 665–674. [19] L. Hooper, P.A. Kroon, E.B. Rimm, J.S. Cohn, I. Harvey, K.A. Le Cornu, J.J. Ryder, W.L. Hall, A. Cassidy, Flavonoids, flavonoid-rich foods, and cardiovascular risk: a metaanalysis of randomized controlled trials, Am. J. Clin. Nutr. 88 (2008) 38–50. [20] P.M. Kris-Etherton, K.D. Hecker, A. Bonanome, S.M. Coval, A.E. Binkoski, K.F. Hilpert, A.E. Griel, T.D. Etherton, Bioactive compounds in foods: their role in the prevention of cardiovascular disease and cancer, Am. J. Med. 113 (2002) 71–88. [21] A. Dembinska-Kiec, O. Mykkanen, B. Kiec-Wilk, H. Mykkanen, Antioxidant phytochemicals against type 2 diabetes, Br. J. Nutr. 99 E (Suppl. 1) (2008) ES109–ES117. [22] G.L. Russo, Ins and outs of dietary phytochemicals in cancer chemoprevention, Biochem. Pharmacol. 74 (2007) 533–544. [23] Y.J. Surh, Cancer chemoprevention with dietary phytochemicals, Nat. Rev. Cancer 3 (2003) 768–780. [24] O.I. Aruoma, T. Bahorun, L.-S. Jen, Neuroprotection by bioactive components in medicinal and food plant extracts, Mutat. Res. Rev. Mutat. Res. 544 (2003) 203–215. [25] N. Khan, V.M. Adhami, H. Mukhtar, Apoptosis by dietary agents for prevention and treatment of cancer, Biochem. Pharmacol. 76 (2008) 1333–1339. [26] T. Litman, T.E. Druley, W.D. Stein, S.E. Bates, From MDR to MXR: new understanding of multidrug resistance systems, their properties and clinical significance, Cell. Mol. Life Sci. 58 (2001) 931–959. [27] J. Taipalensuu, H. Tornblom, G. Lindberg, C. Einarsson, F. Sjoqvist, H. Melhus, P. Garberg, B. Sjostrom, B. Lundgren, P. Artursson, Correlation of gene expression of ten drug efflux proteins of the ATP-binding cassette transporter family in normal human jejunum and in human intestinal epithelial Caco-2 cell monolayers, J. Pharmacol. Exp. Ther. 299 (2001) 164–170. [28] G.A. Kullak-Ublick, M.G. Ismair, B. Stieger, L. Landmann, R. Huber, F. Pizzagalli, K. Fattinger, P.J. Meier, B. Hagenbuch, Organic anion-transporting polypeptide B (OATP-B) and its functional comparison with three other OATPs of human liver, Gastroenterology 120 (2001) 525–533. [29] I. Tamai, Oral drug delivery utilizing intestinal OATP transporters, Adv. Drug Deliv. Rev. (2011). [30] J.W. Jonker, E. Wagenaar, C.A. Mol, M. Buitelaar, H. Koepsell, J.W. Smit, A.H. Schinkel, Reduced hepatic uptake and intestinal excretion of organic cations in mice with a targeted disruption of the organic cation transporter 1 (Oct1 [Slc22a1]) gene, Mol. Cell. Biol. 21 (2001) 5471–5477. [31] J.W. Jonker, A.H. Schinkel, Pharmacological and physiological functions of the polyspecific organic cation transporters: OCT1, 2, and 3 (SLC22A1-3), J. Pharmacol. Exp. Ther. 308 (2004) 2–9. [32] X. Ming, D.R. Thakker, Role of basolateral efflux transporter MRP4 in the intestinal absorption of the antiviral drug adefovir dipivoxil, Biochem. Pharmacol. (2009). [33] H.M. Prime-Chapman, R.A. Fearn, A.E. Cooper, V. Moore, B.H. Hirst, Differential multidrug resistance-associated protein 1 through 6 isoform expression and function in human intestinal epithelial Caco-2 cells, J. Pharmacol. Exp. Ther. 311 (2004) 476–484. [34] G.L. Kellett, The facilitated component of intestinal glucose absorption, J. Physiol. 531 (2001) 585–595. [35] F.J. Sharom, ABC multidrug transporters: structure, function and role in chemoresistance, Pharmacogenomics 9 (2008) 105–127. [36] K. Choi, T.O. Frommel, R.K. Stern, C.F. Perez, M. Kriegler, T. Tsuruo, I.B. Roninson, Multidrug resistance after retroviral transfer of the human MDR1 gene correlates with P-glycoprotein density in the plasma membrane and is not affected by cytotoxic selection, Proc. Natl. Acad. Sci. U. S. A. 88 (1991) 7386–7390. [37] I. Tamai, A.R. Safa, Azidopine noncompetitively interacts with vinblastine and cyclosporin A binding to P-glycoprotein in multidrug resistant cells, J. Biol. Chem. 266 (1991) 16796–16800. [38] H.W. van Veen, K. Venema, H. Bolhuis, I. Oussenko, J. Kok, B. Poolman, A.J. Driessen, W.N. Konings, Multidrug resistance mediated by a bacterial homolog of the human multidrug transporter MDR1, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 10668–10672. [39] G. Chen, G.E. Duran, K.A. Steger, N.J. Lacayo, J.P. Jaffrezou, C. Dumontet, B.I. Sikic, Multidrug-resistant human sarcoma cells with a mutant P-glycoprotein, altered phenotype, and resistance to cyclosporins, J. Biol. Chem. 272 (1997) 5974–5982. [40] B. Sikic, G. Fisher, B. Lum, J. Halsey, L. Beketic-Oreskovic, G. Chen, Modulation and prevention of multidrug resistance by inhibitors of P-glycoprotein, Cancer Chemother. Pharmacol. 40 (1997) 13–19 (Suppl.). [41] A.I. Alvarez, M. Perez, J.G. Prieto, A.J. Molina, R. Real, G. Merino, Fluoroquinolone efflux mediated by ABC transporters, J. Pharm. Sci. 97 (2008) 3483–3493. [42] D.S. Cox, K.R. Scott, H. Gao, S. Raje, N.D. Eddington, Influence of multidrug resistance (MDR) proteins at the blood–brain barrier on the transport and brain distribution of enaminone anticonvulsants, J. Pharm. Sci. 90 (2001) 1540–1552. [43] C. Pascaud, M. Garrigos, Multidrug resistance transporter P-glycoprotein has distinct but interactive binding sites for cytotoxic drugs and reversing agents, Biochem. J. 333 (1998) 3351–3358. [44] H. Spahn-Langguth, G. Baktir, A. Radschuweit, A. Okyar, B. Terhaag, P. Ader, A. Hanafy, P. Langguth, 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. 36 (1998) 16–24. [45] M.E. Cavet, M. West, N.L. Simmons, Transport and epithelial secretion of the cardiac glycoside, digoxin, by human intestinal epithelial (Caco-2) cells, Br. J. Pharmacol. 118 (1996) 1389–1396. [46] G. Fricker, H. Gutmann, A. Droulle, J. Drewe, D. Miller, Epithelial transport of anthelmintic ivermectin in a novel model of isolated proximal kidney tubules, Pharm. Res. 16 (1999) 1570–1575.

U

1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138

13

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229

D

P

R O

O

F

[104] M. Jani, P. Szabo, E. Kis, E. Molnar, H. Glavinas, P. Krajcsi, Kinetic characterization of sulfasalazine transport by human ATP-binding cassette G2, Biol. Pharm. Bull. 32 (2009) 497–499. [105] P. Breedveld, D. Pluim, G. Cipriani, F. Dahlhaus, M.A. van Eijndhoven, C.J. de Wolf, A. Kuil, J.H. Beijnen, G.L. Scheffer, G. Jansen, P. Borst, J.H. Schellens, The effect of low pH on breast cancer resistance protein (ABCG2)-mediated transport of methotrexate, 7-hydroxymethotrexate, methotrexate diglutamate, folic acid, mitoxantrone, topotecan, and resveratrol in in vitro drug transport models, Mol. Pharmacol. 71 (2007) 240–249. [106] M. de Bruin, K. Miyake, T. Litman, R. Robey, S.E. Bates, Reversal of resistance by GF120918 in cell lines expressing the ABC half-transporter, MXR, Cancer Lett. 146 (1999) 117–126. [107] J.D. Allen, R.F. Brinkhuis, J. Wijnholds, A.H. Schinkel, The mouse Bcrp1/Mxr/Abcp gene: amplification and overexpression in cell lines selected for resistance to topotecan, mitoxantrone, or doxorubicin, Cancer Res. 59 (1999) 4237–4241. [108] S.K. Rabindran, D. Ross, A. Doyle, W. Yang, L.M. Greenberger, Fumitremorgin C reverses multidrug resistance in cells transfected with the breast cancer resistance protein, Cancer Res. 60 (2000) 47–50. [109] C. Ozvegy-Laczka, T. Hegedus, G. Varady, O. Ujhelly, J.D. Schuetz, A. Varadi, G. Keri, L. Orfi, K. Nemet, B. Sarkadi, High-affinity interaction of tyrosine kinase inhibitors with the ABCG2 multidrug transporter, Mol. Pharmacol. 65 (2004) 1485–1495. [110] P.J. Houghton, G.S. Germain, F.C. Harwood, J.D. Schuetz, C.F. Stewart, E. Buchdunger, P. Traxler, Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro, Cancer Res. 64 (2004) 2333–2337. [111] H. Gutmann, P. Hruz, C. Zimmermann, C. Beglinger, J. Drewe, Distribution of breast cancer resistance protein (BCRP/ABCG2) mRNA expression along the human GI tract, Biochem. Pharmacol. 70 (2005) 695–699. [112] M. Maliepaard, G.L. Scheffer, I.F. Faneyte, M.A. van Gastelen, A.C. Pijnenborg, A.H. Schinkel, M.J. van De Vijver, R.J. Scheper, J.H. Schellens, Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues, Cancer Res. 61 (2001) 3458–3464. [113] A. Sparreboom, H. Gelderblom, S. Marsh, R. Ahluwalia, R. Obach, P. Principe, C. Twelves, J. Verweij, H.L. McLeod, Diflomotecan pharmacokinetics in relation to ABCG2 421CNA genotype, Clin. Pharmacol. Ther. 76 (2004) 38–44. [114] J. Enokizono, H. Kusuhara, Y. Sugiyama, Effect of breast cancer resistance protein (bcrp/abcg2) on the disposition of phytoestrogens, Mol. Pharmacol. 72 (2007) 967–975. [115] T. Karibe, R. Hagihara-Nakagomi, K. Abe, T. Imaoka, T. Mikkaichi, S. Yasuda, M. Hirouchi, N. Watanabe, N. Okudaira, T. Izumi, Evaluation of the usefulness of breast cancer resistance protein (BCRP) knockout mice and BCRP inhibitor-treated monkeys to estimate the clinical impact of BCRP modulation on the pharmacokinetics of BCRP substrates, Pharm. Res. 32 (2015) 1634–1647. [116] S. Dallas, L. Salphati, D. Gomez-Zepeda, T. Wanek, L. Chen, X. Chu, J. Kunta, M. Mezler, M.-C. Menet, S. Chasseigneaux, X. Decleves, O. Langer, E. Pierre, K. DiLoreto, C. Hoft, L. Laplanche, J. Pang, T. Pereira, C. Andonian, D. Simic, A. Rode, J. Yabut, X. Zhang, N. Scheer, Generation and characterization of a breast cancer resistance protein humanized mouse model, Mol. Pharmacol. (2016). [117] A. Dreiseitel, B. Oosterhuis, K.V. Vukman, P. Schreier, A. Oehme, S. Locher, G. Hajak, P.G. Sand, Berry anthocyanins and anthocyanidins exhibit distinct affinities for the efflux transporters BCRP and MDR1, Br. J. Pharmacol. 158 (2009) 1942–1950. [118] G. An, J. Gallegos, M.E. Morris, The bioflavonoid kaempferol is an Abcg2 substrate and inhibits Abcg2-mediated quercetin efflux, Drug Metab. Dispos. 39 (2011) 426–432. [119] J. Jodoin, M. Demeule, R. Beliveau, Inhibition of the multidrug resistance Pglycoprotein activity by green tea polyphenols, Biochim. Biophys. Acta 1542 (2002) 149–159. [120] S. Zhang, M.E. Morris, Effect of the flavonoids biochanin A and silymarin on the Pglycoprotein-mediated transport of digoxin and vinblastine in human intestinal Caco-2 cells, Pharm. Res. 20 (2003) 1184–1191. [121] J.S. Taur, R. Rodriguez-Proteau, Effects of dietary flavonoids on the transport of cimetidine via P-glycoprotein and cationic transporters in Caco-2 and LLC-PK1 cell models, Xenobiotica 38 (2008) 1536–1550. [122] E.J. Wang, M. Barecki-Roach, W.W. Johnson, Elevation of P-glycoprotein function by a catechin in green tea, Biochem. Biophys. Res. Commun. 297 (2002) 412–418. [123] A.F. Castro, G.A. Altenberg, Inhibition of drug transport by genistein in multidrugresistant cells expressing P-glycoprotein, Biochem. Pharmacol. 53 (1997) 89–93. [124] A. Pick, H. Muller, R. Mayer, B. Haenisch, I.K. Pajeva, M. Weigt, H. Bonisch, C.E. Muller, M. Wiese, Structure–activity relationships of flavonoids as inhibitors of breast cancer resistance protein (BCRP), Bioorg. Med. Chem. 19 (2011) 2090–2102. [125] H. Kusuhara, H. Furuie, A. Inano, A. Sunagawa, S. Yamada, C. Wu, S. Fukizawa, N. Morimoto, I. Ieiri, M. Morishita, K. Sumita, H. Mayahara, T. Fujita, K. Maeda, Y. Sugiyama, Pharmacokinetic interaction study of sulfasalazine in healthy subjects and the impact of curcumin as an in vivo inhibitor of BCRP, Br. J. Pharmacol. 166 (2012) 1793–1803. [126] K.W. Tan, Y. Li, J.W. Paxton, N.P. Birch, A. Scheepens, Identification of novel dietary phytochemicals inhibiting the efflux transporter breast cancer resistance protein (BCRP/ABCG2), Food Chem. 138 (2013) 2267–2274. [127] S. Zhang, X. Wang, K. Sagawa, M.E. Morris, Flavonoids chrysin and benzoflavone, potent breast cancer resistance protein inhibitors, have no significant effect on topotecan pharmacokinetics in rats or mdr1a/1b (−/−) mice, Drug Metab. Dispos. 33 (2005) 341–348. [128] C.M. Kruijtzer, J.H. Beijnen, H. Rosing, W.W. ten Bokkel Huinink, M. Schot, R.C. Jewell, E.M. Paul, J.H. Schellens, Increased oral bioavailability of topotecan in combination with the breast cancer resistance protein and P-glycoprotein inhibitor GF120918, J. Clin. Oncol. 20 (2002) 2943–2950. [129] B. Ebert, A. Seidel, A. Lampen, Identification of BCRP as transporter of benzo[a]pyrene conjugates metabolically formed in Caco-2 cells and its induction by Ah-receptor agonists, Carcinogenesis 26 (2005) 1754–1763.

N

C

O

R

R

E

C

T

[78] S. Anuchapreeda, P. Leechanachai, M.M. Smith, S.V. Ambudkar, P.N. Limtrakul, Modulation of P-glycoprotein expression and function by curcumin in multidrug-resistant human KB cells, Biochem. Pharmacol. 64 (2002) 573–582. [79] B.H. Choi, C.G. Kim, Y. Lim, S.Y. Shin, Y.H. Lee, Curcumin down-regulates the multidrug-resistance mdr1b gene by inhibiting the PI3K/Akt/NF kappa B pathway, Cancer Lett. 259 (2008) 111–118. [80] X.L. Hou, K. Takahashi, K. Tanaka, K. Tougou, F. Qiu, K. Komatsu, J. Azuma, Curcuma drugs and curcumin regulate the expression and function of P-gp in Caco-2 cells in completely opposite ways, Int. J. Pharm. 358 (2008) 224–229. [81] W. Zhang, T.M. Tan, L.Y. Lim, Impact of curcumin-induced changes in Pglycoprotein and CYP3A expression on the pharmacokinetics of peroral celiprolol and midazolam in rats, Drug Metab. Dispos. 35 (2007) 110–115. [82] R.K. Bhardwaj, H. Glaeser, L. Becquemont, U. Klotz, S.K. Gupta, M.F. Fromm, Piperine, a major constituent of black pepper, inhibits human P-glycoprotein and CYP3A4, J. Pharmacol. Exp. Ther. 302 (2002) 645–650. [83] Y. Han, T.M. Chin Tan, L.Y. Lim, In vitro and in vivo evaluation of the effects of piperine on P-gp function and expression, Toxicol. Appl. Pharmacol. 230 (2008) 283–289. [84] M.J. Jin, H.K. Han, Effect of piperine, a major component of black pepper, on the intestinal absorption of fexofenadine and its implication on food–drug interaction, J. Food Sci. 75 (2010) H93–H96. [85] W. Zhang, Y. Han, S.L. Lim, L.Y. Lim, Dietary regulation of P-gp function and expression, Expert Opin. Drug Metab. Toxicol. 5 (2009) 789–801. [86] L.A. Doyle, W. Yang, L.V. Abruzzo, T. Krogmann, Y. Gao, A.K. Rishi, D.D. Ross, A multidrug resistance transporter from human MCF-7 breast cancer cells, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 15665–15670. [87] P.A. Fetsch, A. Abati, T. Litman, K. Morisaki, Y. Honjo, K. Mittal, S.E. Bates, Localization of the ABCG2 mitoxantrone resistance-associated protein in normal tissues, Cancer Lett. 235 (2006) 84–92. [88] J.W. Jonker, G. Merino, S. Musters, A.E. van Herwaarden, E. Bolscher, E. Wagenaar, E. Mesman, T.C. Dale, A.H. Schinkel, The breast cancer resistance protein BCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk, Nat. Med. 11 (2005) 127–129. [89] J.W. Jonker, J.W. Smit, R.F. Brinkhuis, M. Maliepaard, J.H. Beijnen, J.H. Schellens, A.H. Schinkel, Role of breast cancer resistance protein in the bioavailability and fetal penetration of topotecan, J. Natl. Cancer Inst. 92 (2000) 1651–1656. [90] L. Zhou, S.B. Naraharisetti, H. Wang, J.D. Unadkat, M.F. Hebert, Q. Mao, The breast cancer resistance protein (Bcrp1/Abcg2) limits fetal distribution of glyburide in the pregnant mouse: an Obstetric-Fetal Pharmacology Research Unit Network and University of Washington Specialized Center of Research Study, Mol. Pharmacol. 73 (2008) 949–959. [91] F. Staud, Z. Vackova, K. Pospechova, P. Pavek, M. Ceckova, A. Libra, L. Cygalova, P. Nachtigal, Z. Fendrich, Expression and transport activity of breast cancer resistance protein (Bcrp/Abcg2) in dually perfused rat placenta and HRP-1 cell line, J. Pharmacol. Exp. Ther. 319 (2006) 53–62. [92] P. Breedveld, D. Pluim, G. Cipriani, P. Wielinga, O. van Tellingen, A.H. Schinkel, J.H. Schellens, 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. 65 (2005) 2577–2582. [93] S. Cisternino, C. Mercier, F. Bourasset, F. Roux, J.M. Scherrmann, Expression, upregulation, and transport activity of the multidrug-resistance protein Abcg2 at the mouse blood–brain barrier, Cancer Res. 64 (2004) 3296–3301. [94] J.D. Allen, A. van Loevezijn, J.M. Lakhai, M. van der Valk, O. van Tellingen, G. Reid, J.H.M. Schellens, G.-J. Koomen, A.H. Schinkel, Potent and specific inhibition of the breast cancer resistance protein multidrug transporter in vitro and in mouse intestine by a novel analogue of fumitremorgin C, Mol. Cancer Ther. 1 (2002) 417–425. [95] S. Marchetti, N.A. de Vries, T. Buckle, M.J. Bolijn, M.A. van Eijndhoven, J.H. Beijnen, R. Mazzanti, O. van Tellingen, J.H. Schellens, 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. 7 (2008) 2280–2287. [96] H. Zaher, A.A. Khan, J. Palandra, T.G. Brayman, L. Yu, J.A. Ware, Breast cancer resistance protein (Bcrp/abcg2) is a major determinant of sulfasalazine absorption and elimination in the mouse, Mol. Pharm. 3 (2006) 55–61. [97] C. Kondo, R. Onuki, H. Kusuhara, H. Suzuki, M. Suzuki, N. Okudaira, M. Kojima, K. Ishiwata, J.W. Jonker, Y. Sugiyama, Lack of improvement of oral absorption of ME3277 by prodrug formation is ascribed to the intestinal efflux mediated by breast cancer resistant protein (BCRP/ABCG2), Pharm. Res. 22 (2005) 613–618. [98] N. Mizuno, M. Suzuki, H. Kusuhara, H. Suzuki, K. Takeuchi, T. Niwa, J.W. Jonker, Y. Sugiyama, Impaired renal excretion of 6-hydroxy-5,7-dimethyl-2-methylamino4-(3-pyridylmethyl) benzothiazole (E3040) sulfate in breast cancer resistance protein (BCRP1/ABCG2) knockout mice, Drug Metab. Dispos. 32 (2004) 898–901. [99] M. Hirano, K. Maeda, S. Matsushima, Y. Nozaki, H. Kusuhara, Y. Sugiyama, Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin, Mol. Pharmacol. 68 (2005) 800–807. [100] T. Ando, H. Kusuhara, G. Merino, A.I. Alvarez, A.H. Schinkel, Y. Sugiyama, Involvement of breast cancer resistance protein (ABCG2) in the biliary excretion mechanism of fluoroquinolones, Drug Metab. Dispos. 35 (2007) 1873–1879. [101] C. Ozvegy, T. Litman, G. Szakacs, Z. Nagy, S. Bates, A. Varadi, B. Sarkadi, Functional characterization of the human multidrug transporter, ABCG2, expressed in insect cells, Biochem. Biophys. Res. Commun. 285 (2001) 111–117. [102] C. de Wolf, R. Jansen, H. Yamaguchi, M. de Haas, K. van de Wetering, J. Wijnholds, J. Beijnen, P. Borst, Contribution of the drug transporter ABCG2 (breast cancer resistance protein) to resistance against anticancer nucleosides, Mol. Cancer Ther. 7 (2008) 3092–3102. [103] S. Kitamura, K. Maeda, Y. Wang, Y. Sugiyama, Involvement of multiple transporters in the hepatobiliary transport of rosuvastatin, Drug Metab. Dispos. 36 (2008) 2014–2023.

U

1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

E

14

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[160] Y. Shirasaka, M. Shichiri, T. Mori, T. Nakanishi, I. Tamai, Major active components in grapefruit, orange, and apple juices responsible for OATP2B1-mediated drug interactions, J. Pharm. Sci. 102 (2013) 280–288. [161] H. Fuchikami, H. Satoh, M. Tsujimoto, S. Ohdo, H. Ohtani, Y. Sawada, Effects of herbal extracts on the function of human organic anion-transporting polypeptide OATP-B, Drug Metab. Dispos. 34 (2006) 577–582. [162] Y. Meier, J.J. Eloranta, J. Darimont, M.G. Ismair, C. Hiller, M. Fried, G.A. Kullak-Ublick, S.R. Vavricka, Regional distribution of solute carrier mRNA expression along the human intestinal tract, Drug Metab. Dispos. 35 (2007) 590–594. [163] M. Drozdzik, C. Groer, J. Penski, J. Lapczuk, M. Ostrowski, Y. Lai, B. Prasad, J.D. Unadkat, W. Siegmund, S. Oswald, Protein abundance of clinically relevant multidrug transporters along the entire length of the human intestine, Mol. Pharm. 11 (2014) 3547–3555. [164] C. Groer, S. Bruck, Y. Lai, A. Paulick, A. Busemann, C.D. Heidecke, W. Siegmund, S. Oswald, LC–MS/MS-based quantification of clinically relevant intestinal uptake and efflux transporter proteins, J. Pharm. Biomed. Anal. 85 (2013) 253–261. [165] I. Tamai, J. Nezu, H. Uchino, Y. Sai, A. Oku, M. Shimane, A. Tsuji, Molecular identification and characterization of novel members of the human organic anion transporter (OATP) family, Biochem. Biophys. Res. Commun. 273 (2000) 251–260. [166] Y. Yamakawa, A. Hamada, T. Shuto, M. Yuki, T. Uchida, H. Kai, T. Kawaguchi, H. Saito, Pharmacokinetic impact of SLCO1A2 polymorphisms on imatinib disposition in patients with chronic myeloid leukemia, Clin. Pharmacol. Ther. 90 (2011) 157–163. [167] Y. Shirasaka, Y. Li, Y. Shibue, E. Kuraoka, H. Spahn-Langguth, Y. Kato, P. Langguth, I. Tamai, Concentration-dependent effect of naringin on intestinal absorption of β1adrenoceptor antagonist talinolol mediated by P-glycoprotein and organic anion transporting polypeptide (Oatp), Pharm. Res. 26 (2009) 560–567. [168] H. Daniel, G. Kottra, The proton oligopeptide cotransporter family SLC15 in physiology and pharmacology, Pflugers Arch. - Eur. J. Physiol. 447 (2004) 610–618. [169] Y. Jia, Z. Liu, X. Huo, C. Wang, Q. Meng, Q. Liu, H. Sun, P. Sun, X. Yang, X. Shu, K. Liu, Enhancement effect of resveratrol on the intestinal absorption of bestatin by regulating PEPT1, MDR1 and MRP2 in vivo and in vitro, Int. J. Pharm. 495 (2015) 588–598. [170] U. Wenzel, S. Kuntz, H. Daniel, Flavonoids with epidermal growth factor-receptor tyrosine kinase inhibitory activity stimulate PEPT1-mediated cefixime uptake into human intestinal epithelial cells, J. Pharmacol. Exp. Ther. 299 (2001) 351–357. [171] J. Muller, K.S. Lips, L. Metzner, R.H. Neubert, H. Koepsell, M. Brandsch, Drug specificity and intestinal membrane localization of human organic cation transporters (OCT), Biochem. Pharmacol. 70 (2005) 1851–1860. [172] T. Han, R.S. Everett, W.R. Proctor, C.M. Ng, C.L. Costales, K.L.R. Brouwer, D.R. Thakker, Organic cation transporter 1 (OCT1/mOct1) is localized in the apical membrane of Caco-2 cell monolayers and enterocytes, Mol. Pharmacol. 84 (2013) 182–189. [173] V. Gorboulev, J.C. Ulzheimer, A. Akhoundova, I. Ulzheimer-Teuber, U. Karbach, S. Quester, C. Baumann, F. Lang, A.E. Busch, H. Koepsell, Cloning and characterization of two human polyspecific organic cation transporters, DNA Cell Biol. 16 (1997) 871–881. [174] L. Zhang, M. Dresser, A. Gray, S. Yost, S. Terashita, K. Giacomini, Cloning and functional expression of a human liver organic cation transporter, Mol. Pharmacol. 51 (1997) 913–921. [175] M. Hayer-Zillgen, M. Bruss, H. Bonisch, Expression and pharmacological profile of the human organic cation transporters hOCT1, hOCT2 and hOCT3, Br. J. Pharmacol. 136 (2002) 829–836. [176] L. Zhang, M.E. Schaner, K.M. Giacomini, Functional characterization of an organic cation transporter (hOCT1) in a transiently transfected human cell line (HeLa), J. Pharmacol. Exp. Ther. 286 (1998) 354–361. [177] X. Ming, W. Ju, H. Wu, R.R. Tidwell, J.E. Hall, D.R. Thakker, Transport of dicationic drugs pentamidine and furamidine by human organic cation transporters, Drug Metab. Dispos. 37 (2009) 424–430. [178] L. Wang, A. Giannoudis, S. Lane, P. Williamson, M. Pirmohamed, R.E. Clark, Expression of the uptake drug transporter hOCT1 is an important clinical determinant of the response to imatinib in chronic myeloid leukemia, Clin. Pharmacol. Ther. 83 (2008) 258–264. [179] M.V. Tzvetkov, S.V. Vormfelde, D. Balen, I. Meineke, T. Schmidt, D. Sehrt, I. Sabolić, H. Koepsell, J. Brockmöller, The effects of genetic polymorphisms in the organic cation transporters OCT1, OCT2, and OCT3 on the renal clearance of metformin, Clin. Pharmacol. Ther. 86 (2009) 299–306. [180] Y. Shu, C. Brown, R.A. Castro, R.J. Shi, E.T. Lin, R.P. Owen, S.A. Sheardown, L. Yue, E.G. Burchard, C.M. Brett, K.M. Giacomini, Effect of genetic variation in the organic cation transporter 1, OCT1, on metformin pharmacokinetics, Clin. Pharmacol. Ther. 83 (2008) 273–280. [181] M. Zhou, L. Xia, J. Wang, Metformin transport by a newly cloned proton-stimulated organic cation transporter (plasma membrane monoamine transporter) expressed in human intestine, Drug Metab. Dispos. 35 (2007) 1956–1962. [182] T. Han, W.R. Proctor, C.L. Costales, H. Cai, R.S. Everett, D.R. Thakker, Four cationselective transporters contribute to apical uptake and accumulation of metformin in Caco-2 cell monolayers, J. Pharmacol. Exp. Ther. 352 (2015) 519–528. [183] T. Dujic, K. Zhou, L.A. Donnelly, R. Tavendale, C.N.A. Palmer, E.R. Pearson, Association of organic cation transporter 1 with intolerance to metformin in type 2 diabetes: a godarts study, Diabetes 64 (2015) 1786–1793. [184] J.E. van Montfoort, M. Muller, G.M. Groothuis, D.K. Meijer, H. Koepsell, P.J. Meier, Comparison of “type I” and “type II” organic cation transport by organic cation transporters and organic anion-transporting polypeptides, J. Pharmacol. Exp. Ther. 298 (2001) 110–115. [185] D.H. Sweet, D.S. Miller, J.B. Pritchard, Ventricular choline transport: a role for organic cation transporter 2 expressed in choroid plexus, J. Biol. Chem. 276 (2001) 41611–41619. [186] M. Rumsby, L. Drew, J. Warr, Protein kinases and multidrug resistance, Cytotechnology 27 (1998) 203–224. [187] J.A. Sprowl, S.S. Ong, A.A. Gibson, S. Hu, G. Du, W. Lin, L. Li, S. Bharill, R.A. Ness, A. Stecula, S.M. Offer, R.B. Diasio, A.T. Nies, M. Schwab, G. Cavaletti, E. Schlatter, G.

E

T

[130] L. Zhang, Y. Zheng, M.S.S. Chow, Z. Zuo, Investigation of intestinal absorption and disposition of green tea catechins by Caco-2 monolayer model, Int. J. Pharm. 287 (2004) 1–12. [131] L. Zhang, Z. Zuo, G. Lin, Intestinal and hepatic glucuronidation of flavonoids, Mol. Pharm. 4 (2007) 833–845. [132] M.E. Juan, E. Gonzalez-Pons, J.M. Planas, Multidrug resistance proteins restrain the intestinal absorption of trans-resveratrol in rats, J. Nutr. 140 (2010) 489–495. [133] L.S. Gan, M.A. Moseley, B. Khosla, P.F. Augustijns, T.P. Bradshaw, R.W. Hendren, D.R. Thakker, CYP3A-like cytochrome P450-mediated metabolism and polarized efflux of cyclosporin A in Caco-2 cells, Drug Metab. Dispos. 24 (1996) 344–349. [134] P.B. Watkins, The barrier function of CYP3A4 and P-glycoprotein in the small bowel, Adv. Drug Deliv. Rev. 27 (1997) 161–170. [135] V.J. Wacher, L. Salphati, L.Z. Benet, Active secretion and enterocytic drug metabolism barriers to drug absorption, Adv. Drug Deliv. Rev. 46 (2001) 89–102. [136] L.Z. Benet, The drug transporter-metabolism alliance: uncovering and defining the interplay, Mol. Pharm. 6 (2009) 1631–1643. [137] D.F. Lewis, Y. Ito, Human cytochromes P450 in the metabolism of drugs: new molecular models of enzyme–substrate interactions, Expert Opin. Drug Metab. Toxicol. 4 (2008) 1181–1186. [138] J.S. Choi, H.K. Choi, S.C. Shin, Enhanced bioavailability of paclitaxel after oral coadministration with flavone in rats, Int. J. Pharm. 275 (2004) 165–170. [139] J.S. Choi, B.W. Jo, Y.C. Kim, Enhanced paclitaxel bioavailability after oral administration of paclitaxel or prodrug to rats pretreated with quercetin, Eur. J. Pharm. Biopharm. 57 (2004) 313–318. [140] Y. Zhang, L.Z. Benet, The gut as a barrier to drug absorption: combined role of cytochrome P450 3A and P-glycoprotein, Clin. Pharmacokinet. 40 (2001) 159–168. [141] E.J. Jeong, H. Lin, M. Hu, Disposition mechanisms of raloxifene in the human intestinal Caco-2 model, J. Pharmacol. Exp. Ther. (2004) (jpet.103.063925). [142] T. Kosoglou, P. Statkevich, A.O. Johnson-Levonas, J.F. Paolini, A.J. Bergman, K.B. Alton, Ezetimibe: a review of its metabolism, pharmacokinetics and drug interactions, Clin. Pharmacokinet. 44 (2005) 467–494. [143] C.D. Santi, A. Pietrabissa, R. Spisni, F. Mosca, G.M. Pacifici, Sulphation of resveratrol, a natural product present in grapes and wine, in the human liver and duodenum, Xenobiotica 30 (2000) 609–617. [144] G. Kuhnle, J.P.E. Spencer, G. Chowrimootoo, H. Schroeter, E.S. Debnam, S.K.S. Srai, C. Rice-Evans, U. Hahn, Resveratrol is absorbed in the small intestine as resveratrol glucuronide, Biochem. Biophys. Res. Commun. 272 (2000) 212–217. [145] U.K. Walle, A. Galijatovic, T. Walle, Transport of the flavonoid chrysin and its conjugated metabolites by the human intestinal cell line Caco-2, Biochem. Pharmacol. 58 (1999) 431–438. [146] T. Walle, Y. Otake, J.A. Brubaker, U.K. Walle, P.V. Halushka, Disposition and metabolism of the flavonoid chrysin in normal volunteers, Br. J. Clin. Pharmacol. 51 (2001) 143–146. [147] K. van de Wetering, A. Burkon, W. Feddema, A. Bot, H. de Jonge, V. Somoza, P. Borst, Intestinal breast cancer resistance protein (BCRP)/Bcrp1 and multidrug resistance protein 3 (MRP3)/Mrp3 are involved in the pharmacokinetics of resveratrol, Mol. Pharmacol. 75 (2009) 876–885. [148] S. Oswald, S. Haenisch, C. Fricke, T. Sudhop, C. Remmler, T. Giessmann, G. Jedlitschky, U. Adam, E. Dazert, R. Warzok, W. Wacke, I. Cascorbi, H.K. Kroemer, W. Weitschies, K. von Bergmann, W. Siegmund, Intestinal expression of Pglycoprotein (ABCB1), multidrug resistance associated protein 2 (ABCC2), and uridine diphosphate-glucuronosyltransferase 1A1 predicts the disposition and modulates the effects of the cholesterol absorption inhibitor ezetimibe in humans, Clin. Pharmacol. Ther. 79 (2006) 206–217. [149] B. Hagenbuch, P.J. Meier, The superfamily of organic anion transporting polypeptides, Biochim. Biophys. Acta Biomembr. 1609 (2003) 1–18. [150] M. Roth, B.N. Timmermann, B. Hagenbuch, Interactions of green tea catechins with organic anion-transporting polypeptides, Drug Metab. Dispos. 39 (2011) 920–926. [151] K. Mandery, K. Bujok, I. Schmidt, M. Keiser, W. Siegmund, B. Balk, J. Konig, M.F. Fromm, H. Glaeser, Influence of the flavonoids apigenin, kaempferol, and quercetin on the function of organic anion transporting polypeptides 1A2 and 2B1, Biochem. Pharmacol. 80 (2010) 1746–1753. [152] C. Banfield, S. Gupta, M. Marino, J. Lim, M. Affrime, Grapefruit juice reduces the oral bioavailability of fexofenadine but not desloratadine, Clin. Pharmacokinet. 41 (2002) 311–318. [153] G.K. Dresser, D.G. Bailey, B.F. Leake, U.I. Schwarz, P.A. Dawson, D.J. Freeman, R.B. Kim, Fruit juices inhibit organic anion transporting polypeptide-mediated drug uptake to decrease the oral availability of fexofenadine, Clin. Pharmacol. Ther. 71 (2002) 11–20. [154] U.I. Schwarz, D. Seemann, R. Oertel, S. Miehlke, E. Kuhlisch, M.F. Fromm, R.B. Kim, D.G. Bailey, W. Kirch, Grapefruit juice ingestion significantly reduces talinolol bioavailability, Clin. Pharmacol. Ther. 77 (2005) 291–301. [155] J.J. Lilja, J.T. Backman, J. Laitila, H. Luurila, P.J. Neuvonen, Itraconazole increases but grapefruit juice greatly decreases plasma concentrations of celiprolol, Clin. Pharmacol. Ther. 73 (2003) 192–198. [156] S. Vaidyanathan, G. Camenisch, H. Schuetz, C. Reynolds, C.M. Yeh, M.N. Bizot, H.A. Dieterich, D. Howard, W.P. Dole, Pharmacokinetics of the oral direct renin inhibitor aliskiren in combination with digoxin, atorvastatin, and ketoconazole in healthy subjects: the role of P-glycoprotein in the disposition of aliskiren, J. Clin. Pharmacol. 48 (2008) 1323–1338. [157] T. Tapaninen, P.J. Neuvonen, M. Niemi, Grapefruit juice greatly reduces the plasma concentrations of the OATP2B1 and CYP3A4 substrate aliskiren, Clin. Pharmacol. Ther. 88 (2010) 339–342. [158] E.B. Mougey, H. Feng, M. Castro, C.G. Irvin, J.J. Lima, Absorption of montelukast is transporter mediated: a common variant of OATP2B1 is associated with reduced plasma concentrations and poor response, Pharmacogenet. Genomics 19 (2009) 129–138. [159] D. Fujita, Y. Saito, T. Nakanishi, I. Tamai, Organic anion transporting polypeptide (OATP)2B1 contributes to gastrointestinal toxicity of anticancer drug SN-38, active metabolite of irinotecan hydrochloride, Drug Metab. Dispos. 44 (2016) 1–7.

U

1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502

15

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593

[197]

[198]

[199]

[200]

[201]

[202]

[203]

[204]

[205]

[206]

[207] [208]

[209]

[210]

[211]

F

O

R O

P

D

[196]

[212] H. Takane, M. Miyata, N. Burioka, J. Kurai, Y. Fukuoka, H. Suyama, Y. Shigeoka, K. Otsubo, I. Ieiri, E. Shimizu, Severe toxicities after irinotecan-based chemotherapy in a patient with lung cancer: a homozygote for the SLCO1B1*15 allele, Ther. Drug Monit. 29 (2007) 666–668. [213] N.F. Smith, M.R. Acharya, W.D. Figg, A. Sparreboom, Identification of OATP1B3 as a high-affinity hepatocellular transporter of paclitaxel, Cancer Res. 65 (2005) 970. [214] S. Hu, R.M. Franke, K.K. Filipski, C. Hu, S.J. Orwick, E.A. de Bruijn, H. Burger, S.D. Baker, A. Sparreboom, Interaction of imatinib with human organic ion carriers, Clin. Cancer Res. 14 (2008) 3141–3148. [215] X. Wang, A.W. Wolkoff, M.E. Morris, Flavonoids as a novel class of human organic anion-transporting polypeptide OATP1B1 (OATP-C) modulators, Drug Metab. Dispos. 33 (2005) 1666–1672. [216] M. Roth, J.J. Araya, B.N. Timmermann, B. Hagenbuch, Isolation of modulators of the liver specific organic anion transporting polypeptides (OATPs) 1B1 and 1B3 from Rollinia emarginata Schlecht (Annonaceae), J. Pharmacol. Exp. Ther. (2011). [217] K. Mandery, B. Balk, K. Bujok, I. Schmidt, M.F. Fromm, H. Glaeser, Inhibition of hepatic uptake transporters by flavonoids, Eur. J. Pharm. Sci. (2012). [218] L.X. Wu, C.X. Guo, W.Q. Chen, J. Yu, Q. Qu, Y. Chen, Z.R. Tan, G. Wang, L. Fan, Q. Li, W. Zhang, H.H. Zhou, Inhibition of the organic anion-transporting polypeptide 1B1 by quercetin: an in vitro and in vivo assessment, Br. J. Clin. Pharmacol. 73 (2012) 750–757. [219] I. Bachmakov, H. Glaeser, M.F. Fromm, J. Konig, Interaction of oral antidiabetic drugs with hepatic uptake transporters: focus on organic anion transporting polypeptides and organic cation transporter 1, Diabetes 57 (2008) 1463–1469. [220] A.T. Nies, K. Damme, E. Schaeffeler, M. Schwab, Multidrug and toxin extrusion proteins as transporters of antimicrobial drugs, Expert Opin. Drug Metab. Toxicol. 8 (2012) 1565–1577. [221] H. Motohashi, K. Inui, Multidrug and toxin extrusion family SLC47: physiological, pharmacokinetic and toxicokinetic importance of MATE1 and MATE2-K, Mol. Asp. Med. 34 (2013) 661–668. [222] B. Astorga, S. Ekins, M. Morales, S.H. Wright, Molecular determinants of ligand selectivity for the human multidrug and toxin extruder proteins MATE1 and MATE2K, J. Pharmacol. Exp. Ther. 341 (2012) 743–755. [223] J.O. Soyinka, C.O. Onyeji, S.I. Omoruyi, A.R. Owolabi, P.V. Sarma, J.M. Cook, Pharmacokinetic interactions between ritonavir and quinine in healthy volunteers following concurrent administration, Br. J. Clin. Pharmacol. 69 (2010) 262–270. [224] K. Toyama, A. Yonezawa, S. Masuda, R. Osawa, M. Hosokawa, S. Fujimoto, N. Inagaki, K. Inui, T. Katsura, Loss of multidrug and toxin extrusion 1 (MATE1) is associated with metformin-induced lactic acidosis, Br. J. Pharmacol. 166 (2012) 1183–1191. [225] G.J.E.J. Hooiveld, J.E. van Montfoort, D.K.F. Meijer, M. Muller, Function and regulation of ATP-binding cassette transport proteins involved in hepatobiliary transport, Eur. J. Pharm. Sci. 12 (2000) 13–30. [226] U. Mayer, E. Wagenaar, J.H. Beijnen, J.W. Smit, D.K. Meijer, J. van Asperen, P. Borst, A.H. Schinkel, Substantial excretion of digoxin via the intestinal mucosa and prevention of long-term digoxin accumulation in the brain by the mdr 1a Pglycoprotein, Br. J. Pharmacol. 119 (1996) 1038–1044. [227] T.H. Tsai, C.H. Lee, P.H. Yeh, Effect of P-glycoprotein modulators on the pharmacokinetics of camptothecin using microdialysis, Br. J. Pharmacol. 134 (2001) 1245–1252. [228] B.R. Green, L.J. Bain, Mrp2 is involved in the efflux and disposition of fosinopril, J. Appl. Toxicol. 33 (2013) 458–465. [229] Y. Akamine, M. Miura, S. Sunagawa, H. Kagaya, N. Yasui-Furukori, T. Uno, Influence of drug-transporter polymorphisms on the pharmacokinetics of fexofenadine enantiomers, Xenobiotica 40 (2010) 782–789. [230] R.A. van Waterschoot, R. ter Heine, E. Wagenaar, C.M. van der Kruijssen, R.W. Rooswinkel, A.D. Huitema, J.H. Beijnen, A.H. Schinkel, Effects of cytochrome P450 3A (CYP3A) and the drug transporters P-glycoprotein (MDR1/ABCB1) and MRP2 (ABCC2) on the pharmacokinetics of lopinavir, Br. J. Pharmacol. 160 (2010) 1224–1233. [231] K. Myint, Y. Li, J. Paxton, M. McKeage, Multidrug resistance-associated protein 2 (MRP2) mediated transport of oxaliplatin-derived platinum in membrane vesicles, PLoS ONE 10 (2015), e0130727. [232] M.J. Zamek-Gliszczynski, K.A. Hoffmaster, K. Nezasa, M.N. Tallman, K.L. Brouwer, Integration of hepatic drug transporters and phase II metabolizing enzymes: mechanisms of hepatic excretion of sulfate, glucuronide, and glutathione metabolites, Eur. J. Pharm. Sci. 27 (2006) 447–486. [233] X.Y. Chu, Y. Kato, K. Niinuma, K.I. Sudo, H. Hakusui, Y. Sugiyama, Multispecific organic anion transporter is responsible for the biliary excretion of the camptothecin derivative irinotecan and its metabolites in rats, J. Pharmacol. Exp. Ther. 281 (1997) 304–314. [234] V. Keitel, J. Kartenbeck, A.T. Nies, H. Spring, M. Brom, D. Keppler, Impaired protein maturation of the conjugate export pump multidrug resistance protein 2 as a consequence of a deletion mutation in Dubin–Johnson syndrome, Hepatology 32 (2000) 1317–1328. [235] J.J. van Zanden, H.M. Wortelboer, S. Bijlsma, A. Punt, M. Usta, P.J. Bladeren, I.M. Rietjens, N.H. Cnubben, Quantitative structure activity relationship studies on the flavonoid mediated inhibition of multidrug resistance proteins 1 and 2, Biochem. Pharmacol. 69 (2005) 699–708. [236] H.M. Wortelboer, M. Usta, A.E. van der Velde, M.G. Boersma, B. Spenkelink, J.J. van Zanden, I.M.C.M. Rietjens, P.J. van Bladeren, N.H.P. Cnubben, Interplay between MRP inhibition and metabolism of MRP inhibitors: the case of curcumin, Chem. Res. Toxicol. 16 (2003) 1642–1651. [237] N. Dhillon, B.B. Aggarwal, R.A. Newman, R.A. Wolff, A.B. Kunnumakkara, J.L. Abbruzzese, C.S. Ng, V. Badmaev, R. Kurzrock, Phase II trial of curcumin in patients with advanced pancreatic cancer, Clin. Cancer Res. 14 (2008) 4491–4499. [238] S.L. Lim, L.Y. Lim, Effects of citrus fruit juices on cytotoxicity and drug transport pathways of Caco-2 cell monolayers, Int. J. Pharm. 307 (2006) 42–50. [239] J.H. Choi, B.M. Ahn, J. Yi, J.H. Lee, S.W. Nam, C.Y. Chon, K.H. Han, S.H. Ahn, I.J. Jang, J.Y. Cho, Y. Suh, M.O. Cho, J.E. Lee, K.H. Kim, M.G. Lee, MRP2 haplotypes confer

T

[195]

C

[194]

E

[193]

R

[192]

R

[191]

O

[190]

C

[189]

N

[188]

Ciarimboli, J.H.M. Schellens, E.Y. Isacoff, A. Sali, T. Chen, S.D. Baker, A. Sparreboom, N. Pabla, A phosphotyrosine switch regulates organic cation transporters, Nat. Commun. 7 (2016). P.J. Meier, U. Eckhardt, A. Schroeder, B. Hagenbuch, B. Stieger, Substrate specificity of sinusoidal bile acid and organic anion uptake systems in rat and human liver, Hepatology 26 (1997) 1667–1677. Y. Shu, S.A. Sheardown, C. Brown, R.P. Owen, S. Zhang, R.A. Castro, A.G. Ianculescu, L. Yue, J.C. Lo, E.G. Burchard, C.M. Brett, K.M. Giacomini, Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action, J. Clin. Invest. 117 (2007) 1422–1431. J. Matthaei, D. Kuron, F. Faltraco, T. Knoch, J.N. Dos Santos Pereira, M. Abu Abed, T. Prukop, J. Brockmöller, M.V. Tzvetkov, OCT1 mediates hepatic uptake of sumatriptan and loss-of-function OCT1 polymorphisms affect sumatriptan pharmacokinetics, Clin. Pharmacol. Ther. (2016) (n/a-n/a). B. Hsiang, Y. Zhu, Z. Wang, Y. Wu, V. Sasseville, W.-P. Yang, T.G. Kirchgessner, A novel human hepatic organic anion transporting polypeptide (oatp2). identification of a liver-specific human organic anion transporting polypeptide and identification of rat and human hydroxymethylglutaryl-COA reductase inhibitor transporters, J. Biol. Chem. 274 (1999) 37161–37168. K. Kopplow, K. Letschert, J. Konig, B. Walter, D. Keppler, Human hepatobiliary transport of organic anions analyzed by quadruple-transfected cells, Mol. Pharmacol. 68 (2005) 1031–1038. R.G. Tirona, B.F. Leake, A.W. Wolkoff, R.B. Kim, Human organic anion transporting polypeptide-C (SLC21A6) is a major determinant of rifampin-mediated pregnane X receptor activation, J. Pharmacol. Exp. Ther. 304 (2003) 223–228. M. Nakakariya, T. Shimada, M. Irokawa, T. Maeda, I. Tamai, Identification and species similarity of OATP transporters responsible for hepatic uptake of beta-lactam antibiotics, Drug Metab. Pharmacokinet. 23 (2008) 347–355. R. Nakagomi-Hagihara, D. Nakai, K. Kawai, Y. Yoshigae, T. Tokui, T. Abe, T. Ikeda, OATP1B1, OATP1B3, and mrp2 are involved in hepatobiliary transport of olmesartan, a novel angiotensin II blocker, Drug Metab. Dispos. 34 (2006) 862–869. W. Yamashiro, K. Maeda, M. Hirouchi, Y. Adachi, Z.H. Hu, Y. Sugiyama, Involvement of transporters in the hepatic uptake and biliary excretion of valsartan, a selective antagonist of the angiotensin II AT1-receptor, in humans, Drug Metab. Dispos. 34 (2006) 1247–1254. R.C. Hartkoorn, W.S. Kwan, V. Shallcross, A. Chaikan, N. Liptrott, D. Egan, E.S. Sora, C.E. James, S. Gibbons, P.G. Bray, D.J. Back, S.H. Khoo, A. Owen, HIV protease inhibitors are substrates for OATP1A2, OATP1B1 and OATP1B3 and lopinavir plasma concentrations are influenced by SLCO1B1 polymorphisms, Pharmacogenet. Genomics 20 (2010) 112–120. M. Cvetkovic, B. Leake, M.F. Fromm, G.R. Wilkinson, R.B. Kim, OATP and Pglycoprotein transporters mediate the cellular uptake and excretion of fexofenadine, Drug Metab. Dispos. 27 (1999) 866–871. T. Abe, M. Unno, T. Onogawa, T. Tokui, T.N. Kondo, R. Nakagomi, H. Adachi, K. Fujiwara, M. Okabe, T. Suzuki, K. Nunoki, E. Sato, M. Kakyo, T. Nishio, J. Sugita, N. Asano, M. Tanemoto, M. Seki, F. Date, K. Ono, Y. Kondo, K. Shiiba, M. Suzuki, H. Ohtani, T. Shimosegawa, K. Iinuma, H. Nagura, S. Ito, S. Matsuno, LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers, Gastroenterology 120 (2001) 1689–1699. T. Nozawa, H. Minami, S. Sugiura, A. Tsuji, I. Tamai, Role of organic anion transporter OATP1B1 (OATP-C) in hepatic uptake of irinotecan and its active metabolite, 7ethyl-10-hydroxycamptothecin: in vitro evidence and effect of single nucleotide polymorphisms, Drug Metab. Dispos. 33 (2005) 434–439. H. Shen, J. Dai, T. Liu, Y. Cheng, W. Chen, C. Freeden, Y. Zhang, W.G. Humphreys, P. Marathe, Y. Lai, Coproporphyrins I and III as functional markers of OATP1B activity: in vitro and in vivo evaluation in preclinical species, J. Pharmacol. Exp. Ther. (2016). J. Konig, Y. Cui, A.T. Nies, D. Keppler, Localization and genomic organization of a new hepatocellular organic anion transporting polypeptide, J. Biol. Chem. 275 (2000) 23161–23168. Y. Cui, J. Konig, I. Leier, U. Buchholz, D. Keppler, Hepatic uptake of bilirubin and its conjugates by the human organic anion transporter SLC21A6, J. Biol. Chem. 276 (2001) 9626–9630. J. Konig, Y. Cui, A.T. Nies, D. Keppler, A novel human organic anion transporting polypeptide localized to the basolateral hepatocyte membrane, Am. J. Physiol. Gastrointest. Liver Physiol. 278 (2000) G156–G164. I. Ieiri, S. Suwannakul, K. Maeda, H. Uchimaru, K. Hashimoto, M. Kimura, H. Fujino, M. Hirano, H. Kusuhara, S. Irie, S. Higuchi, Y. Sugiyama, SLCO1B1 (OATP1B1, an uptake transporter) and ABCG2 (BCRP, an efflux transporter) variant alleles and pharmacokinetics of pitavastatin in healthy volunteers, Clin. Pharmacol. Ther. 82 (2007) 541–547. I. Ieiri, S. Higuchi, Y. Sugiyama, Genetic polymorphisms of uptake (OATP1B1, 1B3) and efflux (MRP2, BCRP) transporters: implications for inter-individual differences in the pharmacokinetics and pharmacodynamics of statins and other clinically relevant drugs, Expert Opin. Drug Metab. Toxicol. 5 (2009) 703–729. A. Kalliokoski, M. Niemi, Impact of OATP transporters on pharmacokinetics, Br. J. Pharmacol. 158 (2009) 693–705. J. Mwinyi, A. Johne, S. Bauer, I. Roots, T. Gerloff, Evidence for inverse effects of OATP-C (SLC21A6) 5 and 1b haplotypes on pravastatin kinetics, Clin. Pharmacol. Ther. 75 (2004) 415–421. W.A. Teft, S. Welch, J. Lenehan, J. Parfitt, Y.H. Choi, E. Winquist, R.B. Kim, OATP1B1 and tumour OATP1B3 modulate exposure, toxicity, and survival after irinotecanbased chemotherapy, Br. J. Cancer 112 (2015) 857–865. J.Y. Han, H.S. Lim, E.S. Shin, Y.K. Yoo, Y.H. Park, J.E. Lee, H.T. Kim, J.S. Lee, Influence of the organic anion-transporting polypeptide 1B1 (OATP1B1) polymorphisms on irinotecan-pharmacokinetics and clinical outcome of patients with advanced non-small cell lung cancer, Lung Cancer 59 (2008) 69–75. F. Innocenti, D.L. Kroetz, E. Schuetz, M.E. Dolan, J. Ramirez, M. Relling, P. Chen, S. Das, G.L. Rosner, M.J. Ratain, Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics, J. Clin. Oncol. 27 (2009) 2604–2614.

U

1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 1615 1616 1617 1618 1619 1620 1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 1664 1665 1666 1667 1668 1669 1670 1671 1672 1673 1674 1675 1676 1677 1678 1679 1680 1681 1682 1683 1684

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

E

16

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698 1699 1700 1701 1702 1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726 1727 1728 1729 1730 1731 1732 1733 1734 1735 1736 1737 1738 1739 1740 1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768 1769 1770 1771 1772 1773 1774 1775

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[266] Y.J. Lee, H. Kusuhara, J.W. Jonker, A.H. Schinkel, Y. Sugiyama, 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. 312 (2005) 44–52. [267] K.A. Youdim, M.Z. Qaiser, D.J. Begley, C.A. Rice-Evans, N.J. Abbott, Flavonoid permeability across an in situ model of the blood–brain barrier, Free Radic. Biol. Med. 36 (2004) 592–604. [268] M. Ichikawa, A. Yoshimura, T. Sumizawa, N. Shudo, Y. Kuwazuru, T. Furukawa, S. Akiyama, Interaction of organic chemicals with P-glycoprotein in the adrenal gland, kidney, and a multidrug-resistant KB cell, J. Biol. Chem. 266 (1991) 903–908. [269] E.M. Leslie, R.G. Deeley, S.P. Cole, Multidrug resistance proteins: role of Pglycoprotein, MRP1, MRP2, and BCRP (ABCG2) in tissue defense, Toxicol. Appl. Pharmacol. 204 (2005) 216–237. [270] M. Huls, C.D. Brown, A.S. Windass, R. Sayer, J.J. van den Heuvel, S. Heemskerk, F.G. Russel, R. Masereeuw, The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane, Kidney Int. 73 (2008) 220–225. [271] R.A. van Aubel, P.H. Smeets, J.G. Peters, R.J. Bindels, F.G. Russel, The MRP4/ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: putative efflux pump for urinary cAMP and cGMP, J. Am. Soc. Nephrol. 13 (2002) 595–603. [272] M. Hosoyamada, T. Sekine, Y. Kanai, H. Endou, Molecular cloning and functional expression of a multispecific organic anion transporter from human kidney, Am. J. Phys. 276 (1999) F122–F128. [273] H.C. Liu, N. Jamshidi, Y. Chen, S.A. Eraly, S.Y. Cho, V. Bhatnagar, W. Wu, K.T. Bush, R. Abagyan, B.O. Palsson, S.K. Nigam, Construction and evaluation of an organic anion transporter 1 (OAT1)-centered metabolic network, J. Biol. Chem. (2016). [274] W. Wu, N. Jamshidi, S.A. Eraly, H.C. Liu, K.T. Bush, B.O. Palsson, S.K. Nigam, Multispecific drug transporter Slc22a8 (Oat3) regulates multiple metabolic and signaling pathways, Drug Metab. Dispos. 41 (2013) 1825–1834. [275] G. Burckhardt, Drug transport by Organic Anion Transporters (OATs), Pharmacol. Ther. 136 (2012) 106–130. [276] G. An, X. Wang, M.E. Morris, Flavonoids are inhibitors of human organic anion transporter 1 (OAT1)-mediated transport, Drug Metab. Dispos. 42 (2014) 1357–1366. [277] S.S. Hong, M.-J. Jin, H.-K. Han, Enhanced systemic availability of methotrexate in the presence of morin in rats, Biopharm. Drug Dispos. 29 (2008) 189–193. [278] M.L. Frenia, K.S. Long, E.A. Hartshorn, Methotrexate and nonsteroidal antiinflammatory drug interactions, Ann. Pharmacother. 26 (1992) 234–237. [279] S.C. Lim, Y.B. Im, C.S. Bae, S.I. Han, S.E. Kim, H.K. Han, Protective effect of morin on the imipenem-induced nephrotoxicity in rabbits, Arch. Pharm. Res. 31 (2008) 1060–1065. [280] C.C. Wong, N.P. Botting, C. Orfila, N. Al-Maharik, G. Williamson, Flavonoid conjugates interact with organic anion transporters (OATs) and attenuate cytotoxicity of adefovir mediated by organic anion transporter 1 (OAT1/SLC22A6), Biochem. Pharmacol. 81 (2011) 942–949. [281] D. Xu, H. Wang, G. You, Posttranslational regulation of organic anion transporters by ubiquitination: known and novel, Med. Res. Rev. (2016) (n/a-n/a). [282] G. You, K. Kuze, R.A. Kohanski, K. Amsler, S. Henderson, Regulation of mOATmediated organic anion transport by okadaic acid and protein kinase c in llc-pk1 cells, J. Biol. Chem. 275 (2000) 10278–10284. [283] D. Xu, H. Wang, C. Gardner, Z. Pan, P.L. Zhang, J. Zhang, G. You, The role of Nedd4-1 WW domains in binding and regulating human organic anion transporter 1, Am. J. Physiol. Ren. Physiol. 311 (2016) F320–F329. [284] K. Osanai, K.R. Landis-Piwowar, Q.P. Dou, T.H. Chan, A para-amino substituent on the D-ring of green tea polyphenol epigallocatechin-3-gallate as a novel proteasome inhibitor and cancer cell apoptosis inducer, Bioorg. Med. Chem. 15 (2007) 5076–5082. [285] T. Fujita, T.J. Urban, M.K. Leabman, K. Fujita, K.M. Giacomini, Transport of drugs in the kidney by the human organic cation transporter, OCT2 and its genetic variants, J. Pharm. Sci. 95 (2006) 25–36. [286] L. Li, S. Sun, Y. Weng, F. Song, S. Zhou, M. Bai, H. Zhou, S. Zeng, H. Jiang, Interaction of six protoberberine alkaloids with human organic cation transporters 1, 2 and 3, Xenobiotica 46 (2016) 175–183. [287] S.C. Gan, J. Barr, A.I. Arieff, R.G. Pearl, Biguanide-associated lactic acidosis. Case report and review of the literature, Arch. Intern. Med. 152 (1992) 2333–2336. [288] Y. Tanigawara, N. Okamura, M. Hirai, M. Yasuhara, K. Ueda, N. Kioka, T. Komano, R. Hori, Transport of digoxin by human P-glycoprotein expressed in a porcine kidney epithelial cell line (LLC-PK1), J. Pharmacol. Exp. Ther. 263 (1992) 840–845. [289] Y. Li, J. Lu, J.W. Paxton, The role of ABC and SLC transporters in the pharmacokinetics of dietary and herbal phytochemicals and their interactions with xenobiotics, Curr. Drug Metab. 13 (2012) 624–639. [290] C.H. Versantvoort, G.J. Schuurhuis, H.M. Pinedo, C.A. Eekman, C.M. Kuiper, J. Lankelma, H.J. Broxterman, Genistein modulates the decreased drug accumulation in non-P-glycoprotein mediated multidrug resistant tumour cells, Br. J. Cancer 68 (1993) 939–946. [291] C.H. Versantvoort, H.J. Broxterman, J. Lankelma, N. Feller, H.M. Pinedo, Competitive inhibition by genistein and ATP dependence of daunorubicin transport in intact MRP overexpressing human small cell lung cancer cells, Biochem. Pharmacol. 48 (1994) 1129–1136. [292] C.H. Versantvoort, T. Rhodes, P.R. Twentyman, Acceleration of MRP-associated efflux of rhodamine 123 by genistein and related compounds, Br. J. Cancer 74 (1996) 1949–1954. [293] P. Borst, R. Evers, M. Kool, J. Wijnholds, The multidrug resistance protein family, Biochim. Biophys. Acta 1461 (1999) 347–357. [294] S.F. Zhou, L.L. Wang, Y.M. Di, C.C. Xue, W. Duan, C.G. Li, Y. Li, Substrates and inhibitors of human multidrug resistance associated proteins and the implications in drug development, Curr. Med. Chem. 15 (2008) 1981–2039. [295] C.P. Wu, A.M. Calcagno, S.B. Hladky, S.V. Ambudkar, M.A. Barrand, Modulatory effects of plant phenols on human multidrug-resistance proteins 1, 4 and 5 (ABCC1, 4 and 5), FEBS J. 272 (2005) 4725–4740.

E

T

differential susceptibility to toxic liver injury, Pharmacogenet. Genomics 17 (2007) 403–415. [240] S.R. Miranda, J.K. Lee, K.L. Brouwer, Z. Wen, P.C. Smith, R.L. Hawke, Hepatic metabolism and biliary excretion of silymarin flavonolignans in isolated perfused rat livers: role of multidrug resistance-associated protein 2 (Abcc2), Drug Metab. Dispos. 36 (2008) 2219–2226. [241] A. Kumar, D.R. Soprano, H.K. Parekh, Cross-resistance to the synthetic retinoid CD437 in a paclitaxel-resistant human ovarian carcinoma cell line is independent of the overexpression of retinoic acid receptor-gamma, Cancer Res. 61 (2001) 7552–7555. [242] W. Chearwae, S. Shukla, P. Limtrakul, S.V. Ambudkar, Modulation of the function of the multidrug resistance-linked ATP-binding cassette transporter ABCG2 by the cancer chemopreventive agent curcumin, Mol. Cancer Ther. 5 (2006) 1995–2006. [243] M.-H. Pan, T.-M. Huang, J.-K. Lin, Biotransformation of curcumin through reduction and glucuronidation in mice, Drug Metab. Dispos. 27 (1999) 486–494. [244] J.E. Ros, T.A. Roskams, M. Geuken, R. Havinga, P.L. Splinter, B.E. Petersen, N.F. LaRusso, D.M. van der Kolk, F. Kuipers, K.N. Faber, M. Muller, P.L. Jansen, ATP binding cassette transporter gene expression in rat liver progenitor cells, Gut 52 (2003) 1060–1067. [245] R.J. Scialis, I.L. Csanaky, M.J. Goedken, J.E. Manautou, Multidrug resistanceassociated protein 3 plays an important role in protection against acute toxicity of diclofenac, Drug Metab. Dispos. 43 (2015) 944–950. [246] W.M. Pardridge, The Blood–Brain Barrier: Cellular and Molecular Biology, Raven Press, New York, 1993. [247] T. Terasaki, K. Hosoya, The blood–brain barrier efflux transporters as a detoxifying system for the brain, Adv. Drug Deliv. Rev. 36 (1999) 195–209. [248] I. Tamai, A. Tsuji, Transporter-mediated permeation of drugs across the blood– brain barrier, J. Pharm. Sci. 89 (2000) 1371–1388. [249] S. Ohtsuki, T. Terasaki, Contribution of carrier-mediated transport systems to the blood–brain barrier as a supporting and protecting interface for the brain; importance for CNS drug discovery and development, Pharm. Res. 24 (2007) 1745–1758. [250] D.S. Miller, Regulation of ABC transporters at the blood–brain barrier, Clin. Pharmacol. Ther. 97 (2015) 395–403. [251] E.M. Cornford, S. Hyman, Blood–brain barrier permeability to small and large molecules, Adv. Drug Deliv. Rev. 36 (1999) 145–163. [252] B. Gao, B. Hagenbuch, G.A. Kullak-Ublick, D. Benke, A. Aguzzi, P.J. Meier, Organic anion-transporting polypeptides mediate transport of opioid peptides across blood–brain barrier, J. Pharmacol. Exp. Ther. 294 (2000) 73–79. [253] W. Lee, H. Glaeser, L.H. Smith, R.L. Roberts, G.W. Moeckel, G. Gervasini, B.F. Leake, R.B. Kim, Polymorphisms in human organic anion-transporting polypeptide 1A2 (OATP1A2): implications for altered drug disposition and central nervous system drug entry, J. Biol. Chem. 280 (2005) 9610–9617. [254] H. Bronger, J. Konig, K. Kopplow, H.H. Steiner, R. Ahmadi, C. Herold-Mende, D. Keppler, A.T. Nies, ABCC drug efflux pumps and organic anion uptake transporters in human gliomas and the blood-tumor barrier, Cancer Res. 65 (2005) 11419–11428. [255] H. Liu, N. Yu, S. Lu, S. Ito, X. Zhang, B. Prasad, E. He, X. Lu, Y. Li, F. Wang, H. Xu, G. An, J.D. Unadkat, H. Kusuhara, Y. Sugiyama, J. Sahi, Solute carrier family of the organic anion-transporting polypeptides 1a2 — Madin–Darby canine kidney ii: a promising in vitro system to understand the role of organic anion-transporting polypeptide 1A2 in blood–brain barrier drug penetration, Drug Metab. Dispos. 43 (2015) 1008–1018. [256] S. Dauchy, F. Miller, P.O. Couraud, R.J. Weaver, B. Weksler, I.A. Romero, J.M. Scherrmann, I. De Waziers, X. Decleves, Expression and transcriptional regulation of ABC transporters and cytochromes P450 in hCMEC/D3 human cerebral microvascular endothelial cells, Biochem. Pharmacol. 77 (2009) 897–909. [257] Y. Uchida, S. Ohtsuki, Y. Katsukura, C. Ikeda, T. Suzuki, J. Kamiie, T. Terasaki, Quantitative targeted absolute proteomics of human blood–brain barrier transporters and receptors, J. Neurochem. 117 (2011) 333–345. [258] H. Kubota, H. Ishihara, T. Langmann, G. Schmitz, B. Stieger, H.G. Wieser, Y. Yonekawa, K. Frei, Distribution and functional activity of P-glycoprotein and multidrug resistance-associated proteins in human brain microvascular endothelial cells in hippocampal sclerosis, Epilepsy Res. 68 (2006) 213–228. [259] A.H. Schinkel, J.J. Smit, O. van Tellingen, J.H. Beijnen, E. Wagenaar, L. van Deemter, C.A. Mol, M.A. van der Valk, E.C. Robanus-Maandag, H.P.J. te Riele, A.J.M. Berns, P. Borst, Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood–brain barrier and to increased sensitivity to drugs, Cell 77 (1994) 491–502. [260] A.H. Schinkel, E. Wagenaar, C.A. Mol, L. van Deemter, P-glycoprotein in the blood– brain barrier of mice influences the brain penetration and pharmacological activity of many drugs, J. Clin. Invest. 97 (1996) 2517–2524. [261] K. Linnet, T.B. Ejsing, A review on the impact of P-glycoprotein on the penetration of drugs into the brain. Focus on psychotropic drugs, Eur. Neuropsychopharmacol. 18 (2008) 157–169. [262] X.Y. Yu, S.G. Lin, X. Chen, Z.W. Zhou, J. Liang, W. Duan, B. Chowbay, J.Y. Wen, E. Chan, J. Cao, C.G. Li, S.F. Zhou, Transport of cryptotanshinone, a major active triterpenoid in Salvia miltiorrhiza Bunge widely used in the treatment of stroke and Alzheimer's disease, across the blood–brain barrier, Curr. Drug Metab. 8 (2007) 365–378. [263] X.Y. Yu, S.G. Lin, Z.W. Zhou, X. Chen, J. Liang, X.Q. Yu, B. Chowbay, J.Y. Wen, W. Duan, E. Chan, X.T. Li, J. Cao, C.G. Li, C.C. Xue, S.F. Zhou, Role of P-glycoprotein in limiting the brain penetration of glabridin, an active isoflavan from the root of Glycyrrhiza glabra, Pharm. Res. 24 (2007) 1668–1690. [264] X. Wang, C.R. Campos, J.C. Peart, L.K. Smith, J.L. Boni, R.E. Cannon, D.S. Miller, Nrf2 upregulates ATP binding cassette transporter expression and activity at the blood– brain and blood–spinal cord barriers, J. Neurosci. Off. J. Soc. Neurosci. 34 (2014) 8585–8593. [265] B. Bauer, X. Yang, A.M.S. Hartz, E.R. Olson, R. Zhao, J.C. Kalvass, G.M. Pollack, D.S. Miller, In vivo activation of human pregnane x receptor tightens the blood–brain barrier to methadone through P-glycoprotein up-regulation, Mol. Pharmacol. 70 (2006) 1212–1219.

U

1776 1777 1778 1779 1780 1781 1782 1783 1784 1785 1786 1787 1788 1789 1790 1791 1792 1793 1794 1795 1796 1797 1798 1799 1800 1801 1802 1803 1804 1805 1806 1807 1808 1809 1810 1811 1812 1813 1814 1815 1816 1817 1818 1819 1820 1821 1822 1823 1824 1825 1826 1827 1828 1829 1830 1831 1832 1833 1834 1835 1836 1837 1838 1839 1840 1841 1842 1843 1844 1845 1846 1847 1848 1849 1850 1851 1852 1853 1854 1855 1856 1857 1858 1859 1860 1861 1862 1863 1864 1865 1866

17

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

1867 1868 1869 1870 1871 1872 1873 1874 1875 1876 1877 1878 1879 1880 1881 1882 1883 1884 1885 1886 1887 1888 1889 1890 1891 1892 1893 1894 1895 1896 1897 1898 1899 1900 1901 1902 1903 1904 1905 1906 1907 1908 1909 1910 1911 1912 1913 1914 1915 1916 1917 1918 1919 1920 1921 1922 1923 1924 1925 1926 1927 1928 1929 1930 1931 1932 1933 1934 1935 1936 1937 1938 1939 1940 1941 1942 1943 1944 1945 1946 1947 1948 1949 1950 1951 1952 1953 1954 1955 1956 1957

P

R O

O

F

[308] S. Ohtsuki, T. Kikkawa, S. Mori, S. Hori, H. Takanaga, M. Otagiri, T. Terasaki, Mouse reduced in osteosclerosis transporter functions as an organic anion transporter 3 and is localized at abluminal membrane of blood–brain barrier, J. Pharmacol. Exp. Ther. 309 (2004) 1273–1281. [309] D.H. Sweet, D.S. Miller, J.B. Pritchard, Y. Fujiwara, D.R. Beier, S.K. Nigam, Impaired organic anion transport in kidney and choroid plexus of organic anion transporter 3 (Oat3 (Slc22a8)) knockout mice, J. Biol. Chem. 277 (2002) 26934–26943. [310] S. Zhang, M.E. Morris, Effects of the flavonoids biochanin A, morin, phloretin, and silymarin on P-glycoprotein-mediated transport, J. Pharmacol. Exp. Ther. 304 (2003) 1258–1267. [311] S.Z. Zhang, X.N. Yang, M.E. Morris, Flavonoids are inhibitors of breast cancer resistance protein (ABCG2)-mediated transport, Mol. Pharmacol. 65 (2004) 1208–1216. [312] X. Sun, J. Li, C. Guo, H. Xing, J. Xu, Y. Wen, Z. Qiu, Q. Zhang, Y. Zheng, X. Chen, D. Zhao, Pharmacokinetic effects of curcumin on docetaxel mediated by OATP1B1, OATP1B3 and CYP450s, Drug Metab. Pharmacokinet. [313] J. Hong, J.D. Lambert, S.H. Lee, P.J. Sinko, C.S. Yang, Involvement of multidrug resistance-associated proteins in regulating cellular levels of (−)-epigallocatechin-3-gallate and its methyl metabolites, Biochem. Biophys. Res. Commun. 310 (2003) 222–227. [314] Y. Shirasaka, E. Kuraoka, H. Spahn-Langguth, T. Nakanishi, P. Langguth, I. Tamai, Species difference in the effect of grapefruit juice on intestinal absorption of talinolol between human and rat, J. Pharmacol. Exp. Ther. 332 (2010) 181–189. [315] T. Walle, U.K. Walle, The beta-D-glucoside and sodium-dependent glucose transporter 1 (SGLT1)-inhibitor phloridzin is transported by both SGLT1 and multidrug resistance-associated proteins 1/2, Drug Metab. Dispos. 31 (2003) 1288–1291. [316] T. Nabekura, S. Kamiyama, S. Kitagawa, Effects of dietary chemopreventive phytochemicals on P-glycoprotein function, Biochem. Biophys. Res. Commun. 327 (2005) 866–870. [317] J. Riha, S. Brenner, M. Böhmdorfer, B. Giessrigl, M. Pignitter, K. Schueller, T. Thalhammer, B. Stieger, V. Somoza, T. Szekeres, W. Jäger, Resveratrol and its major sulfated conjugates are substrates of organic anion transporting polypeptides (OATPs): Impact on growth of ZR-75-1 breast cancer cells, Mol. Nutr. Food Res. 58 (2014) 1830–1842. [318] H.C. Cooray, T. Janvilisri, H.W. van Veen, S.B. Hladky, M.A. Barrand, Interaction of the breast cancer resistance protein with plant polyphenols, Biochem. Biophys. Res. Commun. 317 (2004) 269–275.

N

C

O

R

R

E

C

T

E

[296] Y. Li, J.L. Revalde, G. Reid, J.W. Paxton, Modulatory effects of curcumin on multidrug resistance-associated protein 5 in pancreatic cancer cells, Cancer Chemother. Pharmacol. 68 (2011) 603–610. [297] M.L. Holland, D.T. Lau, J.D. Allen, J.C. Arnold, The multidrug transporter ABCG2 (BCRP) is inhibited by plant-derived cannabinoids, Br. J. Pharmacol. 152 (2007) 815–824. [298] D.G. Bailey, G.K. Dresser, B.F. Leake, R.B. Kim, Naringin is a major and selective clinical inhibitor of organic anion-transporting polypeptide 1A2 (OATP1A2) in grapefruit juice, Clin. Pharmacol. Ther. 81 (2007) 495–502. [299] T. Abe, M. Kakyo, T. Tokui, R. Nakagomi, T. Nishio, D. Nakai, H. Nomura, M. Unno, M. Suzuki, T. Naitoh, S. Matsuno, H. Yawo, Identification of a novel gene family encoding human liver-specific organic anion transporter LST-1, J. Biol. Chem. 274 (1999) 17159–17163. [300] K. Köck, Y. Xie, R.L. Hawke, N.H. Oberlies, K.L.R. Brouwer, Interaction of silymarin flavonolignans with organic anion-transporting polypeptides, Drug Metab. Dispos. 41 (2013) 958–965. [301] D. Kobayashi, T. Nozawa, K. Imai, J. Nezu, A. Tsuji, I. Tamai, Involvement of human organic anion transporting polypeptide OATP-B (SLC21A9) in pH-dependent transport across intestinal apical membrane, J. Pharmacol. Exp. Ther. 306 (2003) 703–708. [302] M. Grube, K. Kock, S. Oswald, K. Draber, K. Meissner, L. Eckel, M. Bohm, S.B. Felix, S. Vogelgesang, G. Jedlitschky, W. Siegmund, R. Warzok, H.K. Kroemer, Organic anion transporting polypeptide 2B1 is a high-affinity transporter for atorvastatin and is expressed in the human heart, Clin. Pharmacol. Ther. 80 (2006) 607–620. [303] M.V. St-Pierre, B. Hagenbuch, B. Ugele, P.J. Meier, T. Stallmach, Characterization of an organic anion-transporting polypeptide (OATP-B) in human placenta, J. Clin. Endocrinol. Metab. 87 (2002) 1856–1863. [304] H. Satoh, F. Yamashita, M. Tsujimoto, H. Murakami, N. Koyabu, H. Ohtani, Y. Sawada, Citrus juices inhibit the function of human organic anion-transporting polypeptide OATP-B, Drug Metab. Dispos. 33 (2005) 518–523. [305] A.C. Whitley, D.H. Sweet, T. Walle, The dietary polyphenol ellagic acid is a potent inhibitor of hOAT1, Drug Metab. Dispos. 33 (2005) 1097–1100. [306] S.S. Hong, K. Seo, S.C. Lim, H.K. Han, Interaction characteristics of flavonoids with human organic anion transporter 1 (hOAT1) and 3 (hOAT3), Pharmacol. Res. 56 (2007) 468–473. [307] S.H. Cha, T. Sekine, J.I. Fukushima, Y. Kanai, Y. Kobayashi, T. Goya, H. Endou, Identification and characterization of human organic anion transporter 3 expressing predominantly in the kidney, Mol. Pharmacol. 59 (2001) 1277–1286.

U

1958 1959 1960 1961 1962 1963 1964 1965 1966 1967 1968 1969 1970 1971 1972 1973 1974 1975 1976 1977 1978 1979 1980 1981 1982 1983 1984 1985 1986 1987 1988 1989 1990 1991 1992 1993 1994 1995

Y. Li et al. / Advanced Drug Delivery Reviews xxx (2016) xxx–xxx

D

18

Please cite this article as: Y. Li, et al., The effects of dietary and herbal phytochemicals on drug transporters, Adv. Drug Deliv. Rev. (2016), http:// dx.doi.org/10.1016/j.addr.2016.09.004

1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008 2009 Q2 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 2027 2028 2029 2030 2031 2032 2033