Abstract
Background:
In clinical practice chloroquine and hydroxychloroquine are often co-administered with other drugs in the treatment of malaria, chronic inflammatory diseases, and COVID-19. Therefore, their metabolic properties and the effects on activity of cytochrome P450 (P450, CYP) enzymes and drug transporters should be considered into when developing the most efficient treatments for patients.
Methods:
Scientific literature on the interactions of chloroquine and hydroxychloroquine, with human P450 enzymes and drug transporters was searched using PUBMED.Gov (https://pubmed.ncbi.nlm.nih.gov/) and the ADME database (https://life-science.kyushu.fujitsu.com/admedb/).
Results:
Chloroquine and hydroxychloroquine are metabolized by P450 1A2, 2C8, 2C19, 2D6, and 3A4/5 in vitro and by P450s 2C8 and 3A4/5 in vivo by N-deethylation. Chloroquine effectively inhibited P450 2D6 in vitro; however, in vivo inhibition was not apparent except in individuals with limited P450 2D6 activity. Chloroquine is both an inhibitor and inducer of the transporter MRP1 and is also a substrate of the Mate and MRP1 transport systems. Hydroxychloroquine also inhibited P450 2D6 and the transporter OATP1A2.
Conclusions:
Chloroquine caused a statistically significant decrease of P450 2D6 activity in vitro and in vivo, inhibiting also its own metabolism by the enzyme. The inhibition indicates a potential for clinical drug-drug interactions when taken with other drugs that are predominant substrates of the P450 2D6. When chloroquine and hydroxychloroquine are used clinically with other drugs, substrates of P450 2D6 enzyme, attention should be given to substrate specific metabolism by P450 2D6 alleles present in individuals taking the drugs.
Keywords: Chloroquine, hydroxychloroquine, P450s, transporters, metabolism, inhibition, induction
1. METABOLIC PROPERTIES OF CHLOROQUINE AND HYDROXYCHLOROQUINE
Chloroquine and its derivative hydroxychloroquine have been used for many years in the treatment and prevention of malaria and chronic inflammatory diseases (rheumatoid arthritis and systemic lupus erythrematosus) [1–6]. These drugs are also in vitro inhibitors of various viruses, including human immunodeficiency virus (HIV-1) [7, 8], and have strong antiviral effects on SARS-CoV infection of cultured primate cells [9]. Chloroquine and hydroxychloroquine have recently been proposed to be used for prevention and treatment of COVID-19 alone or in combination with other drugs (e.g., azithromycin, lopinavir) [10–15]. While some clinical trials have suggested some beneficial effects of chloroquine and hydroxychloroquine in COVID-19 patients, most of the reported data are still preliminary and need further elaboration. Clinical experience has shown that chloroquine has a narrow safety margin, as three times the adult therapeutic dosage for malaria can be lethal when given as a single dose, particularly in children, who are particularly sensitive to chloroquine toxicity, with one to two tablets being potentially fatal [16].
When given orally to treat rheumatic diseases, chloroquine and hydroxychloroquine are well absorbed (70–80% bioavailability), and between 21 and 47% of the dose is excreted unchanged. Chloroquine is well absorbed and distributed extensively, resulting in a large volume of distribution with apparent and terminal half-lives of 1.6 days and 2 weeks, respectively. There is great variability of plasma concentrations, with an 11-fold range of drug concentrations found following similar doses in treated patients, and the variation may depend on the presence of food, antacids, and iron salts. Protein binding ranges between 30 and 40%, with binding to both albumin and P-glycoprotein. Chloroquine phosphate binds to human serum albumin (HSA), and the number of binding sites and affinity constant were estimated to be 33 and 7.7 × 103 M−1, respectively. [17–19]. Chloroquine binding to plasma favors the (S)-(+)-chloroquine enantiomer (67% vs 35% for the (R)-(−)-enantiomer). Hence, unbound plasma concentrations are higher for (R-)-(−)-chloroquine. Following separate administration of the individual enantiomers, (R)-(−)-chloroquine produced higher and more sustained blood concentrations. The shorter half-life of (S)-(+)-chloroquine is related to its faster clearance. Blood concentrations of the (S)-(+)-forms of desethylchloroquine always exceeded those of the (R-)-(−)-forms, pointing to a preferential metabolism of (S)-(+)-chloroquine. However, both chloroquine and hydroxychloroquine are clinically administered as racemates [20, 21].
Following administration of chloroquine to patients and volunteers, its N-dealkylated metabolites (monodesethylchloroquine, didesethylchloroquine, 7-chloro-4-aminoquinoline), and in addition the chloroquine side chain N-oxide, and chloroquine di-N-oxide were identified by gas chromatography, thin-layer chromatography, and gas chromatography/mass spectrometry [22, 23]. The secondary amine metabolites may be of particular interest as they could be substrates in flavin monooxygenase (FMO)-catalyzed reactions, with formation of hydroxylamine metabolites which may contribute to inhibition of the target P-450 enzymes. However, no FMO reactions have bene reported to date.
Besides a number of potential adverse effects that might be caused by the use of chloroquine and hydroxychloroquine, alone or in combinations with other drugs [24–26], there are also other factors that might contribute to the clinically significant drug-drug interactions. For instance, it has been reported that inflammatory diseases such as Crohn’s disease increase mRNA expression of P450s 3A4 and 3A5 and the drug transporter P-glycoprotein (P-gp). Conversely, acute inflammation (an illness treated with chloroquine and hydroxychloroquine) and morbid obesity can lead to a decrease in P450 3A4 activity. In addition, morbid obesity my lead also to increased P450 2E1 activity and risk of toxic reactions [27–29]. These examples illustrate that changes of activity and/or expression of drug metabolism enzymes and/or transporters might result in clinically significant pharmacokinetic drug-drug interactions and/or adverse effects of drugs.
The present paper summarizes data published on the metabolism of chloroquine and hydroxychloroquine by human cytochrome P450 enzymes, their interactions with drug transporters, and the potential to cause drug-drug interactions in clinical practice.
2. CHLOROQUINE AND HYDROXYCHLOROQUINE METABOLISM BY P450 ENZYMES
In addition to factors that might influence the fate of chloroquine and hydroxychloroquine in the body, of particular interest is the interactions of these drugs with cytochrome P450 enzymes. It has been reported that P450 2C19 is, in addition to P450 3A4, the major enzyme that participates in chloroquine metabolism in vivo. In vitro, however, P450 2D6 was reported to catalyze chloroquine metabolism with only a minor contribution in vivo (N-dealkylation reaction). In addition, both chloroquine and hydroxychloroquine are inhibitors of P450 2D6 both in vivo and in vitro. The potency of inhibition has ranged from weak to medium (Tables 1 and 2 and references therein). Inhibition of P450 2D6 by chloroquine might be associated with minor participation of P450 2D6 in metabolism of chloroquine in vivo.
Table 1.
Metabolism of Chloroquine and Hydroxychloroquine with Cytochrome P450 Enzymes.
| Enzyme | Drug/metabolite | N-Deethylation | References | ||
|---|---|---|---|---|---|
| In vivo | In vitro | ||||
| Human liver microsomes | Chloroquine | In vitro: Km 444 ± 121 μM, Vmax 617 ± 128 pmol/min/mg protein, reaction diminished when coincubated with quercetin (20–40% inhibition), ketoconazole, or troleandomycin (20–30% inhibition) and strongly inhibited (80% inhibition) by a combination of ketoconazole and quercetin [32]; Km 0.21 mM for low and Km 3.43 mM for high Km components, Vmax1.02 nmol/min/mg protein and 10.5 nmol/min/mg protein for low and high Vmax components, respectively [36] | [32,35,36] | ||
| P450 1A1 | Chloroquine | Recombinant enzymes: microsomes, formation of desethylchloroquine | [32] | ||
| P450 1A2 | Chloroquine | Recombinant enzymes: medium Km, high activity | [36] | ||
| P450 2C19 | Chloroquine | Recombinant enzymes | [36] | ||
| P450 2C8 | Chloroquine | Human liver microsomes and recombinant enzymes: 2C8 major enzyme in vivo suggested in addition to P450 3A4 | [32,35, 36] | ||
| P450 2C8 | Hydroxychloroquine | Patients taking drug for >3 months, genetic polymorphism discussed | [34] | ||
| P450 2D6 | Chloroquine | Human liver microsomes and recombinant enzymes: low Km, low Vmax; minor participation in vivo expected [32] | [32,35, 36] | ||
| P450 2D6 | Hydroxychloroquine | patients taking drug for >3 months, [desethylhydrochloroquine]: [hydroxychloroquine] ratio was related to CYP2D6 polymorphisms in Korean lupus patients | [34] | ||
| P450 3A4 | Chloroquine | Human liver microsomes and recombinant enzymes: high Km, high Vmax system, significant participation and as major enzyme in human liver microsomes in addition to P450 2C8 proposed [32,36] | [32,35, 36] | ||
| P450 3A4 | Hydroxychloroquine | Patients taking drug for >3 months, effect of genetic polymorphism could not be determined | [34] | ||
| P450 3A5 | Hydroxychloroquine | Patients taking drug for >3 months, effect of genetic polymorphism could not be determined | [34] | ||
Table 2.
Inhibition of Cytochrome P450 Catalyzed Metabolic Reactions by Chloroquine and Hydroxychloroquine.
| Enzyme | Drug/metabolite | Method | References | |
|---|---|---|---|---|
| In vivo | In vitro | |||
| P450 1A2 | Chloroquine | No effect on caffeine metabolism [33] | Fluorescence-based high throughput screening assays with recombinant P450, no inhibition | [33,42] |
| P450 1A2 | Chloroquine (+)-(S)- | Fluorescence-based high throughput screening assays with recombinant P450: no inhibition | [42] | |
| P450 1A2 | Chloroquine (−)-(R)- | Fluorescence-based high throughput screening assays with recombinant P450: no inhibition | [42] | |
| P450 2C19 | Chloroquine | No effect on mephenytoin metabolism | [33,45] | |
| P450 2C19 | Chloroquine (+)-(S)- | Fluorescence-based high throughput screening assays with recombinant P450: no inhibition | [46] | |
| P450 2C19 | Chloroquine (−)-(R)- | Fluorescence-based high throughput screening assays with recombinant P450: no inhibition | [42] | |
| P450 2C8 | Chloroquine | Recombinant enzymes: no inhibition or very weak inhibition | [35,36,42] | |
| P450 2C9 | Chloroquine | Human liver microsomes and recombinant enzymes: no inhibition or very weak inhibition | [38] | |
| P450 2C9 | Chloroquine (−)-(R)- | Fluorescence-based high throughput screening assays with recombinant P450: no inhibition | [42] | |
| P450 2C9 | Chloroquine (+)-(S)- | Fluorescence-based high throughput screening assays with recombinant P450: no inhibition | [42] | |
| P450 2D6 | Chloroquine | In black Zimbabweans and 12 white Swedish healthy volunteers no P450 2D6 inhibition was observed no statistically significant difference on the urinary debrisoquine to 4-hydroxydebrisoquine metabolic ratio [45]; chloroquine inhibited P450 2D6 debrisoquine metabolism in healthy Zambians, medium and concentration dependent inhibition observed [47]; chloroquine inhibited metabolism of debrisoquine by 2D6 in humans, the effect was modest and progressive with repeated doses, clinically significant pharmacokinetic drug-drug interaction proposed [33] |
Fluorescence-based high throughput screening assays with recombinant P450 Ki 12 μM [42] inhibition of bufuralol hydroxylation Ki 13 μM, human liver microsomes competitive inhibition [42]; moderate inhibition of primaquine metabolism [31]; weak inhibition IC50 127 μM for bufuralol 1´-hydroxylation in extensive metabolizer liver [44]; Ki 0.18 μM for inhibition of metoprolol hydroxylation in rat liver microsomes [40]; Ki 15 μM in human liver microsomes [43]; IC50 17 μM with Echerichia coli recombinant enzyme, IC50 39 μM with β-lymphoblastoid microsomes, IC50 21 μM with human liver microsomes [30] | [30,31,33,40,42,43,44,45,47,48] |
| P450 2D6 | Chloroquine (−)-R- | Fluorescence-based high throughput screening assays with recombinant P450s, Ki 20 μM | [45] | |
| P450 2D6 | Chloroquine (+)-S- | Fluorescence-based high throughput screening assays with recombinant P450s, Ki 6.2 μM | [45] | |
| P450 2D6 | Monodesethylchloroquine | Human liver microsomes and recombinant enzymes, medium inhibition | [30,42] | |
| P450 2D6 | Hydroxychloroquine | Increased bioavailability of metoprolol observed, one individual, who was heterozygous for a mutant CYP2D6 allele, was converted to a poor metabolizer by hydroxychloroquine administration | [46] | |
| P450 2E1 | Chloroquine | No effect on chlorzoxazone metabolism | [33] | |
| P450 3A4 | Chloroquine | No effect on dapsone metabolism [33] | Human liver microsomes, inhibition of quinine 3-hydroxylation IC50 >200 μM [48], inhibition of midazolam hydroxylations [49] | [33,48,49] |
Major in vitro approaches to predicting roles of individual P450s in the metabolism of chloroquine and hydroxychloroquine include (i) comparison of rates measured with recombinant P450s (normalized to P450 content in liver), (ii) correlation of the catalytic activity of interest with marker activities of individual P450s (or immunochemically determined levels) in multiple human tissue samples, and (iii) analysis of the extent of inhibition of the catalytic activity in liver (or other tissue) microsomes with or without selective chemical inhibitors and/or P450-specific antibodies. In vivo methods include non-invasive pharmacokinetic measurements and their relationship with (i) genetics, (ii) treatment with an established P450 inducer, or (iii) treatment with a selective P450 inducer or inhibitor [30, 31].
The major metabolites of chloroquine and hydroxychloroquine in humans are formed by the same metabolic reactions both in vitro and in vivo, i.e. N-delakylation(s) (Fig. 1, Table 1). Following administration chloroquine is rapidly N-dealkylated to the pharmacologically active desethylchloroquine and bis-desethylchloroquine by P450 enzymes. Desethylchloroquine and bis-desethylchloroquine concentrations reach 40% and 10% of chloroquine concentrations, respectively, and both chloroquine and desethylchloroquine concentrations decline slowly, with elimination half-lives of 20 to 60 days. Parent drug and metabolite can be detected in urine months after a single dose. It was also reported that ratio of desethylhydroxychloroquine:hydroxychloroquine is in patents with systemic lupus erythematosus related to P450 2D6 polymorphism [21,32,33,34,35]. In vitro metabolism of chloroquine and hydroxychloroquine was investigated by using both human liver microsomes and recombinant human enzymes (Table 1). Using microsomes from a panel of 16 human livers phenotyped for 10 different P450 enzymes, desethylchloroquine formation from chloroquine was highly correlated with testosterone 6β-hydroxylation (r = 0.80; p < 0.001), a P450 3A4-mediated reaction, and P450 2C8-mediated paclitaxel 6β-hydroxylation (r = 0.82; p < 0.001). In human liver microsomes the deethylation pathway was inhibited by quercetin (20–40% inhibition), ketoconazole, and troleandomycin (20–30% inhibition); additional inhibition was seen by using a combination of ketoconazole and quercetin, which strongly inhibited (80%) the reaction [32,35]. It was also shown that desethylchloroquine formation was significantly correlated with P450 3A4/5-catalyzed midazolam 1´-hydroxylation (r2= 0.76) and P450 2C8-catalyzed paclitaxel 6β-hydroxylation (r2= 0.81). Experiments using human liver microsomes confirmed that the major metabolite of chloroquine is desethylchloroquine. Eadie-Hofstee plots were biphasic and suggested the potential involvement of multiple enzymes, with apparent Km and Vmax values of 0.21 mM and 1.02 nmol/min/mg protein, and 3.4 mM and 10.5 nmol/min/mg for low and high Km components, respectively. Of the cDNA-expressed P450s examined, P450s 1A2, 2C8, 2C19, 2D6, and 3A4/5 had significant activity for desethylchloroquine formation [32,36]. The major contribution to metabolism of chloroquine in vivo is attributed to P450s 2C8 and 3A4, which have both been characterized as high Vmax/high Km systems. P450 2D6, which also catalyzes the deethylation of chloroquine, had a lower Km but also a significantly lower Vmax. Therefore, P450 2C8, P450 3A4, and, to a much lesser extent also P450 2D6, are expected to account for most of the in vivo chloroquine N-deethylation [32] and in human liver microsomes P450s 2C8 and 3A4/5 are the major enzymes responsible for the chloroquine N-deethylation (Table 1 and references therein).
Figure (1).
Structures of Chloroquine, Hydroxychloroquine, and Metabolites Formed by Cytochrome P450 Enzymes.
Hydroxychloroquine is a substrate of P450s 2C8, 2D6, and 3A enzymes in vivo (Table 1 and references therein). The metabolites of hydroxychloroquine reported are desethylhydroxychloroquine, desethylchloroquine, and bis-desethylchloroquine [34, 37]. As already mentioned, it was shown that formation of desethylhydroxychloroquine a major metabolite and pharmacologically active form of hydroxychloroquine was, in Korean lupus patients taking drug orally, related to CYP2D6 polymorphisms. It was suggested, therefore, that the role of an individual’s CYP polymorphisms should be considered when prescribing oral hydroxychloroquine. However, the polymorphisms related to CYP3A5*3 and CYP3A4*18B did not show any significant association with the ratios of blood concentrations of hydroxychloroquine to desethylhydroxychloroquine, or desethylhydroxychloroquine to hydroxychloroquine [34]. For example, the mean maximal blood concentrations of hydroxychloquine in rheumatoid arhrtritis patients after 6 weeks of aplication was ~1400 ng/ml, ~2600 ng/ml, ~5400 ng/ml, following application of 400 mg, 800 mg, or 1200 mg of the drug, respectively. For the same time period maximal blood concentrations of the metabolite desethylhydroxychloroquine were ~750 ng/ml, ~1200 ng/ml and ~1450ng/ml with large interindividual variations [5].
3. CHLOROQUINE AND HYDROXYCHLOROQUINE AS INHIBITORS OF P450 ENZYMES
Chloroquine has been tested for its inhibitory potency in P450-catalyzed reactions (Table 2). An in vitro study with the substrates 17α-ethinylestradiol and tolbutamide (test/marker substrates for P450 3A4 and 2C9 enzymes, respectively), revealed that chloroquine has essentially no effect on metabolism of these drugs [38,39]. However, in vivo studies that included metoprolol [40,41,42], bufuralol [43], and debrisoquine [33] (as test/marker compounds for P450 2D6) indicated that chloroquine and hydroxychloroquine inhibit P450 2D6-mediated reactions both in vitro and in vivo. In one study using human liver microsomes, chloroquine only weakly inhibited 1´-hydroxylation of bufuralol (IC50 127 ± 12 μM) [44].
The inhibitory effects of chloroquine on P450 2D6 and 2C19 activity have been difficult to demonstrate in vivo, and contradictory results have been obtained. An experiment using a cocktail of drugs consisting of caffeine (substrate of P450 1A2), mephenytoin (substrate of P450 2C19), debrisoquine (substrate of P450 2D6), chlorzoxazone (substrate of P450 2E1), and dapsone (substrate of P450 3A4) showed that chloroquine caused a progressive and statistically significant decrease in P450 2D6 activity over 14 days following chloroquine administration. These results showed that chloroquine, being a substrate of P450 2D6, is also an inhibitor of the enzyme. The effect was modest but indicates the potential for drug-drug interactions when taken with other drugs that are predominant substrates of 2D6 enzyme, inhibiting also its own metabolism [33]. An in vivo study in healthy black Zambians showed that an inhibitory effect was sustained for one week after therapeutic doses of the drugs and the authors suggested that the drug should not be used in combination with other drugs that prolong the QT time interval, particularly those that are metabolized by P450 2D6. In addition, the median debrisoquine/4-hydroxydebrisoquine (0–8 h urinary) ratio in healthy Zambians was increased (from 1.96 to 3.91; P <0.01; 95% confidence intervals 1.34–2.66) when debrisoquine was given 2 h after treatment with chloroquine, causing significant inhibition of P450 2D6 activity [47]. However, the effects of chloroquine on debrisoquine hydroxylation (catalyzed by P450 2D6) and S-mephenytoin hydroxylation (catalyzed by P450 2C19), as assessed in 11 black Zimbabwean and 12 white Swedish healthy volunteers, indicated that chloroquine does not inhibit either P450 2D6 or P450 2C19 in vivo and that it is unlikely to compromise the metabolism of substrates for these two enzymes [45]. It was also reported that chloroquine inhibited recombinant P450 2D6-catalyzed metabolism of primaquine in cryopreserved human hepatocytes [31], but not quinine 3-hydroxylation catalyzed by P450 3A4 [48]. However, chloroquine inhibited quinine 3-hydroxylation in rat and dog liver microsomes but not in human liver microsomes [49].
Hydroxychloroquine has been shown to increase the bioavailability of metoprolol in homozygous P450 2D6 extensive metabolizers (EMs) in that significant increases in the area under the plasma concentration-time curve (AUC 65 ± 5%) and maximal plasma concentration (72 ± 7%) of metoprolol were observed. It was concluded that hydroxychloroquine inhibits metoprolol metabolism, most probably by inhibiting P450 2D6. However, the inhibitory effect of hydroxychloroquine on dextromethorphan metabolism was not apparent except in an individual with low P450 2D6 activity [46].
These studies indicate that chloroquine is low to medium potency inhibitor of P450 2D6 in vitro (Ki ~12 μM for recombinant enzyme), but the IC50 ranged from 39 μM (for β-lympoblastoid-generated P450 2D6) to 127 μM (for liver microsomes) showing that the data are at least somewhat dependent on the experimental model used. Inhibition of P450 2D6 activity in vitro by chloroquine enantiomers was stereoselective (Ki 6.21 μM for the (S)-(+)-enantiomer vs. Ki 20 μM) for the R-(−)-enantiomer [42]. No other P450 enzymes tested were significantly affected (Table 2). In vivo P450 2D6 inhibitory activity has been reported, ranging from medium, concentration-dependent inhibition to no inhibition. In individuals showing progressive inhibition with repeated doses, clinically significant pharmacokinetic drug-drug interaction potential was reported and it was suggested that inhibition was not apparent except in individuals with limited P450 2D6 activity [39]. These results may be connected to substrate-specific metabolism by polymorphic P450 2D6 alleles present in individuals participating in the experiments, in that the activity profile of the P450 2D6 alleles can be substrate specific and manifest in pharmacokinetics consequences for individuals [50]. This information should be considered in the context of the participation of P450 enzymes, and in partcular P450 2D6 in the metabolism of marketed drugs [51,52].
4. INTERACTION OF CHLOROQUINE AND HYDROXYCHLOROQUINE WITH DRUG TRANSPORTERS
The multiple drug resistance (MDR) transporter was first discovered for its role in imparting resistance to drugs in cancer cells by pumping them out [53]. As knowledge about this system, together with P450 3A4, developed in the late 1980s, the overlap in substrate specificity was recognized and the role of the MDR1 protein (also called P-170, P-glycoprotein, P-gp, or ABCB1) as a drug export protein was realized in the late 1990s. With time the number of proteins involved in these processes has grown. The overall view today is that passive diffusion of drugs in and out of cells does occur in many cases but that transporters can have important roles in many cases and can have major effects on tissue selectivity of drugs. The transporters can be considered in the context of their own activities and also their interactions with P450s in that the regulation of many transporters involves the same systems as P450s, e.g. P-gp and some of the P450 3A and 2C subfamily enzymes are regulated by the pregnane X receptor (PXR). Another issue is that P-gp export transporter can also lower the level of a drug in a cell and accordingly slow the rate of its metabolism [27,54].
Transporters discussed in the present paper belong to the ATP binding cassette (ABC) transporter family and solute carrier family (SLC) transporters (Table 3). ABC transporters are membrane-bound proteins and are present in all prokaryotes, as well as plants, fungi, yeast, and animals. In humans the ABC transporters participate in transport of a wide variety of compounds (drugs, environmental, and physiological compounds) with the use of the energy of ATP. The most important drug transporters of the ABC protein family are P-glycoprotein (P-gp/ABCB1), bile salt export pump (BSEP/ABCB11), multidrug resistance-associated proteins (MRP1–4/ABCC1–4), breast cancer resistance protein (BCRP/ABCG2), and the organic anion transporter polypeptide family (OATPs). OATP1B1, OATP2B1, and OATP1B3 are influx transporters expressed by hepatoytes, facilitating drug access to the liver, while P-glycoprotein (P-gp) and BCRP are efflux transporters in intestinal epithelial cells, liver, kidney, and brain. ABCC1 and multidrug resistance-associated proteins 1 and 2 (MRP1 and MRP2) are efflux transporters. MRP1 has wide tissue distribution, and MRP2 is an efflux transporter in hepatocytes. MRP1 has wide substrate transport specificity, including important drugs. The main roles of the MRP1 transporter are the efflux of xenobiotics and endogenous metabolites, transport of inflammatory mediators (e.g., LTC4), and defense against oxidative stress. In addition, MRP1 plays a role in the development of drug resistance of various types of cancer and contributes to inflammatory responses [55,56].
Table 3.
Interaction of Chloroquine and Hydroxychloroquine with human transporters.
| Transporter | Compound | Substrate | Inhibition | Induction | Remarks/Model | References | |||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| BCRP1; BCRP; ABCG2; MRX; MXR; ABCP; BMDP; MXR1; ABC15; CD338; CDw338; EST157481; MGC102821 | Chloroquine | X | Chloroquine (100 μM) caused reduced transport of estrone-sulfate in membrane vesicles isolated from HEK293 cells over expressing transport protein, reduced to 69% | [60] | |||||||||
| BSEP, ABCB11 | Chloroquine | X | No significant inhibition; 100 μM induced transport of taurocholic acid in membrane vesicles isolated from HEK293 cells over expressing transport protein, increased to 117% [61] | [61,62,63] | |||||||||
| hOCT2, SLC22A2 | Chloroquine | X | Inhibition of [3H]MPP(+)-uptake in HEK293 cells stably expressing human OCT2 | [64] | |||||||||
| hSERT; SLC6A4; HTT; 5HTT; OCD1; 5HTT; 5HTTLPR | Chloroquine | X | Ki values for hSERT using HeLa cells to inhibit [3H]5-HT uptake was 122 μM [66] | [65,66] | |||||||||
| MATE1, SLC47A1 | Chloroquine | X | X | Substrate and competitive inhibitor of MATE1-mediated metformin uptake (Ki 2.8 μM) in HEK293-MATE1 cells, suggest drug-drug interaction potential | [67] | ||||||||
| MRP1, MRP, | Chloroquine | X | X | X | Chloroquine/protease inhibitor combinations exerted combined inhibitory effects on P-gp and MRP1 function [72]; inhibited the photoaffinity labeling of MRP by IAAQ, cell growth assays showed MRP-expressing multidrug-resistant cells (H69/AR and HL60/AR) to be more resistant to chloroquine than their parental cells (i.e., IC50 of 121 versus 28 μM chloroquine for H69/AR and H69MK 571, an LTD4 receptor antagonist reversed the resistance of H69/AR cells to chloroquine, direct binding to MRP suggested [68]; induction of expression after long-term exposure of CEM T cells, resulting in resistance to chloroquine and co-administered drugs [70]; molar excesses of chloroquine inhibited the photoaffinity labeling of MRP1 by IAAQ [69]; chloroquine resistance was associated with the specific over expression of MRP1 [68,69,70]; rate constant for MRP1-mediated calcein efflux from L23/R cells 12 x 10−4 s−1 at 50 μM chloroquine, control value 11 x 10−4 s−1; little effect on MRP1-associated ATPase activity, inhibition of uptake of 3.3 μM [3H]cGMP into inside-out human erythrocyte membrane vesicles IC50 86 μM [71] | [68,69,70, 71,72] | |||||||
| MRP4, MOATB, MOATB, ABCC4, EST170205 | Chloroquine | X | Little or no effect on MRP4-associated ATPase activity | [71] | |||||||||
| OATP1A2, OATP-A, OATP1, OATP, SLC21A3, SLCO1A2 | Chloroquine | X | IC50 1.0 μM for estrone-3-sulfate using transporter-over expressing Madin-Darby canine kidney (MDCKII) cells [73]; potently inhibited the uptake activity of all-trans-retinol in overexpressing human embryonic kidney (HEK293) cells and human retinal pigment epithelium cells [60] | [60.73] | |||||||||
| OATP1A2, OATP-A, OATP1, OATP, SLC21A3, SLCO1A2 | Hydroxychloroquine | X | Potent inhibitor of OATP1A2 function, 1.0 μM using MDCKII cells and the model substrate estrone-3-sulfate [72]; potently inhibited the uptake activity of all-trans-retinol in over expressing HEK293 cells and in primary human retinal pigment epithelium cells [60] | [60,72] | |||||||||
| OATP1B1, OATP-C, OATP2, LST-1, SLC21A6, SLCO1B1 | Chloroquine | X | Pre-incubation with chloroquine at clinically relevant concentration(s) significantly decreased [3H]E217G and [3H]pitavastatin accumulation in HEK293-OATP1B1 cells and [3H]pitavastatin accumulation in human SCH cells; chloroquine pretreatment (25 μM, 2 h) resulted in increased OATP1B1 total protein levels and ∼1.9-fold decrease in Vmax without affecting the Km for OATP1B1-mediated [3H]E217G transport in HEK293-OATP1B1 cells; potential to cause OATP-mediated drug-drug interactions downregulating transport activity | [74] | |||||||||
| OATP1B3; SLCO1B3; LST3; OATP8; SLC21A8; LST-3TM1 | Chloroquine | X | Treatment with chloroquine (25 μM, 5 h) increased OATP1B3 protein levels to 1.85-fold vehicle in HEK293-OATP1B3 cells and human SCH cells | [75] | |||||||||
| OATP2B1, OATP-B, SLC21A9, SLCO2B1 | Chloroquine | X | No effect or only moderate inhibitor effects observed using MDCKII cells and the model substrate estrone-3-sulfate | [73] | |||||||||
| P-glycoprotein (P-gp), ABCB1 | Chloroquine | X | Chloroquine (100 μM) reduced methyl quinidine transport to 50% in membrane vesicles isolated from HEK293 cells overexpressing ABC transport proteins [61]; chloroquine competed poorly for the binding site of photoreactive analogs of verapamil on P-glycoprotein in membranes of human tumor cells of the CEM/VBL100 line [62]; Ki,app 54 μM for taxol transport inhibition in Caco-2 cells [63]; weak inhibitor of Calcein-AM in MDR1-MDCKII cells [76]; poor inhibitor of photolabeling with vinblastine analog, partial reversal activity on drug resistance [79]; enhanced vinblastine cytotoxicity in CEM/VLB1K cells by 10- to 15-fold, inhibited [125I]vinblastine analog labeling of P-gp [80]; chloroquine/protease inhibitors combination exerts combined inhibitory effects on P-gp function [72] | [61,62,63,72,75,76,77,78,79,80] |
A compound (drug) may interact with a transport protein as a substrate, inducer (by induction of the transporter expression), and/or inhibition of protein function (Table 3). In the clinic, as a consequence, the pharmacokinetics of co-administered drugs can be critically altered to result in drug-drug interactions. The result of drug-drug interactions on the level of drug transport may lead to either an unexpected high plasma concentrations and potential toxic effects or, alternativley, subtherapeutic concentrations at the site of action lowering therapeutic effects. Drug-drug interactions at the level of transporters may reduce systemic exposure to drug by blocking influx transporters in the intestine but increase it by modulating influx and efflux transporters in the liver and efflux transporters in the small intestine [56].
In clinical practice chloroquine is often co-administerd with other drugs in the treatment of malaria [57,58], as well as for treatment of COVID-19 [59]. Therefore, the effect of chloroquine and hydroxychloroquine and co-administered drugs on ABC-mediated transport should be considered in order to provide the most effective treatments for patients.
Reported data on effects of chloroquine and hydroxychloroquine on the activities and properties of drug transporters are presented in Table 3 and references therein. The results presented were obtained either by using human cells or recombinant transporters in different in vitro assays. The drugs were tested for their activitiess as substates, inhibitors, or inducers of a particular transporter activity. The in vitro assays used have involved various types of cells overexpressing transporters, e.g. HEK293 (embryonic human kidney cell line), MDCKII (epithelial Madin-Darby canine kidney cell line), human tumor cells, Caco-2 cells (human epithelial colorectal adenocarcinoma cells), CEM/VLB1K (human leukemic cell line9), CEM T cells (human T lymphoblastic leukemia cell line), HeLa (immortal cell line derived from cervical cancer cells), and human SCH cells (human gastric choriocarcinoma cell line) (Table 3).
The investigations cover effects of chloroquine on P-gp-mediated transport of drugs that are substrates of the transporter and/or on expression of the transporter. Depending on the model and competing drug used, chloroquine was found to be a weak or poor inibitor of P-gp-mediated transport (quinidine, taxol) or of the binding of photoreactive analogs of verapamil and vinblastine (methyl) to the transporter. However, enhanced vinblastine toxicity was observed in CEM/VLB1K cells. In addition, using peripheral blood mononuclear cells, combinatory inhibition effects of chloroquine/protease inhibitors (Table 3) was observed. Chloroquine increased the blockage of the dye Rh123 and also carboxyfluorescein efflux activity exerted by protease inhibitors (Table 3). These results show that chloroquine itself is a weak inhibitor of P-gp transport; however, in combination with other drugs (i.e., inhibitors of the transport system) inhibitory effects might be significantly enhanced.
Studies with MRP-type transporters [68–71] showed that chloroquine is substrate of the MRP1 transporter because MRP1-expressing multidrug-resistant cells accumulate less drug than the parental drug-sensitive cells, and direct binding to the transporter was suggested [68]. It was also shown that long term exposure of CEM T cells to chloroquine induced expression of the transporter and resulted in resistence to chloroquine and coadministered drugs [70]. However, a molar excess of chloroquine inhibited photolabeling of MRP1 by the reagent IAAQ and chloroquine resistance was associated with specific overexpression of MRP-1 [68–70]. As with P-gp, a combination of chloroquine and protease inibitor drugs exerted combined inhibitory effects on MRP1. These data show that chloroquine can acts as a substrate, inducer, and inhibitor of MRP1. However, it was reported that chloroquine excerts little or no effect on MRP4-associated ATPase activity [71].
Potent inhibition of OATP1A2 by chloroquine and hydroxychloroquine [60,73] but only moderate inhibition (BCRP1, hSERT, OATP2B1) or no inhibition (BSEP) of other transporters has been reported [61–63]. Increased protein levels of OATP1B1 and OATP1B3 were reported after treatment of the cells with chloroquine, indicating induction of expression [74,75]. For details see Table 3.
As already discussed, in addition to induction and/or inhibition of activity of P450 enzymes and drug transporters, plymorphism might have additional effect on pharmacokinetic and consequently therapeutic properties of chloroquine and hydroxychloroquine [81,82].
The present data show the complexity of interactions of chloroquine with drug transporting systems, in that it can exert both inhibitory and inducing activity, but in some cases chloroquine is also substrate of the transporters (Mate and MRP1) (Table 3). Co-administration of chloroquine with other drug types is highly anticipated when treating patients for human immunodeficiency virus (HIV) and other viral infections (including COVID-19), and malaria co-infections, and the effect of chloroquine on ABC-mediated transport systems should be of particular attention in order to develop the most effective treatment strategies for patients receiving multiple drug regimens.
CONCLUSION
Major and pharmacologicaly active metabolites of chloroquine and hydroxychloroquine are formed by N-dealkylation reactions catalyzed by P450 enzymes both in vivo and in vitro. P450s 1A2, 2C8, 2C19, 2D6, and 3A4/5 exhibited significant formation of desethyl metabolites with chloroquine as substrate in vitro. The major contributions to in vivo metabolism of chloroquine is attributed to P450s 2C8 and 3A4, and that of hydroxychloroquine to P450 2C8, 2D6, and 3A4/5. Chloroquine and its desethyl metabolite inhibited P450 2D6 both in vivo and in vitro although not strongly. Chloroquine, in interaction with drug transporting systems, can exert both inhibitory and inducing activity (BSEP, MRP1), but in some cases chloroquine is also substrate of the transporters (Mate, MRP1).
Formation of N-desethylhydroxychloroquine from hydroxychloroquine in vivo has been related to CYP2D6 polymorphisms, but hydroxychloroquine also inhibited P450 2D6 in vivo as shown by inhibiting metoprolol bioavailability. Hydroxychloroquine was reported to inhibit also organic anion transporter OATP1A2, shown by inhibition of the uptake activity of all-trans-retinol.
The interaction of chloroquine and hydroxychloroquine with both, P450 enzymes (2D6) and drug transporter systems may by inhibiting their activity require particular attention when these drugs are clinically applied in order to develop the most effective treatment strategies for patients receiving multiple drug regimens.
ACKNOWLEDGEMENTS
Declared none.
Footnotes
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
CONSENT FOR PUBLICATION
Not applicable.
REFERENCES
- 1.Fox RI Mechanism of action of hydroxychloroquine as an antirheumatic drug. Semin. Arthritis Rheum, 1993, 23(2)(Suppl. 1), 82–91. 10.1016/S0049-0172(10)80012-5 [DOI] [PubMed] [Google Scholar]
- 2.White NJ The treatment of malaria. N. Engl. J. Med, 1996, 335(11), 800–806. 10.1056/NEJM199609123351107 [DOI] [PubMed] [Google Scholar]
- 3.Shippey EA; Wagler VD; Collamer AN Hydroxychloroquine: An old drug with new relevance. Cleve. Clin. J. Med, 2018, 85(6), 459–467. 10.3949/ccjm.85a.17034 [DOI] [PubMed] [Google Scholar]
- 4.Plantone D; Koudriavtseva T Current and future use of chloroquine and hydroxychloroquine in infectious, immune, neoplastic, and neurological diseases: a mini-review. Clin. Drug Investig, 2018, 38(8), 653–671. 10.1007/s40261-018-0656-y [DOI] [PubMed] [Google Scholar]
- 5.Munster T; Gibbs JP; Shen D; Baethge BA; Botstein GR; Caldwell J; Dietz F; Ettlinger R; Golden HE; Lindsley H; McLaughlin GE; Moreland LW; Roberts WN; Rooney TW; Rothschild B; Sack M; Sebba AI; Weisman M; Welch KE; Yocum D; Furst DE Hydroxychloroquine concentration-response relationships in patients with rheumatoid arthritis. Arthritis Rheum., 2002, 46(6), 1460–1469. 10.1002/art.10307 [DOI] [PubMed] [Google Scholar]
- 6.Tanaka E; Taniguchi A; Urano W; Yamanaka H; Kamatani N Pharmacogenetics of disease-modifying anti-rheumatic drugs. Best Pract. Res. Clin. Rheumatol, 2004, 18(2), 233–247. 10.1016/j.berh.2004.02.006 [DOI] [PubMed] [Google Scholar]
- 7.Romanelli F; Smith KM; Hoven AD Chloroquine and hydroxychloroquine as inhibitors of human immunodeficiency virus (HIV-1) activity Curr Pharm Des, 2004, 10(21), 2643–2648. 10.2174/1381612043383791 [DOI] [PubMed] [Google Scholar]
- 8.Wang M; Cao R; Zhang L; Yang X; Liu J; Xu M; Shi Z; Hu Z; Zhong W; Xiao G Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res, 2020, 30(3), 269–271. 10.1038/s41422-020-0282-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Vincent MJ; Bergeron E; Benjannet S; Erickson BR; Rollin PE; Ksiazek TG; Seidah NG; Nichol ST Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol. J, 2005, 2, 69. 10.1186/1743-422X-2-69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Huang C; Wang Y; Li X; Ren L; Zhao J; Hu Y; Zhang L; Fan G; Xu J; Gu X; Cheng Z; Yu T; Xia J; Wei Y; Wu W; Xie X; Yin W; Li H; Liu M; Xiao Y; Gao H; Guo L; Xie J; Wang G; Jiang R; Gao Z; Jin Q; Wang J; Cao B Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet, 2020, 395(10223), 497–506. 10.1016/S0140-6736(20)30183-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yao X; Ye F; Zhang M In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) Clin Infect Dis 2020. 10.1093/cid/ciaa237 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Barnabas RV; Brown E; Bershteyn A; Miller RS; Wener M; Celum C; Wald A; Chu H; Wesche D; Baeten JM Efficacy of hydroxychloroquine for post-exposure prophylaxis to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection among adults exposed to coronavirus disease (COVID-19): a structured summary of a study protocol for a randomised controlled trial. Trials, 2020, 21(1), 475. 10.1186/s13063-020-04446-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gautret P; Lagier JC; Parola P; Hoang VT; Meddeb L; Mailhe M; Doudier B; Courjon J; Giordanengo V; Vieira VE; Tissot Dupont H.; Honoré S; Colson P; Chabrière E; La Scola B; Rolain JM; Brouqui P; Raoult D Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents, 2020, 56(1)105949 10.1016/j.ijantimicag.2020.105949 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 14.Meo SA; Klonoff DC; Akram J Efficacy of chloroquine and hydroxychloroquine in the treatment of COVID-19. Eur. Rev. Med. Pharmacol. Sci, 2020, 24(8), 4539–4547. 10.26355/eurrev_202004_21038 [DOI] [PubMed] [Google Scholar]
- 15.Marzolini C; Stader F; Stoeckle M Effect of systemic inflammatory response to SARS-CoV-2 on lopinavir and hydroxychloroquine plasma concentrations Antimicrob Agents Chemother, 2020. AAC.01177–20 10.1128/AAC.01177-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Smit C; Peeters MYM; van den Anker JN; Knibbe CAJ Chloroquine for SARS-CoV-2: implications of its unique pharmacokinetic and safety properties. Clin. Pharmacokinet, 2020, 59(6), 659–669. 10.1007/s40262-020-00891-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Rainsford KD; Parke AL; Clifford-Rashotte M; Kean WF Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology, 2015, 23(5), 231–269. 10.1007/s10787-015-0239-y [DOI] [PubMed] [Google Scholar]
- 18.Tett SE Clinical pharmacokinetics of slow-acting antirheumatic drugs. Clin. Pharmacokinet, 1993, 25(5), 392–407. 10.2165/00003088-199325050-00005 [DOI] [PubMed] [Google Scholar]
- 19.Huang Y; Pan W; Guo M; Yao S Capillary electrophoresis with end-column electrochemiluminescence for the analysis of chloroquine phosphate and the study on its interaction with human serum albumin. J. Chromatogr. A, 2007, 1154(1–2), 373–378. 10.1016/j.chroma.2007.02.029 [DOI] [PubMed] [Google Scholar]
- 20.Müller D; Blaschke G Enantioselective assay of chloroquine and its main metabolite deethyl chloroquine in human plasma by capillary electrophoresis. J. Chromatogr. Sci, 2000, 38(10), 435–440. 10.1093/chromsci/38.10.435 [DOI] [PubMed] [Google Scholar]
- 21.Ducharme J; Farinotti R Clinical pharmacokinetics and metabolism of chloroquine. Focus on recent advancements. Clin. Pharmacokinet, 1996, 31(4), 257–274. 10.2165/00003088-199631040-00003 [DOI] [PubMed] [Google Scholar]
- 22.Köppel C; Tenczer J; Ibe K Urinary metabolism of chloroquine. Arzneimittelforschung, 1987, 37(2), 208–211. [PubMed] [Google Scholar]
- 23.Ette EI; Essien EE; Thomas WO; Brown-Awala EA Pharmacokinetics of chloroquine and some of its metabolites in healthy volunteers: a single dose study. J. Clin. Pharmacol, 1989, 29(5), 457–462. 10.1002/j.1552-4604.1989.tb03362.x [DOI] [PubMed] [Google Scholar]
- 24.Juurlink DN Safety considerations with chloroquine, hydroxychloroquine and azithromycin in the management of SARS-CoV-2 infection. CMAJ, 2020, 192(17), E450–E453. 10.1503/cmaj.200528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Seftel MD Additional safety consideration for azithromycin in the management of SARS-CoV-2 infection. CMAJ, 2020, 192(18)E483 10.1503/cmaj.75427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ferner RE; Aronson JK Chloroquine and hydroxychloroquine in covid-19. Brit Med J., 2020, 369:m1432 10.1136/bmj.m1432 [DOI] [PubMed] [Google Scholar]
- 27.Guengerich FP; Rendic S Update information on drug metabolism systems−-2009, part I. Curr. Drug Metab, 2010, 11(1), 1–3. 10.2174/138920010791110908 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Rendic S; Guengerich FP Update information on drug metabolism systems−-2009, part II: summary of information on the effects of diseases and environmental factors on human cytochrome P450 (CYP) enzymes and transporters. Curr. Drug Metab, 2010, 11(1), 4–84. 10.2174/138920010791110917 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Peter Guengerich F; Avadhani NG Roles of cytochrome P450 in metabolism of ethanol and carcinogens. Adv. Exp. Med. Biol, 2018, 1032, 15–35. 10.1007/978-3-319-98788-0_2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Moody GC; Griffin SJ; Mather AN; McGinnity DF; Riley RJ Fully automated analysis of activities catalysed by the major human liver cytochrome P450 (CYP) enzymes: assessment of human CYP inhibition potential. Xenobiotica, 1999, 29(1), 53–75. 10.1080/004982599238812 [DOI] [PubMed] [Google Scholar]
- 31.Fasinu PS; Tekwani BL; Avula B; Chaurasiya ND; Nanayakkara NP; Wang YH; Khan IA; Walker LA Pathway-specific inhibition of primaquine metabolism by chloroquine/quinine. Malar. J, 2016, 15(1), 466. 10.1186/s12936-016-1509-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Projean D; Baune B; Farinotti R; Flinois JP; Beaune P; Taburet AM; Ducharme J In vitro metabolism of chloroquine: identification of CYP2C8, CYP3A4, and CYP2D6 as the main isoforms catalyzing N-desethylchloroquine formation. Drug Metab. Dispos, 2003, 31(6), 748–754. 10.1124/dmd.31.6.748 [DOI] [PubMed] [Google Scholar]
- 33.Adedoyin A; Frye RF; Mauro K; Branch RA Chloroquine modulation of specific metabolizing enzymes activities: investigation with selective five drug cocktail. Br. J. Clin. Pharmacol, 1998, 46(3), 215–219. [published correction appears in Br J Clin Pharmacol 1998 Dec;46(6):616]. 10.1046/j.1365-2125.1998.00765.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lee JY; Vinayagamoorthy N; Han K; Kwok SK; Ju JH; Park KS; Jung SH; Park SW; Chung YJ; Park SH Association of polymorphisms of cytochrome p450 2D6 with blood hydroxychloroquine levels in patients with systemic lupus erythematosus. Arthritis Rheumatol, 2016, 68(1), 184–190. 10.1002/art.39402 [DOI] [PubMed] [Google Scholar]
- 35.Li XQ; Björkman A; Andersson TB; Gustafsson LL; Masimirembwa CM Identification of human cytochrome P(450)s that metabolise anti-parasitic drugs and predictions of in vivo drug hepatic clearance from in vitro data. Eur. J. Clin. Pharmacol, 2003, 59(5–6), 429–442. 10.1007/s00228-003-0636-9 [DOI] [PubMed] [Google Scholar]
- 36.Kim KA; Park JY; Lee JS; Lim S Cytochrome P450 2C8 and CYP3A4/5 are involved in chloroquine metabolism in human liver microsomes. Arch. Pharm. Res, 2003, 26(8), 631–637. 10.1007/BF02976712 [DOI] [PubMed] [Google Scholar]
- 37.Charlier B; Pingeon M; Dal Piaz F.; Conti V; Valentini G; Filippelli A; Izzo V Development of a novel ion-pairing HPLC-FL method for the separation and quantification of hydroxychloroquine and its metabolites in whole blood. Biomed. Chromatogr, 2018, 32(8)e4258 10.1002/bmc.4258 [DOI] [PubMed] [Google Scholar]
- 38.Riviere JH; Back DJ Inhibition of ethinyloestradiol and tolbutamide metabolism by quinoline derivatives in vitro. Chem. Biol. Interact, 1986, 59(3), 301–308. 10.1016/S0009-2797(86)80075-8 [DOI] [PubMed] [Google Scholar]
- 39.Tirkkonen T; Laine K Drug interactions with the potential to prevent prodrug activation as a common source of irrational prescribing in hospital inpatients. Clin. Pharmacol. Ther, 2004, 76(6), 639–647. 10.1016/j.clpt.2004.08.017 [DOI] [PubMed] [Google Scholar]
- 40.Lancaster DL; Adio RA; Tai KK; Simooya OO; Broadhead GD; Tucker GT; Lennard MS Inhibition of metoprolol metabolism by chloroquine and other antimalarial drugs. J. Pharm. Pharmacol, 1990, 42(4), 267–271. 10.1111/j.2042-7158.1990.tb05405.x [DOI] [PubMed] [Google Scholar]
- 41.Furst DE Pharmacokinetics of hydroxychloroquine and chloroquine during treatment of rheumatic diseases. Lupus, 1996, 5(Suppl. 1), S11–S15. 10.1177/0961203396005001041 [DOI] [PubMed] [Google Scholar]
- 42.Bapiro TE; Egnell AC; Hasler JA; Masimirembwa CM Application of higher throughput screening (HTS) inhibition assays to evaluate the interaction of antiparasitic drugs with cytochrome P450s. Drug Metab. Dispos, 2001, 29(1), 30–35. [PubMed] [Google Scholar]
- 43.Masimirembwa CM; Hasler JA; Johansson I Inhibitory effects of antiparasitic drugs on cytochrome P450 2D6. Eur. J. Clin. Pharmacol, 1995, 48(1), 35–38. 10.1007/BF00202169 [DOI] [PubMed] [Google Scholar]
- 44.Halliday RC; Jones BC; Smith DA; Kitteringham NR; Park BK An investigation of the interaction between halofantrine, CYP2D6 and CYP3A4: studies with human liver microsomes and heterologous enzyme expression systems. Br. J. Clin. Pharmacol, 1995, 40(4), 369–378. 10.1111/j.1365-2125.1995.tb04559.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Masimirembwa CM; Gustafsson LL; Dahl ML; Abdi YA; Hasler JA Lack of effect of chloroquine on the debrisoquine (CYP2D6 and S-mephenytoin (CYP2C19) hydroxylation phenotypes. Br. J. Clin. Pharmacol, 1996, 41(4), 344–346. 10.1046/j.1365-2125.1996.30713.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Somer M; Kallio J; Pesonen U; Pyykkö K; Huupponen R; Scheinin M Influence of hydroxychloroquine on the bioavailability of oral metoprolol. Br. J. Clin. Pharmacol, 2000, 49(6), 549–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Simooya OO; Sijumbil G; Lennard MS; Tucker GT Halofantrine and chloroquine inhibit CYP2D6 activity in healthy Zambians. Br. J. Clin. Pharmacol, 1998, 45(3), 315–317. 10.1046/j.1365-2125.1998.00671.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhao XJ; Ishizaki T Metabolic interactions of selected antimalarial and non-antimalarial drugs with the major pathway (3-hydroxylation) of quinine in human liver microsomes. Br. J. Clin. Pharmacol, 1997, 44(5), 505–511. 10.1046/j.1365-2125.1997.t01-1-00619.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhao XJ; Ishizaki T The In vitro hepatic metabolism of quinine in mice, rats and dogs: comparison with human liver microsomes. J. Pharmacol. Exp. Ther, 1997, 283(3), 1168–1176. [PubMed] [Google Scholar]
- 50.Ingelman-Sundberg M Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J, 2005, 5(1), 6–13. 10.1038/sj.tpj.6500285 [DOI] [PubMed] [Google Scholar]
- 51.Rendic S; Guengerich FP Survey of human oxidoreductases and cytochrome P450 enzymes involved in the metabolism of xenobiotic and natu+ral chemicals. Chem. Res. Toxicol, 2015, 28(1), 38–42. 10.1021/tx500444e [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Stringer RA; Strain-Damerell C; Nicklin P; Houston JB Evaluation of recombinant cytochrome P450 enzymes as an in vitro system for metabolic clearance predictions. Drug Metab. Dispos, 2009, 37(5), 1025–1034. 10.1124/dmd.108.024810 [DOI] [PubMed] [Google Scholar]
- 53.Bradley G; Juranka PF; Ling V Mechanism of multidrug resistance. Biochim. Biophys. Acta, 1988, 948(1), 87–128. 10.1016/0304-419x(88)90006-6 [DOI] [PubMed] [Google Scholar]
- 54.Kerb R; Fux R; Mörike K; Kremsner PG; Gil JP; Gleiter CH; Schwab M Pharmacogenetics of antimalarial drugs: effect on metabolism and transport. Lancet Infect. Dis, 2009, 9(12), 760–774. 10.1016/S1473-3099(09)70320-2 [DOI] [PubMed] [Google Scholar]
- 55.Cole SP Multidrug resistance protein 1 (MRP1, ABCC1), a “multitasking” ATP-binding cassette (ABC) transporter. J. Biol. Chem, 2014, 289(45), 30880–30888. 10.1074/jbc.R114.609248 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Nakanishi T; Tamai I Interaction of drug or food with drug transporters in intestine and liver. Curr. Drug Metab, 2015, 16(9), 753–764. 10.2174/138920021609151201113537 [DOI] [PubMed] [Google Scholar]
- 57.Obua C; Ntale M; Lundblad MS; Mahindi M; Gustafsson LL; Ogwal-Okeng JW; Anokbonggo WW; Hellgren U Pharmacokinetic interactions between chloroquine, sulfadoxine and pyrimethamine and their bioequivalence in a generic fixed-dose combination in healthy volunteers in Uganda. Afr. Health Sci, 2006, 6(2), 86–92. 10.5555/afhs.2006.6.2.86 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ogungbamigbe TO; Ojurongbe O; Ogunro PS; Okanlawon BM; Kolawole SO Chloroquine resistant Plasmodium falciparum malaria in Osogbo Nigeria: efficacy of amodiaquine + sulfadoxine-pyrimethamine and chloroquine + chlorpheniramine for treatment. Mem. Inst. Oswaldo Cruz, 2008, 103(1), 79–84. 10.1590/S0074-02762008000100012 [DOI] [PubMed] [Google Scholar]
- 59.Karalis V; Ismailos G; Karatza E Chloroquine dosage regimens in patients with COVID-19: Safety risks and optimization using simulations. Saf. Sci, 2020, 129104842 10.1016/j.ssci.2020.104842 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Xu C; Zhu L; Chan T; Lu X; Shen W; Madigan MC; Gillies MC; Zhou F Chloroquine and hydroxychloroquine are novel inhibitors of human organic anion transporting polypeptide 1A2. J. Pharm. Sci, 2016, 105(2), 884–890. 10.1002/jps.24663 [DOI] [PubMed] [Google Scholar]
- 61.Rijpma SR; van den Heuvel JJ; van der Velden M; Sauerwein RW; Russel FG; Koenderink JB Atovaquone and quinine anti-malarials inhibit ATP binding cassette transporter activity. Malar. J, 2014, 13, 359. 10.1186/1475-2875-13-359 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Riffkin CD; Chung R; Wall DM; Zalcberg JR; Cowman AF; Foley M; Tilley L Modulation of the function of human MDR1 P-glycoprotein by the antimalarial drug mefloquine. Biochem. Pharmacol, 1996, 52(10), 1545–1552. 10.1016/S0006-2952(96)00556-4 [DOI] [PubMed] [Google Scholar]
- 63.Hayeshi R; Masimirembwa C; Mukanganyama S; Ungell AL The potential inhibitory effect of antiparasitic drugs and natural products on P-glycoprotein mediated efflux. Eur. J. Pharm. Sci, 2006, 29(1), 70–81. 10.1016/j.ejps.2006.05.009 [DOI] [PubMed] [Google Scholar]
- 64.Zolk O; Solbach TF; König J; Fromm MF Structural determinants of inhibitor interaction with the human organic cation transporter OCT2 (SLC22A2). Naunyn Schmiedebergs Arch. Pharmacol, 2009, 379(4), 337–348. 10.1007/s00210-008-0369-5 [DOI] [PubMed] [Google Scholar]
- 65.Beckman ML; Pramod AB; Perley D; Henry LK Stereoselective inhibition of serotonin transporters by antimalarial compounds. Neurochem. Int, 2014, 73, 98–106. 10.1016/j.neuint.2013.10.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bridges JM; Baldini M Effect of quinidine and related compounds on uptake and release of serotonin by blood platelets. Nature, 1966, 210(5043), 1364–1365. 10.1038/2101364a0 [DOI] [PubMed] [Google Scholar]
- 67.Müller F; König J; Glaeser H; Schmidt I; Zolk O; Fromm MF; Maas R Molecular mechanism of renal tubular secretion of the antimalarial drug chloroquine. Antimicrob. Agents Chemother, 2011, 55(7), 3091–3098. 10.1128/AAC.01835-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Vezmar M; Georges E Direct binding of chloroquine to the multidrug resistance protein (MRP): possible role for MRP in chloroquine drug transport and resistance in tumor cells. Biochem. Pharmacol, 1998, 56(6), 733–742. 10.1016/S0006-2952(98)00217-2 [DOI] [PubMed] [Google Scholar]
- 69.Vezmar M; Georges E Reversal of MRP-mediated doxorubicin resistance with quinoline-based drugs. Biochem. Pharmacol, 2000, 59(10), 1245–1252. 10.1016/S0006-2952(00)00270-7 [DOI] [PubMed] [Google Scholar]
- 70.Oerlemans R; van der Heijden J; Vink J; Dijkmans BA; Kaspers GJ; Lems WF; Scheffer GL; Ifergan I; Scheper RJ; Cloos J; Assaraf YG; Jansen G Acquired resistance to chloroquine in human CEM T cells is mediated by multidrug resistance-associated protein 1 and provokes high levels of cross-resistance to glucocorticoids. Arthritis Rheum, 2006, 54(2), 557–568. 10.1002/art.21569 [DOI] [PubMed] [Google Scholar]
- 71.Wu CP; Klokouzas A; Hladky SB; Ambudkar SV; Barrand MA Interactions of mefloquine with ABC proteins, MRP1 (ABCC1) and MRP4 (ABCC4) that are present in human red cell membranes. Biochem. Pharmacol, 2005, 70(4), 500–510. 10.1016/j.bcp.2005.05.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Savarino A; Lucia MB; Rastrelli E; Rutella S; Golotta C; Morra E; Tamburrini E; Perno CF; Boelaert JR; Sperber K; Cauda R Anti-HIV effects of chloroquine: inhibition of viral particle glycosylation and synergism with protease inhibitors. J. Acquir. Immune Defic. Syndr, 2004, 35(3), 223–232. 10.1097/00126334-200403010-00002 [DOI] [PubMed] [Google Scholar]
- 73.Hubeny A; Keiser M; Oswald S; Jedlitschky G; Kroemer HK; Siegmund W; Grube M Expression of organic anion transporting polypeptide 1A2 in red blood cells and its potential impact on antimalarial therapy. Drug Metab. Dispos, 2016, 44(10), 1562–1568. [DOI] [PubMed] [Google Scholar]
- 74.Alam K; Pahwa S; Wang X; Zhang P; Ding K; Abuznait AH; Li L; Yue W Downregulation of organic anion transporting polypeptide (OATP) 1B1 transport function by lysosomotropic drug chloroquine: implication in OATP-Mediated Drug-Drug Interactions. Mol. Pharm, 2016, 13(3), 839–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Alam K; Farasyn T; Crowe A; Ding K; Yue W Treatment with proteasome inhibitor bortezomib decreases organic anion transporting polypeptide (OATP) 1B3-mediated transport in a substrate-dependent manner PLoS One, 2017, 12(11), e0186924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Tiberghien F; Loor F Ranking of P-glycoprotein substrates and inhibitors by a calcein-AM fluorometry screening assay. Anticancer Drugs, 1996, 7(5), 568–578. 10.1097/00001813-199607000-00012 [DOI] [PubMed] [Google Scholar]
- 77.Avendaño C; Menéndez JC Inhibitors of multidrug resistance to antitumor agents (MDR). Curr. Med. Chem, 2002, 9(2), 159–193. 10.2174/0929867023371175 [DOI] [PubMed] [Google Scholar]
- 78.Polli JW; Wring SA; Humphreys JE; Huang L; Morgan JB; Webster LO; Serabjit-Singh CS Rational use of in vitro P-glycoprotein assays in drug discovery. J. Pharmacol. Exp. Ther, 2001, 299(2), 620–628. [PubMed] [Google Scholar]
- 79.Akiyama S; Cornwell MM; Kuwano M; Pastan I; Gottesman MM Most drugs that reverse multidrug resistance also inhibit photoaffinity labeling of P-glycoprotein by a vinblastine analog. Mol. Pharmacol, 1988, 33(2), 144–147. [PubMed] [Google Scholar]
- 80.Beck WT; Cirtain MC; Glover CJ; Felsted RL; Safa AR Effects of indole alkaloids on multidrug resistance and labeling of P-glycoprotein by a photoaffinity analog of vinblastine. Biochem. Biophys. Res. Commun, 1988, 153(3), 959–966. 10.1016/S0006-291X(88)81321-4 [DOI] [PubMed] [Google Scholar]
- 81.Elewa H; Wilby KJ A Review of pharmacogenetics of antimalarials and associated *clinical implications. Eur. J. Drug Metab. Pharmacokinet, 2017, 42(5), 745–756. 10.1007/s13318-016-0399-1 [DOI] [PubMed] [Google Scholar]
- 82.Fakhouri EW; Peterson SJ; Kothari J; Alex R; Shapiro JI; Abraham NG Genetic polymorphisms complicate COVID-19 therapy: pivotal role of HO-1 in cytokine storm. Antioxidants, 2020, 9(7)E636 10.3390/antiox9070636 [DOI] [PMC free article] [PubMed] [Google Scholar]


