Pharmacokinetic Drug-Drug Interactions:
A Systematic Review of the Cytochrome P450 (CYP) Isoenzyme 3A4
Ahmad Ainurofiq1*, Lutvi Ismaya2
1Department of Pharmacy, Sebelas Maret University, Ir. Sutami 36A, Surakarta 57126, Indonesia.
2Pharmacy Program, School of Vocational, Sebelas Maret University,
Kolonel Sutarto 150K, Surakarta 57126, Indonesia.
*Corresponding Author E-mail: rofiq@mipa.uns.ac.id
ABSTRACT:
The cytochrome P450 (CYP) isoenzyme 3A4 or CYP3A4 is a major drug-metabolizing enzyme that has the potential to cause pharmacokinetic drug-drug interactions. Primary research studies have demonstrated CYP3A4-mediated drug-drug interactions through a variety of mechanisms. However, there has been no review during the last 10 years of pharmacokinetic drug-drug interactions mediated by CYP3A4 isoenzymes. It is necessary to systematically review the pharmacokinetic drug-drug interactions mediated by CYP3A4. Source review of articles were retrieved from the PubMed and Scopus databases. The preparation of keywords through the population, intervention, comparison, and outcomes (PICO) method written based on the Boolean operator. Reporting the results of the paper search is presented in the Prisma version 1 2020 flowchart. The risk of bias assessment used COHORT tools and Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) tools. Data was analyzed narratively. Pharmacokinetic drug interactions are mediated by CYP3A4 through the mechanism of induction, activation, inhibition, and inactivation. Induction or activation of CYP3A4 can cause an increase in CYP3A4 expression, so that the drug is metabolized more quickly and has the potential to lose drug efficacy. Inhibition and inactivation of CYP3A4 causes plasma drug levels to increase and drug elimination time to last longer. CYP3A4 plays a major role in the bioactivation of drugs that cause hepatotoxicity through the formation of reactive metabolites. The use of drugs needs to be monitored to avoid pharmacokinetic drug interactions.
KEYWORDS: Drug-drug interaction, CYP3A4, Mechanism, Pharmacokinetic, Drug metabolism.
INTRODUCTION:
Drug interactions are the effects or actions of drugs that change or are modified due to interactions with one or more other drugs1-3. Adverse drug reactions are drug effects that are mild to toxic, including hypersensitivity and anaphylaxis. Drug incompatibility is a chemical or physical action that occurs between two or more in vitro conditions4. There are three categories of drug interaction mechanisms5, which include pharmaceutical interactions (in vitro)6,7, pharmacokinetics8-10, and pharmacodynamics11,12. Pharmacokinetic drug interactions involve processes of absorption, distribution, metabolism, and excretion, all of which are associated with treatment failure or toxicity13.
Pharmacokinetic drug-drug interactions occur when a drug changes the disposition (absorption, distribution, metabolism, elimination) of the co-administered drug. Pharmacokinetic interactions lead to an increase or decrease in the plasma concentration of the drug. The intensity of these modifications varies, but may cause association contraindications14. Drug interactions in the distribution process consist of three mechanisms, namely metabolism inhibition, metabolism induction, and changes in hepatic blood flow15. Inhibition of drug metabolism applies to drugs or substances that are substrates for cytochrome P450 (CYP) enzymes16.
CYP is an enzyme that can cause drug toxicity due to enzyme induction factors, enzyme inhibition (both reversible and irreversible), and pharmacogenetics. Pharmacogenetic approaches that take into account CYP genotype and/or phenotype by individualizing treatment regimen17. Inhibition or induction of the CYP system, especially CYP3A4, play an important role in the pharmacokinetic interaction of drugs by changing the concentration of drugs or toxic metabolites in the target tissue of action18. CYP enzymes belong to a superfamily of monooxygenases whose structures are interrelated and hydrolyze many physiological molecules (such as steroids and fatty acids) and xenobiotic compounds (such as drugs, carcinogens, and environmental substances). In humans, there are approximately 100 CYP isoenzymes with differing but overlapping specificities. These isoenzymes generally have two main components, namely an electron donor reductase system that uses an electron transfer from NADPH and a substrate-bound CYP19. Its catalytic activity plays an important role in various fields such as drug-drug interactions and endocrine function20.
CYP is a major source of variability in pharmacokinetics and drug responses. These enzymes are responsible for the biotransformation of most foreign substances, accounting for the metabolism of 70-80% of all drugs in clinical use. The highest expression forms in the liver were CYPs 3A4, 2C9, 2C8, 2E1, and 1A2, while 2A6, 2D6, 2B6, 2C19, and 3A5 were less abundant, and CYPs 2J2, 1A1, and 1B1 were expressed in extrahepatic tissues. The expression of each CYP is influenced by a unique combination of mechanisms and factors including genetic polymorphisms, xenobiotic induction, cytokine regulation, hormones, disease, gender, age, and others21. In addition, polypharmacy (prescribing more than five types of drugs) has the potential to cause drug-drug interactions22.
Based on the background description above, it is known that there is a potential for drug interactions based on pharmacokinetic properties mediated by CYP3A4 isoenzymes. However, there are no studies that conclude the pharmacokinetic mechanism of drug interactions mediated by CYP3A4 isoenzymes. Therefore, it is necessary to review the pharmacokinetic drug interactions mediated by the CYP3A4 isoenzyme based on research data from the results of a literature review that often occurs with systematic assessment. The urgency this study is expected to provide a systematic description of the pharmacokinetic drug interactions mediated by the CYP3A4 isoenzyme. The benefit of this research for the development of science, especially in the field of pharmaceutical technology and clinical pharmacy. For the community, this research is expected to be the basis for the decision to consume a certain drug because it can cause pharmacokinetic interactions mediated by the CYP3A4 isoenzyme.
MATERIALS AND METHODS:
This systematic review summarizes the results of primary research related to pharmacokinetic drug-drug interactions mediated by CYP3A4 for the period 2010–2020. The systematic review of the literature was conducted December 2020–December 2021. The collection of papers sourced from Scopus and PubMed were carried out fromJanuary to October 2021. This study aims to summarize the primary research on the pharmacokinetic mechanism of drug interactions mediated by the CYP3A4 isoenzyme. The initial search approach was to compile keywords through the population, intervention, comparison, and outcome (PICO) method, because keywords obtained in this manner are used to obtain papers that are in accordance with the theme and as a reference in research discussions. The arrangement of keywords is written based on the Boolean operator with the conjunction "AND" to get a wider range of results. The search used the obtained keywords as follows: (drug-drug interaction) AND (P450 (CYP) 3A4) AND (mechanism). The inclusion criteria were as follows: each paper was written in English, was published 2010–2020, and followed a primary research study design examining in vitro and in vivo pharmacokinetic drug interactions mediated by CYP3A423-25.
The risk of bias assessment for the preclinical in vitro studies used the Consolidated Standards of Reporting Trials (CONSORT) protocol. Paper quality was assessed using CONSORT, which consists of 15 items to assess individual parts of a paper (abstract, introduction, methods, results, discussion, and other information). The CONSORT checklist was originally developed to assess the quality of dental studies in vitro and has been used in the assessment of mechanistic studies on SARS-CoV-infected cells in vitro with modifications according to the study26,27.
The risk of bias assessment for the in vivo studies used the Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) method. The SYRCLE checklist consists of 10 questions to assess different types of bias (selection bias, detection bias, performance bias, attrition bias, reporting bias, and others). The possible answers for each item are yes, no, and unclear, which indicate low, high, and unclear risk of bias. The SYRCLE tool was developed for animal studies and adapted from Cochrane's Risk of Bias Tool. The assessed bias risks are selection, performance, detection, attrition, and reporting bias. Selection bias results from inadequate randomization and allocation concealment. Performance bias depends on placement of animals in random cages and concealment from caregivers and operators. Detection bias occurs when selection of animals is inadequate for outcome assessment and concealment from examiners. Friction bias is related to animal omissions and incomplete or uneven exclusions. Reporting bias occurs due to the selection of reported results28.
Figure 1: PRISMA flow chart for papers selection and evaluation
RESULT:
The search resulted in 396 (339 from Scopus and 57 from PubMed) studies. The initially identified papers duplicated by one search engine or by both search engines, so 13 studies were removed. Papers (163) whose title and abstract did not match the research topic were excluded. Eighteen papers that could not be accessed and two papers with results that did not match the research topic were excluded. Thus, 41 papers met the inclusion criteria for further analysis. The results of the search are presented in the Prisma flow chart in Figure 1
DISCUSSION:
The primary consequence of pharmacokinetic drug-drug interactions is increasing or decreasing drug levels in plasma and tissues. The inhibition of drug-metabolizing enzymes results in elevated plasma drug levels. The effect of the resulting increase in pharmacological activity depends on the therapeutic window. Inhibiting enzyme activation and prodrug induction reduces the effectiveness of treatment. Inhibiting the primary pathway of drug metabolism diverts metabolism to the secondary pathway, which is problematic if toxic products are created29.
Drug metabolism involves the catalysis of a class of mixed-function enzymes (CYP). One of the most abundant drug-metabolizing enzymes in this class is CYP3A4. The CYP3A4 isoenzyme is a member of the CYP3A enzyme family, consisting of four genes (CYP3A4, CYP3A5, CYP3A7, and CYP3A43) arranged on the CYP3A locus. An increase in CYP3A4 expression indicates an increase in the production and metabolic activity of the enzyme, so that the drug level in the plasma decreases. Likewise, a decrease in CYP3A4 expression indicates lower enzyme levels and decreased metabolic activity, resulting in higher plasma drug level30.
The purpose of this systematic review was to determine which drugs cause pharmacokinetic drug-drug interactions mediated by the CYP3A4 isoenzyme and to determine the pharmacokinetic mechanism of drug interactions mediated by the CYP3A4 isoenzyme based on the results of a search for journal articles published 2010–2020. The focus was on drug interactions mediated by CYP3A4 because it is the most highly expressed form in the liver31. CYP3A4 is a Phase I enzyme found in large quantities in the liver and intestines which metabolizes approximately 50% of therapeutic drugs32. The pharmacokinetic drug-drug interactions mediated by CYP3A4 revealed by the research results are the mechanisms of induction, activation, inhibition, and inactivation.
Induction and Activation:
Drug interactions through CYP3A4 induction can lead to decreased bioavailability (Table 1) and toxic effects (Table 2). Table 1 lists CYP3A4 inducers and activators that cause a decrease in bioavailability.
Table 1. CYP3A4 inducers and activators that cause a decrease in drug bioavailability
Drug Name(s) |
Test Media |
Description |
Author (Year) |
Carbamaze-pine |
HepG2 cells, primary cultured human hepatocytes |
Inducer CYP3A4 via HDAC1 |
Wu et al . (2012)33 |
Mitotane |
Human hepatocytes |
Inducer of CYP3A4 via SXR activation |
Takeshita et al. (2013)34 |
Metolazone |
Human hepatocytes, human intestinal cells |
Inducer of CYP3A4 via hPXR activation |
Banerjee and Chen, (2014)35 |
Aflutinib |
Human liver microsomes |
Strong inducer of CYP3A4 |
Liu et al. (2020)36 |
Derivatives of acridinone (C-1305 and C-1311) |
HCT116 cells (colon cancer cells) |
Very strong activator of CYP3A4 and UGT1A10 |
Pawlowska et al. (2020)37 |
HDAC1, histone deacetylase 1; CYP, cytochrome P450, SXR, steroid and xenobiotic receptor; hPXR, human pregnane X receptor; UGT1A10, UDP-glucuronosyltransferase family 1 member A10)
As seen in Table 1, carbamazepine33, mitotane34, metolazone35, aflutinib36, and the acridinone derivatives C-1305 and C-131137 induce the expression of CYP3A4 or activate CYP3A4 activity. CYP3A4 occurs through several mechanisms. For example, its induction by carbamazepine is mediated by histone deacetylase 1 (HDAC1)33 and its induction by mitotane34 and metolazone35 is mediated by the steroid and xenobiotic receptor (SXR), commonly called the human pregnane X receptor (hPXR). An inducer of a metabolizing enzyme (i.e., CYP) increases the synthesis of that enzyme. The combination of an inducing drug with drugs that are CYP3A4 substrates can increase the rate of substrate drug metabolism, so that drug levels and efficacy decrease32.
The systematic review revealed drug-drug interactions that cause hepatotoxicity due substrate drugs being combined with CYP3A4 inducers. As seen in Table 2, certain combinations of drugs with CYP3A4 inducers cause toxic effects.
Table 2. Combinations of drugs with CYP3A4 inducers that cause toxic effects
Drug name |
Test media |
Mechanism of interaction |
Author (Year) |
Clozapine |
Human liver microsomes |
CYP3A4 as the major enzyme in the bioactivation of clozapine that causes hepatotoxicity |
Dragovic et al. (2013)38 |
Saquinavir and rifampicin (CYP3A4 inducers) |
Mice, human liver microsomes |
Formation of two metabolites (α-hydroxyaldehyde and a primary amine); a three-fold increase in the toxicity of saquinavir |
Li et al. (2014)39 |
The inhibition or induction of enzymes in the CYP system, particularly CYP3A4, plays an important role in drug interaction pharmacokinetics by altering the concentration of the drug or toxic metabolite in the target tissue18. CYP induction can also lead to the formation of toxic reactive metabolites32. As seen in Table 2, the combination of clozapine and CYP3A4 inducers forms reactive metabolites38. Thus, CYP3A4 is the main enzyme involved in clozapine bioactivation, with an eight-fold increase in the reactive metabolite that causes hepatotoxicity. In addition, drug-drug interactions occur between saquinavir and the CYP3A4 inducer rifampicin. The combination of saquinavir with a CYP3A4 inducer increases the formation of the reactive metabolite α-hydroxyaldehyde(M229), which can increase the toxicity of saquinavir39. From these studies, it is known that toxic effects can occur due to the formation of reactive metabolites from drugs combined with CYP3A4 inducers.
Inhibition:
Pharmacokinetic drug interactions mediated by CYP3A4 may also occur through inhibition. Inhibition of CYP3A4 activity is divided into two main mechanisms, namely reversible inhibition and mechanism-based inactivation (including quasi-irreversible and irreversible inhibition). Reversible inhibition is classified as competitive, noncompetitive, and mixed32. Table 3 shows the mechanism of pharmacokinetic drug-drug interaction through reversible inhibition of CYP3A4.
Table 3. Inhibition of CYP3A4 through a reversible mechanism
Drug Name(s) |
Test Media |
Description |
Author (Year) |
Competitive reversible inhibition of CYP3A4 and other enzymes |
|||
Clarithromycin |
Human hepatocyte suspension |
Weak inhibitor of CYP3A4 and CYP3A5 |
Michaud and Turgeon (2010)40 |
Artemisinin (antimalarial) |
Human liver microsomes |
Strong inhibitor of CYP2B6 and weak inhibitor of CYP3A4 |
Xing et al. (2012)41 |
Ethambutol |
Human liver microsomes |
Strong inhibitor of CYP1A1 and CYP2E1 and a moderate inhibitor of CYP2A6, CYP2C9, and CYP3A4 |
Lee et al. (2014)42 |
MDEA and DDEA |
Human liver microsomes |
Inhibitor of CYP3A4 |
McDonald et al. (2015)43 |
AM-2201 synthetic cannabinoid (narcotic) |
Human liver microsomes |
Potent inhibitor of CYP3A4, CYP2C9, UGT1A3, and UGT2B7 |
Kim et al. (2017)44 |
Noncompetitive reversible inhibition of CYP3A4 and other enzymes |
|||
Norendoxifen |
Incubation microsomes |
Noncompetitive inhibitor of CYP3A19, CYP3A19, and CYP2 and competitive inhibitor of CYP2A6 and CYP2C19 |
Liu et al. (2013)45 |
MAM-2201 synthetic cannabinoid (narcotics) |
Human liver microsomes |
Noncompetitive inhibitor CYP3A4, competitive inhibitor of CYP2C9, potent inhibitor of CYP2C8, and reversible inhibitor of UGT1A3 |
Kong et al. (2017)46 |
Asenapine (atypical antipsychotic) |
Microsomes,human liver supersomes |
Weak noncompetitive inhibitor of CYP3A4, mixed-mechanism CYP1A2 inhibitor, and competitive CYP2D6 inhibitor |
Wojcikowski et al. (2020)47 |
Reversible inhibition of CYP3A4 via a mixed mechanism |
|||
Levomepromazine (phenothiazine neuroleptic) |
Human liver microsomes |
Competitive inhibitor of CYP3A4 and CYP2D6; mixed-mechanism inhibitor of CYP13A4 |
Basińska-Ziobroń et al. (2015)48 |
MDEA, mono-N-desethylamiodarone; DDEA, N-didesethylamiodarone; AM-2201, [1-(5-Fluoropentyl)-1H-indol-3-yl](1-naphthyl)methanone; MAM-2201, 4'-methyl-AM-2201
As seen in Table 3, several CYP3A4 inhibitors use a reversible mechanism. Competitive inhibitors compete for the active site of the enzyme with the substrate32. Drugs that are competitive inhibitors of CYP3A4 include clarithromycin40, artemisinin41, ethambutol42, mono-N-desethylamiodarone (MDEA), N-didesethylamiodarone (DDEA)43, and [1-(5-Fluoropentyl)-1H-indol-3-yl](1-naphthyl)methanone (AM-2201)44. Noncompetitive inhibition occurs when the drug binds to the enzyme at a site other than the active site and does not compete for the active site32.Noncompetitive CYP3A4 inhibitors include norendoxifen45, the synthetic cannabinoid 4'-methyl-AM-2201 (MAM-2201)46, and asenapine47. In the case of mixed inhibition, the inhibitor and substrate can bind to the active site of the enzyme simultaneously32.CYP3A4 inhibition by a mixed mechanism occurs with levomepromazine. In the case of mixed inhibition, the Km (values of Michaelis-Menten constants) and Vmax (the maximum velocities of the reactions) values changed at different inhibitors concentrations. In the case of competitive inhibition, the Vmax value did not significantly change, while the Km was altered at different inhibitor concentrations. The interaction between levomepromazine and CYP3A4 substrates, such as benzodiazepines, CCB antagonists, macrolides, and testosterone, needs carefully monitored48.
Mechanism-based CYP3A4 inhibition depends on time, concentration, and nicotinamide dinucleotide phosphate (NADPH). Time-dependent inhibition of CYP refers to a change in potency during an in vitro incubation or in vivo. Concentration-dependent inhibition is drug concentration in the in vitro or in vivo after a sufficient of incubation time. Table 4 describes the mechanism of pharmacokinetic drug-drug interactions through mechanism-based CYP3A4 inhibition.
Table 4. Mechanism-based CYP3A4 inhibition
Drug Name(s) |
Test Media |
Description |
Author (Year) |
|||
Time-dependent inhibitors of CYP3A4 |
||||||
Oncology drugs (docetaxel, dasatinib, erlotinib, everolimus, gefitinib, imatinib, lapatinib, nilotinib, pazopanib, sirolimus, sorafenib, sunitinib, and temsirolimus) |
Human liver microsomes |
Inhibitors of CYP3A (CYP3A4 and CYP3A5) |
Kenny et al. (2012)49 |
|||
Sertraline (selective serotonin reuptake inhibitor) |
Human liver microsomes |
Time-dependent inhibitor of CYP3A4 |
Masubuchi and Kawaguchi (2013)50 |
|||
Propiverine (antimuscarinic) |
Human liver microsomes |
Time-dependent inhibitor of CYP3A4 and CYP2D6 |
Dahlinger et al. (2017)51 |
|||
Panobinostat |
Human liver microsomes |
Competitive inhibitor of CYP2D6, time-dependent inhibitor of CYP3A4/5, and weak inhibitor of CYP2C19 |
Einolf et al. (2017)52 |
|||
Concentration-dependent inhibitors of CYP3A4 |
||||||
Nateglinide |
Human liver microsomes |
Very weak inhibitor of CYP1A1/2, CYP2C9, CYP2D6, CYP2E1, and CYP3A4 |
Takanobashi et al. (2010)53 |
|||
Tanespimycin and 17-amino-17-demethoxygeldanamycin |
Human liver microsomes |
Moderate inhibitor of CYP3A4/5 and CYP2C19 |
Gan et al. (2011)54 |
|||
β-lapachone (candidate anticancer) |
Human liver microsomes |
Inhibitors of CYP1A2, CYP2A6, CYP2C9, CYPC19, CYP2D6, and CYP3A4 |
Kim et al. (2013)55 |
|||
Reversible inhibition CYP3A4 via a mixed mechanism |
||||||
Levomepromazine (Phenothiazine neuroleptic) |
Human liver microsomes |
Competitive inhibitor of CYP2D6, mixed-mechanism inhibitor of CYP1A2 and CYP3A4 |
Basińska-Ziobroń et al. (2015)48 |
|||
Table 5: Drugs as CYP3A4 inhibitors
Drug Name(s) |
Test Media |
Description |
Author (Year) |
INH, PAS, and macrolides |
Human hepatocytes (HepaRG cells) |
Weak inhibitors of CYP3A4 |
Horita and Doi (2014)56 |
5α-androstane-3α, 17β-diol androgen derivatives |
Recombinant cell-enzymatic dibenzylfluoroscein |
Weak inhibitor of CYP3A4 and CYP2D6 |
Ayan et al. (2014)57 |
Gevokizumab |
Human hepatocytes |
Attenuates, but does not abolish, IL-1β-mediated functional repression of CYP3A4 |
Moreau et al. (2017)58 |
INH, isoniazid/isonicotinic acid hydrazide; PAS, para-aminosalicylic acid;
*Study did not explain the mechanism of inhibition of CYP3A4
As seen in Table 4, time-dependent CYP3A4 inhibitors include oncological drugs (such as docetaxel, dasatinib, erlotinib, everolimus, imatinib, lapatinib, nilotinib, pazopanib, sirolimus, sorafenib, sunitinib, and temsirolimus)49, sertraline50, propiverine51, and panobinostat52. Concentration-dependent inhibitors of CYP3A4 include nateglinide53,tanespimycin, 17-amino-17-demethoxygeldanamycin54, and β-lapachone55.
Several drugs have not been studied further regarding the mechanism of CYP3A4 inhibition because they are weak inhibitors of CYP3A4 and the potential for drug-drug interactions is not clinically significant. These drugs include isoniazid (isonicotinic acid hydrazide, INH), para-aminosalicylic acid (PAS), macrolide antibiotics56, the androgen derivative 5α-androstane-3α, 17β-diol57, and gevokizumab58. Seen in Table 5 is a list of drugs that are CYP3A4 inhibitors (Table 5).
Irreversible Inhibition or Irreversible Inactivation:
Irreversible inhibition of CYP3A4 due to inactivation or enzyme complexation occurs when some therapeutic drugs are converted by CYP enzymes into reactive metabolites capable of covalently binding to the CYP3A4 protein or heme moiety32. The drugs listed in Table 6 cause CYP3A4 inactivation.
Irreversible inhibition leads to time-dependent loss of CYP3A4 activity and cannot be recovered over the desired time span32. As seen in Table 6, irreversible inhibitors or CYP3A4 inactivators include mertansine59, 2-methylindole (2-MI)60, zolpidem61, amiodarone, dronedarone, N-desethylamiodarone (NDEA), and N-desbutyldronedarone (NDBD) combined with rivaroxaban62.
Table 6: Drugs that cause irreversible inhibition or inactivation of CYP3A4
Drug Name(s) |
Test Media |
Description |
Author (Year) |
Inactivation of CYP3A4 through an irreversible mechanism |
|||
Mertansine (cancer therapy) |
Human liver microsomes |
Irreversible inhibitor of CYP3A4, competitive inhibitor of CYP2D6, CYP2C8, UGT1A3, UGT1A4, and UGT1A1 |
Choi et al. (2020)59 |
2-MI |
Human liver microsomes, Sprague–Dawley rats |
Inactivation of CYP3A (CYP3A4 and CYP3A5) due to reactive intermediates irreversibly forming covalent bonds |
Wong et al. (2010)60 |
Zolpidem |
Human liver microsomes |
Inactivation of CYP3A (CYP3ACYP3A4 and CYP3A5) irreversibly |
Polasek et al. (2010)61 |
Amiodarone, dronedarone, NDEA, and NDBD-Rivaroxaban (substrate probe CYP3A4) |
Supersomes with human recombinant CYP |
Inhibitor reversibly and inactivator based on the mechanism of irreversible CYP3A4 |
Cheong et al. (2017)62 |
Inactivation of CYP3A4-based mechanisms |
|||
SCH 66712 (human dopamine D4 receptor antagonist) and EMTPP |
Human CYP3A4 recombinant |
Inactivation potent of CYP3A dependent of concentration, time, and NADPH |
Bolles et al. (2014)63 |
Dronedarone (antiarrhythmic agent), NDBD |
Human liver microsomes |
Mechanism-based inactivation of CYP3A4 and CYP3A5 |
Hong et al. (2016)64 |
C-1311 (antitumor) |
Human recombinant cells |
Selective mechanism-based CYP1A2 and CYP3A4 inactivators |
Potęga et al. (2016a)65 |
C-1305 (antitumor) |
Human recombinant cells |
Selective mechanism-based CYP1A2 and CYP3A4 inactivators |
Potęga et al. (2016b)66 |
2-MI, 2-methylindole; NDEA, N-desethylamiodarone (NDEA), NDBD, N-desbutyldronedarone, SCH 66712, 5-fluoro-2-[4-[(2-phenyl-1H-imidazol-5-yl)methyl]-1-piperazinyl]pyrimidine; EMTPP, 1-[(2-ethyl-4-methyl-1H-imidazol-5-yl)methyl]-4-[4-(trifluoromethyl)-2-pyridinyl]piperazine; NADPH, nicotinamide dinucleotide phosphate
Table 7: Drug interactions with CYP3A4 inhibitor and inactivator
Drug Name(s) |
Test Media |
Description |
Author (Year) |
Arteether α/β-ketoconazole (CYP3A4 inhibitors) |
Liver of healthy Swiss mice |
Ketoconazole (a potent inhibitor of CYP3A4) slows Arteether α/β metabolism and prolongs increases in blood plasma drug levels |
Tripathi et al. (2013)67 |
Pioglitazone-domperidone (CYP3A4 inhibitor) |
Human liver microsomes |
Pioglitazone inhibits domperidone metabolism depending on concentration and time |
Youssef et al. (2014)68 |
Sunitinib (tyrosine kinase inhibitor)-Ketoconazole (CYP 3A4 inhibitor) |
Mice (liver, kidney, and brain tissue) |
Ketoconazole inhibits metabolism sunitinib |
Chee et al. (2015)69 |
Verapamil-HCPT |
Sprague–Dawley rats |
Verapamil inhibits the activity of CYP3A4 and P-gp, increases the plasma peak, and decreases the oral clearance |
Xing et al. (2020)70 |
Bedaquiline-CYP3A4 |
Plasma from Wistar rats |
Ciprofloxacin and fluconazole inhibit the pharmacokinetics of bedaquiline |
Kotwal et al. (2019)71 |
Triazolam-diltiazem |
Mechanistic physiology (gut and liver) |
Diltiazem (CYP3A4 inactivator) competitively increases triazolam levels depending on time |
Yeo et al. (2010)72 |
HCPT, hydroxycamptothecin
Mechanism-based CYP3A4 inactivators include 5-fluoro-2-[4-[(2-phenyl-1H-imidazol-5-yl)methyl]-1-piperazinyl]pyrimidine (SCH 66712), 1-[(2-ethyl-4-methyl-1H-imidazol-5-yl)methyl]-4-[4-(trifluoromethyl)-2-pyridinyl]piperazine (EMTPP)63, dronedarone, and NDBD64. Long-term use the of the acridinone derivatives 5-dimethylaminopropylamino-8-hydroxytriazoloacridinone (C-1305) and 5-diethylaminoethylamino-8-hydroxyimidazoacridinone (C-1311) in colon cancer cells (HCT116) increases CYP3A4 activity37, indicating that C-1305 and C-1311 are strong inducers of CYP3A4. In contrast to the research of Potęga et al., which uses a non-cellular system65,66, this study revealed that C-1305 and C-1311 are selective mechanism-based inactivators of the ability of CYP3A4 to catalyze 6β-hydroxylation of testosterone (Table 6). It can be concluded that inhibition of the catalytic activity of CYP3A4 is not generated through a direct interaction between the compound and the CYP enzyme. Inhibition of the catalytic activity of CYP3A4 by C-1305 and C-1311 through cellular regulatory pathways is stronger than the direct inhibitory feature of the drug37. Drug interactions with CYP3A4 inhibitors or inactivators in are shown in Table 7.
As seen in Table 7, there are interactions between the following drugs and inhibitors of CYP3A4: Arteether α/β67, pioglitazone68, sunitinib69, hydroxycamptothecin (HCPT)70, and bedaquiline71. The combination of these drugs with CYP3A4 inhibitors causes an increase in peak plasma levels and a decrease in oral clearance70, thereby prolonging increases in blood plasma levels of the drug67. Interactions between triazolam and the CYP3A4 inactivator diltiazem also occur, competitively inactivating CYP3A4 expression and leading to a time-dependent increase in plasma triazolam levels72.
CONCLUSION:
The systematic review of the literature reveals pharmacokinetic drug interactions that have been tested preclinically. Drugs can interact with other drugs through CYP3A4 inhibition or inactivation, leading to increased bioavailability or increased plasma drug levels and longer drug clearance times. Likewise, CYP3A4 induction or activation can lead to an increase in CYP3A4 activity, resulting in decreased bioavailability and loss of drug efficacy. The toxic effects of drugs can be caused by inhibition or increased expression of CYP3A4 through the formation of reactive metabolites. The use of combinations of drugs metabolized by CYP3A4 needs to be monitored carefully because it can cause adverse drug interactions.
CONFLICT OF INTEREST:
The authors have no conflicts of interest regarding this investigation.
ACKNOWLEDGMENTS:
The authors would like to thank Sebelas Maret University for kind support during research.
REFERENCES:
1. Haque S, Alam NN, Ahmed M, Sultana N, Mushroor S. Detection of Drug Interactions by using an Automated Tool-A Prospective Study. Research Journal of Pharmacy and Technology. 2018;11(6):2345-2350.
2. Jagadeesan M, Manikandan R, Sudha NSS. The Drug-Drug Interactions: Affecting the Rationality of Prescriptions. Research Journal of Pharmacy and Technology. 2018;11(7):3077-3080.
3. Jose C, Antony M, Kumar JG, Maheswari C, Venkatanarayanan R, Chander SJU. Conceptual and Practical Update on Drug-Drug Interactions. Research Journal of Pharmacy and Technology. 2016;9(1):60-68.
4. Kee JL, Hayes ER. Pharmacology: Nursing Process Approach. Jakarta: EGC; 1996.
5. Gitawati R. Drug interactions and some of their implications. Health Research and Development Media. 2008;18(4):175-184.
6. Rastogi H, Sharma G, Sharma A, Jana S. In-Vitro Assessment of the Inhibitory effects of Dietary flavones on the regulated metabolism of CYP 450: Potential for Herb-drug interactions. Research Journal of Pharmacy and Technology. 2020; 13(12): 6086-6092.
7. Shahnaz H, Arayne MS, Sultana M, Haider A. In vitro drug interaction studies of Fexofenadine with Enoxacin, Levofloxacin and Sparfloxacin. Asian Journal of Research in Chemistry. 2012; 5(5): 687-696.
8. Murthy TEGK, Mayuren C. Effect of Ramipril on the Pharmacodynamics of Gliclazide in Diabetic Rats. Research Journal of Pharmacy and Technology. 2009;2(1):120-122.
9. Nitin M, Firdous A, Begum A, Sana S. Pharmacodynamic Drug Interaction of Ethionamide with Glibenclamide in Normal and Diabetic Rats. Research Journal of Pharmacology and Pharmacodynamics. 2013; 5(4): 227-231.
10. Nitin M, Krunal S, Rooman H, Girish M, Chetan M. Pharmacodynamic Drug Interaction of Imipramine with Glibenclamide in Normal Rabbits. Research Journal of Pharmacology and Pharmacodynamics. 2011; 3(3): 129-133.
11. Mutalib NA, Rafi MAAM, Latip NA. Revisiting CYP2C9-Mediated drug-drug Interactions: A Review. Research Journal of Pharmacy and Technology. 2021;14(11):6166-2.
12. Siddiqui MAH, Roshan S. Pharmacokinetic Drug Interactions between HMG-COA Reductase Inhibitors Fluvastatin with Concomitantly administered Ticagrelor. Research Journal of Pharmacy and Technology. 2018; 11(1): 179-182
13. Palleria C, Di Paolo A, Giofrč C, Caglioti C, Leuzzi G, Siniscalchi A, et al. Pharmacokinetic drug – drug interaction and their implication in clinical management. Journal of Research in Medical Sciences. 2013; 18(7): 601-610.
14. Levęque D, Lemachatti J, Nivoix Y, Coliat P, Santucci R, Ubeaud-Sequier G, et al. Mechanisms of pharmacokinetic drug-drug interactions. La Revue De Medecine Interne. 2010; 31(2): 170-179.
15. Pelkonen O, Mäeenpäeä J, Taavitsainen P, Rautio A, Raunio H. Inhibition and induction of human cytochrome P450 (CYP) enzymes. Xenobiotica. 1998; 28(12): 1203-1253.
16. Walsky RL, Obach RS. Validated assays for human cytochrome P450 activities. Drug Metabolism and Disposition. 2004; 32(6): 647-660.
17. Guengerich FP. A history of the roles of cytochrome P450 enzymes in the toxicity of drugs. Toxicological Research, 2020; 1-23.
18. Macit C, Taner N, Ozbay L, Behzetoglu Y, Mercanoglu G. Clinical impacts of CYP3A4 isoenzyme on drugs used in treatment of cardiovascular diseases: drug-drug interactions. Cardiology and Cardiovascular Medicine, 2016; 1(1): 43-49.
19. Nuralih N, Churiyah C, Pambudi S, Tamat SR, Meila O. Effect of ethanol extract of mulberry leaves (Morus Alba L.) with glibenclamide on CYP3A4 gene expression in HepG2 cell culture. Pharmacon: JurnalFarmasi Indonesia. 2019; 15(1): 29-36.
20. Marks DB, Marks AD, Smith CM. Basic medical biochemistry: a clinical approach, Jakarta: EGC; 2000.
21. Guengerich FP, Waterman MR, Egli M. Recent structural insights cytochrome P450 function. Trends in Pharmacological Sciences. 2016; 37(8): 625-640.
22. Zanger UM, Schwab M. 2013, Cytochrome P450 enzymes in drug metabolism, regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacology and Therapeutics. 2013; 138(1): 103-141.
23. Dewi CAK, Athiyah U, Mufarrihah YN. Drug therapy problems in patients receiving polypharmacy prescriptions, Jurnal Farmasi Komunitas, 2014;1(1):18-23.
24. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ (Clinical Research ed.). 2021;10(89):1-11.
25. Siddaway AP, Wood AM, Hedges LV. How to do a systematic review, a best practice guide for conducting and reporting narrative reviews, meta-analyses, and meta-synthesis. Annual Review of Psychology. 2019;70(1):747–770.
26. Singh S. How to conduct and interpret systematic reviews and meta analyses. Clinical and Translational Gastroenterology. 2017; 8(5): e93.
27. Faggion CM. Guidelines for reporting pre-clinical in vitro studies on dental materials. Journal of Evidence-Based Dental Practice. 2012; 12(4):182–189.
28. Sayedpour S, Khodaei B, Loghman AH, Sayedpour N, Kisomi MF, Balibegloo M, et al. Targeted theraphy strategies against SARS-CoV-2 cell entry mechanism: a systematic review of in vitro and in vivo studies. Journal Cellular Physiology. 2020; 236, 2364-2392.
29. Hooijmans CR, Rovers MM, de Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE's risk of bias tool for animal studies. BMC Medical Research Methodology. 2014; 14: 43.
30. Guengerich FP. Role off cytochrome P450 enzymes in drug-drug interaction. Advances in Pharmacology. 1997; 43:7-35.
31. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism, regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacology and Therapeutics. 2013; 138(1): 103-141.
32. Zhou SF. Drug behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. Current Drug-Drug Metabolism. 2008; 9(4): 310-322.
33. Wu Y, Shi X, Liu Y, Zhang X, Wang J, Luo X, et al. Histone deacetylase 1 is required for carbamazepine-induced CYP3A4 expression. Journal of Pharmaceutical and Biomedical Analysis. 2012;5m8:78–82.
34. Takeshita A, Igarashi-Migitaka J, Koibuchi N, Takeuchi Y. 2013, Mitotane induces CYP3A4 expression via activation of the steroid and xenobiotic receptor. Journal of Endocrinology. 2013; 216(3): 297–305.
35. Banerjee M, Chen T. thiazide-like diuretic drug metolazone activates human pregnane X receptor to induce cytochrome 3A4 and multidrug-resistance protein 1. Biochemical Pharmacology. 2014; 92(2): 389-402.
36. Liu XY, Guo ZT, Chen ZD, Zhang YF, Zhou JL, Jiang Y, et al. Alflutinib (AST2818), primarily metabolized by CYP3A4, is a potent CYP3A4 inducer. Acta Pharmacologica Sinica, 2020; 41(10): 1366-1376.
37. Pawłowska M, Kwaśniewska A, Mazerska Z, Augustin E. Enhanced activity of P4503A4 and UGT1A10 induced by acridinone derivatives C-1305 and C-1311 in MCF-7 and HCT116 cancer cells, consequences for the drugs cytotoxicity, metabolism and cellular response. International Journal of Molecular Sciences. 2020; 21(11): 3954.
38. Dragovic S, Gunness P, Ingelman-Sundberg M, Vermeulen NP, Commandeur JN. Characterization of human cytochrome P450s involved in the bioactivation of clozapine. Drug Metabolism and Disposition. 2013; 41(3): 651-658.
39. Li F, Lu J, Ma X. CPY3A4-mediated-hydroxyaldehyde formation in saquinavir metabolism. Drug Metabolism and Disposition, 2014; 42(2): 213–220.
40. Michaud V, Turgeon J. Assessment of competitive and mechanism-based inhibition by clarithromycin, use of domperidone as a CYP3A probe-drug substrate and various enzymatic sources including a new cell-based assay with freshly isolated human hepatocytes. Drug Metabolism Letters. 2010; 4(2): 69–76.
41. Xing J, Kirby BJ, Whittington D, Wan Y, Goodlett DR. Evaluation of P450 inhibition and induction by artemisinin antimalarials in human liver microsomes and primary human hepatocytes. Drug Metabolism and Disposition. 2012; 40(9):1757-1764.
42. Lee SY, Jang H, Lee JY, Kwon KI, Oh SJ, Kim SK. Inhibition of cytochrome P450 by ethambutol in human liver microsomes. Toxicology Letters. 2014; 229(1), 33-40.
43. Mcdonald MG, Au NT, Rettie AE. P450-based drug-drug interactions of amiodarone and its metabolites, diversity of inhibitory mechanisms. Drug Metabolism and Disposition. 2015; 43(11): 1661–1669.
44. Kim J, Kwon S, Kong TY, Cheong JC, Kim HS, In MK, et al. AM-2201 inhibits multiple cytochrome P450 and uridine 5`diphospho-glucuronosyltransferase enzyme activities in human liver microsomes. Molecules. 2017; 22(443):1-11.
45. Liu J, Flockhart PJ, Lu D, Lu W, Lu WJ, Han X, et al. Inhibition of cytochrome P450 enzymes by the E- and Z-isomers of norendoxifen. Drug Metabolism and Disposition. 2013; 41(9): 1715–1720.
46. Kong TY, Kim JH, Kwon SS, Cheong JC, Kim HS, In MK, Lee HS. Inhibition of cytochrome P450 and uridine 5-diphospho-glucuronosyltransferases by MAM-2201 in human liver microsomes. Archives of Pharmacal Research. 2017; 40(6), 727–735.
47. Wójcikowski J, Danek PJ, Basińska-Ziobroń A, Pukło R, Daniel WA. In vitro inhibition of human cytochrome P450 enzymes by the novel atypical antipsychotic drug asenapine, a prediction of possible drug–drug interactions. Pharmacological Reports. 2020; 72(3): 612-621.
48. Basińska-Ziobroń A, Daniel WA, Wójcikowski J. Inhibition of human cytochrome P450 isoenzymes by a phenothiazine neuroleptic levomepromazine: an in vitro study. Pharmacological Reports. 2015 ;67(6): 1178-1182.
49. Kenny JR, Mkadam S, Zhang C, Tay S, Collins C, Galetin A, et al. Drug–drug interaction potential of marketed oncology drugs: in vitro assessment of time-dependent cytochrome P450 inhibition, reactive metabolite formation and drug–drug interaction prediction. Pharmaceutical Research. 2012; 29(7): 1960-1976.
50. Masubuchi Y, Kawaguchi Y. Time-dependent inhibition of CYP3A4 by sertraline, a selective serotonin reuptake inhibitor. Biopharmaceutics and Drug Disposition. 2013; 34(8): 423-430.
51. Dahlinger D, Aslan S, Pietsch M, Frechen S, Fuhr U. Assessment of inhibitory effects on major human cytochrome P450 enzymes by spasmolytics used in the treatment of overactive bladder syndrome. Therapeutic Advances In Urology. 2017; 9(7): 163-177.
52. Einolf HJ, Lin W, Won CS, Wang L, Gu H, Chun DY, et al. Physiologically based pharmacokinetic model predictions of panobinostat (LBH589) as a victim and perpetrator of drug-drug interactions. Drug Metabolism and Disposition. 2017; 45(12): 1304-1316.
53. Takanohashi T, Kubo S, Nakayama A, Mihara R, Hayashi M. Inhibition of human liver microsomal CYP by nateglinide. Journal of Pharmacy and Pharmacology. 2010; 62(5):592–597.
54. Gan J, Liu-Kreyche P, Humphreys WG. In vitro assessment of cytochrome P450 inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin. Cancer Chemotherapy and Pharmacology. 2011;69(1): 51–56.
55. Kim IS, Kim Y, Kwak TH, Yoo HH. 2013, Effects of β-lapachone, a new anticancer candidate, on cytochrome P450-mediated drug metabolism. Cancer Chemotherapy and Pharmacology. 2013; 72(3): 699-702.
56. Horita Y, Doi N. Comparative study of the effects of antituberculosis drugs and antiretroviral drugs on cytochrome P450 3A4 and p-glycoprotein. Antimicrobial Agents and Chemotherapy. 2014; 58(6): 3168-3176.
57. Ayan D, Maltais R, Hospital A, Poirier D. Chemical synthesis, cytotoxicity, selectivity and bioavailability of 5α-androstane-3α,17 β-diol derivatives. Bioorganic and Medicinal Chemistry. 2014; 22(21): 5847–5859.
58. Moreau A, Le VM, Jouan E, Denizot C, Parmentier Y, Fardel O. Effect of gevokizumab on interleukin-1β-mediated cytochrome P450 3A4 and drug transporter repression in cultured human hepatocytes. European Journal of Drug Metabolism and Pharmacokinetics. 2017; 42(5), 871-878.
59. Choi WG, Park R, Kim DK, Shin Y, Cho YY, Lee HS. Mertansine inhibits mRNA expression and enzyme activities of cytochrome P450s and uridine 5-diphospho-glucuronosyltransferases in human hepatocytes and liver microsomes. Pharmaceutics. 2020; 12(3): 220.
60. Wong SG, Fan PW, Subramanian R, Tonn GR, Henne KR, Johnson MG., et al. Bioactivation of a novel 2-methylindole-containing dual chemoattractant receptor-homologous molecule expressed on T-helper type-2 cells/d-prostanoid receptor antagonist leads to mechanism-based CYP3A inactivation, glutathione adduct characterization and prediction of in vivo drug-drug interaction. Drug Metabolism and Disposition. 2010; 38(5): 841–850.
61. Polasek TM, Sadagopal JS, Elliot DJ, Miners JO. In vitro-in vivo extrapolation of zolpidem as a perpetrator of metabolic interactions involving CYP3A. European Journal of Clinical Pharmacology. 2010; 66(3): 275–283.
62. Cheong EJY, Goh JJN, Hong Y, Venkatesan G, Liu Y, Chiu GNC, et al. Application of static modeling in the prediction of in vivo drug–drug interactions between rivaroxaban and antiarrhythmic agents based on in vitro inhibition studies. Drug Metabolism and Disposition. 2017; 45(3): 260-268.
63. Bolles AK, Fujiwara R, Briggs ED, Nomeir AA, Furge LL. Mechanism-based inactivation of human cytochrome P450 3A4 by two piperazine-containing compounds. Drug Metabolism and Disposition. 2014; 42(12): 2087-2096.
64. Hong Y, Chia YMF, Yeo RH, Venkatesan G, Koh SK, Chai CLL, et al. Inactivation of human cytochrome P450 3A4 and 3A5 by dronedarone and n-desbutyl dronedarone. Molecular Pharmacology. 2016; 89(1):1-13.
65. Potęga A, Fedejko-Kap B, Mazerska Z. Imidazoacridinone antitumor agent C-1311 as a selective mechanism-based inactivator of human cytochrome P450 1A2 and 3A4 isoenzymes. Pharmacological Reports. 2016a; 68(4): 663-670.
66. Potęga A, Fedejko-Kap B, Mazerska Z. Mechanism-based inactivation of human cytochrome P450 1A2 and 3A4 isoenzymes by antitumor triazoloacridinone C-1305. Xenobiotica. 2016b; 46(12): 1056-1065.
67. Tripathi R, Rizvi A, Pandey SK, Dwivedi H, Saxena JK. Ketoconazole, a cytochrome P450 inhibitor can potentiate the antimalarial action of α/β arteether against MDR Plasmodium YoeliiNigeriensis. Acta Tropica. 2013; 126(2): 150-155.
68. Youssef AS, Parkman HP, Nagar S. Domperidone interacts with pioglitazone but not with ondansetron via common CYP metabolism in vitro. Xenobiotica. 2014; 44(9): 792-803.
69. Chee ELC, Lim AYL, Modamio P, Fernandez-Lastra C, Segarra I. Sunitinib tissue distribution changes after coadministration with ketoconazole in mice. European Journal of Drug Metabolism and Pharmacokinetics. 2015; 41(3): 309-319.
70. Xing H, Luo X, Li Y, Fan C, Liu N, Cui C, et al. Effect of verapamil on the pharmacokinetics of hydroxycamptothecin and its potential mechanism. Pharmaceutical Biology. 2020; 58(1): 152-156.
71. Kotwal P, Magotra A, Dogra A, Sharma S, Gour A, Bhatt S, et al. Assessment of preclinical drug interactions of bedaquiline by a highly sensitive LC-ESI-MS/MS based bioanalytical method. Journal of Chromatography B. 2019; 1112: 48-55.
72. Yeo KR, Jamei M, Yang J, Tucker GT, Rostami-Hodjegan A. Physiologically based mechanistic modelling to predict complex drug-drug interactions involving simultaneous competitive and time-dependent enzyme inhibition by parent compound and its metabolite in both liver and gut - the effect of diltiazem on the time-course of exposure to triazolam. European Journal of Pharmaceutical Sciences. 2010; 39(5): 298–309.
Received on 24.07.2022 Modified on 04.10.2022
Accepted on 01.12.2022 © RJPT All right reserved
Research J. Pharm. and Tech 2023; 16(6):3016-3024.
DOI: 10.52711/0974-360X.2023.00498