Skip to main content
Journal of Antimicrobial Chemotherapy logoLink to Journal of Antimicrobial Chemotherapy
. 2014 Apr 11;69(8):2175–2182. doi: 10.1093/jac/dku110

Secondary metabolism pathway polymorphisms and plasma efavirenz concentrations in HIV-infected adults with CYP2B6 slow metabolizer genotypes

David W Haas 1,*, Awewura Kwara 2, Danielle M Richardson 1, Paxton Baker 1, Ioannis Papageorgiou 3, Edward P Acosta 4, Gene D Morse 5, Michael H Court 3
PMCID: PMC4100708  PMID: 24729586

Abstract

Objectives

Efavirenz is widely prescribed for HIV-1 infection, and CYP2B6 polymorphisms 516G→T and 983T→C define efavirenz slow metabolizer genotypes. To identify genetic predictors of higher plasma efavirenz concentrations beyond these two common functional alleles, we characterized associations with mid-dosing interval efavirenz concentrations in 84 HIV-infected adults, all carrying two copies of these major loss-of-function CYP2B6 alleles.

Methods

Study participants had been randomized to efavirenz-containing regimens in prospective clinical trials and had available plasma efavirenz assay data. Analyses focused on secondary metabolism pathway polymorphisms CYP2A6 -48T→G (rs28399433), UGT2B7 735A→G (rs28365062) and UGT2B7 802T→C (rs7439366). Exploratory analyses also considered 196 polymorphisms and 8 copy number variants in 41 drug metabolism/transport genes. Mid-dosing interval efavirenz concentrations at steady-state were obtained ≥8 h but <19 h post-dose. Linear regression was used to test for associations between polymorphisms and log-transformed efavirenz concentrations.

Results

Increased efavirenz concentrations were associated with CYP2A6 -48T→G in all subjects (P = 3.8 × 10−4) and in Black subjects (P = 0.027) and White subjects (P = 0.0011) analysed separately; and with UGT2B7 735 G/G homozygosity in all subjects (P = 0.006) and in Black subjects (P = 0.046) and White subjects (P = 0.062) analysed separately. In a multivariable model, CYP2A6 -48T→G and UGT2B7 735 G/G homozygosity remained significant (P < 0.05 for each). No additional polymorphisms or copy number variants were significantly associated with efavirenz concentrations.

Conclusions

Among individuals with a CYP2B6 slow metabolizer genotype, CYP2A6 and possibly UGT2B7 polymorphisms contribute to even higher efavirenz concentrations.

Keywords: pharmacogenomics, pharmacogenetics, pharmacokinetics, antiretroviral therapy, non-nucleoside reverse transcriptase inhibitor

Introduction

The once-daily non-nucleoside reverse transcriptase inhibitor efavirenz is one of the most frequently prescribed antiretrovirals worldwide. It is included among recommended first-line regimens for HIV-1 infection,1 based on data from many prospective, randomized clinical trials.27 Efavirenz is metabolized primarily by cytochrome P450 (CYP) 2B6, with minor metabolism by CYP2A6 and CYP3A4/5,8,9 and direct N-glucuronidation by UDP-glucuronosyltransferase (UGT) 2B7.10

Three CYP2B6 polymorphisms, 516G→T (rs3745274),1116 983T→C (rs28399499)1619 and 15582C→T (rs4803419), have consistently been associated with increased plasma efavirenz exposure.16 The greater frequency of CYP2B6 516G→T with African ancestry than with European ancestry20 largely explains the somewhat greater mean plasma efavirenz trough concentrations (Cmin) with African ancestry.21 The per allele effect of CYP2B6 983T→C on efavirenz concentrations is somewhat greater than that of 516G→T,16 although 983T→C is far less frequent and appears only to be found with African ancestry.20 The per allele effect of CYP2B6 15582C→T, which is frequent with European and Asian ancestry,20 is modest compared with 516G→T.16 These three polymorphisms stratify patients into 10 plasma trough concentration subgroups with medians that span an ∼10-fold range.16 The top three strata (i.e. CYP2B6 slow metabolizer genotypes) are defined by either 516 T/T homozygosity, dual 516 G/T–983 C/T heterozygosity, or 983 C/C homozygosity. Because CYP2B6 516T, 983C and 15582T reside on mutually exclusive haplotypes, 15582T is absent in CYP2B6 slow metabolizer genotypes. These three polymorphisms explained ∼35% of overall interindividual variability in efavirenz estimated Cmin.16

Additional CYP2B6 polymorphisms suggested to affect CYP2B6 activity either have not predicted plasma efavirenz exposure22,23 or have been extremely infrequent.18,22 Polymorphisms in genes beyond CYP2B6 reported to affect interindividual variability in efavirenz pharmacokinetics include CYP2A6,24,25 UGT2B7,25 CYP3A511 and NR1I326,27 (which encodes the constitutive androstane receptor), but findings have seemed to be inconsistent, perhaps a consequence of differences in study design. Kwara et al.25,28 reported that among 94 HIV-infected Ghanaian patients, increased mid-dosing interval plasma efavirenz concentrations were associated with CYP2A6 -48T→G (rs28399433, *9) or *17 carrier status and UGT2B7*1a status, in addition to CYP2B6 516G→T and 983T→C. Similarly, di Iulio et al.24 reported that in a predominantly Caucasian cohort of 169 HIV-infected individuals, CYP2A6 loss-of-function alleles (primarily CYP2A6 -48T→G) had an effect on efavirenz exposure, but only with concomitant CYP2B6 loss-of-function genotypes. However, in a genome-wide association study (GWAS) of efavirenz estimated Cmin values, which involved 856 ACTG protocol participants of various ancestries,16 no independent associations were found with additional polymorphisms in or beyond CYP2B6, including CYP2A6 -48T→G (P = 0.82). Similarly, a recent report by Sarfo et al.29 indicated that among 473 HIV-infected Ghanaians, CYP2A6 -48T→G was associated with mid-dose plasma efavirenz concentrations on univariate analysis only (P = 0.002), but not after controlling for 516G→T and 983T→C (P = 0.6), suggesting that the CYP2A6 -48T→G association was not independent of CYP2B6 polymorphisms.

There were several limitations to the above GWAS. The method used to estimate efavirenz Cmin did not allow differences between some concentrations within the top concentration strata to be discriminated; subgroup analyses were not performed solely among subjects with a CYP2B6 slow metabolizer genotype; and previously implicated UGT2B7 polymorphisms were not genotyped.16 To assess whether higher plasma efavirenz concentrations are affected by polymorphisms in genes relevant to drug absorption, distribution, metabolism and elimination (ADME) beyond CYP2B6, the present study focused on ACTG protocol participants with a CYP2B6 slow metabolizer genotype. Plasma efavirenz concentrations in this group should be exquisitely sensitive to loss-of-function polymorphisms in minor pathways of efavirenz metabolism, as previously suggested.24 We only considered efavirenz concentrations from 8 to 19 h post-dose, and performed targeted genotyping of UGT2B7 polymorphisms as well as an additional 194 polymorphisms and 8 gene copy number variants in 40 ADME genes.

Materials and methods

Study participants

Most individuals in these analyses were also included in the previously reported GWAS,16 which involved treatment-naive individuals who were randomized to efavirenz-containing regimens in ACTG studies 384,30 A5095 (including its neurologic substudy A5097s)3,31 and A5202,7 with DNA obtained under protocol A512832 and with available plasma efavirenz assay data. We limited the present analyses to individuals with a CYP2B6 slow metabolizer genotype (516TT, 516T/983C or 983CC). Self-identified race/ethnicity categories ‘white, non-Hispanic’, ‘black, non-Hispanic’ and ‘Hispanic’ are hereafter referred to as White, Black and Hispanic, respectively. This study complied with the Helsinki Declaration, was approved by institutional review boards for each site and subjects gave written informed consent.

Genotyping

Genotypes for CYP2B6 516G→T, 983T→C and CYP2A6 -48T→G were available from a custom-designed MassARRAY® iPLEX Gold assay (Sequenom Inc.), as previously described,16 and confirmed by genotyping with the iPLEX ADME PGx panel (Sequenom Inc.). Genotypes of five UGT2B7 polymorphisms, including 735A→G (rs28365062), 801A→T (rs7438284), 802T→C (rs7439366), 870 + 115G→A (rs7441750) and 870 + 148G→A (rs7441774) were determined using genomic PCR amplification and direct sequencing as described previously,28 with minor modifications using Platinum® PCR SuperMix, High Fidelity (Life Technologies Inc.). The PCR reactions were denatured initially at 94°C for 2 min, then 35 cycles of 94°C for 30 s, 55°C for 30 s and 68°C for 1 min, followed by 68°C for 5 min. Four of the five UGT2B7 polymorphisms (rs7438284, rs7439366, rs7441750, rs7441774) were in complete linkage disequilibrium in our subjects, so analyses only included 735A→G (rs28365062) and 802T→C (rs7439366). These polymorphisms were chosen for analysis since they discriminate the three most common UGT2B7 alleles identified to date, including UGT2B7*1a (reference), UGT2B7*1c (735A→G) and UGT2B7*2 (802T→C).28 Furthermore, 802T→C is non-synonymous, causing a histidine to tyrosine transition at codon 268, while 735A→G was associated with altered clearance and glucuronidation of zidovudine, a specific UGT2B7 substrate.33 Two NR1I3 polymorphisms previously associated with efavirenz plasma concentrations, rs230742426 and rs3003596,27 were genotyped by TaqManTM assay with ABI PRISM 7900 HT Sequence Detection System (Applied Biosystems Inc., Foster City, CA). From iPLEX ADME PGx, we only included data from specimens with >95% genotyping efficiency and polymorphisms with >95% genotyping efficiency. Laboratory personnel with no knowledge of the clinical data performed the genotyping. Within each race/ethnicity group, each iPLEX ADME PGx polymorphism was in Hardy–Weinberg equilibrium after Bonferroni correction for multiple comparisons. All assays were run in duplicate.

Plasma efavirenz concentrations

Plasma efavirenz concentrations were assayed by HPLC at treatment weeks 1, 4, 12 and 24, as described elsewhere.34 Sampling times were not pre-specified, and time of prior dose was by self-report. We only included mid-dosing interval efavirenz concentrations obtained ≥8 h but <19 h post-dose. Efavirenz is typically taken at bedtime to minimize CNS side effects, and mid-dose concentrations are more convenient for therapeutic drug monitoring than AUC. At steady-state, efavirenz mid-dose concentrations (∼12 h post-dose) very strongly correlate with AUC values.35 We also only included subjects with at least two efavirenz determinations within this window, and with relatively consistent values. For subjects with only two such determinations, subjects with a difference between log10 efavirenz concentrations ≥0.3 μg/mL were excluded. For subjects with more than two determinations, subjects with a standard deviation of log10 efavirenz concentrations ≥0.2 μg/mL were excluded. These standard deviation cut-offs, chosen based on visual inspection of frequency distribution plots, only excluded a few outlier subjects with extreme inter-assay variability.

Statistical analysis

For each subject, the mean of log10-transformed efavirenz concentrations was used in analyses. Linear regression was used to test for association between polymorphisms and efavirenz concentrations. Primary analyses used additive genetic models. Exploratory post hoc analyses also considered recessive models. For CYP2A6 -48T→G, UGT2B7 735A→G, 802T→C and composite CYP2B6 516/983 genotype, nominal two-way P values <0.05 were considered significant. For other polymorphisms, P values were Bonferroni corrected for multiple comparisons. Statistical analyses were performed with PLINK software v1.0736 and with STATA software v13.0.37

Results

Study subjects

These analyses included 84 subjects, of which 73 (86.9%) were male, 44 (52.4%) Black, 24 (28.6%) White and 16 (19.1%) Hispanic. Mean age (±SD) was 38.3 ± 9.2 years. The mean of log10 plasma efavirenz concentrations (of each subject's mean value) was 0.79 log10 ± 0.19 log10 μg/mL. On a linear scale, median was 5.8 μg/mL, minimum 2.4 μg/mL, IQR 4.3–8.5 μg/mL and maximum 17.4 μg/mL. Mean time post-dose was 12.8 ± 2.0 h (based on within-subject means).

Genetic associations with mid-dose efavirenz concentrations

All 84 subjects had a CYP2B6 slow metabolizer genotype, including 71 (84.5%) homozygous for CYP2B6 516TT, 12 (14.3%) heterozygous for CYP2B6 516GT with 983TC and 1 (1.2%) homozygous for CYP2B6 983CC. There was a trend toward association between increasing CYP2B6 983C allele copy number and higher efavirenz concentrations in all subjects (P = 0.12). To address possible confounding by genetic substructure, we analysed each race/ethnicity group separately. This relationship was also apparent in Black subjects analysed separately. The one Hispanic subject with a 983C allele self-identified as ‘Black, Hispanic’. No White subject had a 983C allele (Figure 1).

Figure 1.

Figure 1.

Plasma efavirenz (EFV) concentrations by CYP2B6 516/983 slow metabolizer genotype subgroup. On each graph, each marker represents a different subject, each with a CYP2B6 slow metabolizer genotype. Panels represent all subjects (top left) and self-identified Black subjects (top right), White subjects (bottom left) and Hispanic subjects (bottom right). Each marker represents the median of at least two log10-transformed efavirenz determinations on plasma samples obtained 8–19 h post-dose. Horizontal bars are medians and IQRs. Median values are shown below the markers.

Among all subjects, CYP2A6 -48T→G was significantly associated with increased efavirenz concentrations by univariate analysis (P = 3.8 × 10−4). This association was also apparent in Black subjects (P = 0.027) and White subjects (P = 0.0011) analysed separately, but not in Hispanic subjects (P = 0.70), perhaps due to small sample size (Figure 2). Of note, the only subject with apparent homozygosity for CYP2A6 -48 G/G was also heterozygous for deletion of the CYP2A6 gene, and so this subject had only one CYP2A6 -48G allele (see later regarding iPLEX ADME PGx genotypes). Among all subjects, UGT2B7 735A→G was not associated with increased efavirenz concentrations by univariate analysis (P = 0.32) or in Black, White and Hispanic subjects analysed separately (P > 0.1 for each). However, among all subjects, using a recessive genetic model, UGT2B7 735 G/G homozygosity was associated with higher efavirenz concentrations than the combined A/A and A/G genotypes (P = 0.0063), although only three subjects were homozygous for G/G. This association appeared to be consistent in Black subjects (P = 0.046) and White subjects (P = 0.062) analysed separately (Figure 3). There was no significant association between either UGT2B7 802T→C, NR1I3 rs2307424 or NR1I3 rs3003596 and efavirenz concentrations by univariate analysis in all subjects, or in Black, White or Hispanic subjects analysed separately (P > 0.3 for each analysis; Figures S1 to S3, available as Supplementary data at JAC Online).

Figure 2.

Figure 2.

Plasma efavirenz (EFV) concentrations by CYP2A6 -48T→G genotype. On each graph, each marker represents a different subject. Panels represent all subjects (top left) and self-identified Black subjects (top right), White subjects (bottom left) and Hispanic subjects (bottom right). All have a CYP2B6 slow metabolizer genotype (i.e. 516TT, 516T/983C or 983CC). Genotypes for CYP2A6 -48T→G (rs28399433) are shown. Each marker represents the median of at least two log10-transformed efavirenz determinations on plasma samples obtained 8–19 h post-dose. Horizontal bars are medians and IQRs. Median values are shown below the markers.

Figure 3.

Figure 3.

Plasma efavirenz (EFV) concentrations by UGT2B7 735A→G genotype. On each graph, each marker represents a different subject. Panels represent all subjects (top left) and self-identified Black subjects (top right), White subjects (bottom left) and Hispanic subjects (bottom right). All have a CYP2B6 slow metabolizer genotype (i.e. 516TT, 516T/983C or 983CC). Each marker represents the median of at least two log10-transformed efavirenz determinations on plasma samples obtained 8–19 h post-dose. Genotypes for UGT2B7 735A→G (rs28365062) are shown. Horizontal bars are medians and IQRs. Median values are shown below the markers.

Multivariable models were used to test for independent associations. Among all 84 subjects, in additive genetic models that controlled for CYP2B6 slow metabolizer subgroup (516TT, 516T/983C and 983CC), efavirenz concentrations remained significantly associated with CYP2A6 -48T→G (P = 0.0010) but not with UGT2B7 735A→G (P = 0.29), UGT2B7 802T→C (P = 0.89), NR1I3 rs2307424 (P = 0.49) or rs3003596 (P = 0.43); in a recessive genetic model, UGT2B7 735 G/G homozygosity was associated with efavirenz concentrations (P = 0.0085). In a multivariable model that controlled for both CYP2B6 slow metabolizer subgroup and CYP2A6 -48T→G genotype, CYP2A6 -48T→G (P = 0.0036) and UGT2B7 735A→G (P = 0.025, recessive model), but not CYP2B6 516/983 subgroup (P = 0.33), were significantly associated with efavirenz concentrations.

In separate univariate linear regression models, CYP2A6 -48T→G explained 15% of interindividual variance in log10 plasma efavirenz concentrations, UGT2B7 735 (G/G homozygosity) explained 9%, CYP2B6 983T→C explained 3% and time post-dose explained 2%. A multivariable model that included CYP2A6 -48T→G and UGT2B7 735 (G/G homozygosity) explained 21% of interindividual variance in log10 plasma efavirenz concentrations. Inclusion of CYP2B6 983T→C only increased this to 22%, and time post-dose only to 24%. The final model, which included CYP2A6 -48T→G, UGT2B7 735 G/G homozygosity and CYP2B6 983T→C, is shown in Table 1.

Table 1.

Multivariate linear regression model for association between genetic polymorphisms and log10 plasma efavirenz concentrations

Variant Coefficient 95% CI Standard error t P value
CYP2A6 -48T→G 0.135 0.045, 0.224 0.045 3.00 0.004
UGT2B7 735 G/Ga 0.232 0.030, 0.434 0.102 2.29 0.025
CYP2B6 983T→C 0.046 −0.047, 0.140 0.047 0.99 0.326

aUGT2B7 735A→G was analysed for recessive effect (i.e. G/G versus combined A/A and A/G).

Associations with additional polymorphisms were explored based on iPLEX ADME PGx genotypes, which were available for 68 subjects (21 White, 39 Black and 8 Hispanic) representing 194 loci in 40 genes (of which 103 were polymorphic and 74 had minor allele frequencies >5%). This platform also assayed for copy number variants of CYP2A6, CYP2B6, CYP2D6, GSTM1, GSTT1, GSTT2B, SULT1A1 and UGT2B17. Information regarding iPLEX ADME PGx genes, polymorphisms and minor allele frequencies are shown in Table S1 (available as Supplementary data at JAC Online). In univariate analyses with correction for multiple comparisons, only CYP2A6 -48T→G in the iPLEX ADME PGx panel was significantly associated with plasma efavirenz concentrations (P = 5.8 × 10−4). This association was also apparent in Whites (P = 0.0063) and Blacks (P = 0.024) analysed separately. Considering nominal significance (without correcting for multiple comparisons), six polymorphisms in CYP2C9, CYP2C19, CYP3A5, GSTM1 and SULT1A1 had P values between 0.02 and 0.05, but associations were not consistent in White and Black subjects analysed separately. These included the CYP3A5 loss-of-function polymorphism rs776746 (CYP3A5*3), which had nominal significance for association in all subjects (P = 0.048) but not in White subjects (P = 0.54) or Black subjects (P = 0.80) analysed separately. In a multivariable analysis that controlled for CYP2A6 -48T→G, the lowest nominal P value was in GSTM1 (P = 0.040). None was significant after correcting for multiple comparisons. Association analysis results for iPLEX ADME PGx genotypes and gene copy number variants are shown in Tables S2 to S6 (available as Supplementary data at JAC Online). Frequencies of gene copy number variants are shown in Table S7.

For three subjects heterozygous for a CYP2A6 gene deletion (one with a CYP2A6 -48G allele, as noted above), there was no significant association with plasma efavirenz concentrations. (This gene deletion is also known as CYP2A6*4).38 The two individuals with a single CYP2A6 -48T allele gene copy did not have particularly high plasma efavirenz concentrations (4.4 μg/mL and 9.3 μg/mL). Similarly, two subjects heterozygous for CYP2A6 rs28399454 A/G (also known as CYP2A6*17)39 did not have particularly high plasma efavirenz concentrations (4.0 μg/mL and 5.3 μg/mL). A Q–Q plot of observed versus expected –log10 P values showed that only the CYP2A6 -48T→G P value was substantially less than expected by chance (Figure 4).

Figure 4.

Figure 4.

Q–Q plot of observed versus expected –log10 P values for iPLEX ADME PGx panel polymorphisms. Each marker represents a different polymorphism. The arrowhead identifies CYP2A6 -48T→G (rs28399433).

Sensitivity analyses assessed the effect of censoring single plasma efavirenz concentration values from selected analyses. With removal of the one subject with a single CYP2A6 -48G allele, the univariate association with CYP2A6 -48T→G remained significant (P = 9.4 × 10−4). With removal of the one subject with the highest efavirenz value (heterozygous for CYP2A6 -48 G/T, homozygous for UGT2B7 735 G/G), the association with CYP2A6 -48T→G again remained significant (P = 0.0014), but the association with UGT2B7 735A→G did not (recessive model, P = 0.11).

Discussion

Efavirenz is one of the most frequently prescribed antiretrovirals worldwide. The present study shows that, among individuals with already increased efavirenz concentrations due to a CYP2B6 slow metabolizer genotype, CYP2A6 -48T→G is associated with even greater increases in plasma efavirenz concentrations. This polymorphism, which defines the CYP2A6*9 haplotype, disrupts the TATA box and is associated with reduced CYP2A6 expression.40,41 This association was seen in all subjects and in Black subjects and White subjects analysed separately. In multivariable analyses, the association with CYP2A6 -48T→G was independent of CYP2B6 slow metabolizer genotype subgroup (i.e. 516TT, 516T/983C and 983CC). The validity of this association is further supported by the Q–Q plot, which clearly shows that CYP2A6 -48T→G stands out among the many ADME polymorphisms genotyped. A possible association between UGT2B7 735A→G and higher plasma efavirenz concentrations was only seen with 735 G/G homozygosity, which was independent of both CYP2A6 -48T→G and CYP2B6 slow metabolizer genotype subgroup in multivariable analyses. However, loss of UGT2B7 735A→G significance in a sensitivity analysis that censored a single subject shows the tenuousness of this association. In addition, increasing number of CYP2B6 983C alleles tended toward association with higher plasma efavirenz concentrations, consistent with previous reports.16

The present analyses only included individuals with a CYP2B6 slow metabolizer genotype, because only in the setting of markedly reduced CYP2B6 activity are minor metabolism pathway effects most apparent, as previously suggested.24 Although UGT2B7 can directly N-glucuronidate efavirenz in vitro,10 our finding that an association with UGT2B7 735A→G was only seen with G/G homozygosity suggests that the effect (if any) of this polymorphism is weak. This is consistent with data from a study of 10 HIV-negative healthy volunteers (eight Caucasians and two African Americans), which suggested that the contribution of UGT2B7 to efavirenz metabolism in vivo is minimal.42 However, there were likely no CYP2B6 slow metabolizer genotypes in the latter study (although genotyping was not done), and the contribution of UGT2B7 might be more substantial in such individuals.

We found no additional ADME polymorphisms in or beyond CYP2B6, CYP2A6 and UGT2B7 associated with plasma efavirenz concentrations. This includes CYP3A5*3 (rs776746), which markedly reduces CYP3A5 expression and was very frequent in our study subjects. We conclude that CYP3A5 either does not contribute substantially to efavirenz metabolism in vivo or that a compensatory increase in CYP3A4 activity (or of some other enzyme) in these subjects offsets the decreased CYP3A5 activity. We also did not replicate previously reported associations with NR1I3 polymorphisms.26,27 This may reflect the fact that we only studied individuals with a CYP2B6 slow metabolizer genotype, in whom gene expression may be less influenced by nuclear receptor activity. We cannot explain the lack of association between heterozygosity for either CYP2A6*4 or CYP2A6*17 and plasma efavirenz concentrations, but this may reflect low statistical power, with only five subjects carrying these alleles. It is conceivable that the association between CYP2A6 -48T→G and decreased plasma efavirenz concentrations is somehow mediated through a further reduction in CYP2B6 (rather than CYP2A6) expression and/or activity among individuals with a CYP2B6 slow metabolizer genotype, as this polymorphism is ∼140 kb upstream of CYP2B6 on chromosome 19. This would be consistent with the lack of association with CYP2A6*4 and CYP2A6*17 in the present study, but would not be consistent with the lack of association between CYP2A6 -48T→G and plasma efavirenz concentrations in subjects with a CYP2B6 extensive or intermediate metabolizer genotype in the previous GWAS that involved 856 subjects.16

This study has potential implications for efavirenz side effects. Higher plasma efavirenz concentrations have been associated with CNS side effects in some13,29,43,44 but not all studies.4547 In ACTG protocol A5097s (a double-blinded, placebo-controlled study specifically designed to assess efavirenz CNS symptoms), efavirenz was significantly associated with increased CNS symptoms compared with placebo within the first week of treatment initiation but not at weeks 4, 12 and 24.31 Changes in efavirenz-associated neurological symptoms within the first week of treatment initiation were correlated with week 1 plasma efavirenz plasma concentrations31 and with CYP2B6 516G→T genotype.11 A larger ACTG analysis suggested an association between increased CNS side effects and a CYP2B6 slow metabolizer genotype in Whites but not in Blacks or Hispanics.19 A subsequent Swiss HIV Cohort Study analysis involving a largely Caucasian population suggested increased risk of efavirenz discontinuation in 13 individuals with various combinations of CYP2B6, CYP2A6 and CYP3A4 polymorphisms.48

The present study also has potential implications for efavirenz dosing-reduction strategies, as has been proposed to reduce side effects and/or cost.49,50 The present study suggests that among individuals with a CYP2B6 slow metabolizer genotype and concomitant CYP2A6 -48T→G (and possibly UGT2B7 735 G/G homozygosity), marked dose reduction will still maintain ample plasma efavirenz exposure to control HIV-1 replication.

This study had limitations. We only studied individuals with a CYP2B6 slow metabolizer genotype. However, our previous GWAS showed no significant association between CYP2A6 -48T→G and plasma efavirenz exposure among ACTG protocol participants in multivariable analyses that controlled for 516G→T, 983T→C and 15582C→T.16 Thus, CYP2A6 -48T→G, and possibly UGT2B7 735A→G, are only relevant to efavirenz exposure in individuals with markedly reduced CYP2B6 activity, as previously suggested.24 Also, because this study included few Hispanic subjects and no Asian subjects, we cannot generalize these findings to other populations.

In summary, knowledge of CYP2A6 -48T→G genotype improves stratification of plasma efavirenz concentrations in individuals with a CYP2B6 slow metabolizer genotype. Stratification may also be improved by UGT2B7 genotype, although this association was less robust. These findings support the importance of minor metabolism pathways of efavirenz metabolism in CYP2B6 slow metabolizers, and suggest that individuals with CYP2A6 -48T→G, and possibly UGT2B7 735 G/G, are at increased risk for considerably higher plasma efavirenz exposure.

Funding

This project was supported by Award Number U01AI068636 from the National Institute of Allergy and Infectious Diseases and supported by National Institute of Mental Health (NIMH), National Institute of Dental and Craniofacial Research (NIDCR).

Grant support included AI-077505, TR-000445, AI-54999 (D. W. H.), BRS-ACURE-06-00140-T001 (G. D. M.), R01-GM061834 and R01-GM102130 (M. H. C.). Study drugs were provided by Bristol-Myers Squibb Company (Princeton, NJ), Gilead Sciences (Foster City, CA), GlaxoSmithKline, Inc (Research Triangle Park, NC), and Boehringer Ingelheim (Ridgefield, CT).

Clinical Research Sites that participated in ACTG protocols ACTG 384, A5095, or A5202, and collected DNA under protocol A5128, were supported by the following grants from NIH National Institute of Allergy and Infectious Diseases (NIAID): AI-069532, AI-069484, AI-069432, AI-069450, AI-069495, AI-069434, AI-069424, AI-069439, AI-069467, AI-069423, AI-069513, AI-069477, AI-069465, AI-069419, AI-069502, AI-069474, AI-069472, AI-069501, AI-069418, AI-069494, AI-069471, AI-069511, AI-069452, AI-069428, AI-069556, AI-069415, AI-032782, AI-046376, AI-046370, AI-038858, AI-034853, AI-027661, AI-025859, AI-069470, AI-027675, AI-073961, AI-050410, AI-045008, AI-050409, AI-072626, AI-069447, AI-027658, AI-027666, AI-058740 and AI-025868, and by the following grants from NIH National Center for Research Resources (NCRR): RR-000046, RR-000425, RR-025747, RR-025777, RR-025780, RR-024996, RR-024160, RR-023561, RR-024156, RR-024160 and RR-024160.

Transparency declarations

D. W. H. has been principal investigator on a research grant to Vanderbilt from Merck, and has been a consultant to Merck. The remaining authors have none to declare.

Supplementary data

Tables S1 to S7 and Figures S1 to S3 are available as Supplementary data at JAC Online (http://jac.oxfordjournals.org/).

Disclaimer

The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute of Allergy and Infectious Diseases or the National Institutes of Health.

Supplementary Material

Supplementary Data

Acknowledgements

The authors are grateful to the many persons with HIV infection who volunteered for ACTG clinical trials and A5128. The authors also acknowledge the contributions of study teams and site staff for protocols, especially ACTG 384, A5095, A5097s, A5202 and A5128.

References

  • 1.The Panel on Clinical Practices for Treatment of HIV Infection. Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents. http://www.aidsinfo.nih.gov/contentfiles/lvguidelines/adultandadolescentgl.pdf . Updated February 12, 2013 (8 January 2014, date last accessed)
  • 2.van Leth F, Phanuphak P, Ruxrungtham K, et al. Comparison of first-line antiretroviral therapy with regimens including nevirapine, efavirenz, or both drugs, plus stavudine and lamivudine: a randomised open-label trial, the 2NN Study. Lancet. 2004;363:1253–63. doi: 10.1016/S0140-6736(04)15997-7. [DOI] [PubMed] [Google Scholar]
  • 3.Gulick RM, Ribaudo HJ, Shikuma CM, et al. Three- vs four-drug antiretroviral regimens for the initial treatment of HIV-1 infection: a randomized controlled trial. JAMA. 2006;296:769–81. doi: 10.1001/jama.296.7.769. [DOI] [PubMed] [Google Scholar]
  • 4.Riddler SA, Haubrich R, DiRienzo AG, et al. Class-sparing regimens for initial treatment of HIV-1 infection. N Engl J Med. 2008;358:2095–106. doi: 10.1056/NEJMoa074609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lennox JL, DeJesus E, Lazzarin A, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009;374:796–806. doi: 10.1016/S0140-6736(09)60918-1. [DOI] [PubMed] [Google Scholar]
  • 6.Cooper DA, Heera J, Goodrich J, et al. Maraviroc versus efavirenz, both in combination with zidovudine-lamivudine, for the treatment of antiretroviral-naive subjects with CCR5-tropic HIV-1 infection. J Infect Dis. 2010;201:803–13. doi: 10.1086/650697. [DOI] [PubMed] [Google Scholar]
  • 7.Daar ES, Tierney C, Fischl MA, et al. Atazanavir plus ritonavir or efavirenz as part of a 3-drug regimen for initial treatment of HIV-1. Ann Intern Med. 2011;154:445–56. doi: 10.1059/0003-4819-154-7-201104050-00316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ward BA, Gorski JC, Jones DR, et al. 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]
  • 9.Desta Z, Saussele T, Ward B, et al. Impact of CYP2B6 polymorphism on hepatic efavirenz metabolism in vitro. Pharmacogenomics. 2007;8:547–58. doi: 10.2217/14622416.8.6.547. [DOI] [PubMed] [Google Scholar]
  • 10.Belanger AS, Caron P, Harvey M, et al. Glucuronidation of the antiretroviral drug efavirenz by UGT2B7 and an in vitro investigation of drug-drug interaction with zidovudine. Drug Metab Dispos. 2009;37:1793–6. doi: 10.1124/dmd.109.027706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Haas DW, Ribaudo HJ, Kim RB, et al. Pharmacogenetics of efavirenz and central nervous system side effects: an Adult AIDS Clinical Trials Group study. AIDS. 2004;18:2391–400. [PubMed] [Google Scholar]
  • 12.Tsuchiya K, Gatanaga H, Tachikawa N, et al. 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;319:1322–6. doi: 10.1016/j.bbrc.2004.05.116. [DOI] [PubMed] [Google Scholar]
  • 13.Rotger M, Colombo S, Furrer H, et al. Influence of CYP2B6 polymorphism on plasma and intracellular concentrations and toxicity of efavirenz and nevirapine in HIV-infected patients. Pharmacogenet Genomics. 2005;15:1–5. doi: 10.1097/01213011-200501000-00001. [DOI] [PubMed] [Google Scholar]
  • 14.Haas DW, Smeaton LM, Shafer RW, et al. Pharmacogenetics of long-term responses to antiretroviral regimens containing efavirenz and/or nelfinavir: an Adult AIDS Clinical Trials Group Study. J Infect Dis. 2005;192:1931–42. doi: 10.1086/497610. [DOI] [PubMed] [Google Scholar]
  • 15.Rodriguez-Novoa S, Barreiro P, Rendon A, et al. Influence of 516G>T polymorphisms at the gene encoding the CYP450–2B6 isoenzyme on efavirenz plasma concentrations in HIV-infected subjects. Clin Infect Dis. 2005;40:1358–61. doi: 10.1086/429327. [DOI] [PubMed] [Google Scholar]
  • 16.Holzinger ER, Grady B, Ritchie MD, et al. Genome-wide association study of plasma efavirenz pharmacokinetics in AIDS Clinical Trials Group protocols implicates several CYP2B6 variants. Pharmacogenet Genomics. 2012;22:858–67. doi: 10.1097/FPC.0b013e32835a450b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wyen C, Hendra H, Vogel M, et al. Impact of CYP2B6 983T>C polymorphism on non-nucleoside reverse transcriptase inhibitor plasma concentrations in HIV-infected patients. J Antimicrob Chemother. 2008;61:914–8. doi: 10.1093/jac/dkn029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wang J, Sonnerborg A, Rane A, et al. Identification of a novel specific CYP2B6 allele in Africans causing impaired metabolism of the HIV drug efavirenz. Pharmacogenet Genomics. 2006;16:191–8. doi: 10.1097/01.fpc.0000189797.03845.90. [DOI] [PubMed] [Google Scholar]
  • 19.Ribaudo HJ, Liu H, Schwab M, et al. Effect of CYP2B6, ABCB1, and CYP3A5 polymorphisms on efavirenz pharmacokinetics and treatment response: an AIDS Clinical Trials Group study. J Infect Dis. 2010;202:717–22. doi: 10.1086/655470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.NCBI. dbSNP: Short Genetic Variations. http://www.ncbi.nlm.nih.gov/projects/SNP/ (9 December 2011, date last accessed)
  • 21.Barrett JS, Joshi AS, Chai M, et al. Population pharmacokinetic meta-analysis with efavirenz. Int J Clin Pharmacol Ther. 2002;40:507–19. doi: 10.5414/cpp40507. [DOI] [PubMed] [Google Scholar]
  • 22.Rotger M, Tegude H, Colombo S, et al. Predictive value of known and novel alleles of CYP2B6 for efavirenz plasma concentrations in HIV-infected individuals. Clin Pharmacol Ther. 2007;81:557–66. doi: 10.1038/sj.clpt.6100072. [DOI] [PubMed] [Google Scholar]
  • 23.Rotger M, Saumoy M, Zhang K, et al. Partial deletion of CYP2B6 owing to unequal crossover with CYP2B7. Pharmacogenet Genomics. 2007;17:885–90. doi: 10.1097/FPC.0b013e3282ef5cd1. [DOI] [PubMed] [Google Scholar]
  • 24.di Iulio J, Fayet A, Arab-Alameddine M, et al. In vivo analysis of efavirenz metabolism in individuals with impaired CYP2A6 function. Pharmacogenet Genomics. 2009;19:300–9. doi: 10.1097/FPC.0b013e328328d577. [DOI] [PubMed] [Google Scholar]
  • 25.Kwara A, Lartey M, Sagoe KW, et al. CYP2B6 (c.516G→T) and CYP2A6 (*9B and/or *17) polymorphisms are independent predictors of efavirenz plasma concentrations in HIV-infected patients. Br J Clin Pharmacol. 2009;67:427–36. doi: 10.1111/j.1365-2125.2009.03368.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cortes CP, Siccardi M, Chaikan A, et al. Correlates of efavirenz exposure in Chilean patients affected with human immunodeficiency virus reveals a novel association with a polymorphism in the constitutive androstane receptor. Ther Drug Monit. 2013;35:78–83. doi: 10.1097/FTD.0b013e318274197e. [DOI] [PubMed] [Google Scholar]
  • 27.Swart M, Whitehorn H, Ren Y, et al. PXR and CAR single nucleotide polymorphisms influence plasma efavirenz levels in South African HIV/AIDS patients. BMC Med Genet. 2012;13:112. doi: 10.1186/1471-2350-13-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kwara A, Lartey M, Sagoe KW, et al. CYP2B6, CYP2A6 and UGT2B7 genetic polymorphisms are predictors of efavirenz mid-dose concentration in HIV-infected patients. AIDS. 2009;23:2101–6. doi: 10.1097/QAD.0b013e3283319908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sarfo FS, Zhang Y, Egan D, et al. Pharmacogenetic associations with plasma efavirenz concentrations and clinical correlates in a retrospective cohort of Ghanaian HIV-infected patients. J Antimicrob Chemother. 2014;69:491–9. doi: 10.1093/jac/dkt372. [DOI] [PubMed] [Google Scholar]
  • 30.Robbins GK, DeGruttola V, Shafer RW, et al. Comparison of sequential three-drug regimens as initial therapy for HIV-1 infection. N Engl J Med. 2003;349:2293–303. doi: 10.1056/NEJMoa030264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Clifford DB, Evans S, Yang Y, et al. Impact of efavirenz on neuropsychological performance and symptoms in HIV-infected individuals. Ann Intern Med. 2005;143:714–21. doi: 10.7326/0003-4819-143-10-200511150-00008. [DOI] [PubMed] [Google Scholar]
  • 32.Haas DW, Wilkinson GR, Kuritzkes DR, et al. A multi-investigator/institutional DNA bank for AIDS-related human genetic studies: AACTG Protocol A5128. HIV Clin Trials. 2003;4:287–300. doi: 10.1310/MUQC-QXBC-8118-BPM5. [DOI] [PubMed] [Google Scholar]
  • 33.Kwara A, Lartey M, Boamah I, et al. Interindividual variability in pharmacokinetics of generic nucleoside reverse transcriptase inhibitors in TB/HIV-coinfected Ghanaian patients: UGT2B7*1c is associated with faster zidovudine clearance and glucuronidation. J Clin Pharmacol. 2009;49:1079–90. doi: 10.1177/0091270009338482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Turner ML, Reed-Walker K, King JR, et al. Simultaneous determination of nine antiretroviral compounds in human plasma using liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci. 2003;784:331–41. doi: 10.1016/s1570-0232(02)00822-x. [DOI] [PubMed] [Google Scholar]
  • 35.Kwara A, Lartey M, Sagoe KW, et al. Pharmacokinetics of efavirenz when co-administered with rifampin in TB/HIV co-infected patients: pharmacogenetic effect of CYP2B6 variation. J Clin Pharmacol. 2008;48:1032–40. doi: 10.1177/0091270008321790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Purcell S, Neale B, Todd-Brown K, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–75. doi: 10.1086/519795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.StataCorp. Stata Statistical Software: Release 13. College Station, TX: StataCorp LP; 2013. [Google Scholar]
  • 38.Nunoya K, Yokoi T, Kimura K, et al. A new deleted allele in the human cytochrome P450 2A6 (CYP2A6) gene found in individuals showing poor metabolic capacity to coumarin and (+)-cis-3,5-dimethyl-2-(3-pyridyl)thiazolidin-4-one hydrochloride (SM-12502) Pharmacogenetics. 1998;8:239–49. doi: 10.1097/00008571-199806000-00006. [DOI] [PubMed] [Google Scholar]
  • 39.Fukami T, Nakajima M, Yoshida R, et al. A novel polymorphism of human CYP2A6 gene CYP2A6*17 has an amino acid substitution (V365M) that decreases enzymatic activity in vitro and in vivo. Clin Pharmacol Ther. 2004;76:519–27. doi: 10.1016/j.clpt.2004.08.014. [DOI] [PubMed] [Google Scholar]
  • 40.Pitarque M, von Richter O, Oke B, et al. Identification of a single nucleotide polymorphism in the TATA box of the CYP2A6 gene: impairment of its promoter activity. Biochem Biophys Res Commun. 2001;284:455–60. doi: 10.1006/bbrc.2001.4990. [DOI] [PubMed] [Google Scholar]
  • 41.Yoshida R, Nakajima M, Nishimura K, et al. Effects of polymorphism in promoter region of human CYP2A6 gene (CYP2A6*9) on expression level of messenger ribonucleic acid and enzymatic activity in vivo and in vitro. Clin Pharmacol Ther. 2003;74:69–76. doi: 10.1016/S0009-9236(03)00090-0. [DOI] [PubMed] [Google Scholar]
  • 42.Cho DY, Ogburn ET, Jones D, et al. Contribution of N-glucuronidation to efavirenz elimination in vivo in the basal and rifampin-induced metabolism of efavirenz. Antimicrob Agents Chemother. 2011;55:1504–9. doi: 10.1128/AAC.00883-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Marzolini C, Telenti A, Decosterd LA, et al. Efavirenz plasma levels can predict treatment failure and central nervous system side effects in HIV-1-infected patients. AIDS. 2001;15:71–5. doi: 10.1097/00002030-200101050-00011. [DOI] [PubMed] [Google Scholar]
  • 44.Gutierrez F, Navarro A, Padilla S, et al. Prediction of neuropsychiatric adverse events associated with long-term efavirenz therapy, using plasma drug level monitoring. Clin Infect Dis. 2005;41:1648–53. doi: 10.1086/497835. [DOI] [PubMed] [Google Scholar]
  • 45.Read TR, Carey D, Mallon P, et al. Efavirenz plasma concentrations did not predict cessation of therapy due to neuropsychiatric symptoms in a large randomized trial. AIDS. 2009;23:2222–3. doi: 10.1097/QAD.0b013e32832e95f8. [DOI] [PubMed] [Google Scholar]
  • 46.Takahashi M, Ibe S, Kudaka Y, et al. No observable correlation between central nervous system side effects and EFV plasma concentrations in Japanese HIV type 1-infected patients treated with EFV containing HAART. AIDS Res Hum Retroviruses. 2007;23:983–7. doi: 10.1089/aid.2006.0193. [DOI] [PubMed] [Google Scholar]
  • 47.Fumaz CR, Munoz-Moreno JA, Molto J, et al. Long-term neuropsychiatric disorders on efavirenz-based approaches: quality of life, psychologic issues, and adherence. J Acquir Immune Defic Syndr. 2005;38:560–5. doi: 10.1097/01.qai.0000147523.41993.47. [DOI] [PubMed] [Google Scholar]
  • 48.Lubomirov R, Colombo S, di Iulio J, et al. Association of pharmacogenetic markers with premature discontinuation of first-line anti-HIV therapy: an observational cohort study. J Infect Dis. 2011;203:246–57. doi: 10.1093/infdis/jiq043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gatanaga H, Hayashida T, Tsuchiya K, et al. Successful efavirenz dose reduction in HIV type 1-infected individuals with cytochrome P450 2B6 *6 and *26. Clin Infect Dis. 2007;45:1230–7. doi: 10.1086/522175. [DOI] [PubMed] [Google Scholar]
  • 50.Puls R Group tES. A daily dose of 400 mg efavirenz (EFV) is non-inferior to the standard 600 mg dose: week 48 data from the ENCORE1 study, a randomised, double-blind, placebo controlled, non-inferiority trial. Abstracts of the Seventh IAS Conference on HIV Pathogenesis, Treatment and Prevention; Kuala Lumpur, Malaysia. 2013. Abstract WELBB01. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

Articles from Journal of Antimicrobial Chemotherapy are provided here courtesy of Oxford University Press

RESOURCES