Skip to main content
Acta Pharmaceutica Sinica. B logoLink to Acta Pharmaceutica Sinica. B
. 2016 Aug 9;6(5):413–425. doi: 10.1016/j.apsb.2016.07.016

Insights into CYP2B6-mediated drug–drug interactions

William D Hedrich 1, Hazem E Hassan 1, Hongbing Wang 1,
PMCID: PMC5045548  PMID: 27709010

Abstract

Mounting evidence demonstrates that CYP2B6 plays a much larger role in human drug metabolism than was previously believed. The discovery of multiple important substrates of CYP2B6 as well as polymorphic differences has sparked increasing interest in the genetic and xenobiotic factors contributing to the expression and function of the enzyme. The expression of CYP2B6 is regulated primarily by the xenobiotic receptors constitutive androstane receptor (CAR) and pregnane X receptor (PXR) in the liver. In addition to CYP2B6, these receptors also mediate the inductive expression of CYP3A4, and a number of important phase II enzymes and drug transporters. CYP2B6 has been demonstrated to play a role in the metabolism of 2%–10% of clinically used drugs including widely used antineoplastic agents cyclophosphamide and ifosfamide, anesthetics propofol and ketamine, synthetic opioids pethidine and methadone, and the antiretrovirals nevirapine and efavirenz, among others. Significant inter-individual variability in the expression and function of the human CYP2B6 gene exists and can result in altered clinical outcomes in patients receiving treatment with CYP2B6-substrate drugs. These variances arise from a number of sources including genetic polymorphism, and xenobiotic intervention. In this review, we will provide an overview of the key players in CYP2B6 expression and function and highlight recent advances made in assessing clinical ramifications of important CYP2B6-mediated drug–drug interactions.

Abbreviations: CAR, constitutive androstane receptor; C/EBP, CCAAT/enhancer-binding protein; CHOP, cyclophosphamide–doxorubicin–vincristine–prednisone; CITCO, (6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime); COUP-TF, chicken ovalbumin upstream promoter-transcription factor; CPA, cyclophosphamide; 4-OH-CPA, 4-hydroxycyclophosphamide; CYP, cytochrome P450; DDI, drug–drug interaction; DEX, dexamethasone; EFV, efavirenz; E2, estradiol; ERE, estrogen responsive element; GR, glucocorticoid receptor; GRE, glucocorticoid responsive element; HAART, highly active antiretroviral therapy; HNF, hepatocyte nuclear factor; IFA, Ifosfamide; MAOI, monoamine oxidase inhibitor; NNRTI, non-nucleotide reverse-transcriptase inhibitor; NR1/2, nuclear receptor binding site 1/2; NVP, nevirapine; PB, phenobarbital; PBREM, phenobarbital-responsive enhancer module; PCN, pregnenolone 16 alpha-carbonitrile; PXR, pregnane X receptor; RIF, rifampin; SNP, single nucleotide polymorphism; TCPOBOP, 1,4-bis[3,5-dichloropyridyloxy]benzene; UGT, UDP-glucuronosyl transferase

Key Words: CYP2B6, CAR, PXR, Polymorphism, Drug–drug interaction, Cyclophosphamide, Efavirenz

Graphical abstract

CYP2B6 is a highly inducible and polymorphic enzyme which plays a significant role in human drug metabolism. Variations in the expression and function of CYP2B6 significantly alter the metabolism and pharmacokinetics of many drugs. These alterations may result in significant drug–drug interactions which may lead to improved therapy or toxicity.

fx1

1. Introduction

The human cytochrome P450 (CYP) superfamily is made up of 18 families and 43 subfamilies containing 57 genes and 59 pseudogenes1, 2, 3. CYP2B6 is expressed primarily in the liver and represents one of the approximately fifteen CYP enzymes, distributed amongst P450 families 1–4, predominantly responsible for xenobiotic metabolism4, 5. Alongside CYP2B7, a related pseudogene, CYP2B6 is located on the long arm of chromosome 19 within a CYP2B cluster3, 6. Orthologs of the human CYP2B6 genes can be found in other species including rats, mice, and dogs, which are termed Cyp2b1, Cyp2b10, and CYP2B11, respectively3. Notably, unlike in other species, CYP2B6 is the only isozyme of the CYP2B subfamily with metabolic function in humans3.

Historically, CYP2B6 has been believed to be relatively inconsequential with respect to human xenobiotic metabolism7, 8. However, in recent years, the discovery of important substrates, robust chemical-mediated induction, and genetic polymorphisms of this CYP isozyme has triggered significant academic and industrial research interests. The number of drugs known to be metabolized by this enzyme has drastically increased since the development of effective monoclonal antibodies, the establishment of bupropion as a selective marker of CYP2B6 catalytic activity, and the utilization of recombinant DNA techniques9, 10. Current estimates indicate that CYP2B6 accounts for 2%–10% of total hepatic CYP content and is, in fact, involved in the metabolism of a significant number of drugs in humans, estimated to be around 8% of all commercially available drugs11, 12, 13, 14, 15. Known CYP2B6 substrates include but are not limited to a number of clinically utilized therapeutic agents such as cyclophosphamide (CPA), artemisinin, bupropion, ketamine, pethidine, propofol, methadone, nevirapine (NVP), and efavirenz (EFV) (Table 1), as well as endogenous chemicals and environmental compounds10, 13, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59.

Table 1.

Clinically utilized CYP2B6-substrate drugs.

Class Substrate Contribution of CYPs Ref.
Anesthetic Ketamine Major, CYP3A4; Minor, CYP2B6, 2C9 25, 29
Lidocaine Major, CYP2B6, 2A6; Minor, CYP2B6 30
Propofol Major, CYP2B6; Minor, CYP2C9 19, 31
Antiarrhythmic Mexiletine Major, CYP2A1; Minor, CYP2B6, 2E1 32
Anticoagulant Coumarins Major, CYP2B6; Minor, CYP2E1, 2C19 33, 34
Anticonvulsant Mephenytoin Major, CYP2B6; Minor, CYP2C9 21, 35
Antidepressant Bupropion Major, CYP2B6; Minor, CYP2D6, 3A4 10, 13, 26, 27, 28
Antiepileptic Mephobarbital Major, CYP2B6 36
Valproic Acid Major, CYP2A6; Minor, CYP2B6, 1A1 37
Anti-inflammatory Aminopyrine Major, CYP2B6, 2C19; Minor, CYP2C8, 2D6 38, 39
Antipyrine Major, CYP3A4, 2C; Minor, CYP2B6, 1A2 40
Tazofelone Major, CYP3A4; Minor, CYP2B6 41
Antimalarial Artemether Major, CYP2B6; Minor, CYP3A4 42
Artemisinin Major, CYP2B6; Minor, CYP3A4 18, 43
Antiretroviral Efavirenz Major, CYP2B6; Minor, CYP3A 23, 44, 45
Nevirapine Major, CYP2B6, 3A4; Minor, CYP2D6 46, 47
Chemotherapeutic Cyclophosphamide Major, CYP2B6; Minor, CYP3A4, 2C9 22, 48, 49, 50
Ifosfamide Major, CYP2B6, 3A4; Minor, CYP2C9, 2C19 49, 50, 51
Tamoxifen Major, CYP2E1, 2D6; Minor, CYP2B6, 3A4 52, 53
MAOI Selegiline Major, CYP2B6, 2C19 ; Minor, CYP3A4, 1A2 4, 54
Opioid Methadone Major, CYP2B6, 3A4 20, 55
Pethidine Major, CYP2B6; Minor, CYP3A4, 2C19 21
Psychotropic Clotiazepam Major, CYP2B6, 3A4; Minor, CYP2C18, 2C19 56
Diazepam Major, CYP2B6, 2C19; Minor, CYP3A4 57, 58, 59
Temazepam Major, CYP2B6; Minor, CYP2C, 3A 57, 59
Steroid Testosterone Major, CYP3A4; Minor, CYP2B6 30

Metabolism of the same compounds is often achieved by several CYP enzymes generating similar or various intermediate metabolites, which contribute to the biotransformation of substrates to different extents14, 60. In the case of CYP2B6, although it shares the same substrates with several other CYP enzymes, most notably CYP3A4, there are some biotransformation reactions for which CYP2B6 is the predominant or only known catalyst. For instance, CYP2B6 is the sole enzyme which mediates N-demethylation of mephobarbital, while the 4-hydroxylation biotransformation reaction of this molecule is mediated by the CYP2C family of enzymes36, 61. Further, it was shown that CYP2B6 is the only enzyme capable of mediating both O-demethylation and ortho-hydroxylation of the endocrine disruptor methoxychlor, while other P450 isoforms may contribute only to one biotransformation reaction62, 63, 64.

Predominantly expressed in the liver, CYP2B6 has been estimated to contribute to between 2% and 10% of the overall pool of microsomal P450s, with significant inter-individual variability9, 11, 12, 13, 59, 65, 66, 67. A major contributing factor to the variability observed in CYP2B6 expression and function is induction of the enzyme, which results in de novo synthesis of the protein after exposure to particular chemicals68. The constitutive androstane receptor (CAR, NR1I3) and the pregnane X receptor (PXR, NR1I2) are key modulators governing the inductive expression of CYP2B669, 70. Activation or inhibition of these receptors by known compounds including rifampin (RIF), phenobarbital (PB), dexamethasone (DEX), and phenytoin can have a significant impact on the downstream expression of important drug-metabolizing enzymes and drug transporters71, 72, 73. Studies have illustrated that selective activation of CAR over PXR provides preferential induction of CYP2B6 over CYP3A4, while activation of PXR induces both enzymes with less discernible differences70. Interestingly, the selective transcription of CYP2B6 over CYP3A4 by CAR may have clinical relevance with respect to drugs that are predominantly metabolized by CYP2B6, and activators of CAR may function as co-administered facilitators for such biotransformation74.

Expression of CYP2B6 exhibits significant inter- and intra-individual variability and up to 250-fold of CYP2B6 expression between individuals has been observed11, 12. The highly variable enzyme expression arises from multiple factors including genetic polymorphisms, non-genetic factors such as disease conditions, gender differences, and transcriptional induction or suppression by xenobiotics and cytokines14, 67. Though there are several sources, genetic polymorphisms and transcriptional gene regulation are believed to be the major contributors to the observed variability of CYP2B6 expression.

Single nucleotide polymorphisms (SNPs) within the CYP2B6 gene have been shown to be indicative of drug response and pharmacokinetics of administered CYP2B6-substrate drugs14, 75, 76. The most common such polymorphism is CYP2B6*6 (Q172H, K262R), which occurs at frequencies ranging from 15% to 60% amongst various populations and results in a functionally deficient allele22, 77, 78. To date, up to 63 alleles covering both coding and non-coding regions of CYP2B6 gene have been identified (http://www.cypalleles.ki.se/cyp2b6.htm), including more than 30 non-synonymous SNPs which result in amino-acid replacement.

Given the highly inducible and polymorphic nature of the CYP2B6 gene, dramatic individual variability in hepatic CYP2B6 expression has been recognized in humans. Such variation is closely associated with the variable systemic exposure and therapeutic response to a growing list of CYP2B6 substrates. This review is designed to discuss recent developments in areas which exemplify the potential for clinically significant drug–drug interactions (DDI) that arise from both pharmacological and genetic modulations of CYP2B6.

2. CYP2B6 polymorphisms

Although pharmacogenetics of genes encoding drug-metabolizing enzymes has been the subject of intensive studies for many years, only within the last ten years or so has the analysis of CYP2B6 genetic variations been examined and partially elucidated. The first systematic investigation of genetic polymorphism in the CYP2B6 gene was conducted by Lang et al.66 in 2001 using cDNA derived from 35 German Caucasians. This early study, with the focus on all exons, resulted in the identification of nine novel SNPs, of which five are nonsynonymous mutations in exon 1 (C64T, Arg22Cys), exon 4 (G516T, Gln172His), exon 5 (C777A, Ser259Arg and A785G, Lys262Arg) and exon 9 (C1459T, Arg487Cys) and four are silent mutations (C78T, G216C, G714A and C732T)66, 79. In 2003, a more comprehensive analysis of SNPs in the coding region, introns, or 5ʹ-flanking sequences of CYP2B6 gene from 80 DNA samples of Caucasian, African, and Hispanic Americans found 10 SNPs in the CYP2B6 promoter, seven in the coding region, and one in intron 365. With additional subsequent investigations, a much improved understanding of CYP2B6 genotype–phenotype associations has been achieved. Clearly, polymorphisms of CYP2B6 contribute significantly to a number of clinical important DDI.

2.1. Nonsynonymous SNPs

CYP2B6*6, defined by the 516G>T and 785A>G mutations, has been elucidated as the most clinically relevant polymorphism of CYP2B6. These particular mutations harboring two amino acid (Q172H and K262R) replacements result in decreased levels of expression and function of CYP2B6 protein80. CYP2B6*6 and the anti-HIV EFV probably represent the most convincing gene–drug pair in elucidating the clinical influence of CYP2B6 polymorphisms on drug administration. EFV is a widely used non-nucleoside reverse-transcriptase inhibitor (NNRTI) utilized as part of a highly active anti-retroviral therapy (HAART) for treatment of HIV-1 infections alongside emtricitabine and tenofovir within the Atripla regimen. Compared with other hepatic CYPs, CYP2B6 is the main catalyst of EFV primary and secondary metabolism81. Importantly, individuals expressing this variant of CYP2B6 have demonstrated significantly decreased rates of 8-hydroxylation of EFV and increased circulating plasma concentrations of the parent drug82. Many studies have explored the impact of the 516G>T polymorphism on EFV pharmacokinetics and have associated this mutation with elevated plasma levels resulting in neurotoxicity and CNS side effects83, 84, 85, 86, 87, liver injury88, and acquired drug resistance89, 90, 91. Genotyping for this particular CYP2B6 variant has been proposed as a method to aid in personalizing EFV dosages for individual patients. Genotyping would also assist in identifying individuals who may be classified as poor metabolizers or ultra-rapid metabolizers of EFV, and who may benefit from early therapeutic drug monitoring92, 93. A retrospective study reported that therapeutic drug monitoring and dose reduction in patients with the CYP2B6*6 homozygotes reduced the EFV plasma concentration from toxic levels back into normal therapeutic levels. This study further revealed that those patients with the homozygous CYP2B6*6 genotype receiving lower EFV doses experienced fewer adverse events following treatment, and increased the proportion of patients exhibiting an undetectable HIV viral load94. To date, multiple clinical studies consistently indicate that CYP2B6*6 is associated with high EFV plasma concentration and increased central nervous toxicity93. Thus, it is reasonable to speculate that implementation of CYP2B6 genotyping test clinically would benefit HIV-infected patients receiving an EFV-based regimen.

NVP, another NNRTI, has been associated with significant toxicities including, in some cases, life threatening rashes and/or hepatotoxicity during the early weeks of therapy95, 96. Similarly to EFV, the 516G>T polymorphism of CYP2B6 has been studied with respect to its impact on NVP pharmacokinetics. In individuals with the CYP2B6 *6/*6 or *6/*18 haplotype, NVP clearance is significantly decreased and circulating plasma concentrations are elevated97, 98, 99. The 983T>C nonsynonymous SNP has been shown to affect NVP pharmacokinetics in a similar manner97, 100. While literature regarding chemical alteration of CYP expression and its resulting impact on NVP disposition is currently lacking, it is expected that increased expression of CYP2B6 and CYP3A4 would increase the metabolism and clearance of NVP, potentially resulting in non-therapeutic plasma concentrations, while inhibition of these enzymes may result in increased circulating levels and potential serious toxicities.

Comparatively, the role of CYP2B6*6 in CPA application appears to be less convincing. CPA is an alkylating prodrug requiring hepatic bioactivation and a CYP2B6 substrate. To date, the impact of polymorphisms of CYP2B6 on hepatic metabolism of CPA remains a conflicted topic. It has been demonstrated in vitro that human livers expressing CYP2B6*6 exhibit markedly enhanced catalytic activity in CPA 4-hydroxylation although these individual samples also expressed comparatively low levels of CYP2B6 protein101. Studies performed by Xie et al.22 concluded that although there are differences in CYP2B6 protein expression and function, there is no significant difference in overall 4-hydroxylation of CPA between liver donors. However, clinically, it has been reported that CYP2B6*6 itself is a determinant of poor response to FC (fluradabine, CPA) therapy in the treatment of chronic lymphocytic leukemia102.

Like CPA, ifosfamide (IFA) is another commonly prescribed antitumor prodrug within the oxazaphosphorine class of alkylating agents. It is frequently utilized in the treatment of solid tumors and hematologic malignancies103. The bioactivation of IFA in the liver is catalyzed by multiple CYP isoforms with CYP2B6 and CYP3A4 being the most prevalent contributors to its metabolism; each isoform contributes roughly equivalently to the 4-hydroxylation of IFA to yield its active metabolite, 4-hydroxyifosfamide51, 104. Up to 20-fold interpatient differences have been reported in the pharmacokinetics of IFA and are likely attributable to pharmacogenetic differences105. CYP2B6*6 heterozygous and homozygous carriers have been linked with decreased catalytic activity and hepatic expression of CYP2B6 functional protein, increased IFA plasma concentrations, and increased toxicities as compared with reference phenotypes22, 105.

CYP2B6*5, designated by a C>T SNP in exon 9, has also been examined with respect to its impact on CPA22, 106. Lymphoma patients with this CYP2B6 variant have demonstrated significantly altered remission rates and clinical outcomes. Patients expressing the CYP2B6*1/*5 genotype exhibited an increased 2-year relapse rate and diminished overall survival as compared to those with the reference allele. Bachanova et al.106 suggested that the CYP2B6*5 variant is an independent indicator of a patient׳s chance of successful treatment when utilizing autologous hematopoetic cell transplantation and high dose CPA-based chemotherapy. Similar to CYP2B6*6, Caucasian female carriers of the CYP2B6*5 variant exhibit decreased protein expression which may result in decreased bioactivation of CPA65, 66, 107.

Additionally, CYP2B6*18, defined by the T983C SNP (I328T, exon 7), is found with relative frequency of 4%–12% in African populations, though not in Caucasians and Asians78, 108. This particular allele exhibits a loss of functional protein108, 109. This SNP was associated with a threefold increase in mean plasma EFV concentrations in African HIV patients110. When combined with the A785G SNP, these mutations together make up the CYP2B6*16 allele which has been associated with an even greater increase (5-fold) in mean plasma EFV concentrations, indicating a synergistic effect between the two SNPs110.

2.2. SNPs in the promoter region of CYP2B6

In addition to the identified genetic variations in the coding regions of the CYP2B6 gene, polymorphisms within the non-coding region may influence the overall expression of this gene65, 111, 112. Interestingly, some of the SNPs identified in the promoter region of CYP2B6 lie within the binding sites of several transcription factors113, 114. For example, the SNP at −2320T>C is located in a putative hepatocyte nuclear factor (HNF4)-binding site, the −750T>C and −575C>T are within binding sites for HNF1 and Sp-1, respectively, while the −82T>C generates a novel CCAAT-enhancer–binding protein α (C/EBPα) binding site65, 67, 111. Notably, the −82T>C substitution not only introduces a functional C/EBP binding site into the CYP2B6 promoter, but also shifts the transcriptional starting site approximately 30 base pairs (bp) downstream111. Further analysis revealed that livers genotyped –82T>C were associated with an approximately 2-fold higher CYP2B6 mRNA expression in comparison to the reference −82T/T carriers. In exploring whether this polymorphism could affect drug-induced expression of CYP2B6, Li et al.113 demonstrated a strong synergism between −82T>C mutation and the activation of PXR by ligand binding via cell-based reporter assays in HepG2 and Huh7 cells (Fig. 1). Mechanistic studies revealed that the −82-bound C/EBPα can interact with PXR and loops the PXR bound distal phenobarbital-responsive enhancer module (PBREM) toward the proximal CYP2B6 transcriptional start site. These findings suggest that individuals carrying −82T>C mutant might be hypersensitive to drugs that are CYP2B6 substrates when co-administered with PXR-type inducers. In the CYP2B6 promoter, the most frequent SNPs identified was the −750T>C mutation that occurred in close to 50% or more of all ethnic groups studied65. In a 2007 study, Nakajima et al.115 described the impact of the −750T>C substitution on CPA hydroxylation by CYP2B6. Patients possessing this SNP exhibited significantly decreased area under the concentration–time curve (AUC) ratios of 4-OH-CPA/CPA, indicating decreased enzyme activity. A decrease in CPA hydroxylation and bioactivation by this genetic mutation can significantly alter the potency of CPA and detection of this polymorphism may be valuable as an early predictor of adverse effects or diminished therapy115.

Figure 1.

Fig. 1

Synergistic activation of CYP2B6 reporter by −82T>C mutation and PXR activation. The SNP −82T>C introduced a C/EBPα-binding site in the CYP2B6 promoter (A). The presence of this mutation and RIF-mediated activation of PXR synergistically enhanced the transcriptional activity of CYP2B6 in both HepG2 (B) and Huh7 cells (C). (This figure was adopted from Li et al.113 with permission of the copyright holder, The American Society for Pharmacology and Experimental Therapeutics).

3. Transcriptional regulation of CYP2B6

Transcriptional regulation of CYP2B6 has been implicated as one of the major contributing factors to the observed inter- and intra-individual variations in the expression of this CYP isozyme. For many years, the inducibility of its rodent counterparts, particularly in mice and rats, has been studied as the model gene for the PB-mediated CYP induction phenomenon116, 117. However, significant species differences exist in the induction of CYP enzymes limiting the utility of these models for direct human extrapolation. For example, 1,4-bis[3,5-dichloropyridyloxy]benzene (TCPOBOP) and pregnenolone 16α-carbonitrile (PCN) are known to significantly induce CYP3A and CYP2B in rodents, but have no effects on related humans CYPs. On the other hand, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)-oxime (CITCO) and RIF potently induce human CYP2B6 and CYP3A4 without affecting the expression of their rodent counterparts118, 119, 120, 121.

In humans, cultured primary hepatocytes are widely accepted as the most appropriate in vitro model for assessing the induction of hepatic drug-metabolizing enzymes as nearly all immortalized hepatic cell lines express significantly lower levels of drug-metabolizing enzymes as well as key liver-enriched transcriptional factors122. In human primary hepatocyte (HPH) cultures, expression of CYP2B6 is well-maintained and robust induction in response to prototypical inducers has been observed. In fact, in certain liver donors, CYP2B6 can be induced to such a high level that is comparable to that of CYP3A4, which is widely accepted as the most abundant CYP isoform expressed in the human liver. Utilizing HPH as an in vitro model of the human liver, several known CYP3A4 and CYP2C inducers have been shown to simultaneously augment the expression of CYP2B669, 123, suggesting these CYP enzymes may share common transcriptional regulation mechanisms and coordinate a defensive hepatic responsive network to xenobiotic challenges.

3.1. CAR- and PXR-mediated induction of CYP2B6

Over the years, researchers have observed potent induction of CYP2B genes by barbiturates in the liver of many different species. The first study illustrating molecular mechanisms behind this induction came in 1995 using cultured adult rat hepatocytes124. A functional analysis of the 5ʹ flanking promoter region of rat Cyp2b1 and Cyp2b2 has linked PB-mediated induction to a 163-bp DNA sequence, termed the PB-responsive element (PBRE) or PB-responsive unit (PBRU), located −2155 to 2318 bp from the transcription start site of Cyp2b1/2124, 125. Subsequent investigations led to the localization of a 51-bp similar sequence named PBREM at positions of −2339/−2289 and −1733/−1683 within the promoters of mouse Cyp2b10 and human CYP2B6, respectively126, 127. Later, in the distal upstream region of the CYP2B6 gene, a xenobiotic-responsive enhancer module (XREM) located around −8.5 kb from the transcriptional start site of CYP2B6 was also identified and functionally evaluated128. Together, these response elements coordinate the optimal induction of CYP2B genes by PB-like inducers.

Another important milestone in our understanding of the mechanisms underlying drug-induced CYP2B expression was achieved when the nuclear receptor CAR was functionally linked to CYP2B transcription126. In HepG2 cell-based reporter assays, nuclear receptors including liver X receptor, retinoid X receptor (RXR), CAR, thyroid hormone receptor, HNF4, and chicken ovalbumin upstream promoter-transcription factor were initially screened to examine potential transactivation of a luciferase reporter construct containing the mouse PBREM126. Of these receptors, only CAR demonstrated robust transactivation of the responsive elements identifying it as the first nuclear receptor able to activate PBREM-mediated gene transcription126. CAR exhibits this regulatory function by binding to the nuclear receptor binding site 1 (NR1) and NR2 motifs within the PBREM as a heterodimer with RXR in the nucleus of cells. The affinity of this binding is increased significantly by treatment with PB. In mouse hepatocytes, as well as HepG2 cells, reporter constructs containing the Cyp2b10 reporter or CYP2B6 reporter, respectively, were activated by a myriad of compounds including PB, TCPOBOP, clotrimazole, metyrapone, and chlorpromazine129, 130. Downstream, these compounds induced the expression of Cyp2b10 in mouse hepatocytes and CY2B6 in HPH. The CAR-mediated induction of CYP2B genes by PB-type compounds was definitively established by experiments in Car-null mice, in which loss of CAR completely eliminated the induction of Cyp2b10 by PB and TCPOBOP131.

Around the same time, a novel orphan nuclear receptor, PXR, was cloned and firmly established as the primary modulator for drug-induced expression of CYP3A genes in different species69, 132. Evolutionarily, CAR and PXR represent the two closest members in the whole nuclear receptor superfamily, sharing approximately 40% amino acid identity in their ligand binding domains. Although CYP2B and CYP3A are the primary targets of CAR and PXR, respectively, accumulating evidence reveals that CAR and PXR can regulate each other׳s transcriptional targets through cross-talk131, 133. Like CAR, the PXR–RXR heterodimer binds to the PBREM in the CYP2B promoter with greater affinity to the NR1 site than the NR2 site69. In mice, known PXR ligands such as DEX and PCN significantly increased the expression of Cyp2b10 while this induction was not observed in Pxr-null mice134, 135. Notably, both these nuclear receptors display significant promiscuity in their ligand recognition and downstream target gene regulation, mediating the transcription of numerous genes involving drug metabolism and transport, energy homeostasis, and cell proliferation136, 137, 138. Known CYP2B6 inducers such as PB, RIF, clotrimazole, phenytoin, and carbamazepine have also been shown to induce CYP3A4, CYP2C9, UDP-glucuronosyltransferase 1A1 (UGT1A1), and the important efflux transporter, multidrug resistance protein 1 (MDR1)139, 140, 141, 142.

It is important to note that recognizing the role of CAR and PXR in CYP regulation not only provides a rational explanation for the mechanism by which many drugs can induce the same class of drug-metabolizing genes such as CYP2B6 and CYP3A4, but also supports the existence of a metabolic protection network coordinated by both receptors. Moreover, such findings also offer a mechanistic justification for the observed species-specific induction of CYP2B and CYP3A between human and rodents. For instance, TCPOBOP and PCN are selective activators of mouse CAR and PXR, respectively, and induce the expression of Cyp2b10 and Cyp3a11 but not their human counterparts131, 133. On the other hand, CITCO and RIF activate human CAR and PXR and induce the expression of human but not mouse CYP2B and CYP3A genes, respectively121, 143. In CAR- and PXR-humanized mice with their rodent counterparts being knocked out, induction of Cyp2b10 and Cyp3a11 was achieved by CITCO and RIF but not TCPOBOP and PCN, suggesting CAR and PXR are the xenobiotic dictators that convey the observed species-specific induction of CYP2B and CYP3A genes144, 145. Interestingly, human (h) PXR appears to have evolved into an extremely promiscuous xenobiotic sensor and almost all known hCAR activators activate hPXR as well. For example, PB is a rather selective activator of rodent CAR not PXR, while it exhibits effective activation of both human CAR and PXR143, 146. Importantly, although each of these nuclear receptors holds an impact on the expression of these target genes, their respective contributions to the induction of individual genes can vary. It has been well-recognized that the selective activation of CAR preferentially induces the expression of CYP2B6 over CYP3A4 while activation of PXR induces both P450 enzymes in concert70.

3.2. Induction of CYP2B6 expression by other nuclear receptors

To date, it has been clearly established that induction of the CYP2B6 gene by xenobiotics is mediated predominantly by hCAR and hPXR through interactions with the PBREM and XREM located upstream of the CYP2B transcriptional start site69, 126, 132. However, the dramatic inter-individual variability in CYP2B6 gene induction cannot be fully explained by a simple PXR/CAR-based induction model. For instance, the majority of PXR/CAR target genes (e.g., CYP2Cs and UGT1A1) are induced relatively moderately. This is in stark contrast to the potent induction of CYP2B6 and CYP3A4 genes observed clinically and in HPH cultures. On the other hand, over-expression of CAR and/or PXR alone failed to fully restore the basal and inductive expression of CYP2B6 in non-hepatic or hepatoma cell lines147. Accumulating evidence suggests that other nuclear receptors and liver-enriched transcriptional factors may also be involved in the transcription of CYP2B6 and contribute to the large individual variations of CYP2B6 expression in the human population.

The role of the glucocorticoid receptor (GR) in CYP2B regulation has been more firmly established in rodents than in humans. DEX, a synthetic glucocorticoid and GR activator, efficiently induced the expression of rat Cyp2b2 and mouse Cyp2b10 both in vivo and in cultured primary hepatocytes118. Importantly, in GR-deficient mice, not only did treatment with DEX fail to induce Cyp2b10 expression, but the basal level of Cyp2b10 was also significantly decreased148. Further in silico analysis resulted in the identification of putative glucocorticoid responsive elements (GRE) in the promoters of mouse Cyp2b10 and rat Cyp2b1/2, but not in the promoter of human CYP2B6. Although sub-micromolar concentrations of DEX dose-dependently induce the expression of CYP3A4 but not CYP2B6 in HPH, co-treatment of PB and RIF with the same concentration range of DEX enhanced the induction of both CYP enzymes123, 140, 149. Interestingly, DEX increases the expression of CAR and PXR in a GR-dependent manner and a functional GRE was later located in the promoter of CAR itself, suggesting GR activation may indirectly regulate CYP2B6 by facilitating the availability of CAR and PXR.

Initial screening for potential endogenous CAR activators by Negishi and colleagues resulted in the identification of estradiol (E2) and estrone as effective mouse CAR (mCAR) activators at pharmacological concentrations150. In mouse primary hepatocytes, these estrogens increased the expression of Cyp2b10 and nuclear accumulation of mCAR, the first step of CAR activation. It appears that this estrogen-dependent induction of Cyp2b10 is specific to mice and most likely ER-independent given that there is no estrogen responsive element (ERE) identified in the Cyp2b10 promoter. Further, not all ER agonists enhance Cyp2b10 expression. However, such contention may not apply to the case for human CYP2B6. It has been known that a greater level of CYP2B6 is expressed in ERα-positive compared to ERα-negative breast tumor tissues151. In a chromatin immunoprecipitation and promoter focused microarray (ChIP-on-chip)-based screening in T-47D human breast cancer cells, multiple ERα-bound regions were located in the upstream regulatory sequences of the CYP2B gene cluster152. Further analysis revealed a functional ERE located at −1669/−1657 right next to the PBREM of CYP2B6. Luciferase reporter assays demonstrated that both ERα and ERβ are capable of stimulating CYP2B6 transactivation, while such activation was completely abolished when the ERE was deleted. Moreover, physiological levels of E2 significantly induced the expression of CYP2B6 in T-47D cells152. Compared with extrahepatic cells, E2 was rapidly metabolized in primary hepatocytes with a first order elimination half-life of 37 min153. To overcome this rapid clearance, Koh and colleagues153 replenished E2 regularly during the treatment of HPH to achieve an average concentration of ~100 nmol/L, which reflects the plasma concentration reached at term pregnancy. Under such experimental condition, the authors observed that E2 robustly increased the expression of CYP2B6 and activation of both CAR and ER. Moreover, concurrent activation of both ER and CAR by E2 enhanced CYP2B6 expression in a synergistic manner, suggesting a positive cross-talk between these two receptors153.

Knowledge of transcriptional regulation of CYP2B6 expression has grown substantially in the past two decades. In addition to its known transcriptional regulators such as nuclear receptors including CAR, PXR, GR, and ER, recently, several studies demonstrated that expression of the CYP2B6 gene can be influenced by interactions between nuclear receptors and liver-enriched transcriptional factors such as HNF4α, C/EBPα, and HNF3β154, 155, 156. Multiple responsive elements for different liver-enriched transcriptional factors have been identified in the promoter of CYP2B6. Importantly, with the presence of CAR/PXR agonists or other transcription factors like early growth response 1, the distally recruited nuclear receptor can be efficiently looped to the proximate promoter of CYP2B6 and synergistically enhance the CYP2B6 transcription156.

4. Implications for clinical drug–drug interactions and adverse events

CYP2B6 shares an overlapping substrate spectrum with other CYP enzymes including, in particular, CYP3A4. Its pharmacological/toxicological significance is, however, distinguished by a distinct affinity for specific drug substrates and unique enzymatic biotransformation reactions. As increasing numbers of substrates of CYP2B6 are identified, it becomes more likely that we will uncover significant DDI mediated by this enzyme. It is not uncommon for multiple drugs to be administered simultaneously to an individual. Combination therapies have proven to be rather effective in combatting cancers, autoimmune disorders, and other prevalent diseases. As such, it is important to understand the impact drugs may have on the expression and function of genes responsible not only for their own disposition, but the metabolism and clearance of any co-administered agents.

Two of the most well-studied and better understood drugs with respect to CYP2B6 metabolism are EFV and CPA. Both of these widely used drugs have very narrow therapeutic indices, associated toxicities, and variations in CYP2B6 expression and function lead to significantly altered drug plasma concentrations of each agent45, 115. In the case of CPA increased expression or function of CYP2B6 may be beneficial as it may result in an increase in circulating concentrations of the active moiety74, 157. However, in the case of EFV, increased metabolism may lead to non-therapeutic concentrations in circulation45. Conversely, decreased metabolic capacity of the enzyme may result in toxic concentrations of EFV in circulation or non-therapeutic concentrations of the active CPA moiety.

CPA has been used extensively for the treatment of various cancers and autoimmune disorders for more than half a century. CPA is metabolized to its active form, 4-OH-CPA, in the liver primarily by CYP2B6, with moderate contributions from CYP2C9, CYP2C19, and CYP3A422, 158, 159. Following the metabolism of CPA to 4-OH-CPA, it produces a DNA alkylating phosphoramide mustard which yields therapeutic cytotoxicity. Alternatively, CPA may be metabolized via N-dechloroethylation exclusively by CYP3A4 yielding a neurotoxic metabolite, chloroacetaldehyde, which contributes to the narrow therapeutic index of CPA159, 160. Thus, it has been hypothesized that selective induction of CYP2B6 over CYP3A4 could significantly increase the beneficial biotransformation of CPA to 4-OH-CPA without concomitant augmentation of the formation of the toxic chloroacetaldehyde (Fig. 2)74, 161. Recently, we have demonstrated that selective activation of CAR and downstream preferential induction of CYP2B6 over other enzymes and transporters with a selective small molecule activator can facilitate the bioactivation of CPA to 4-OH-CPA and improve the therapeutic index of CHOP chemotherapy (cyclophosphamide–doxorubicin–vincristine–prednisone) for the treatment of non-Hodgkin lymphoma157. It is expected that if such interactions held true in vivo, inclusion of a selective hCAR activator in the CHOP regimen may significantly reduce the dose of the chemotherapeutic agents and side toxicity without sacrificing therapeutic efficacy. By manipulating the expression of the CYP2B6 isozyme, we may be able to alter the front-line strategies employed to treat hematopoetic malignancies74, 157.

Figure 2.

Fig. 2

Schematic illustration of CPA metabolism and the potential role of CAR in CPA bioactivation. (This figure was adopted from Wang et al.161 with minor modification).

EFV is a frequently prescribed NNRTI utilized as a treatment for HIV-1 infections. EFV has a very narrow therapeutic index as increased plasma concentrations of EFV have been shown to result in toxicities while insufficient plasma concentrations do not achieve anti-viral therapy23, 93, 162. CYP2B6 is the primary catalyst of EFV metabolism and the function of this enzyme, as well as its induction or inhibition, plays an important role in maintaining therapeutic yet non-toxic concentrations of the drug in circulation23. EFV is thought to auto-induce its own metabolism by increasing the expression of CYP2B6 via activation of CAR and PXR163. Further, EFV has been shown to competitively inhibit bupropion metabolism by CYP2B6 and to inhibit several CYP2C isoforms including CYP2C8, 2C9, and 2C19164. Together, the impact EFV has on important metabolizing enzymes can result in significant DDI with other antiretrovirals or medications commonly taken concurrently with EFV therapy.

Artemisinin is an extract obtained from the Chinese herb Artemisia annua and is utilized as an antimalarial agent, though poor bioavailability limits its efficacy165. The metabolism of artemisinin is mediated primarily by CYP2B6 in the liver with some contribution from CYP3A4, though it has been proposed that their relative contributions are reversed in patients with low levels of functional CYP2B618. Inhibition of CYP2B6 in vitro by orphenadrine has been shown to decrease artemisinin disappearance rates by 75%166. This result indicates that inhibition of CYP2B6 may result in increased plasma concentrations of drug which would, in turn, increase the risk of adverse events. While there is currently no literature available regarding the impact of CYP2B6 pharmacogenetics on artemisinin disposition, it is reasonable to anticipate that polymorphisms which result in decreased expression or function of CYP2B6 may potentially contribute to decreased metabolism and clearance of the drug, and potentially increased toxicities.

Bupropion, an antidepressant which is often utilized as a non-nicotine aid to quit smoking, is metabolized to hydroxybupropion in human liver microsomes predominantly by CYP2B6 with only negligible contribution from CYP2E110, 13, 114. Long-term use of bupropion has been associated with select toxicities including seizures167. In vivo, plasma concentrations of bupropion are typically less than that of hydroxybupropion, indicating that it may be the metabolite which is responsible for the associated toxicity of this drug. Thus, chemical activation or genetic variations resulting in increased CYP2B6 activity that enhances the metabolism of bupropion and, in turn, increased circulation of the hydroxylated moiety could lead to increased risk for adverse events13. Further, bupropion has been demonstrated to be an effective inhibitor of other important CYP isoforms in vitro including CYP2D6 which is responsible for an estimated 25% of clinically utilized drugs168, 169. This indicates that co-administration of bupropion alongside a drug that is a CYP2D6 substrate could result in harmful DDI due to varied circulating drug levels which may cause unexpected toxicities.

Ketamine has multiple clinical uses including analgesia and moderate stimulation of the cardiovascular system. CYP2B6 is the primary enzyme responsible for the N-demethylation of ketamine enantiomers to pharmacologically active products25. Currently, limited literature is available regarding DDI involving ketamine in humans. However, it has been demonstrated that co-administration of ketamine with diazepam, a substrate of CYP2C19 and CYP3A4, or secobarbital, a CYP2B6 inhibitor, significantly increased the plasma half-life of ketamine170, 171.

Methadone is a synthetic opioid, which is administered as a racemic mixture for the treatment of chronic pain. CYP2B6 mediates a stereo-selective metabolism reaction of methadone towards the (S)-enantiomer172. The (R)-enantiomer of methadone produces the analgesic effects of the drug by binding to and activating the μ-opioid receptor, while the (S)-enantiomer produces undesirable cardiotoxicity by inhibiting the cardiac potassium channel172, 173. As such, decreased CYP2B6 activity is associated with decreased metabolism of the (S)-enantiomer of methadone and increased plasma concentrations of this enantiomer. Elevated levels of (S)-methadone in circulation are associated with a greater risk of cardiac side effects and death174.

Pethidine, also known as meperidine, another synthetic opioid, is also metabolized in the human liver by CYP2B6, CYP3A4 and CYP2C19 accounting for 57%, 28%, and 15% of its total intrinsic clearance, respectively21. The major metabolite of pethidine, norpethidine, can accumulate in the brain and lead to significant central nervous toxicities when pethidine is administered at high dosage175, 176. The rates of formation and clearance of norpethidine from pethidine can be difficult to anticipate due to the highly polymorphic and inducible nature of CYP2B621. Increased expression of CYP2B6 can result in an increase in the formation of norpethidine and a resultant increase in adverse events, most frequently manifesting as convulsions177. Due to the unpredictable nature of pethidine metabolism and disposition, it is often withheld from elderly patients or patients with compromised liver or kidney function21, 177.

Selegiline is frequently used in the treatment of Parkinson׳s disease. Sridar et al.178 have shown selegiline to be a strong inhibitor of CYP2B6-mediated metabolism of bupropion in vitro, increasing the Km of bupropion from 10 to 92 µmol/L and decreasing the kcat by approximately 50%178. This strong inhibition of CYP2B6 by selegiline highlights a serious potential of DDI for combination therapies involving bupropion.

Collectively, along with increased understanding of the transcriptional regulation of CYP2B6 and its pharmacogenetics, the potential clinical implication of CYP2B6 in the context of DDIs is escalating. Altered expression of CYP2B6 could result in unexpected drug–drug and gene–drug interactions which may be either harmful or beneficial.

5. Concluding remarks

Although historically believed to be relatively inconsequential with respect to human drug metabolism, over the past two decades CYP2B6 has been identified as a catalyst for many biotransformation reactions. CYP2B6 is both highly inducible and polymorphic resulting in widely varied expression and function of the enzyme between individuals leading to differential drug metabolism and disposition. Polymorphisms of CYP2B6 are often associated with loss-of-function and can result in elevated plasma concentrations of drugs and enhanced toxicity.

Many drugs and chemicals have demonstrated the ability to either induce or inhibit the expression of CYP2B6 whether directly or through the transcriptional activation of nuclear receptors. Recent studies have begun to explore the potential of these nuclear receptors as targets for combination therapies in the hopes of altering the expression of drug-metabolizing enzymes and transporters in a manner that is beneficial for the treatment of cancers and other disorders.

As more substrates of CYP2B6 are identified, greater interest is generated in the impact of both genetic and pharmacological modulation of CYP2B6 expression on the disposition of drugs. In this review, we have highlighted the impact of CYP2B6 modulation and its potential for clinically significant DDI. It is important to point out that although many drugs exhibit the potential for CYP2B6-associated DDI based mostly on in vitro experimental results, clinically significant DDI mediated by CYP2B6 are limited. To this end, the role of CYP2B6*6 in the therapeutic efficacy and toxicity of EFV appears to be the only CYP2B6–drug pair that is supported by compelling clinical evidence across different ethnic groups. Given that EFV continues to be in the front line for HIV therapy, clinical implementation of a CYP2B6 genotyping test would eventually benefit patients undergoing EFV-based treatment.

Acknowledgments

The authors are grateful to Dr. Maria R. Baer (University of Maryland School of Medicine and Stewart and Marlene Greenebaum Cancer Center) for discussions regarding the content of this manuscript. We apologize to the scientists who made contributions to this field, but have not been cited due to space limitations. This work was supported by research grants from the U. S. National Institute of Health (DK061652 and GM107058).

Footnotes

Peer review under responsibility of Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical Association.

References

  • 1.Estabrook R.W. A passion for P450s (remembrances of the early history of research on cytochrome P450) Drug Metab Dispos. 2003;31:1461–1473. doi: 10.1124/dmd.31.12.1461. [DOI] [PubMed] [Google Scholar]
  • 2.Nelson D.R., Koymans L., Kamataki T., Stegeman J.J., Feyereisen R., Waxman D.J. P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics. 1996;6:1–42. doi: 10.1097/00008571-199602000-00002. [DOI] [PubMed] [Google Scholar]
  • 3.Nelson D.R., Zeldin D.C., Hoffman S.M., Maltais L.J., Wain H.M., Nebert D.W. Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants. Pharmacogenetics. 2004;14:1–18. doi: 10.1097/00008571-200401000-00001. [DOI] [PubMed] [Google Scholar]
  • 4.Hidestrand M., Oscarson M., Salonen J.S., Nyman L., Pelkonen O., Turpeinen M. CYP2B6 and CYP2C19 as the major enzymes responsible for the metabolism of selegiline, a drug used in the treatment of Parkinson׳s disease, as revealed from experiments with recombinant enzymes. Drug Metab Dispos. 2001;29:1480–1484. [PubMed] [Google Scholar]
  • 5.Purnapatre K., Khattar S.K., Saini K.S. Cytochrome P450s in the development of target-based anticancer drugs. Cancer Lett. 2008;259:1–15. doi: 10.1016/j.canlet.2007.10.024. [DOI] [PubMed] [Google Scholar]
  • 6.Hoffman S.M., Nelson D.R., Keeney D.S. Organization, structure and evolution of the CYP2 gene cluster on human chromosome 19. Pharmacogenetics. 2001;11:687–698. doi: 10.1097/00008571-200111000-00007. [DOI] [PubMed] [Google Scholar]
  • 7.Shimada T., Yamazaki H., Mimura M., Inui Y., Guengerich F.P. Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther. 1994;270:414–423. [PubMed] [Google Scholar]
  • 8.Mimura M., Baba T., Yamazaki H., Ohmori S., Inui Y., Gonzalez F.J. Characterization of cytochrome P-450 2B6 in human liver microsomes. Drug Metab Dispos. 1993;21:1048–1056. [PubMed] [Google Scholar]
  • 9.Stresser D.M., Kupfer D. Monospecific antipeptide antibody to cytochrome P-450 2B6. Drug Metab Dispos. 1999;27:517–525. [PubMed] [Google Scholar]
  • 10.Faucette S.R., Hawke R.L., Lecluyse E.L., Shord S.S., Yan B., Laethem R.M. Validation of bupropion hydroxylation as a selective marker of human cytochrome P450 2B6 catalytic activity. Drug Metab Dispos. 2000;28:1222–1230. [PubMed] [Google Scholar]
  • 11.Code E.L., Crespi C.L., Penman B.W., Gonzalez F.J., Chang T.K., Waxman D.J. Human cytochrome P4502B6: interindividual hepatic expression, substrate specificity, and role in procarcinogen activation. Drug Metab Dispos. 1997;25:985–993. [PubMed] [Google Scholar]
  • 12.Hanna I.H., Reed J.R., Guengerich F.P., Hollenberg P.F. Expression of human cytochrome P450 2B6 in Escherichia coli: characterization of catalytic activity and expression levels in human liver. Arch Biochem Biophys. 2000;376:206–216. doi: 10.1006/abbi.2000.1708. [DOI] [PubMed] [Google Scholar]
  • 13.Hesse L.M., Venkatakrishnan K., Court M.H., von Moltke L.L., Duan S.X., Shader R.I. CYP2B6 mediates the in vitro hydroxylation of bupropion: potential drug interactions with other antidepressants. Drug Metab Dispos. 2000;28:1176–1183. [PubMed] [Google Scholar]
  • 14.Wang H., Tompkins L.M. CYP2B6: new insights into a historically overlooked cytochrome P450 isozyme. Curr Drug Metab. 2008;9:598–610. doi: 10.2174/138920008785821710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34:83–448. doi: 10.1081/dmr-120001392. [DOI] [PubMed] [Google Scholar]
  • 16.Hodgson E., Rose R.L. The importance of cytochrome P450 2B6 in the human metabolism of environmental chemicals. Pharmacol Ther. 2007;113:420–428. doi: 10.1016/j.pharmthera.2006.10.002. [DOI] [PubMed] [Google Scholar]
  • 17.Yamada H., Ishii Y., Yamamoto M., Oguri K. Induction of the hepatic cytochrome P450 2B subfamily by xenobiotics: research history, evolutionary aspect, relation to tumorigenesis, and mechanism. Curr Drug Metab. 2006;7:397–409. doi: 10.2174/138920006776873508. [DOI] [PubMed] [Google Scholar]
  • 18.Svensson U.S., Ashton M. Identification of the human cytochrome P450 enzymes involved in the in vitro metabolism of artemisinin. Br J Clin Pharmacol. 1999;48:528–535. doi: 10.1046/j.1365-2125.1999.00044.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Court M.H., Duan S.X., Hesse L.M., Venkatakrishnan K., Greenblatt D.J. Cytochrome P-450 2B6 is responsible for interindividual variability of propofol hydroxylation by human liver microsomes. Anesthesiology. 2001;94:110–119. doi: 10.1097/00000542-200101000-00021. [DOI] [PubMed] [Google Scholar]
  • 20.Kharasch E.D., Hoffer C., Whittington D., Sheffels P. Role of hepatic and intestinal cytochrome P450 3A and 2B6 in the metabolism, disposition, and mitotic effects of methadone. Clin Pharmacol Ther. 2004;76:250–269. doi: 10.1016/j.clpt.2004.05.003. [DOI] [PubMed] [Google Scholar]
  • 21.Ramírez J., Innocenti F., Schuetz E.G., Flockhart D.A., Relling M.V., Santucci R. CYP2B6, CYP3A4, and CYP2C19 are responsible for the in vitro N-demethylation of meperidine in human liver microsomes. Drug Metab Dispos. 2004;32:930–936. [PubMed] [Google Scholar]
  • 22.Xie H.J., Yasar Ü., Lundgren S., Griskevicius L., Terelius Y., Hassan M. Role of polymorphic human CYP2B6 in cyclophosphamide bioactivation. Pharmacogenomics J. 2003;3:53–61. doi: 10.1038/sj.tpj.6500157. [DOI] [PubMed] [Google Scholar]
  • 23.Ward B.A., Gorski J.C., Jones D.R., Hall S.D., Flockhart D.A., Desta Z. The cytochrome P450 2B6 (CYP2B6) is the main catalyst of efavirenz primary and secondary metabolism: implication for HIV/AIDS therapy and utility of efavirenz as a substrate marker of CYP2B6 catalytic activity. J Pharmacol Exp Ther. 2003;306:287–300. doi: 10.1124/jpet.103.049601. [DOI] [PubMed] [Google Scholar]
  • 24.Saitoh A., Sarles E., Capparelli E., Aweeka F., Kovacs A., Burchett S.K. CYP2B6 genetic variants are associated with nevirapine pharmacokinetics and clinical response in HIV-1-infected children. AIDS. 2007;21:2191–2199. doi: 10.1097/QAD.0b013e3282ef9695. [DOI] [PubMed] [Google Scholar]
  • 25.Yanagihara Y., Kariya S., Ohtani M., Uchino K., Aoyama T., Yamamura Y. Involvement of CYP2B6 in N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2001;29:887–890. [PubMed] [Google Scholar]
  • 26.Turpeinen M., Jouko U., Jorma J., Olavi P. Multiple P450 substrates in a single run: rapid and comprehensive in vitro interaction assay. Eur J Pharm Sci. 2005;24:123–132. doi: 10.1016/j.ejps.2004.10.006. [DOI] [PubMed] [Google Scholar]
  • 27.Loboz K.K., Gross A.S., Williams K.M., Liauw W.S., Day R.O., Blievernicht J.K. Cytochrome P450 2B6 activity as measured by bupropion hydroxylation: effect of induction by rifampin and ethnicity. Clin Pharmacol Ther. 2006;80:75–84. doi: 10.1016/j.clpt.2006.03.010. [DOI] [PubMed] [Google Scholar]
  • 28.Turpeinen M., Nieminen R., Juntunen T., Taavitsainen P., Raunio H., Pelkonen O. Selective inhibition of CYP2B6-catalyzed bupropion hydroxylation in human liver microsomes in vitro. Drug Metab Dispos. 2004;32:626–631. doi: 10.1124/dmd.32.6.626. [DOI] [PubMed] [Google Scholar]
  • 29.Hijazi Y., Boulieu R. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30:853–858. doi: 10.1124/dmd.30.7.853. [DOI] [PubMed] [Google Scholar]
  • 30.Imaoka S., Yamada T., Hiroi T., Hayashi K., Sakaki T., Yabusaki Y. Multiple forms of human P450 expressed in Saccharomyces cerevisiae: systematic characterization and comparison with those of the rat. Biochem Pharmacol. 1996;51:1041–1050. doi: 10.1016/0006-2952(96)00052-4. [DOI] [PubMed] [Google Scholar]
  • 31.Oda Y., Hamaoka N., Hiroi T., Imaoka S., Hase I., Tanaka K. Involvement of human liver cytochrome P4502B6 in the metabolism of propofol. Br J Clin Pharmacol. 2001;51:281–285. doi: 10.1046/j.1365-2125.2001.00344.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Labbè L., Abolfathi Z., Lessard È., Pakdel H., Beaune P., Turgeon J. Role of specific cytochrome P450 enzymes in the N-oxidation of the antiarrhythmic agent mexiletine. Xenobiotica. 2003;33:13–25. doi: 10.1080/0049825021000017948. [DOI] [PubMed] [Google Scholar]
  • 33.Ekins S., VandenBranden M., Ring B.J., Wrighton S.A. Examination of purported probes of human CYP2B6. Pharmacogenetics. 1997;7:165–179. doi: 10.1097/00008571-199706000-00001. [DOI] [PubMed] [Google Scholar]
  • 34.Lake B.G. Coumarin metabolism, toxicity and carcinogenicity: relevance for human risk assessment. Food Chem Toxicol. 1999;37:423–453. doi: 10.1016/s0278-6915(99)00010-1. [DOI] [PubMed] [Google Scholar]
  • 35.Ko J.W., Desta Z., Flockhart D.A. Human N-demethylation of (S)-mephenytoin by cytochrome P450s 2C9 and 2B6. Drug Metab Dispos. 1998;26:775–778. [PubMed] [Google Scholar]
  • 36.Kobayashi K., Abe S., Nakajima M., Shimada N., Tani M., Chiba K. Role of human CYP2B6 in S-mephobarbital N-demethylation. Drug Metab Dispos. 1999;27:1429–1433. [PubMed] [Google Scholar]
  • 37.Kiang T.K., Ho P.C., Anari M.R., Tong V., Abbott F.S., Chang T.K. Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Toxicol Sci. 2006;94:261–271. doi: 10.1093/toxsci/kfl096. [DOI] [PubMed] [Google Scholar]
  • 38.Niwa T., Sato R., Yabusaki Y., Ishibashi F., Katagiri M. Contribution of human hepatic cytochrome P450s and steroidogenic CYP17 to the N-demethylation of aminopyrine. Xenobiotica. 1999;29:187–193. doi: 10.1080/004982599238731. [DOI] [PubMed] [Google Scholar]
  • 39.Niwa T., Tsutsui M., Kishimoto K., Yabusaki Y., Ishibashi F., Katagiri M. Inhibition of drug-metabolizing enzyme activity in human hepatic cytochrome P450s by bisphenol A. Biol Pharm Bull. 2000;23:498–501. doi: 10.1248/bpb.23.498. [DOI] [PubMed] [Google Scholar]
  • 40.Engel G., Hofmann U., Heidemann H., Cosme J., Eichelbaum M. Antipyrine as a probe for human oxidative drug metabolism: identification of the cytochrome P450 enzymes catalyzing 4-hydroxyantipyrine, 3-hydroxymethylantipyrine, and norantipyrine formation. Clin Pharmacol Ther. 1996;59:613–623. doi: 10.1016/S0009-9236(96)90001-6. [DOI] [PubMed] [Google Scholar]
  • 41.Surapaneni S.S., Clay M.P., Spangle L.A., Paschal J.W., Lindstrom T.D. In vitro biotransformation and identification of human cytochrome P450 isozyme-dependent metabolism of tazofelone. Drug Metab Dispos. 1997;25:1383–1388. [PubMed] [Google Scholar]
  • 42.Honda M., Muroi Y., Tamaki Y., Saigusa D., Suzuki N., Tomioka Y. Functional characterization of CYP2B6 allelic variants in demethylation of antimalarial artemether. Drug Metab Dispos. 2011;39:1860–1865. doi: 10.1124/dmd.111.040352. [DOI] [PubMed] [Google Scholar]
  • 43.Burk O., Arnold K.A., Nussler A.K., Schaeffeler E., Efimova E., Avery B.A. Antimalarial artemisinin drugs induce cytochrome P450 and MDR1 expression by activation of xenosensors pregnane X receptor and constitutive androstane receptor. Mol Pharmacol. 2005;67:1954–1965. doi: 10.1124/mol.104.009019. [DOI] [PubMed] [Google Scholar]
  • 44.Tsuchiya K., Gatanaga H., Tachikawa N., Teruya K., Kikuchi Y., Yoshino M. Homozygous CYP2B6*6 (Q172H and K262R) correlates with high plasma efavirenz concentrations in HIV-1 patients treated with standard efavirenz-containing regimens. Biochem Biophys Res Commun. 2004;319:1322–1326. doi: 10.1016/j.bbrc.2004.05.116. [DOI] [PubMed] [Google Scholar]
  • 45.Desta Z., Saussele T., Ward B., Blievernicht J., Li L., Klein K. Impact of CYP2B6 polymorphism on hepatic efavirenz metabolism in vitro. Pharmacogenomics. 2007;8:547–558. doi: 10.2217/14622416.8.6.547. [DOI] [PubMed] [Google Scholar]
  • 46.Rotger M., Colombo S., Furrer H., Bleiber G., Buclin T., Lee B.L. Influence of CYP2B6 polymorphism on plasma and intracellular concentrations and toxicity of efavirenz and nevirapine in HIV-infected patients. Pharmacogenet Genom. 2005;15:1–5. doi: 10.1097/01213011-200501000-00001. [DOI] [PubMed] [Google Scholar]
  • 47.Erickson D.A., Mather G., Trager W.F., Levy R.H., Keirns J.J. Characterization of the in vitro biotransformation of the HIV-1 reverse transcriptase inhibitor nevirapine by human hepatic cytochromes P-450. Drug Metab Dispos. 1999;27:1488–1495. [PubMed] [Google Scholar]
  • 48.Lindley C., Hamilton G., McCune J.S., Faucette S., Shord S.S., Hawke R.L. The effect of cyclophosphamide with and without dexamethasone on cytochrome P450 3A4 and 2B6 in human hepatocytes. Drug Metab Dispos. 2002;30:814–822. doi: 10.1124/dmd.30.7.814. [DOI] [PubMed] [Google Scholar]
  • 49.Huang Z., Roy P., Waxman D.J. Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Biochem Pharmacol. 2000;59:961–972. doi: 10.1016/s0006-2952(99)00410-4. [DOI] [PubMed] [Google Scholar]
  • 50.Roy P., Yu L.J., Crespi C.L., Waxman D.J. Development of a substrate-activity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. Drug Metab Dispos. 1999;27:655–666. [PubMed] [Google Scholar]
  • 51.Roy P., Tretyakov O., Wright J., Waxman D.J. Stereoselective metabolism of ifosfamide by human P-450s 3A4 and 2B6. Favorable metabolic properties of R-enantiomer. Drug Metab Dispos. 1999;27:1309–1318. [PubMed] [Google Scholar]
  • 52.Coller J.K., Krebsfaenger N., Klein K., Endrizzi K., Wolbold R., Lang T. The influence of CYP2B6, CYP2C9 and CYP2D6 genotypes on the formation of the potent antioestrogen Z-4-hydroxy-tamoxifen in human liver. Br J Clin Pharmacol. 2002;54:157–167. doi: 10.1046/j.1365-2125.2002.01614.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Styles J.A., Davies A., Lim C.K., De Matteis F., Stanley L.A., White I.N. Genotoxicity of tamoxifen, tamoxifen epoxide and toremifene in human lymphoblastoid cells containing human cytochrome P450s. Carcinogenesis. 1994;15:5–9. doi: 10.1093/carcin/15.1.5. [DOI] [PubMed] [Google Scholar]
  • 54.Salonen J.S., Nyman L., Boobis A.R., Edwards R.J., Watts P., Lake B.G. Comparative studies on the cytochrome p450-associated metabolism and interaction potential of selegiline between human liver-derived in vitro systems. Drug Metab Dispos. 2003;31:1093–1102. doi: 10.1124/dmd.31.9.1093. [DOI] [PubMed] [Google Scholar]
  • 55.Gerber J.G., Rhodes R.J., Gal J. Stereoselective metabolism of methadone N-demethylation by cytochrome P4502B6 and 2C19. Chirality. 2004;16:36–44. doi: 10.1002/chir.10303. [DOI] [PubMed] [Google Scholar]
  • 56.Niwa T., Shiraga T., Ishii I., Kagayama A., Takagi A. Contribution of human hepatic cytochrome p450 isoforms to the metabolism of psychotropic drugs. Biol Pharm Bull. 2005;28:1711–1716. doi: 10.1248/bpb.28.1711. [DOI] [PubMed] [Google Scholar]
  • 57.Onof S., Hatanaka T., Miyazawa S., Tsutsui M., Aoyama T., Gonzalez F.J. Human liver microsomal diazepam metabolism using cDNA-expressed cytochrome P450s: role of CYP2B6, 2C19 and the 3A subfamily. Xenobiotica. 1996;26:1155–1166. doi: 10.3109/00498259609050260. [DOI] [PubMed] [Google Scholar]
  • 58.Yang T.J., Krausz K.W., Shou M., Yang S.K., Buters J.T., Gonzalez F.J. Inhibitory monoclonal antibody to human cytochrome P450 2B6. Biochem Pharmacol. 1998;55:1633–1640. doi: 10.1016/s0006-2952(98)00018-5. [DOI] [PubMed] [Google Scholar]
  • 59.Ekins S., Vandenbranden M., Ring B.J., Gillespie J.S., Yang T.J., Gelboin H.V. Further characterization of the expression in liver and catalytic activity of CYP2B6. J Pharmacol Exp Ther. 1998;286:1253–1259. [PubMed] [Google Scholar]
  • 60.Zhou S.F. Drugs behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. Curr Drug Metab. 2008;9:310–322. doi: 10.2174/138920008784220664. [DOI] [PubMed] [Google Scholar]
  • 61.Kobayashi K., Kogo M., Tani M., Shimada N., Ishizaki T., Numazawa S. Role of CYP2C19 in stereoselective hydroxylation of mephobarbital by human liver microsomes. Drug Metab Dispos. 2001;29:36–40. [PubMed] [Google Scholar]
  • 62.Hu Y., Kupfer D. Enantioselective metabolism of the endocrine disruptor pesticide methoxychlor by human cytochromes P450 (P450s): major differences in selective enantiomer formation by various P450 isoforms. Drug Metab Dispos. 2002;30:1329–1336. doi: 10.1124/dmd.30.12.1329. [DOI] [PubMed] [Google Scholar]
  • 63.Hu Y., Kupfer D. Metabolism of the endocrine disruptor pesticide-methoxychlor by human P450s: pathways involving a novel catechol metabolite. Drug Metab Dispos. 2002;30:1035–1042. doi: 10.1124/dmd.30.9.1035. [DOI] [PubMed] [Google Scholar]
  • 64.Dehal S.S., Kupfer D. Metabolism of the proestrogenic pesticide methoxychlor by hepatic P450 monooxygenases in rats and humans. Dual pathways involving novel ortho ring-hydroxylation by CYP2B. Drug Metab Dispos. 1994;22:937–946. [PubMed] [Google Scholar]
  • 65.Lamba V., Lamba J., Yasuda K., Strom S., Davila J., Hancock M.L. Hepatic CYP2B6 expression: gender and ethnic differences and relationship to CYP2B6 genotype and CAR (constitutive androstane receptor) expression. J Pharmacol Exp Ther. 2003;307:906–922. doi: 10.1124/jpet.103.054866. [DOI] [PubMed] [Google Scholar]
  • 66.Lang T., Klein K., Fischer J., Nüssler A.K., Neuhaus P., Hofmann U. Extensive genetic polymorphism in the human CYP2B6 gene with impact on expression and function in human liver. Pharmacogenetics. 2001;11:399–415. doi: 10.1097/00008571-200107000-00004. [DOI] [PubMed] [Google Scholar]
  • 67.Hesse L.M., He P., Krishnaswamy S., Hao Q., Hogan K., von Moltke L.L. Pharmacogenetic determinants of interindividual variability in bupropion hydroxylation by cytochrome P450 2B6 in human liver microsomes. Pharmacogenetics. 2004;14:225–238. doi: 10.1097/00008571-200404000-00002. [DOI] [PubMed] [Google Scholar]
  • 68.Remmer H., Schoene B., Fleischmann R.A. Induction of the unspecific microsomal hydroxylase in the human liver. Drug Metab Dispos. 1973;1:224–230. [PubMed] [Google Scholar]
  • 69.Goodwin B., Moore L.B., Stoltz C.M., McKee D.D., Kliewer S.A. Regulation of the human CYP2B6 gene by the nuclear pregnane X receptor. Mol Pharmacol. 2001;60:427–431. [PubMed] [Google Scholar]
  • 70.Faucette S.R., Sueyoshi T., Smith C.M., Negishi M., LeCluyse E.L., Wang H. Differential regulation of hepatic CYP2B6 and CYP3A4 genes by constitutive androstane receptor but not pregnane X receptor. J Pharmacol Exp Ther. 2006;317:1200–1209. doi: 10.1124/jpet.105.098160. [DOI] [PubMed] [Google Scholar]
  • 71.Honkakoski P., Sueyoshi T., Negishi M. Drug-activated nuclear receptors CAR and PXR. Ann Med. 2003;35:172–182. doi: 10.1080/07853890310008224. [DOI] [PubMed] [Google Scholar]
  • 72.Chai X., Zeng S., Xie W. Nuclear receptors PXR and CAR: implications for drug metabolism regulation, pharmacogenomics and beyond. Expert Opin Drug Metab Toxicol. 2013;9:253–266. doi: 10.1517/17425255.2013.754010. [DOI] [PubMed] [Google Scholar]
  • 73.Willson T.M., Kliewer S.A. PXR, CAR and drug metabolism. Nat Rev Drug Discov. 2002;1:259–266. doi: 10.1038/nrd753. [DOI] [PubMed] [Google Scholar]
  • 74.Lu J., Huang Y., Zhao W., Marquez R.T., Meng X., Li J. PEG-derivatized embelin as a nanomicellar carrier for delivery of paclitaxel to breast and prostate cancers. Biomaterials. 2013;34:1591–1600. doi: 10.1016/j.biomaterials.2012.10.073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ekins S., Wrighton S.A. The role of CYP2B6 in human xenobiotic metabolism. Drug Metab Rev. 1999;31:719–754. doi: 10.1081/dmr-100101942. [DOI] [PubMed] [Google Scholar]
  • 76.Zanger U.M., Klein K., Saussele T., Blievernicht J., Hofmann M.H., Schwab M. Polymorphic CYP2B6: molecular mechanisms and emerging clinical significance. Pharmacogenomics. 2007;8:743–759. doi: 10.2217/14622416.8.7.743. [DOI] [PubMed] [Google Scholar]
  • 77.Guan S., Huang M., Li X., Chen X., Chan E., Zhou S.F. Intra- and inter-ethnic differences in the allele frequencies of cytochrome P450 2B6 gene in Chinese. Pharm Res. 2006;23:1983–1990. doi: 10.1007/s11095-006-9083-5. [DOI] [PubMed] [Google Scholar]
  • 78.Mehlotra R.K., Bockarie M.J., Zimmerman P.A. CYP2B6 983T>C polymorphism is prevalent in West Africa but absent in Papua New Guinea: implications for HIV/AIDS treatment. Br J Clin Pharmacol. 2007;64:391–395. doi: 10.1111/j.1365-2125.2007.02884.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lang T., Klein K., Richter T., Zibat A., Kerb R., Eichelbaum M. Multiple novel nonsynonymous CYP2B6 gene polymorphisms in Caucasians: demonstration of phenotypic null alleles. J Pharmacol Exp Ther. 2004;311:34–43. doi: 10.1124/jpet.104.068973. [DOI] [PubMed] [Google Scholar]
  • 80.Hofmann M.H., Blievernicht J.K., Klein K., Saussele T., Schaeffeler E., Schwab M. Aberrant splicing caused by single nucleotide polymorphism c.516G>T [Q172H], a marker of CYP2B6*6, is responsible for decreased expression and activity of CYP2B6 in liver. J Pharmacol Exp Ther. 2008;325:284–292. doi: 10.1124/jpet.107.133306. [DOI] [PubMed] [Google Scholar]
  • 81.Ogburn E.T., Jones D.R., Masters A.R., Xu C., Guo Y., Desta Z. Efavirenz primary and secondary metabolism in vitro and in vivo: identification of novel metabolic pathways and cytochrome P450 2A6 as the principal catalyst of efavirenz 7-hydroxylation. Drug Metab Dispos. 2010;38:1218–1229. doi: 10.1124/dmd.109.031393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Xu C., Ogburn E.T., Guo Y., Desta Z. Effects of the CYP2B6*6 allele on catalytic properties and inhibition of CYP2B6 in vitro: implication for the mechanism of reduced efavirenz metabolism and other CYP2B6 substrates in vivo. Drug Metab Dispos. 2012;40:717–725. doi: 10.1124/dmd.111.042416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Haas D.W., Ribaudo H.J., Kim R.B., Tierney C., Wilkinson G.R., Gulick R.M. Pharmacogenetics of efavirenz and central nervous system side effects: an Adult AIDS Clinical Trials Group study. AIDS. 2004;18:2391–2400. [PubMed] [Google Scholar]
  • 84.King J., Aberg J.A. Clinical impact of patient population differences and genomic variation in efavirenz therapy. AIDS. 2008;22:1709–1717. doi: 10.1097/QAD.0b013e32830163ad. [DOI] [PubMed] [Google Scholar]
  • 85.Lubomirov R., di Iulio J., Fayet A., Colombo S., Martinez R., Marzolini C. ADME pharmacogenetics: investigation of the pharmacokinetics of the antiretroviral agent lopinavir coformulated with ritonavir. Pharmacogenet Genom. 2010;20:217–230. doi: 10.1097/FPC.0b013e328336eee4. [DOI] [PubMed] [Google Scholar]
  • 86.Maimbo M., Kiyotani K., Mushiroda T., Masimirembwa C., Nakamura Y. CYP2B6 genotype is a strong predictor of systemic exposure to efavirenz in HIV-infected Zimbabweans. Eur J Clin Pharmacol. 2012;68:267–271. doi: 10.1007/s00228-011-1118-0. [DOI] [PubMed] [Google Scholar]
  • 87.Ribaudo H.J., Liu H., Schwab M., Schaeffeler E., Eichelbaum M., Motsinger-Reif A.A. Effect of CYP2B6, ABCB1, and CYP3A5 polymorphisms on efavirenz pharmacokinetics and treatment response: an AIDS Clinical Trials Group study. J Infect Dis. 2010;202:717–722. doi: 10.1086/655470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Yimer G., Amogne W., Habtewold A., Makonnen E., Ueda N., Suda A. High plasma efavirenz level and CYP2B6*6 are associated with efavirenz-based HAART-induced liver injury in the treatment of naïve HIV patients from Ethiopia: a prospective cohort study. Pharmacogenom J. 2012;12:499–506. doi: 10.1038/tpj.2011.34. [DOI] [PubMed] [Google Scholar]
  • 89.Ribaudo H.J., Haas D.W., Tierney C., Kim R.B., Wilkinson G.R., Gulick R.M. Pharmacogenetics of plasma efavirenz exposure after treatment discontinuation: an Adult AIDS Clinical Trials Group Study. Clin Infect Dis. 2006;42:401–407. doi: 10.1086/499364. [DOI] [PubMed] [Google Scholar]
  • 90.Lubomirov R., Colombo S., di Iulio J., Ledergerber B., Martinez R., Cavassini M. Association of pharmacogenetic markers with premature discontinuation of first-line anti-HIV therapy: an observational cohort study. J Infect Dis. 2011;203:246–257. doi: 10.1093/infdis/jiq043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wyen C., Hendra H., Siccardi M., Platten M., Jaeger H., Harrer T. Cytochrome P450 2B6 (CYP2B6) and constitutive androstane receptor (CAR) polymorphisms are associated with early discontinuation of efavirenz-containing regimens. J Antimicrob Chemother. 2011;66:2092–2098. doi: 10.1093/jac/dkr272. [DOI] [PubMed] [Google Scholar]
  • 92.Cabrera Figueroa S., Fernández de Gatta M., Hernández García L., Domínguez-Gil Hurlé A., Bustos Bernal C., Sepúlveda Correa R. The convergence of therapeutic drug monitoring and pharmacogenetic testing to optimize efavirenz therapy. Ther Drug Monit. 2010;32:579–585. doi: 10.1097/FTD.0b013e3181f0634c. [DOI] [PubMed] [Google Scholar]
  • 93.Nolan D., Phillips E., Mallal S. Efavirenz and CYP2B6 polymorphism: implications for drug toxicity and resistance. Clin Infect Dis. 2006;42:408–410. doi: 10.1086/499369. [DOI] [PubMed] [Google Scholar]
  • 94.Martín A.S., Gómez A.I., García-Berrocal B., Figueroa S.C., Sánchez M.C., Calvo Hernández M.V. Dose reduction of efavirenz: an observational study describing cost-effectiveness, pharmacokinetics and pharmacogenetics. Pharmacogenomics. 2014;15:997–1006. doi: 10.2217/pgs.14.48. [DOI] [PubMed] [Google Scholar]
  • 95.Rivero A., Mira J.A., Pineda J.A. Liver toxicity induced by non-nucleoside reverse transcriptase inhibitors. J Antimicrob Chemother. 2007;59:342–346. doi: 10.1093/jac/dkl524. [DOI] [PubMed] [Google Scholar]
  • 96.Ciccacci C., Borgiani P., Ceffa S., Sirianni E., Marazzi M.C., Altan A.M. Nevirapine-induced hepatotoxicity and pharmacogenetics: a retrospective study in a population from Mozambique. Pharmacogenomics. 2010;11:23–31. doi: 10.2217/pgs.09.142. [DOI] [PubMed] [Google Scholar]
  • 97.Schipani A., Wyen C., Mahungu T., Hendra H., Egan D., Siccardi M. Integration of population pharmacokinetics and pharmacogenetics: an aid to optimal nevirapine dose selection in HIV-infected individuals. J Antimicrob Chemother. 2011;66:1332–1339. doi: 10.1093/jac/dkr087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Heil S.G., van der Ende M.E., Schenk P.W., van der Heiden I., Lindemans J., Burger D. Associations between ABCB1, CYP2A6, CYP2B6, CYP2D6, and CYP3A5 alleles in relation to efavirenz and nevirapine pharmacokinetics in HIV-infected individuals. Ther Drug Monit. 2012;34:153–159. doi: 10.1097/FTD.0b013e31824868f3. [DOI] [PubMed] [Google Scholar]
  • 99.Brown K.C., Hosseinipour M.C., Hoskins J.M., Thirumaran R.K., Tien H.C., Weigel R. Exploration of CYP450 and drug transporter genotypes and correlations with nevirapine exposure in Malawians. Pharmacogenomics. 2012;13:113–121. doi: 10.2217/pgs.11.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wyen C., Hendra H., Vogel M., Hoffmann C., Knechten H., Brockmeyer N.H. Impact of CYP2B6 983 T>C polymorphism on non-nucleoside reverse transcriptase inhibitor plasma concentrations in HIV-infected patients. J Antimicrob Chemother. 2008;61:914–918. doi: 10.1093/jac/dkn029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ariyoshi N., Ohara M., Kaneko M., Afuso S., Kumamoto T., Nakamura H. Q172H replacement overcomes effects on the metabolism of cyclophosphamide and efavirenz caused by CYP2B6 variant with Arg262. Drug Metab Dispos. 2011;39:2045–2048. doi: 10.1124/dmd.111.039586. [DOI] [PubMed] [Google Scholar]
  • 102.Johnson G.G., Lin K., Cox T.F., Oates M., Sibson D.R., Eccles R. CYP2B6*6 is an independent determinant of inferior response to fludarabine plus cyclophosphamide in chronic lymphocytic leukemia. Blood. 2013;122:4253–4258. doi: 10.1182/blood-2013-07-516666. [DOI] [PubMed] [Google Scholar]
  • 103.Chugh R., Wagner T., Griffith K.A., Taylor J.M., Thomas D.G., Worden F.P. Assessment of ifosfamide pharmacokinetics, toxicity, and relation to CYP3A4 activity as measured by the erythromycin breath test in patients with sarcoma. Cancer. 2007;109:2315–2322. doi: 10.1002/cncr.22669. [DOI] [PubMed] [Google Scholar]
  • 104.Zhang J., Tian Q., Yung Chan S., Chuen Li S., Zhou S., Duan W. Metabolism and transport of oxazaphosphorines and the clinical implications. Drug Metab Rev. 2005;37:611–703. doi: 10.1080/03602530500364023. [DOI] [PubMed] [Google Scholar]
  • 105.Aleksa K., Matsell D., Krausz K., Gelboin H., Ito S., Koren G. Cytochrome P450 3A and 2B6 in the developing kidney: implications for ifosfamide nephrotoxicity. Pediatr Nephrol. 2005;20:872–885. doi: 10.1007/s00467-004-1807-3. [DOI] [PubMed] [Google Scholar]
  • 106.Bachanova V., Shanley R., Malik F., Chauhan L., Lamba V., Weisdorf D.J. Cytochrome P450 2B6*5 increases relapse after cyclophosphamide-containing conditioning and autologous transplantation for lymphoma. Biol Blood Marrow Transpl. 2015;21:944–948. doi: 10.1016/j.bbmt.2015.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Helsby N.A., Hui C.Y., Goldthorpe M.A., Coller J.K., Soh M.C., Gow P.J. The combined impact of CYP2C19 and CYP2B6 pharmacogenetics on cyclophosphamide bioactivation. Br J Clin Pharmacol. 2010;70:844–853. doi: 10.1111/j.1365-2125.2010.03789.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Klein K., Lang T., Saussele T., Barbosa-Sicard E., Schunck W.H., Eichelbaum M. Genetic variability of CYP2B6 in populations of African and Asian origin: allele frequencies, novel functional variants, and possible implications for anti-HIV therapy with efavirenz. Pharmacogenet Genom. 2005;15:861–873. doi: 10.1097/01213011-200512000-00004. [DOI] [PubMed] [Google Scholar]
  • 109.Li J., Menard V., Benish R.L., Jurevic R.J., Guillemette C., Stoneking M. Worldwide variation in human drug-metabolism enzyme genes CYP2B6 and UGT2B7: implications for HIV/AIDS treatment. Pharmacogenomics. 2012;13:555–570. doi: 10.2217/pgs.11.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wang J., Sönnerborg A., Rane A., Josephson F., Lundgren S., Ståhle L. Identification of a novel specific CYP2B6 allele in Africans causing impaired metabolism of the HIV drug efavirenz. Pharmacogenet Genom. 2006;16:191–198. doi: 10.1097/01.fpc.0000189797.03845.90. [DOI] [PubMed] [Google Scholar]
  • 111.Zukunft J., Lang T., Richter T., Hirsch-Ernst K.I., Nussler A.K., Klein K. A natural CYP2B6 TATA box polymorphism (−82T>C) leading to enhanced transcription and relocation of the transcriptional start site. Mol Pharmacol. 2005;67:1772–1782. doi: 10.1124/mol.104.008086. [DOI] [PubMed] [Google Scholar]
  • 112.He M.L., Lin C.K., Tong K., Xu B., Sung J.J., Kung H.F. Absence of CYP2B6 promoter −82T>C mutation in Chinese as an additional factor for slow metabolism of drugs commonly used in infections. Eur J Clin Pharmacol. 2006;62:585–586. doi: 10.1007/s00228-006-0163-6. [DOI] [PubMed] [Google Scholar]
  • 113.Li H., Ferguson S.S., Wang H. Synergistically enhanced CYP2B6 inducibility between a polymorphic mutation in CYP2B6 promoter and pregnane X receptor activation. Mol Pharmacol. 2010;78:704–713. doi: 10.1124/mol.110.065185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Zhang H., Sridar C., Kenaan C., Amunugama H., Ballou D.P., Hollenberg P.F. Polymorphic variants of cytochrome P450 2B6 (CYP2B6.4-CYP2B6.9) exhibit altered rates of metabolism for bupropion and efavirenz: a charge-reversal mutation in the K139E variant (CYP2B6.8) impairs formation of a functional cytochrome p450-reductase complex. J Pharmacol Exp Ther. 2011;338:803–809. doi: 10.1124/jpet.111.183111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Nakajima M., Komagata S., Fujiki Y., Kanada Y., Ebi H., Itoh K. Genetic polymorphisms of CYP2B6 affect the pharmacokinetics/pharmacodynamics of cyclophosphamide in Japanese cancer patients. Pharmacogenet Genom. 2007;17:431–445. doi: 10.1097/FPC.0b013e328045c4fb. [DOI] [PubMed] [Google Scholar]
  • 116.Hardwick J.P., Gonzalez F.J., Kasper C.B. Transcriptional regulation of rat liver epoxide hydratase, NADPH-cytochrome P-450 oxidoreductase, and cytochrome P-450b genes by phenobarbital. J Biol Chem. 1983;258:8081–8085. [PubMed] [Google Scholar]
  • 117.Sueyoshi T., Negishi M. Phenobarbital response elements of cytochrome P450 genes and nuclear receptors. Annu Rev Pharmacol Toxicol. 2001;41:123–143. doi: 10.1146/annurev.pharmtox.41.1.123. [DOI] [PubMed] [Google Scholar]
  • 118.Honkakoski P., Moore R., Gynther J., Negishi M. Characterization of phenobarbital-inducible mouse Cyp2b10 gene transcription in primary hepatocytes. J Biol Chem. 1996;271:9746–9753. doi: 10.1074/jbc.271.16.9746. [DOI] [PubMed] [Google Scholar]
  • 119.Lake B.G., Renwick A.B., Cunninghame M.E., Price R.J., Surry D., Evans D.C. Comparison of the effects of some CYP3A and other enzyme inducers on replicative DNA synthesis and cytochrome P450 isoforms in rat liver. Toxicology. 1998;131:9–20. doi: 10.1016/s0300-483x(98)00085-7. [DOI] [PubMed] [Google Scholar]
  • 120.Kolars J.C., Schmiedlin-Ren P., Schuetz J.D., Fang C., Watkins P.B. Identification of rifampin-inducible P450IIIA4 (CYP3A4) in human small bowel enterocytes. J Clin Invest. 1992;90:1871–1878. doi: 10.1172/JCI116064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Maglich J.M., Parks D.J., Moore L.B., Collins J.L., Goodwin B., Billin A.N. Identification of a novel human constitutive androstane receptor (CAR) agonist and its use in the identification of CAR target genes. J Biol Chem. 2003;278:17277–17283. doi: 10.1074/jbc.M300138200. [DOI] [PubMed] [Google Scholar]
  • 122.LeCluyse E.L. Human hepatocyte culture systems for the in vitro evaluation of cytochrome P450 expression and regulation. Eur J Pharm Sci. 2001;13:343–368. doi: 10.1016/s0928-0987(01)00135-x. [DOI] [PubMed] [Google Scholar]
  • 123.Pascussi J.M., Gerbal-Chaloin S., Fabre J.M., Maurel P., Vilarem M.J. Dexamethasone enhances constitutive androstane receptor expression in human hepatocytes: consequences on cytochrome P450 gene regulation. Mol Pharmacol. 2000;58:1441–1450. doi: 10.1124/mol.58.6.1441. [DOI] [PubMed] [Google Scholar]
  • 124.Trottier E., Belzil A., Stoltz C., Anderson A. Localization of a phenobarbital-responsive element (PBRE) in the 5ʹ-flanking region of the rat Cyp2b2 gene. Gene. 1995;158:263–268. doi: 10.1016/0378-1119(94)00916-g. [DOI] [PubMed] [Google Scholar]
  • 125.Park Y., Li H., Kemper B. Phenobarbital induction mediated by a distal CYP2B2 sequence in rat liver transiently transfected in situ. J Biol Chem. 1996;271:23725–23728. doi: 10.1074/jbc.271.39.23725. [DOI] [PubMed] [Google Scholar]
  • 126.Honkakoski P., Zelko I., Sueyoshi T., Negishi M. The nuclear orphan receptor CAR-retinoid X receptor heterodimer activates the phenobarbital-responsive enhancer module of the CYP2B gene. Mol Cell Biol. 1998;18:5652–5658. doi: 10.1128/mcb.18.10.5652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Inoue K., Negishi M. Nuclear receptor CAR requires early growth response 1 to activate the human cytochrome P450 2B6 gene. J Biol Chem. 2008;283:10425–10432. doi: 10.1074/jbc.M800729200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Wang H., Faucette S., Sueyoshi T., Moore R., Ferguson S., Negishi M. A novel distal enhancer module regulated by pregnane X receptor/constitutive androstane receptor is essential for the maximal induction of CYP2B6 gene expression. J Biol Chem. 2003;278:14146–14152. doi: 10.1074/jbc.M212482200. [DOI] [PubMed] [Google Scholar]
  • 129.Honkakoski P., Moore R., Washburn K.A., Negishi M. Activation by diverse xenochemicals of the 51-base pair phenobarbital-responsive enhancer module in the CYP2B10 gene. Mol Pharmacol. 1998;53:597–601. doi: 10.1124/mol.53.4.597. [DOI] [PubMed] [Google Scholar]
  • 130.Sueyoshi T., Kawamoto T., Zelko I., Honkakoski P., Negishi M. The repressed nuclear receptor CAR responds to phenobarbital in activating the human CYP2B6 gene. J Biol Chem. 1999;274:6043–6046. doi: 10.1074/jbc.274.10.6043. [DOI] [PubMed] [Google Scholar]
  • 131.Wei P., Zhang J., Egan-Hafley M., Liang S., Moore D.D. The nuclear receptor CAR mediates specific xenobiotic induction of drug metabolism. Nature. 2000;407:920–923. doi: 10.1038/35038112. [DOI] [PubMed] [Google Scholar]
  • 132.Smirlis D., Muangmoonchai R., Edwards M., Phillips I.R., Shephard E.A. Orphan receptor promiscuity in the induction of cytochromes p450 by xenobiotics. J Biol Chem. 2001;276:12822–12826. doi: 10.1074/jbc.M005930200. [DOI] [PubMed] [Google Scholar]
  • 133.Kodama S., Koike C., Negishi M., Yamamoto Y. Nuclear receptors CAR and PXR cross talk with FOXO1 to regulate genes that encode drug-metabolizing and gluconeogenic enzymes. Mol Cell Biol. 2004;24:7931–7940. doi: 10.1128/MCB.24.18.7931-7940.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Honkakoski P., Negishi M. Regulatory DNA elements of phenobarbital-responsive cytochrome P450 CYP2B genes. J Biochem Mol Toxicol. 1998;12:3–9. doi: 10.1002/(sici)1099-0461(1998)12:1<3::aid-jbt2>3.0.co;2-p. [DOI] [PubMed] [Google Scholar]
  • 135.Maglich J.M., Stoltz C.M., Goodwin B., Hawkins-Brown D., Moore J.T., Kliewer S.A. Nuclear pregnane x receptor and constitutive androstane receptor regulate overlapping but distinct sets of genes involved in xenobiotic detoxification. Mol Pharmacol. 2002;62:638–646. doi: 10.1124/mol.62.3.638. [DOI] [PubMed] [Google Scholar]
  • 136.Li D., Mackowiak B., Brayman T.G., Mitchell M., Zhang L., Huang S.M. Genome-wide analysis of human constitutive androstane receptor (CAR) transcriptome in wild-type and CAR-knockout HepaRG cells. Biochem Pharmacol. 2015;98:190–202. doi: 10.1016/j.bcp.2015.08.087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Zhou J., Zhai Y., Mu Y., Gong H., Uppal H., Toma D. A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway. J Biol Chem. 2006;281:15013–15020. doi: 10.1074/jbc.M511116200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Roth A., Looser R., Kaufmann M., Blättler S.M., Rencurel F., Huang W. Regulatory cross-talk between drug metabolism and lipid homeostasis: constitutive androstane receptor and pregnane X receptor increase Insig-1 expression. Mol Pharmacol. 2008;73:1282–1289. doi: 10.1124/mol.107.041012. [DOI] [PubMed] [Google Scholar]
  • 139.Geick A., Eichelbaum M., Burk O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J Biol Chem. 2001;276:14581–14587. doi: 10.1074/jbc.M010173200. [DOI] [PubMed] [Google Scholar]
  • 140.Wang H., Faucette S.R., Gilbert D., Jolley S.L., Sueyoshi T., Negishi M. Glucocorticoid receptor enhancement of pregnane X receptor-mediated CYP2B6 regulation in primary human hepatocytes. Drug Metab Dispos. 2003;31:620–630. doi: 10.1124/dmd.31.5.620. [DOI] [PubMed] [Google Scholar]
  • 141.Ferguson S.S., LeCluyse E.L., Negishi M., Goldstein J.A. Regulation of human CYP2C9 by the constitutive androstane receptor: discovery of a new distal binding site. Mol Pharmacol. 2002;62:737–746. doi: 10.1124/mol.62.3.737. [DOI] [PubMed] [Google Scholar]
  • 142.Sugatani J., Kojima H., Ueda A., Kakizaki S., Yoshinari K., Gong Q.H. The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR. Hepatology. 2001;33:1232–1238. doi: 10.1053/jhep.2001.24172. [DOI] [PubMed] [Google Scholar]
  • 143.LeCluyse E.L. Pregnane X receptor: molecular basis for species differences in CYP3A induction by xenobiotics. Chem Biol Interact. 2001;134:283–289. doi: 10.1016/s0009-2797(01)00163-6. [DOI] [PubMed] [Google Scholar]
  • 144.Xie W., Barwick J.L., Downes M., Blumberg B., Simon C.M., Nelson M.C. Humanized xenobiotic response in mice expressing nuclear receptor SXR. Nature. 2000;406:435–439. doi: 10.1038/35019116. [DOI] [PubMed] [Google Scholar]
  • 145.Zhang Y.K., Lu H., Klaassen C.D. Expression of human CAR splicing variants in BAC-transgenic mice. Toxicol Sci. 2013;132:142–150. doi: 10.1093/toxsci/kfs321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Kawamoto T., Sueyoshi T., Zelko I., Moore R., Washburn K., Negishi M. Phenobarbital-responsive nuclear translocation of the receptor CAR in induction of the CYP2B gene. Mol Cell Biol. 1999;19:6318–6322. doi: 10.1128/mcb.19.9.6318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Gervot L., Rochat B., Gautier J.C., Bohnenstengel F., Kroemer H., de Berardinis V. Human CYP2B6: expression, inducibility and catalytic activities. Pharmacogenetics. 1999;9:295–306. [PubMed] [Google Scholar]
  • 148.Audet-Walsh É., Anderson A. Dexamethasone induction of murine CYP2B genes requires the glucocorticoid receptor. Drug Metab Dispos. 2009;37:580–588. doi: 10.1124/dmd.108.022772. [DOI] [PubMed] [Google Scholar]
  • 149.Faucette S.R., Wang H., Hamilton G.A., Jolley S.L., Gilbert D., Lindley C. Regulation of CYP2B6 in primary human hepatocytes by prototypical inducers. Drug Metab Dispos. 2004;32:348–358. doi: 10.1124/dmd.32.3.348. [DOI] [PubMed] [Google Scholar]
  • 150.Kawamoto T., Kakizaki S., Yoshinari K., Negishi M. Estrogen activation of the nuclear orphan receptor CAR (constitutive active receptor) in induction of the mouse Cyp2b10 gene. Mol Endocrinol. 2000;14:1897–1905. doi: 10.1210/mend.14.11.0547. [DOI] [PubMed] [Google Scholar]
  • 151.Bièche I., Girault I., Urbain E., Tozlu S., Lidereau R. Relationship between intratumoral expression of genes coding for xenobiotic-metabolizing enzymes and benefit from adjuvant tamoxifen in estrogen receptor alpha-positive postmenopausal breast carcinoma. Breast Cancer Res. 2004;6:R252–R263. doi: 10.1186/bcr784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Lo R., Burgoon L., MacPherson L., Ahmed S., Matthews J. Estrogen receptor-dependent regulation of CYP2B6 in human breast cancer cells. Biochim Biophys Acta. 2010;1799:469–479. doi: 10.1016/j.bbagrm.2010.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Koh K.H., Jurkovic S., Yang K., Choi S.Y., Jung J.W., Kim K.P. Estradiol induces cytochrome P450 2B6 expression at high concentrations: implication in estrogen-mediated gene regulation in pregnancy. Biochem Pharmacol. 2012;84:93–103. doi: 10.1016/j.bcp.2012.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Li L., Li D., Heyward S., Wang H. Transcriptional regulation of CYP2B6 expression by hepatocyte nuclear factor 3β in human liver cells. PLoS One. 2016;11:e0150587. doi: 10.1371/journal.pone.0150587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Jover R., Bort R., Gómez-Lechón M.J., Castell J.V. Re-expression of C/EBPα induces CYP2B6, CYP2C9 and CYP2D6 genes in HepG2 cells. FEBS Lett. 1998;431:227–230. doi: 10.1016/s0014-5793(98)00746-7. [DOI] [PubMed] [Google Scholar]
  • 156.Inoue K., Negishi M. Early growth response 1 loops the CYP2B6 promoter for synergistic activation by the distal and proximal nuclear receptors CAR and HNF4α. FEBS Lett. 2009;583:2126–2130. doi: 10.1016/j.febslet.2009.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Hedrich W.D., Xiao J., Heyward S., Zhang Y., Zhang J., Baer M.R. Activation of the constitutive androstane receptor increases the therapeutic index of CHOP in lymphoma treatment. Mol Cancer Ther. 2016;15:392–401. doi: 10.1158/1535-7163.MCT-15-0667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Griskevicius L., Yasar Ü., Sandberg M., Hidestrand M., Eliasson E., Tybring G. Bioactivation of cyclophosphamide: the role of polymorphic CYP2C enzymes. Eur J Clin Pharmacol. 2003;59:103–109. doi: 10.1007/s00228-003-0590-6. [DOI] [PubMed] [Google Scholar]
  • 159.Clarke L., Waxman D.J. Oxidative metabolism of cyclophosphamide: identification of the hepatic monooxygenase catalysts of drug activation. Cancer Res. 1989;49:2344–2350. [PubMed] [Google Scholar]
  • 160.Yu L.J., Drewes P., Gustafsson K., Brain E.G., Hecht J.E., Waxman D.J. In vivo modulation of alternative pathways of P-450–catalyzed cyclophosphamide metabolism: impact on pharmacokinetics and antitumor activity. J Pharm Exp Ther. 1999;288:928–937. [PubMed] [Google Scholar]
  • 161.Wang D., Li L., Yang H., Ferguson S.S., Baer M.R., Gartenhaus R.B. The constitutive androstane receptor is a novel therapeutic target facilitating cyclophosphamide-based treatment of hematopoietic malignancies. Blood. 2013;121:329–338. doi: 10.1182/blood-2012-06-436691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Rakhmanina N.Y., van den Anker J.N. Efavirenz in the therapy of HIV infection. Expert Opin Drug Metab Toxicol. 2010;6:95–103. doi: 10.1517/17425250903483207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Robertson S.M., Maldarelli F., Natarajan V., Formentini E., Alfaro R.M., Penzak S.R. Efavirenz induces CYP2B6-mediated hydroxylation of bupropion in healthy subjects. J Acquir Immune Defic Syndr. 2008;49:513–519. doi: 10.1097/QAI.0b013e318183a425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Xu C., Desta Z. In vitro analysis and quantitative prediction of efavirenz inhibition of eight cytochrome P450 (CYP) enzymes: major effects on CYPs 2B6, 2C8, 2C9 and 2C19. Drug Metab Pharmacokinet. 2013;28:362–371. doi: 10.2133/dmpk.dmpk-12-rg-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Medhi B., Patyar S., Rao R.S., Byrav D.S., Prakash A. Pharmacokinetic and toxicological profile of artemisinin compounds: an update. Pharmacology. 2009;84:323–332. doi: 10.1159/000252658. [DOI] [PubMed] [Google Scholar]
  • 166.Svensson U.S., Ashton M., Hai T.N., Bertilsson L., Dinh X.H., Nguyen V.H. Artemisinin induces omeprazole metabolism in human beings. Clin Pharmacol Ther. 1998;64:160–167. doi: 10.1016/S0009-9236(98)90149-7. [DOI] [PubMed] [Google Scholar]
  • 167.Davidson J. Seizures and bupropion: a review. J Clin Psychiatry. 1989;50:256–261. [PubMed] [Google Scholar]
  • 168.Kotlyar M., Brauer L.H., Tracy T.S., Hatsukami D.K., Harris J., Bronars C.A. Inhibition of CYP2D6 activity by bupropion. J Clin Psychopharmacol. 2005;25:226–229. doi: 10.1097/01.jcp.0000162805.46453.e3. [DOI] [PubMed] [Google Scholar]
  • 169.Pollock B.G., Sweet R.A., Kirshner M., Reynolds CF 3rd. Bupropion plasma levels and CYP2D6 phenotype. Ther Drug Monit. 1996;18:581–585. doi: 10.1097/00007691-199610000-00010. [DOI] [PubMed] [Google Scholar]
  • 170.Idvall J., Aronsen K.F., Stenberg P., Paalzow L. Pharmacodynamic and pharmacokinetic interactions between ketamine and diazepam. Eur J Clin Pharmacol. 1983;24:337–343. doi: 10.1007/BF00610051. [DOI] [PubMed] [Google Scholar]
  • 171.Lo J.N., Cumming J.F. Interaction between sedative premedicants and ketamine in man in isolated perfused rat livers. Anesthesiology. 1975;43:307–312. doi: 10.1097/00000542-197509000-00007. [DOI] [PubMed] [Google Scholar]
  • 172.Totah R.A., Sheffels P., Roberts T., Whittington D., Thummel K., Kharasch E.D. Role of CYP2B6 in stereoselective human methadone metabolism. Anesthesiology. 2008;108:363–374. doi: 10.1097/ALN.0b013e3181642938. [DOI] [PubMed] [Google Scholar]
  • 173.Karch S.B., Stephens B.G. Toxicology and pathology of deaths related to methadone: retrospective review. West J Med. 2000;172:11–14. doi: 10.1136/ewjm.172.1.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Eap C.B., Crettol S., Rougier J.S., Schläpfer J., Sintra Grilo L., Déglon J.J. Stereoselective block of hERG channel by (S)-methadone and QT interval prolongation in CYP2B6 slow metabolizers. Clin Pharmacol Ther. 2007;81:719–728. doi: 10.1038/sj.clpt.6100120. [DOI] [PubMed] [Google Scholar]
  • 175.Holmberg L., Odar-Cederlöf I., Boréus L.O., Heyner L., Ehrnebo M. Comparative disposition of pethidine and norpethidine in old and young patients. Eur J Clin Pharmacol. 1982;22:175–179. doi: 10.1007/BF00542464. [DOI] [PubMed] [Google Scholar]
  • 176.McHugh G.J. Norpethidine accumulation and generalized seizure during pethidine patient-controlled analgesia. Anaesth Intensive Care. 1999;27:289–291. [PubMed] [Google Scholar]
  • 177.Stone P.A., Macintyre P.E., Jarvis D.A. Norpethidine toxicity and patient controlled analgesia. Br J Anaesth. 1993;71:738–740. doi: 10.1093/bja/71.5.738. [DOI] [PubMed] [Google Scholar]
  • 178.Sridar C., Kenaan C., Hollenberg P.F. Inhibition of bupropion metabolism by selegiline: mechanism-based inactivation of human CYP2B6 and characterization of glutathione and peptide adducts. Drug Metab Dispos. 2012;40:2256–2266. doi: 10.1124/dmd.112.046979. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Acta Pharmaceutica Sinica. B are provided here courtesy of Elsevier

RESOURCES