145
Views
6
CrossRef citations to date
0
Altmetric
Review

Clinical outcomes and management of mechanism-based inhibition of cytochrome P450 3A4

, , &
Pages 3-13 | Published online: 28 Dec 2022

Abstract

Mechanism-based inhibition of cytochrome P450 (CYP) 3A4 is characterized by NADPH-, time-, and concentration-dependent enzyme inactivation, occurring when some drugs are converted by CYPs to reactive metabolites. Such inhibition of CYP3A4 can be due to the chemical modification of the heme, the protein, or both as a result of covalent binding of modified heme to the protein. The inactivation of CYP3A4 by drugs has important clinical significance as it metabolizes approximately 60% of therapeutic drugs, and its inhibition frequently causes unfavorable drug–drug interactions and toxicity. The clinical outcomes due to CYP3A4 inactivation depend on many factors associated with the enzyme, drugs, and patients. Clinical professionals should adopt proper approaches when using drugs that are mechanism-based CYP3A4 inhibitors. These include early identification of drugs behaving as CYP3A4 inactivators, rational use of such drugs (eg, safe drug combination regimen, dose adjustment, or discontinuation of therapy when toxic drug interactions occur), therapeutic drug monitoring, and predicting the risks for potential drug–drug interactions. A good understanding of CYP3A4 inactivation and proper clinical management are needed by clinical professionals when these drugs are used.

Introduction

The human cytochrome P450 (CYP) 3A subfamily, includes CYP3A4, 3A5, 3A7 (CitationNelson et al 1996), and 3A43 (CitationDomanski et al 2001). CYP3A4 is most abundant in the human liver (∼40%) and metabolizes more than 50% of clinically used drugs (CitationShimada et al 1994; CitationRendic and Di Carlo 1997). Significant interindividual variability in the expression and activity of CYP3A4 has also been observed (CitationShimada et al 1994; CitationThummel et al 1994; Citationvon Richter et al 2004; CitationWatanabe et al 2004). Such a substantial variability is considered to be a result of environmental, physiological, and genetic factors (CitationGibson et al 2002). CYP3A4 is known to metabolize a large variety of compounds varying in molecular weight from lidocaine (Mr = 234) to cyclosporine (Mr = 1203) (CitationGuengerich 1999; CitationRendic 2002). It is also subjected to reversible and mechanism-based inhibition by a number of drugs. The latter involves the inactivation of the enzyme via the formation of metabolic intermediates (MIs) that bind tightly and irreversibly to the enzyme (CitationSilverman 1988; CitationKent et al 2001). Mechanism-based inactivation of CYP3A4 by drugs can be due to the chemical modification of the heme, the protein, or both as a result of covalent binding of modified heme to the protein (CitationOsawa and Pohl 1989; CitationOrtiz de Montellano and Correia 1995; CitationSilverman 1998).

A mechanism-based inhibition of CYP3A4 is characterized by NADPH-, time- and concentration-dependent enzyme inactivation and substrate protection (CitationIto et al 1998b; CitationSilverman 1998). Human liver microsomes, cDNA-expressed enzyme, and hepatocytes are commonly used in in-vitro models for the investigation of mechanism-based inhibition of CYP3A4 (CitationSilverman 1998). Important kinetic parameters for mechanism-based inhibition such as KI (the concentration required for half-maximum inactivation), kinact (the rate constant of maximum inactivation at saturation, analogous to Vmax), and partition ratio (Rmax, ratio of moles of substrate activation per mole of enzyme inactivation) can be determined using in vitro models. However, in vivo animal and human studies are usually needed to explore the clinical importance of CYP3A4 inactivation.

CYP3A4 inactivators such as delavirdine (CitationVoorman et al 1998), L-754,394 (CitationLightning et al 2000), 17α-ethynylestradiol (CitationLin et al 2002), and midazolam (CitationSchrag and Wienkers 2001; CitationKhan et al 2002) possibly bind covalently to the CYP apoprotein and inactivate it. On the other hand, certain CYP3A4 inactivators such as macrolides, eg, erythromycin (CitationFranklin 1991), glabridin (CitationKent et al 2002), and nelfinavir (CitationLillibridge et al 1998) bind the heme and inactivate the enzyme. In addition, the reactive intermediates of acetylenic compounds formed by several CYPs have been known to alkylate the prosthetic heme group as well as to bind covalently to the protein (CitationOrtiz de Montellano and Correia 1995). The aim of this review is to highlight the clinical outcomes and management of mechanism-based inhibition of CYP3A4.

Clinical outcomes of mechanism-based inhibition of CYP3A4

Pharmacokinetic drug–drug interactions

Due to the key role of CYP3A4 in drug metabolism, significant inactivation of this enzyme could result in marked pharmacokinetic drug–drug interactions. The in vivo inhibitory effect of a mechanistic inactivator is more prominent after multiple dosing and lasts longer than that of a reversible inhibitor (CitationLin and Lu 1998). The activated species irreversibly alters the enzyme to remove it permanently from the pool of active enzyme. Pharmacokinetic interactions often occur as a result of a change in drug metabolism.

For example, diltiazem as a CYP3A4 inactivator has been shown to potently inhibit the metabolism of a variety of coadministered drugs including carbamazepine (CitationBrodie and Macphee 1986), cyclosporine (CitationBrockmöller et al 1990; CitationSadrieh and Thomas 1994), quinidine (CitationLaganière et al 1996), midazolam (CitationBackman et al 1994), alfentanil (CitationAhonen et al 1996), nifedipine (CitationToyosaki et al 1988), and lovastatin (CitationAzie et al 1998). Inhibition of CYP3A by ritonavir explains, at least in part, the remarkable elevation of blood concentrations and area under the plasma concentration-time curve (AUC) of concomitantly administered drugs including rifabutin (CitationCato et al 1998), clarithromycin (CitationOuellet et al 1998), ketoconazole (CitationHsu et al 1998), saquinavir (CitationHsu et al 1998a), amprenavir (CitationSadler et al 2001; CitationGoujard et al 2003), and nelfinavir (CitationHsu et al 1998; CitationKurowski et al 2002).

Pharmacokinetic studies have indeed revealed specific interactions between erythromycin and cyclosporine (CitationVereerstraeten et al 1987), cisapride (CitationJenkins and Gibson 1996; CitationMichalets and Williams 2000; CitationKyrmizakis et al 2002), pimozide (CitationRubinstein 2001), disopyramide (CitationRagosta et al 1989), lidocaine (CitationIsohanni et al 1998, Citation1999), theophylline (CitationHemsworth and Renton 1981), astemizole (CitationKlausner 1999), carbamazepine (CitationZitelli et al 1987; CitationTagawa et al 1989; CitationTurner and Renton 1989; CitationMota et al 1996), midazolam (CitationYeates et al 1997), triazolam (CitationGreenblatt et al 1998), alprazolam (CitationYasui et al 1996), alfentanil (CitationBartkowski et al 1989), mosapride (CitationKatoh et al 2003), digoxin (CitationTsutsumi et al 2002), and warfarin (CitationBussey et al 1985; CitationWeibert et al 1989).

Inhibition of CYP3A by ritonavir partly explains the remarkable elevation of blood concentrations and AUC of concomitantly administered drugs that are extensively metabolized by CYP3A4 and have intermediate (10–80 L/h) to high (> 80 L/h) intrinsic clearance and significant first-pass metabolism. These drugs include rifabutin (400% increase) (CitationCato et al 1998), clarithromycin (77%) (CitationOuellet et al 1998), ketoconazole (350%) (CitationHsu et al 1998), saquinavir (5000%) (CitationHsu et al 1998a), amprenavir (210%) (CitationSadler et al 2001), nelfinavir (152%) (CitationHsu et al 1998; CitationKurowski et al 2002), ABT-378 (7700%) (CitationSham et al 1998), and indinavir (380%) (CitationHsu et al 1998b). The large increase in the plasma concentration of other protease inhibitors when coadministered with ritonavir provides the basis of rational dual protease inhibitor regimens, resulting in reduced doses and less frequent dosage intervals.

The functional effects of CYP3A4 inactivation on the pharmacokinetic behavior of the inhibitor itself or that of coadministered drugs are complex and depend on factors that are associated both with drugs and patients. The downstream consequences (efficacy or toxicity) of CYP3A4 inhibition may or may not parallel the pharmacokinetic alterations. Importantly, mechanism-based CYP3A4 inactivation causes long-term effects on drug pharmacokinetics, as the inactivated CYP3A4 has to be replaced by newly synthesized CYP3A4 protein.

It is believed that the in vivo inhibitory effect of a mechanistic inactivator is more prominent after multiple dosing and lasts longer than that of a reversible inhibitor (CitationLin and Lu 1998). The activated species irreversibly alters the enzyme to remove it permanently from the pool of active enzyme. Thus, the time-dependent inactivation of CYP3A enzymes would result in nonlinear pharmacokinetics, as exemplified by 50%–100% prolongation of the diltiazem half-life in humans after chronic dosing compared with the single-dose effects (CitationTsao et al 1990). Certainly, in addition to enzyme inactivation, other mechanisms may also play a role in the prolonged t1/2β. These include reversible inhibition of CYP3A by diltiazem and its metabolites, interactions of diltiazem with its metabolites, change of plasma protein binding, and possible involvement of drug transporters such as PgP. By investigating the pharmacokinetic interactions between diltiazem and its metabolites, and with model PgP inhibitors, identifying the contributions of these alternative mechanisms is likely.

The extent of drug interaction due to CYP3A inactivation will be time-dependent in both its onset and offset. For example, erythromycin did not significantly inhibit the clearance of alfentanil on the first day of coadministration but produced a 25% decrease after 7 days (CitationBartkowski et al 1989). As the half-life for the onset of inactivation is inversely proportional to the efficiency [kinact/(KI + [I])] of inactivation, the delayed onset of inhibition by erythromycin is a predictable property of a relatively weak inactivator. The delayed termination of CYP3A inhibition is expected to be independent of the inactivating drug and the extent of inhibition. This time-dependent termination may explain the serious adverse events associated with discontinuation of the irreversible inactivator, mibefradil, and the need for immediate initiation of alternative calcium-channel blocker treatment (CitationMullins et al 1998; CitationPrueksaritanont et al 1999). A mibefradil washout period of 7–14 days is therefore recommended.

Drug toxicities

Mechanism-based inactivation of CYP3A4 may cause severe drug toxicity through the enhanced exposure to coadministered drugs (CitationDresser et al 2000). For example, when irreversible CYP3A4 inhibitors such as erythromycin or clarithromycin are coadministered with terfenadine, astemizole, or pimozide patients may experience Torsades de pointes (a life-threatening ventricular arrhythmia associated with QT prolongation) (CitationSpinler et al 1995; CitationDresser et al 2000; CitationMichalets and Williams 2000). Terfenadine is a CYP3A4 substrate that undergoes extensive first-pass metabolism following oral administration (CitationHonig et al 1992; CitationJurima-Romet et al 1994). Normally, the carboxylate metabolite of terfenadine is the principal circulating entity in plasma, whereas unchanged terfenadine is not present at measurable concentrations (CitationHonig et al 1992, Citation1993). Furthermore, rhabdomyolysis has been observed when simvastatin was combined with erythromycin or ritonavir (CitationWilliams and Feely 2002). Symptomatic hypotension may occur when mechanism-based CYP3A4 inhibitors are combined with some dihydropyridine calcium antagonists (CitationAnderson and Nawarskas 2001), as well as with the phosphodiesterase inhibitor, sildenafil (CitationSimonsen 2002). In addition, ataxia can occur when carbamazepine is coadministered with mechanism-based CYP3A4 inhibitors such as macrolide antibiotics, isoniazid, verapamil, or diltiazem (CitationSpina et al 1996; CitationPatsalos et al 2002).

Mibefradil (Posicor) was launched for the long-term therapy of patients with hypertension in the United States in 1997, but was voluntarily withdrawn from the market in June 1998 due to potential toxic drug interactions. Severe drug interactions of mibefradil (a mechanism-based CYP3A4 inhibitor) have been reported with β-blockers (CitationMullins et al 1998), digoxin (CitationSiepmann et al 1995), methylprednisolone (CitationVaris et al 2000), verapamil (CitationMullins et al 1998; CitationWelker et al 1998), diltiazem (CitationMullins et al 1998; CitationWelker et al 1998), cyclosporine (CitationSpoendlin et al 1998), simvastatin (CitationSchmassmann-Suhijar et al 1998; CitationWelker et al 1998), and tacrolimus (CitationKrahenbuhl et al 1998). For example, 4 cases of life-threatening cardiogenic shock have been reported in patients taking mibefradil and β-blockers, who also began taking dihydropyridine calcium channel blockers (CitationMullins et al 1998). One case resulted in death, and the other 3 survived episodes of cardiogenic shock with intensive support of heart rate and blood pressure. Cases of myopathy, including rhabdomyolysis, have been reported in patients with hypertension taking simvastatin concomitantly with mibefradil (CitationSchmassmann-Suhijar et al 1998; CitationMuck 2000). These drug interactions often resulted in increased plasma concentrations of coadministered drugs and/or severe toxicity.

Immune responses

In addition to toxic drug–drug interactions, the formation of drug-modified CYP3A can also play a role in the initiation of toxicity. The formed adduct can act as a hapten and induce potential immune responses, leading to anti-CYP3A autoantibody production. The formation of drug-CYP3A adducts may thus be either nontoxic, or rarely fatal, depending on the drugs, the kinetics of adduct formation and degradation, other affected target proteins and organs, and the pathological conditions of the patients (CitationChitturi and George 2002). The reactivity of drug intermediates, the subcellular localization of major protein targets, and the primary interactions between metabolite and protein are important determining factors of toxicity (CitationPumford and Halmes 1997; CitationTornqvist et al 2002). Selective protein covalent binding by drugs or metabolites has been associated with target organ/tissue toxicity of drugs (CitationCohen et al 1997), although the underlying mechanisms are still incompletely known for most drugs.

Studies to reveal the role of drug reactive metabolites and their protein-adducts (eg, modified CYP3A4) in the mechanism of drug-induced idiosyncratic reactions are lacking. Mostly the proposed mechanism is based on the so-called hapten hypothesis, which requires drug bioactivation, covalent binding to proteins followed by uptake, antigen processing, and a polyclonal immune response (CitationNaisbitt et al 2001; CitationJu and Uetrecht 2002; CitationPirmohamed et al 2002). The recently proposed “danger hypothesis” by CitationPirmohamed et al (2002; CitationNaisbitt et al 2003), which hypothesizes that the immune system only responds to danger signals, can be considered to be additive to the hapten hypothesis. Thus, stimulation of an immune response to a drug–protein adduct (signal 1) requires the presence of co-stimulatory signals and cytokines (signals 2 and 3), which propagate and determine the type of immune response. For example, in acetaminophen (APAP) toxicity, covalent binding of N-acetyl-p-benzoquinone (NAPQI) to proteins (signal 1) may induce the production of nitric oxide (signal 2) due to the activation of Kupffer cells, which then scavenges superoxide to produce peroxynitrite (signal 3), leading to protein nitration and tissue injury (CitationJaeschke et al 2002). The nature of the danger signal is poorly defined, as some drug–protein adducts are not toxic in vivo.

Factors affecting the clinical outcomes of mechanism-based CYP3A4 inhibition

The clinical outcomes due to CYP3A4 inactivation depend on many factors associated with the enzyme, drugs, and patients. The interindividual variations in enzyme activity, polypharmacy, diseases, and genetic factors are all potentially important factors affecting the outcomes of mechanism-based inactivation of CYP3A4. In most cases, there are remarkable interindividual variations in the mechanism-based inactivation of CYP3A4 and subsequent drug–drug interactions. This is mainly due to the significant differences in the hepatic expression of CYP3A4, based on both in vitro (35–100-fold) studies with a human liver bank (CitationShimada et al 1994) and in vivo (20–50-fold) studies with probe drugs such as erythromycin (CitationWatkins 1994), midazolam (CitationThummel et al 1994), and alfentanil (CitationKharasch et al 2003). The expression of CYP3A can also be influenced by inflammation and proinflammatory cytokines, which are known to affect drug metabolism by downregulating or upregulating expression of several CYPs, including the CYP3A subfamily. However, it appears that there are no marked age and gender differences in CYP3A4 expression (CitationHunt et al 1992; CitationSchmucker 2001; CitationMeibohm et al 2002), although 24%–36% higher activity in females has been reported (CitationWatkins et al 1989). Changes in liver size and blood flow, renal function, and drug–protein binding and distribution with aging may be significant variables that can affect drug metabolism and elimination. Thus, age and gender may be important factors determining the clinical outcome of drug interactions involving CYP3A4 inactivation.

Intestinal CYP3A4 inactivation is also an important factor modulating the clinical outcomes of CYP3A4 inactivation. For example, oral administration of clarithromycin for one week resulted in an AUC ratio (ie, pretreatment AUC/post-treatment AUC) for intravenous midazolam of 0.33, but the ratio was 0.16 for oral midazolam (CitationGorski et al 1998). This greater extent of interaction observed after oral administration is consistent with the intestinal availability of approximately 0.4 for midazolam and a complete inactivation of intestinal CYP3A by clarithromycin (CitationGorski et al 1998). Such a complete inactivation of intestinal CYP3A is consistent with the estimates of kinact and KI and the relatively high concentrations of clarithromycin expected in the intestinal lumen. The impact of inactivation of intestinal wall CYP3A depends on the values of the inactivation parameters, the residence time of the inhibitor at the site of CYP3A, and the relative contribution of intestinal and hepatic metabolism to the first-pass metabolism of a given drug.

The clinical consequences of mechanism-based CYP3A4 inhibition depend upon additional factors. Such factors include variations in concentration-time course, plasma protein binding, atypical substrate kinetics for CYP3A4, existence of multiple inhibitory metabolites, partitioning from plasma to liver, rate-limiting active transport of drug and inhibitor into the hepatocytes, intestinal active efflux of drug, and inhibitor and extrahepatic metabolism of drugs and inhibitors.

Clinical management of mechanism-based inhibition of CYP3A4

The inactivation of CYP3A4 by drugs has important clinical implications since CYP3A4 metabolizes approximately 60% of therapeutic drugs, and its inhibition frequently causes unfavorable drug–drug interactions and toxicity. A good understanding of CYP3A4 inactivation and proper clinical management are needed by clinical professionals when these drugs are used.

Identification of drugs causing irreversible CYP3A4 inhibition

A number of drugs with widely differing structures and therapeutic targets have been reported to be mechanism-based inhibitors of CYP3A4 (). The identification of drugs causing irreversible CYP3A4 inhibition and the mechanisms involved are important in terms of rational use of therapeutic drugs. Mechanism-based inhibition of CYP3A4 can decrease a drug's first-pass clearance in the liver and greatly alter the kinetic behavior. Moreover, the decrease in functional catalytic activity of the damaged CYP can lead to enhanced exposure of other drugs that are normally cleared by CYP3A4, thus setting the stage for potential drug–drug interactions. It is therefore not surprising that during drug development new compounds that are potent CYP3A4 inhibitors are regarded with high skepticism and often withdrawn from further development.

Table 1 Drugs as mechanism-based CYP3A4 inhibitors

Early and timely identification of candidate drugs that inactivate CYP3A4 is important in drug development, as this can save large expenditures in time and money. In cases where new drugs have already been launched, and where a large population of patients has been exposed to the drug, severe interactions and even fatal toxicity may occur. Therefore, lessons should be learned from such cases (eg, mibefradil) where a drug was withdrawn from the market due to an unacceptable risk of potentially dangerous drug–drug interactions resulting from CYP3A inactivation. With the current techniques, it is increasingly possible to predict which new drugs will be associated with the formation of reactive metabolites and CYP3A inactivation. By screening drug candidates for possible formation of reactive metabolic intermediates (MIs) that inhibit CYP3A4 in an irreversible manner, and by establishing structure-activity relationships, it is possible to identify and eliminate such chemicals at an early stage of development. The application of genomic and proteomic approaches to the study of drug–CYP interactions, as well as the use of heterogeneous expression systems for specific CYPs including CYP3A4, has the potential to lead to a more effective screen due to a high-throughput capacity.

So far, the identified clinically important mechanism-based CYP3A4 inhibitors include antibiotics (eg, clarithromycin, erythromycin, and isoniazid), anticancer agents (eg, irinotecan and tamoxifen), anti-HIV agents (eg, ritonavir and delavirdine), antihypertensives (eg, verapamil and diltiazem), and steroids and their modulators (eg, gestodene and mifepristone). Most of these CYP3A4 inactivators are also substrates and inducers of CYPs (in particular CYP3A4). The drugs that inactivate CYP3A4 often possess a number of structural similarities such as a tertiary amine function, a furan ring, and an acetylene functional group. It appears that the chemical properties of a drug critical for CYP3A4 inactivation include: (1) being a CYP substrate; (2) formation of reactive metabolites; (3) preponderance of CYP inducers; and (4) being a P-glycoprotein (PgP) substrate.

Rational use of drugs as mechanism-based CYP3A4 inhibitors

Several drugs (eg, mibefradil) that have been identified as CPY3A4 inactivators have been withdrawn from the market due to reported toxic drug–drug toxicities. However, a number of drugs that behave as mechanism-based CYP3A4 inhibitors are still in use by clinical doctors. Caution should be taken when these drugs are used, particularly when used in combination with other drugs that are CYP3A4 substrates and have narrow therapeutic indices.

Mechanism-based CYP3A4 inhibitors that have been reported to cause significantly increased systemic exposure of coadministered drugs and/or severe drug toxicity should be avoided. For example, it is known that erythromycin can cause Torsades de pointes when coadministered with terfenadine, cisapride, or astemizole (all CYP3A4 substrates) (CitationSpinler et al 1995; CitationDresser et al 2000; CitationMichalets and Williams 2000), and should therefore be avoided in these combinations.

In some instances, proper dose adjustment or alternative drugs may be needed when a CYP3A4 inactivator is combined with another drug. For example, isoniazid, a mechanism-based CYP3A4 inhibitor (CitationWen et al 2002), markedly increased serum phenytoin concentrations, with resultant toxicity in some patients (CitationKutt et al 1968; CitationMiller et al 1979). Isoniazid also potentiated acetaminophen hepatotoxicity (CitationCrippin 1993; CitationNolan et al 1994). One case of increased warfarin toxicity (at 10 mg/day) including increased prothrombin time, hematuria, and bleeding gums caused by isoniazid has also been reported (CitationSelf et al 1999). Such interactions will require a reduced dose of coadministered drugs or replacement of drugs with similar efficacy while maintaining the isoniazid therapy.

Role of therapeutic drug monitoring

Monitoring plasma concentrations of concurrently administered drugs that are CYP3A4 substrates and/or have narrow therapeutic indices, and observing for signs of clinical toxicity are necessary for the early identification of potential drug–drug interactions and subsequent toxicities.

Tamoxifen, a nonsteroidal antiestrogen used for adjuvant chemotherapy of all stages of hormone-dependent breast cancer, is a potent mechanism-based CYP3A4 inhibitor (CitationZhao et al 2002). However, clinical data on tamoxifen–drug interactions are scant. A potential life-threatening interaction (CitationLodwick et al 1987; CitationRitchie and Grant 1989; CitationTenni et al 1989) may occur when tamoxifen is combined with warfarin, a substrate for CYP2C9, 2C19, and 3A4 (CitationKomatsu et al 2000). Prothrombin time doubled in a few cases and subdural hematoma developed. It is believed that CYP inhibition is likely to be the major mechanism for this interaction, while displacement of protein binding may just play a minor role. Thus, the plasma concentration of warfarin and prothrombin time should be carefully monitored when tamoxifen is coadministered with warfarin.

Herbal components may also behave as mechanism-based CYP3A4 inhibitors. Silybin, the major constituent of milk thistle (Sylybum marianum), is a mechanism-based inhibitor of CPY3A4 (CitationSridar et al 2004). Treatment of milk thistle at 175 mg (equivalent to silymarin 153 mg) 3 times per day for 3 weeks caused a 9% and 25% reduction in the AUC and mean trough level of indinavir, respectively (C8, the concentration at 8 hours) in healthy volunteers (CitationPiscitelli et al 2002). Therefore, monitoring plasma indinavir concentrations may be required when patients also take milk thistle.

Predicting risks for drug–drug interactions involving CYP3A4 inactivation

It may be necessary to assess the clinical risks by predicting qualitatively and quantitatively drug–drug interactions involving CYP3A4 inactivation. Generally, it can be anticipated that the inactivation of CYP3A4 by various drugs would increase the bioavailability of coadministered drugs that are primarily metabolized by CYP3A4, due to intestinal and/or hepatic inhibition of CYP3A4. Compared with reversible inhibition of CYP3A4, mechanism-based inhibitors of CYP3A4 more frequently cause pharmacokinetic-pharmacodynamic drug–drug interactions, as the inactivated CYP3A4 has to be replaced by newly synthesized CYP3A4 protein. The resultant drug interactions may lead to adverse drug effects, including some fatal events.

The accurate interpretation and extrapolation of in vitro interaction data requires a good understanding of pharmacokinetic principles. If the elimination of a drug is mainly by the liver, the total clearance is approximately equal to the hepatic clearance. For low-clearance drugs, a metabolism-based drug interaction will result in an almost proportional decrease in their clearance. On the other hand, if the intrinsic clearance is high, then the hepatic clearance is limited by the hepatic blood flow. Thus, for high-clearance drugs, a decrease in the intrinsic clearance caused by drug interaction has little effect on their hepatic clearance. However, the in vitro inhibitory potency of mechanism-based CYP3A4 inhibitors does not necessarily translate directly into relative extents of inhibition in vivo.

Determination of in vitro kinetic parameters is essential for the prediction of drug metabolism by a particular CYP in vivo. The apparent KI values for competitive inhibition determined in vitro, together with its relationship to unbound plasma concentrations of the inhibitor achieved with therapeutic doses in vivo, can be used as a rough guide to predict the possibility of a significant in vivo drug interaction (CitationIto et al 1998a, Citation1998b). Relating the in vitro results to in vivo pharmacokinetics is not straightforward. Generally, to quantitatively predict the degree of interaction observed in clinical cases, it is necessary to investigate the correlation between in vitro inhibitory potency of the inhibitor and in vivo inhibition, taking into account the distribution of the inhibitor into the liver, and extrapolation of data from animal studies (CitationYamano et al 2001).

Conclusions and future perspectives

The broad substrate selectivity makes CYP3A4 susceptible to relatively frequent reversible or irreversible (mechanism-based) inhibition by a wide variety of drugs. Irreversible inhibition of CYP3A4 due to enzyme inactivation or complexation occurs when some therapeutic drugs are converted by CYPs to reactive metabolites capable of covalently binding to CYP3A4 protein or heme moiety. Mechanism-based inhibition of CYP3A4 is characterized by NADPH-, time-, and concentration-dependent enzyme inactivation. A number of therapeutic drugs have been identified as mechanism-based CYP3A4 inhibitors.

Due to the key role of CYP3A4 in drug metabolism, inactivation of this enzyme could result in marked pharmacokinetic drug–drug interactions and/or toxicities. The clinical outcome of any drug–drug interaction due to CYP3A4 inactivation depends on a number of factors that are associated with both the administered drugs and patients, and the pharmacodynamic consequences may or may not closely follow pharmacokinetic alterations. Since many mechanism-based CYP3A4 inhibitors are also inducers of CYPs, and because enzyme inactivation is significantly affected by drug exposure time, the clearance of coadministered drugs may be increased or decreased in vivo, depending on the interplay between substrate, inhibitor, inducer, and CYP3A4. Thus, detailed studies are often needed to characterize these drugs and their potential drug–drug interactions.

Clinicians should adopt proper strategies when using drugs that are mechanism-based CYP3A4 inhibitors. Early identification of drugs behaving as CYP3A4 inactivators and the mechanism involved is important. If these drugs have to be used in some instances, rational use of such drugs becomes necessary, including the use of a safe drug combination regimen, dose adjustment, and discontinuation of therapy when toxic drug interactions occur. When combined with drugs with narrow therapeutic indices, the monitoring of plasma drug concentrations and observing of potential toxicities should be conducted. Predicting the risks for potential drug–drug interactions following proper pharmacokinetic principles and in vitro–in vivo extrapolation is likely. A good understanding of CYP3A4 inactivation and proper clinical management are needed by clinical professionals when these drugs are used.

Abbreviations

AUC=

area under the plasma concentration-time curve

Cmax=

maximum plasma concentration

CL=

clearance

CYP=

cytochrome P450

Einact=

inactivation efficiency

Ki=

apparent inhibition constant

KI=

the concentration required for half-maximum inactivation

Kinact=

the maximum rate of inactivation at saturation

PgP=

P-glycoprotein

t1/2β=

elimination half-life

t1/2inact=

inactivation half-life

Acknowledgements

The authors appreciate the support by the National University of Singapore Academic Research Funds.

References

  • AhonenJOlkkolaKTSalmenperäMEffect of diltiazem on midazolam and alfentanil disposition in patients undergoing coronary artery bypass graftingAnesthesiology1996851246518968170
  • AndersonJRNawarskasJJCardiovascular drug-drug interactionsCardiol Clin2001192153411407107
  • AzieNEBraterDCBeckerPAThe interaction of diltiazem with lovastatin and pravastatinClin Pharmacol Ther199864369779797793
  • BackmanJTOlkkolaKTArankoKDose of midazolam should be reduced during diltiazem and verapamil treatmentsBr J Clin Pharmacol19943722158198928
  • BartkowskiRRGoldbergMELarijaniGEInhibition of alfentanil metabolism by erythromycinClin Pharmacol Ther198946991022501060
  • BrockmöllerJNeumayerH-HWagnerKPharmacokinetic interaction between cyclosporin and diltiazemEur J Clin Pharmacol199038237422340843
  • BrodieMJMacpheeGJACarbamazepine neurotoxicity precipitated by diltiazemBMJ1986292117013085769
  • BusseyHIKnodelLCBoyleDAWarfarin-erythromycin interactionArch Intern Med1985145173674026508
  • CatoArdCavanaughJShiHThe effect of multiple doses of ritonavir on the pharmacokinetics of rifabutinClin Pharmacol Ther199863414219585795
  • ChanWKDelucchiABResveratrol, a red wine constituent, is a mechanism-based inactivator of cytochrome P450 3A4Life Sci20006731031211125847
  • ChatterjeePFranklinMRHuman cytochrome p450 inhibition and metabolic-intermediate complex formation by goldenseal extract and its methylenedioxyphenyl componentsDrug Metab Dispos2003311391714570772
  • ChenQNguiJSDossGACytochrome P450 3A4-mediated bioactivation of raloxifene: irreversible enzyme inhibition and thiol adduct formationChem Res Toxicol2002159071412119000
  • ChibaMNishimeJALinJHPotent and selective inactivation of human liver microsomal cytochrome P-450 isoforms by L-754,394, an investigational human immune deficiency virus protease inhibitorJ Pharmacol Exp Ther19952751527348531125
  • ChitturiSGeorgeJHepatotoxicity of commonly used drugs: nonsteroidal anti-inflammatory drugs, antihypertensives, antidiabetic agents, anticonvulsants, lipid-lowering agents, psychotropic drugsSemin Liver Dis2002221698312016548
  • CohenSDPumfordNRKhairallahEASelective protein covalent binding and target organ toxicityToxicol Appl Pharmacol19971431129073586
  • CrippinJSAcetaminophen hepatotoxicity: potentiation by isoniazidAm J Gastroenterol19938859058470644
  • DomanskiTLFintaCHalpertJRcDNA cloning and initial characterization of CYP3A43, a novel human cytochrome P450Mol Pharmacol2001593869211160876
  • DresserGKSpenceJDBaileyDGPharmacokinetic-pharmacodynamic consequences and clinical relevance of cytochrome P450 3A4 inhibitionClin Pharmacokinet200038415710668858
  • EaglingVAProfitLBackDJInhibition of the CYP3A4-mediated metabolism and P-glycoprotein-mediated transport of the HIV-1 protease inhibitor saquinavir by grapefruit juice componentsBr J Clin Pharmacol1999485435210583025
  • FranklinMRCytochrome P450 metabolic intermediate complexes from macrolide antibiotics and related compoundsMethods Enzymol1991206559731784242
  • GibsonGGPlantNJSwalesKEReceptor-dependent transcriptional activation of cytochrome P4503A genes: induction mechanisms, species differences and interindividual variation in manXenobiotica20023216520611958559
  • GorskiJCJonesDRHaehnerdanielsBDThe contribution of intestinal and hepatic CYP3A to the interaction between midazolam and clarithromycinClin Pharmacol Ther199864133439728893
  • GoujardCVincentIMeynardJLSteady-state pharmacokinetics of amprenavir coadministered with ritonavir in human immunodeficiency virus type 1-infected patientsAntimicrob Agents Chemother2003471182312499178
  • GreenblattDJvon MoltkeLLHarmatzJSInhibition of triazolam clearance by macrolide antimicrobial agents: in vitro correlates and dynamic consequencesClin Pharmacol Ther199864278859757151
  • GuengerichFPMechanism-based inactivation of human liver microsomal cytochrome P-450 IIIA4 by gestodeneChem Res Toxicol19903363712133086
  • GuengerichFPCytochrome P-450 3A4: regulation and role in drug metabolismAnnu Rev Pharmacol Toxicol19993911710331074
  • HaniokaNOzawaSJinnoHInteraction of irinotecan (CPT-11) and its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38) with human cytochrome P450 enzymesDrug Metab Dispos200230391611901092
  • HeKIyerKRHayesRNInactivation of cytochrome P4503A4 by bergamottin, a component of grapefruit juiceChem Res Toxicol19981125299548795
  • HeKWoolfTFHollenbergPFMechanism-based inactivation of cytochrome P-450-3A4 by mifepristone (RU486)J Pharmacol Exp Ther199928879179918590
  • HemsworthTCRentonKWDepression of theophylline metabolism and elimination by troleandomycin and erythromycinBiochem Pharmacol19813012993046973975
  • HonigPKWoosleyRLZamaniKChanges in the pharmacokinetics and electrocardiographic pharmacodynamics of terfenadine with concomitant administration of erythromycinClin Pharmacol Ther19925223181526078
  • HonigPKWorthamDCZamaniKTerfenadine-ketoconazole interaction: pharmacokinetic and electrocardiographic consequencesJAMA19932691513188445813
  • HsuAGrannemanGRBertzRJRitonavir. Clinical pharmacokinetics and interactions with other anti-HIV agentsClin Pharmacokinet199835275919812178
  • HsuAGrannemanGRCaoGPharmacokinetic interactions between two human immunodeficiency virus protease inhibitors, ritonavir and saquinavirClin Pharmacol Ther1998a63453649585800
  • HsuAGrannemanGRCaoGPharmacokinetic interaction between ritonavir and indinavir in healthy volunteersAntimicrob Agents Chemother1998b422784919797204
  • HuntCMWesterkamWRStaveGMEffect of age and gender on the activity of human hepatic CYP3ABiochem Pharmacol199244275831642641
  • IsohanniMHNeuvonenPOlkkolaKTEffect of erythromycin and itraconazole on the pharmacokinetics of oral lignocainePharmacol Toxicol199984143610193676
  • IsohanniMHNeuvonenPJPalkamaVJEffect of erythromycin and itraconazole on the pharmacokinetics of intravenous lignocaineEur J Clin Pharmacol19985456159832299
  • ItoKIwatsuboTKanamitsuSQuantitative prediction of in vivo drug clearance and drug interactions from in vitro data on metabolism, together with binding and transportAnnu Rev Pharmacol Toxicol1998a38461999597163
  • ItoKIwatsuboTKanamitsuSPrediction of pharmacokinetic alterations caused by drug-drug interactions: metabolic interaction in the liverPharmacol Rev1998b503874119755288
  • JacobsenWChristiansUBenetLZIn vitro evaluation of the disposition of A novel cysteine protease inhibitorDrug Metab Dispos20002813435111038163
  • JaeschkeHGoresGJCederbaumAIMechanisms of hepatotoxicityToxicol Sci2002651667611812920
  • JenkinsIRGibsonJCisapride, erythromycin and arrhythmiaAnaesth Intensive Care1996247288971331
  • JonesDRGorskiJCHammanMADiltiazem inhibition of cytochrome P-450 3A activity is due to metabolite intermediate complex formationJ Pharmacol Exp Ther199929011162510454485
  • JuCUetrechtJPMechanism of idiosyncratic drug reactions: reactive metabolite formation, protein binding and the regulation of the immune systemCurr Drug Metab200233677712093356
  • Jurima-RometMCrawfordKCyrTTerfenadine metabolism in human liver. In vitro inhibition by macrolide antibiotics and azole antifungalsDrug Metab Dispos199422849577895601
  • KanamitsuSItoKGreenCEPrediction of in vivo interaction between triazolam and erythromycin based on in vitro studies using human liver microsomes and recombinant human CYP3A4Pharm Res2000174192610870985
  • KatohTSaitohHOhnoNDrug interaction between mosapride and erythromycin without electrocardiographic changesJpn Heart J2003442253412718484
  • KentUMAviramMRosenblatMThe licorice root derived isoflavan glabridin inhibits the activities of human cytochrome P450S 3A4, 2B6, and 2C9Drug Metab Dispos2002307091512019199
  • KentUMJuschyshynMIHollenbergPFMechanism-based inactivators as probes of cytochrome P450 structure and functionCurr Drug Metab200122154311513328
  • KhanKKHeYQDomanskiTLMidazolam oxidation by cytochrome P450 3A4 and active-site mutants: an evaluation of multiple binding sites and of the metabolic pathway that leads to enzyme inactivationMol Pharmacol20026149550611854429
  • KharaschEDHofferCWalkerADisposition and miotic effects of oral alfentanil: a potential noninvasive probe for first-pass cytochrome P4503A activityClin Pharmacol Ther20037319920812621385
  • KlausnerMAAstemizole use with erythromycinAnn Allergy Asthma Immunol19998342210582723
  • KomatsuTYamazakiHAsahiSFormation of a dihydroxy metabolite of phenytoin in human liver microsomes/cytosol: roles of cytochromes P4502C9, 2C19, and 3A4Drug Metab Dispos2000281361811038165
  • KoudriakovaTIatsimirskaiaEUtkinIMetabolism of the human immunodeficiency virus protease inhibitors indinavir and ritonavir by human intestinal microsomes and expressed cytochrome P4503A4/3A5: mechanism-based inactivation of cytochrome P4503A by ritonavirDrug Metab Dispos199826552619616191
  • KrahenbuhlSMenafoglioAGiostraESerious interaction between mibefradil and tacrolimusTransplantation1998661113159808502
  • KurowskiMKaeserBSawyerALow-dose ritonavir moderately enhances nelfinavir exposureClin Pharmacol Ther2002721233212189359
  • KuttHVerebelyKMcDowellFInhibition of diphenylhydantoin metabolism in rats and in rat liver microsomes by antitubercular drugsNeurology196818706105691599
  • KyrmizakisDEChimonaTSKanoupakisEMQT prolongation and torsades de pointes associated with concurrent use of cisapride and erythromycinAm J Otolaryngol200223303712239699
  • LaganièreSDaviesRFCarignanGPharmacokinetic and pharmacodynamic interactions between diltiazem and quinidineClin Pharmacol Ther199660255648841148
  • LightningLKJonesJPFriedbergTMechanism-based inactivation of cytochrome P450 3A4 by L-754,394Biochemistry20003942768710757976
  • LillibridgeJHLiangBHKerrBMCharacterization of the selectivity and mechanism of human cytochrome P450 inhibition by the human immunodeficiency virus-protease inhibitor nelfinavir mesylateDrug Metab Dispos199826609169660842
  • LinHLKentUMHollenbergPFMechanism-based inactivation of cytochrome P450 3A4 by 17 alpha-ethynylestradiol: evidence for heme destruction and covalent binding to proteinJ Pharmacol Exp Ther2002301160711907170
  • LinJHLuAYHInhibition and induction of cytochrome P450 and the clinical implicationsClin Pharmacokinet199835361909839089
  • LodwickRMcConkeyBBrownAMLife threatening interaction between tamoxifen and warfarinBMJ198729511413120919
  • LuoGLinJFiskeWDConcurrent induction and mechanism-based inactivation of CYP3A4 by an L-valinamide derivativeDrug Metab Dispos2003311170512920173
  • MaBPrueksaritanontTLinJHDrug interactions with calcium channel blockers: possible involvement of metabolite-intermediate complexation with CYP3ADrug Metab Dispos2000281253010640508
  • MasubuchiYHorieTMechanism-based inactivation of cytochrome P450s 1A2 and 3A4 by dihydralazine in human liver microsomesChem Res Toxicol19991210283210525281
  • MasubuchiYOseAHorieTDiclofenac-induced inactivation of CYP3A4 and its stimulation by quinidineDrug Metab Dispos2002301143812228192
  • MayhewBSJonesDRHallSDAn in vitro model for predicting in vivo inhibition of cytochrome P450 3A4 by metabolic intermediate complex formationDrug Metab Dispos2000281031710950845
  • MeibohmBBeierleIDerendorfHHow important are gender differences in pharmacokinetics?Clin Pharmacokinet2002413294212036391
  • MichaletsELWilliamsCRDrug interactions with cisapride: clinical implicationsClin Pharmacokinet200039497510926350
  • MillerRRPorterJGreenblattDJClinical importance of the interaction of phenytoin and isoniazid: a report from the Boston Collaborative Drug Surveillance ProgramChest1979753568421578
  • MiuraTIwasakiMKomoriMDecrease in a constitutive form of cytochrome P-450 by macrolide antibioticsJ Antimicrob Chemother19892455192515189
  • MotaCRCarvalhoCMotaCSevere carbamazepine toxicity induced by concurrent erythromycin therapyEur J Pediatr19961553458777933
  • MuckWMetabolic interactions between mibefradil and HMG-CoA reductase inhibitors: linking in vitro with in vivo informationBr J Clin Pharmacol200049879010691326
  • MullinsMEHorowitzZLindenDHJLife-threatening interaction of mibefradil and beta-blockers with dihydropyridine calcium channel blockersJAMA199828015789669789
  • NaisbittDJPirmohamedMParkBKImmunopharmacology of hypersensitivity reactions to drugsCurr Allergy Asthma Rep200332229
  • NaisbittDJWilliamsDPPirmohamedMReactive metabolites and their role in drug reactionsCurr Opin Allergy Clin Immunol200113172511964707
  • NelsonDRKoymansLKamatakiTP450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclaturePharmacogenetics199661428845856
  • NolanCMSandblomREThummelKEHepatotoxicity associated with acetaminophen usage in patients receiving multiple drug therapy for tuberculosisChest1994105408117508362
  • Ortiz de MontellanoPRCorreiaMAOrtiz de MontellanoPRInhibition of cytochrome P450Cytochrome P450: structure, mechanism and biochemistry1995New YorkPlenum Pr30566
  • OsawaYPohlLRCovalent bonding of the prosthetic heme to protein: a potential mechanism for suicide inactivation or activation of hemoproteinsChem Res Toxicol19892131412519716
  • OuelletDHsuAGrannemanGRPharmacokinetic interaction between ritonavir and clarithromycinClin Pharmacol Ther199864355629797791
  • PatsalosPNFroscherWPisaniFThe importance of drug interactions in epilepsy therapyEpilepsia2002433658511952767
  • PessayreDTinelMLarreyDInactivation of cytochrome P-450 by a troleandomycin metabolite. Protective role of glutathioneJ Pharmacol Exp Ther1983224685916600790
  • PirmohamedMNaisbittDJGordonFThe danger hypothesis-potential role in idiosyncratic drug reactionsToxicology2002181–1825563
  • PiscitelliSCFormentiniEBursteinAHEffect of milk thistle on the pharmacokinetics of indinavir in healthy volunteersPharmacotherapy200222551612013352
  • PrueksaritanontTMaBTangCMetabolic interactions between mibefradil and HMG-CoA reductase inhibitors: an in vitro investigation with human liver preparationsBr J Clin Pharmacol199947291810215754
  • PumfordNRHalmesNCProtein targets of xenobiotic reactive intermediatesAnnu Rev Pharmacol Toxicol199737911179131248
  • RagostaMWeihlACRosenfeldLEPotentially fatal interaction between erythromycin and disopyramideAm J Med19898646562467560
  • RendicSSummary of information on human CYP enzymes: human P450 metabolism dataDrug Metab Rev2002348344811996015
  • RendicSDi CarloFJHuman cytochrome P450 enzyme: a status report summarizing their reactions, substrates, induction, and inhibitorsDrug Metab Rev1997294135809187528
  • RitchieLDGrantSMTamoxifen-warfarin interaction: the Aberdeen hospitals drug fileBMJ198929812532502242
  • RubinsteinEComparative safety of the different macrolidesInt J Antimicrob Agents200118S71611574199
  • SadlerBMPilieroPJPrestonSLPharmacokinetics and safety of amprenavir and ritonavir following multiple-dose, co-administration to healthy volunteersAIDS20011510091811399983
  • SadriehNThomasPECharacterization of rat cytochrome P450 isozymes involved in the covalent binding of cyclosporin A to microsomal proteinsToxicol Appl Pharmacol1994127222328048065
  • Schmassmann-SuhijarDBullinghamRGasserRRhabdomyolysis due to interaction of simvastatin with mibefradilLancet19983511929309654265
  • Schmiedlin-RenPEdwardsDJFitzsimmonsMEMechanisms of enhanced oral availability of CYP3A4 substrates by grapefruit constituents. Decreased enterocyte CYP3A4 concentration and mechanism-based inactivation by furanocoumarinsDrug Metab Dispos1997251228339351897
  • SchmuckerDLLiver function and phase I drug metabolism in the elderly: a paradoxDrug Aging20011883751
  • SchragMLWienkersLCCovalent alteration of the CYP3A4 active site: evidence for multiple substrate binding domainsArch Biochem Biophys2001391495511414684
  • SelfTHChrismanCRBaciewiczAMIsoniazid drug and food interactionsAm J Med Sci19993173041110334118
  • ShamHLKempfDJMollaAABT-378, a highly potent inhibitor of the human immunodeficiency virus proteaseAntimicrob Agents Chemother1998423218249835517
  • ShimadaTYamazakiHMimuraMInterindividual variations in human liver cytochrome P450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicalsJ Pharmacol Exp Ther1994270414238035341
  • SiepmannMKleinbloesemCKirchWThe interaction of the calcium antagonist RO 40-5967 with digoxinBr J Clin Pharmacol19953949167669484
  • SilvermanRBMechanism-based enzyme inactivation: chemistry and enzymology1988Boca RatonCRC Pr
  • SilvermanRBMechanism-based enzyme inactivation: chemistry and enzymology1998Boca RatonCRC Pr
  • SimonsenUInteractions between drugs for erectile dysfunction and drugs for cardiovascular diseaseInt J Impot Res2002141788812058245
  • SpinaEPisaniFPeruccaEClinically significant pharmacokinetic drug interactions with carbamazepine. An updateClin Pharmacokinet1996311982148877250
  • SpinlerSAChengJWKindwallKEPossible inhibition of hepatic metabolism of quinidine by erythromycinClin Pharmacol Ther19955789947828386
  • SpoendlinMPetersJWelkerHPharmacokinetic interaction between oral cyclosporin and mibefradil in stabilized post-renal-transplant patientsNephrol Dial Transplant1998131787919681729
  • SridarCGoosenTCKentUMSilybin inactivates cytochromes P450 3A4 and 2C9 and inhibits major hepatic glucuronosyltransferasesDrug Metab Dispos2004325879415155549
  • StupansIMurrayMKirlichAInactivation of cytochrome P450 by the food-derived complex phenol oleuropeinFood Chem Toxicol20013911192411527571
  • TagawaTMimakiTOnoJErythromycin-induced carbamazepine intoxication in two epileptic childrenJpn J Psychiatry Neurol198943513142625794
  • TenniPLalichDLByrneMJLife threatening interaction between tamoxifen and warfarinBMJ1989298932493305
  • ThummelKEShenDDPodollTDUse of midazolam as a human cytochrome P450 3A probe: I. In vitro-in vivo correlations in liver transplant patientsJ Pharmacol Exp Ther1994271549567965755
  • TinelMDescatoireVLarreyDEffects of clarithromycin on cytochrome P-450. Comparison with other macrolidesJ Pharmacol Exp Ther1989250746512527301
  • TornqvistMFredCHaglundJProtein adducts: quantitative and qualitative aspects of their formation, analysis and applicationsJ Chromatogr B Analyt Technol Biomed Life Sci2002778279308
  • ToyosakiNToyo-OkaTNatsumeTCombination therapy with diltiazem and nifedipine in patients with effort angina pectorisCirculation198877137053286041
  • TsaoS-CDickinsonTHAbernethyDRMetabolite inhibition of parent drug biotransformation. Studies of diltiazemDrug Metab Dispos19901818021971570
  • TsutsumiKKotegawaTKuranariMThe effect of erythromycin and clarithromycin on the pharmacokinetics of intravenous digoxin in healthy volunteersJ Clin Pharmacol20024211596412362931
  • TurnerPVRentonKWThe interaction between carbamazepine and erythromycinCan J Physiol Pharmacol19896758262776076
  • VarisTBackmanJTKivistoKTDiltiazem and mibefradil increase the plasma concentrations and greatly enhance the adrenal-suppressant effect of oral methylprednisoloneClin Pharmacol Ther2000672152110741623
  • VereerstraetenPThiryPKinnaertPInfluence of erythromycin on cyclosporine pharmacokineticsTransplantation19874415563603676
  • von MoltkeLLDurolALDuanSXPotent mechanism-based inhibition of human CYP3A in vitro by amprenavir and ritonavir: comparison with ketoconazoleEur J Clin Pharmacol2000562596110952482
  • von RichterOBurkOFrommMFCytochrome P450 3A4 and P-glycoprotein expression in human small intestinal enterocytes and hepatocytes: a comparative analysis in paired tissue specimensClin Pharmacol Ther2004751728315001968
  • VoormanRLMaioSMHauerMJMetabolism of delavirdine, a human immunodeficiency virus type-1 reverse transcriptase inhibitor, by microsomal cytochrome P450 in humans, rats, and other species: probable involvement of CYP2D6 and CYP3ADrug Metab Dispos19982663199660845
  • WatanabeMKumaiTMatsumotoNExpression of CYP3A4 mRNA is correlated with CYP3A4 protein level and metabolic activity in human liverJ Pharmacol Sci2004944596215107587
  • WatkinsPBNoninvasive tests of CYP3A enzymesPharmacogenetics19944171847987401
  • WatkinsPBMurraySAWinkelmanLGErythromycin breath test as an assay of glucocorticoid-inducible liver cytochromes P-450. Studies in rats and patientsJ Clin Invest198983688972913056
  • WeibertRTLorentzSMTownsendRJEffect of erythromycin in patients receiving long-term warfarin therapyClin Pharm19898210142706893
  • WelkerHAWiltshireHBullinghamRClinical pharmacokinetics of mibefradilClin Pharmacokinet199835405239884814
  • WenXWangJSNeuvonenPJIsoniazid is a mechanism-based inhibitor of cytochrome P450 1A2, 2A6, 2C19 and 3A4 isoforms in human liver microsomesEur J Clin Pharmacol20025779980411868802
  • WilliamsDFeelyJPharmacokinetic-pharmacodynamic drug interactions with HMG-CoA reductase inhibitorsClin Pharmacokinet2002413437012036392
  • YamanoKYamamotoKKatashimaMPrediction of midazolam-CYP3A inhibitors interaction in the human liver from in vivo/in vitro absorption, distribution, and metabolism dataDrug Metab Dispos2001294435211259329
  • YasuiNOtaniKKanekoSA kinetic and dynamic study of oral alprazolam with and without erythromycin in humans: in vivo evidence for the involvement of CYP3A4 in alprazolam metabolismClin Pharmacol Ther199659514198646822
  • YeatesRALaufenHZimmermannTPharmacokinetic and pharmacodynamic interaction study between midazolam and the macrolide antibiotics, erythromycin, clarithromycin, and the azalide azithromycinInt J Clin Pharmacol Ther19973557799455717
  • YeoKRYeoWWInhibitory effects of verapamil and diltiazem on simvastatin metabolism in human liver microsomesBr J Clin Pharmacol2001514617011422004
  • ZdravkovicMOlsenAKChristiansenTA clinical study investigating the pharmacokinetic interaction between NN703 (tabimorelin), a potential inhibitor of CYP3A4 activity, and midazolam, a CYP3A4 substrateEur J Clin Pharmacol200358683812610745
  • ZhaoXJJonesDRWangYHReversible and irreversible inhibition of CYP3A enzymes by tamoxifen and metabolitesXenobiotica2002328637812419016
  • ZitelliBJHowrieDLAltmanHErythromycin-induced drug interactions. An illustrative case and review of the literatureClin Pediatr (Phila)198726117193816008