Abstract
Inter-individual variability in efavirenz (EFV) pharmacokinetics and dynamics are dominantly driven by the polymorphism in cytochrome P450 (CYP) isoenzyme 2B6 516G>T. We hypothesized that additional CYP polymorphisms mediate the relationship between CYP2B6 516G>T, EFV metabolism, and clinical events. We investigated 21 SNPs in 814 HIV-infected adults initiating EFV-based therapy in Botswana for population pharmacokinetics, CNS toxicities, and treatment outcomes. Two SNPs (rs28399499 and rs28399433) showed reduced apparent oral EFV clearance. Four SNPs (rs2279345, rs4803417, rs4802101, and rs61663607) showed extensive clearance. Composite CYP2B-mediated EFV metabolism was significantly associated with CNS toxicity (p=0.04), with extensive metabolizers reporting more and slow and very slow metabolizers reporting less toxicity after one month compared to intermediate metabolizers. Composite CYP2B6 metabolism was not associated with composite early treatment failure. In conclusion, our data suggest that CNS-related toxicities might not be solely the result of super-therapeutic parent EFV concentrations in HIV-infected individuals in patients of African ancestry.
Introduction
The number of people with human immunodeficiency virus (HIV) type 1 initiating antiretroviral therapy (ART) in sub-Saharan Africa has dramatically increased over the past decade [1]. Combination therapy containing efavirenz (EFV), a non-nucleoside reverse transcriptase inhibitor (NNRTI), and two nucleoside reserve transcriptase inhibitors (emtricitabine and tenofovir disoproxil fumarate) has been the preferred management strategy in this region. EFV has a high efficacy, low cost, and long elimination half-life with once daily dosing fixed at 600 mg [2]. Plasma EFV concentrations <1 μg/ml are associated with virologic failure whereas super-therapeutic levels of >4 μg/ml have been associated with central nervous system (CNS) adverse effects including vivid dreams, headaches, dizziness, insomnia, and fatigue, that frequently lead to therapy discontinuation [3–6]. Recently, US treatment guidelines have relegated EFV to an alternative choice for first line therapy, primarily due to concerns about tolerability [7]. Yet, EFV remains a mainstay of treatment in resource-limited settings, in part due to the higher cost associated with drugs thought to be better tolerated.
EFV is primarily metabolized by cytochrome P450 (CYP) 2B6 and alternatively by CYP 2A6, 3A4, and 3A5 isoenzymes [8, 9]. Pharmacokinetic (PK) variability in EFV is mainly attributed to single nucleotide polymorphisms (SNPs) present in these highly polymorphic isoenzymes responsible for its metabolism and clearance [10, 11]. Among these variants, CYP2B6 516G>T has a high frequency and functional relevance, as confirmed in several settings including Botswana [12–16]. The presence of the minor slow-metabolizing allele has been implicated with CNS toxicities, but results are controversial. For example, Haas et al found the T-allele to be associated with CNS symptoms after 1 week of treatment and Rotger et al. reported a positive association between CYP2B6 genotype and sleep disorders and fatigue [17]. Studies in African populations showed that the relationship between CYP2B6 and CNS toxicity is not as clearly apparent or non-existent as compared to people of European ancestry [15, 18–21], and there have been reports in European populations as well that did not find this association [22–27]. In light of these differential findings for CYP2B6 genotype among African patients, we aimed to establish robust PK estimates at steady-state for 21 SNPs in CYP 2B6, 2A6, 3A4, and 3A5 in HIV infected individuals of African origin initiating EFV-based therapy. We hypothesized that the presence of additional CYP polymorphisms mediate the association between CYP2B6 516G>T, EFV metabolism, CNS toxicities, and treatment outcomes.
Subjects and Methods
Study Design
Data were used from a cohort study of HIV-infected patients newly initiating their first ART regimen at clinics in and around Gaborone, Botswana. Participants were approached for enrollment in the clinics on the day they were scheduled to initiate antiretroviral therapy from June of 2009 to November of 2013. The inclusion criteria were (i) confirmed HIV infection, (ii) black African origin, (iii) no pregnancy or pregnancy intention, (iv) age 21 years or older, and (v) initiation of a standard three-drug regimen including EFV at 600 mg/day plus two NRTIs. Follow-up visits occurred at month 1 and month 6 and patients were included if they had at least one EFV plasma concentration available for analysis and pharmacokinetics modeling. The study was approved by the Ethics Board of the Health Research and Development Committee of the Botswana Ministry of Health and by the Committee on Human Subjects Research of the University of Pennsylvania, and each study participant provided written informed consent in English or Setswana.
Drug assay
Midpoint plasma samples for measuring drug concentrations were collected at steady-state at 1 month after initiation of therapy and 6 months after initiation. EFV steady-state plasma concentrations are generally reached within a 6–10 day time frame [28]. Dates and times of blood draw and last ingestion were recorded. Samples were prepared using protein precipitation followed by one to one dilution with water. EFV was separated from plasma by high performance liquid chromatography, and concentrations measured with tandem mass spectrometry in negative ionization mode with multiple reaction monitoring having a lower limit of quantification of 1 ng/mL, as described previously [29].
Genotyping
In order to characterize the genetic polymorphisms of CYP2B6, CYP2A6, CYP3A4, and CYP3A5, genomic DNA was extracted from whole blood samples obtained at the baseline visit. A total of 21 SNPs besides CYP2B6 516 G>T were selected in CYP2B6, CYP2A6, CYP3A4, and CYP3A5. SNPs were chosen based on results from previous studies, allele frequencies in people of African ancestry, and the availability of the SNP template on the Taqman OpenArray platform. All SNPs genotyped in the study were identified in the International HapMap project [30]. Allele-specific fluorescent discrimination was performed using Taqman Genotyper software, version 1.3 (Life Technology). All genotype-specific MAFs were compared against populations of African ancestry from the International HapMap project reference panel.
Medication Adherence
EFV adherence was calculated from pharmacy data over the first month and last 3-months before end of follow-up. Refill dates were used to calculate medication possession ratio according to the formula: (number of doses dispensed minus the number of doses returned) divided by (number of days between the initial fill and the fill closest to the target date) [31].
Population PK Model Development
Population pharmacokinetic modeling was performed using nonlinear mixed-effects modeling in NONMEM VII (ICON Development Solutions, Ellicott City, MD, USA). The ADVAN2 subroutine (one compartment linear model with first order absorption) was selected with the first-order conditional estimation (FOCE-I) method with interaction to estimate the typical model parameters. Random inter-individual and inter-occasional variability were estimated and the final model was minimized with successful execution of the covariance step. The baseline EFV PK model parameters were adopted from a previous study in the same cohort, where clearance (CL/F) was the only parameter to be estimated while holding the parameters of volume of distribution (Vd) and absorption rate constant (Ka) constant at 150 L and 0.18 h−1 respectively [13]. We used an exponential variance model to describe the unexplained random variability of parameters across individuals.
To test whether baseline characteristics influence EFV PK, a structural baseline population PK covariate model was established. Models were explored using various inter-individual random effect covariance structures. Inter-individual variability was initially estimated for CL/F, and then subsequently evaluated for the remaining pharmacokinetic parameters. A composite additive and proportional error model was used to describe random residual variability. Covariates included subject weight, gender, EFV adherence, and suspected or confirmed tuberculosis co-occurrence. Weight was included in an allometric model and fixed at 0.75 CL/F and normalized to a reference weight of 70 kg. All baseline covariates were included with power terms into the model. Subsequently, SNPs in CYP2B6, CYP2A6, CYP3A4, and CYP3A5 were tested for model improvement in a univariate fashion.
The final model was identified using stepwise forward selection and backward elimination of covariates using NONMEM. Goodness of fit diagnostic plots were evaluated in R (version 3.2.3). The covariate that resulted in the greatest statistically significant decrease in the Minimum Objective Function Value (MOFV) was kept in the model and remaining significant covariates were added in the same fashion. The ΔMOFV following a Chi-Square distribution with a change of 3.84 (df=1) or 5.99 (df=2) was considered statistically significant at α=0.05. A p-value of 0.20 was used for the backward elimination procedure. SNPs were considered extensive or slow metabolizers if they respectively increased or decreased CL by 20%, and were not in LD with other SNPs with a similar effect on clearance (e.g. multi-colinearity).
Pharmacodynamics
In subsequent pharmacodynamics (PD) analyses patients were classified into composite CYP2B6-mediated metabolizer groups. Patients are classified into ‘Very Extensive’, ‘Extensive’, ‘Intermediate’, ‘Slow’, and ‘Very Slow’ composite metabolizer group based on their CYP2B6 516G>T genotype, 983T>C genotype and presence of an extensive metabolizer allele in 18492T>C, 15817A>C, or -745A>G. The 35-item Subject Experience Questionnaire (SEQ) [22] questionnaire was completed at months 1 and 6 after initiation of therapy [32]. We limited our analysis to the 21 items related to CNS-related adverse effects: difficulty sleeping, abnormal dreams, fatigue, dizziness, nervousness, mood change, irritability, memory loss and impaired concentration. Patients were classified into ordinal categories according to their CNS scores into: none, mild (1-3), moderate (4-7), severe (8-12), and very severe (>12). A proportional odds model for ordinal regression using VGAM R-package was performed to quantify the relationship between CNS symptoms classes and composite CYP2B6 metabolizer class. We also ran the ordinal logistic regression model with CYP2B6 as categorical variable. Treatment outcome measures included a composite treatment endpoint defined as death, detectable viral load (HIV RNA >25 copies/ml at month 6), or loss to follow-up at 6 months. Composite treatment endpoint was modeled using multivariate logistic regression with composite CYP2B6 group where the intermediate composite metabolizer group was considered the reference. Two additional logistic regression analyses were performed to assess whether the associations were more or less strong for the endpoints of detectable viral load or loss to follow-up [33].
Code availability
Programming code for population PK and PD analyses using NONMEM and R are available upon request.
Results
Population Pharmacokinetics
A total of 941 individuals were initially enrolled in the cohort study with a scheduled follow-up of 6 months, in which 814 were successfully genotyped for CYP2B6 516G>T (rs3745274). 742 genotyped participants had at least one midpoint steady-state plasma EFV specimen available at month 1 for drug quantification, of whom 562 also had an available blood sample at month 6 (Table 1). Median plasma EFV concentration dropped from 2.17 μg/ml after one month of therapy initiation to 2.05 μg/ml after six months (p < 0.01). The general characteristics as shown in Table 1 indicate patients with lower CD4 counts were more likely to have dropped out of the study at month 6 (Table 1). The structural baseline population PK model without covariates showed a MOFV of 2314. Inter-occasion variability resulted in 3% higher apparent clearance at month 6 compared to month 1 measurements (7.3 vs. 7.6 L/hr/70kg for month 1 and month 6, respectively, MOFV = 2304). CYP2B6 516G>T genotypes demonstrated an impact on EFV clearance for normal (GG, 9.439 L/hr/70kg), slow (GT, 7.233 L/hr/70kg), and very slow (TT, 4.033 L/hr/70kg) genotypes respectively, with a MOFV of 2085. History of tuberculosis, gender, and drug adherence did not significantly improve MOFV[13]. Therefore, the final baseline covariate model before inclusion of the additional SNPs under investigation included CYP2B6 516G>T genotype, allometrically scaled weight, and visit as covariates (MOFV = 2075) with an inter-individual variability (ω2 CL) of 40.5% coefficient variation, proportional residual variability of 0.0831% coefficient variation, and residual additive variability of 0.0699 SD.
Table 1.
Baseline Characteristics Stratified by Availability of Treatment Outcome Data at Month 6.
Characteristic | Included Patients | Treatment Outcome data available at Month 6 | P | |
---|---|---|---|---|
Yes | No | |||
Total*, n | 941 | 712 | 229 | |
Genotyped at baseline, n | 814 | 624 | 190 | |
Genotyped and provided one EFV sample, n | 742 | 567 | 175 | |
Genotyped and provided two EFV samples, n | 562 | 525 | 37 | |
Plasma EFV at month 1 (μg/ml), median (IQR) | 2.17 (1.62 – 3.88) | 2.14 (1.60 – 3.72) | 2.34 (1.71 – 4.68) | 0.085 |
Age in years, median (IQR) | 37 (33 – 44) | 37 (32 – 44) | 38 (34 – 44) | 0.14 |
Male gender, n (%) | 399 (49.0%) | 280 (44.9%) | 98 (51.5%) | 0.15 |
Body Mass Index, median (IQR) | 22.0 (19.8 – 25.1) | 22.0 (19.8 – 25.1) | 21.7 (20.0 – 25.1) | >0.5 |
CD4 count (cells/mm3), median (IQR) | 196 (112 – 256) | 204 (118 – 271) | 184 (117 – 235) | <0.02 |
Plasma viral load (log10 copies/ml), median (IQR) | 4.9 (4.2 – 5.4) | 4.9 (4.2 – 5.4) | 5.0 (4.3 – 5.4) | >0.5 |
History of tuberculosis, n (%) | 31 (3.8%) | 23 (3.7%) | 8 (4.2%) | >0.5 |
Study cohort included patients were successfully genotyped for CYP2B6 516G>T polymorphism. P-values were calculated with Wilcoxon rank sum test for continuous variables, and chi-square test for categorical covariates and represent statistical differences between patients that have outcome data at Month 6 available versus who do not.
SNPs and Pharmacokinetics
The effects of 21 individual SNPs from CYP isoenzymes on apparent oral EFV clearance are shown in Table 2. In particular, two SNPs were associated with decreased apparent EFV clearance, which were CYP2B6 983T>C (rs28399499, MOFV = 1862) with a MAF of 11% and CYP2A6 -48T>G (rs28399433, MOFV = 2016) with a MAF of 8%. Four SNPs were associated with apparent extensive EFV clearance, namely 1) rs2279345 (CYP2B6 18492T>C, MOFV = 1995), an intronic variant in CYP2B6 with a MAF of 19%, 2) rs4803417 (CYP2B6 15817A>C, MOFV = 2016) located in the UTR-5 region of CYP2B6 with a MAF of 12%, 3) CYP2B6, -745A>G (rs4802101, MOFV = 2037) with a MAF of 4% and 4) finally, a SNP in the 5 prime flanking region of CYP2A6 (-750T>C, rs61663607, MOFV = 2021) were associated with extensive apparent clearance (Table 2).
Table 2.
Apparent EFV Clearance for Genotyped SNPs
CYP | SNP | MAF | LD | No. Participants | Clearance (L/hr/70kg) | |||||
---|---|---|---|---|---|---|---|---|---|---|
Block | Wt | Het | Hom | Wt | Het | Hom | MOFV | |||
3A5 | rs15524 | 0.37 | - | 287 | 353 | 95 | 7.302 | 7.250 | 7.270 | 2027 |
rs41303343 | 0.09 | - | 614 | 113 | 10 | 7.539 | 7.217 | 7.157 | 2009 | |
rs10264272 | 0.21 | - | 462 | 247 | 29 | 7.387 | 7.388 | 7.852 | 2032 | |
rs776746 | 0.18 | - | 494 | 208 | 26 | 7.421 | 7.219 | 7.355 | 2011 | |
3A4 | rs4646437 | 0.14 | - | 554 | 174 | 12 | 7.423 | 7.425 | 7.686 | 2052 |
rs2740574 | 0.24 | A | 418 | 281 | 37 | 7.466 | 7.319 | 7.316 | 2042 | |
rs1851426 | 0.20 | A | 470 | 243 | 26 | 7.439 | 7.306 | 7.321 | 2038 | |
2A6 | rs143731390 | 0.06 | - | 631 | 90 | 0 | 7.481 | 7.788 | - | 2001 |
rs8192726 | 0.07 | B | 643 | 87 | 9 | 7.636 | 5.737 | 4.671 | 2004 | |
rs28399433 | 0.08 | B | 635 | 95 | 8 | 7.598 | 5.926 | 4.939 | 2016 | |
rs61663607 | 0.14 | - | 543 | 171 | 16 | 7.225 | 7.172 | 8.678 | 2021 | |
2B6 | rs4802101 | 0.04 | - | 687 | 54 | 0 | 7.251 | 8.640 | - | 2037 |
rs6508963 | 0.21 | C | 461 | 250 | 29 | 6.734 | 7.238 | 7.537 | 2044 | |
rs8100458 | 0.16 | C | 528 | 190 | 23 | 6.848 | 7.633 | 7.738 | 2041 | |
rs4803417 | 0.12 | C | 572 | 151 | 14 | 6.819 | 8.303 | 9.377 | 2016 | |
rs4803419 | 0.07 | C | 640 | 92 | 7 | 7.225 | 7.431 | 6.739 | 2046 | |
rs2279343 | 0.06 | - | 645 | 96 | 0 | 8.139 | 5.091 | - | 2041 | |
rs2279345 | 0.19 | - | 485 | 224 | 27 | 6.539 | 8.005 | 8.349 | 1995 | |
rs28399499 | 0.11 | D | 586 | 149 | 5 | 7.984 | 4.691 | 1.440 | 1862 | |
rs8192719 | 0.37 | D | 283 | 349 | 99 | 9.546 | 7.377 | 5.600 | 2017 | |
rs707265 | 0.19 | D | 490 | 223 | 26 | 7.719 | 5.614 | 4.729 | 1933 |
LD blocks are denoted by capital letters for SNPs that are in linkage disequilibrium. MOFV of the model prior to evaluating SNPs was 2075, which includes the covariates of CYP2B6 516G>T, allometrically scaled weight, and inter occasion variability. Clearance estimates are corrected for the covariates: CYP2B6 G516T genotype (rs3745274), allometrically scaled weight, and inter-occasional variability between visits. MAF-minor allele frequency, Wt-wild type, het-heterozygote, hom-homozygote variant
SNPs that were not considered for evaluation due to their high linkage disequilibrium with index SNPs were rs707265 (r2 = 0.44, D’ = 1 with rs28399499), rs8192726 (r2 = 0.99, D’ = 1 with rs28399433), rs2279343 (r2 = 0.30, D’ = 1 with rs3745274), and rs8192719 (r2 = 0.82, D’ = 1 with rs3745274). The composite variable of CYP2B6 metabolism was created based on these PK findings, and subsequently tested for associations with CNS toxicities and treatment endpoints.
Pharmacodynamics
Patients reported an average of 1.67 CNS-related symptoms at Month 1 (median 1, IQR 0-2). Among the patients that remained on study, the average number of CNS-related symptoms was 0.79 at Month 6 (median 0, IQR 0-1). However, the majority of the participants (n = 372) reported no CNS adverse experiences. Figure 2 depicts levels of reported CNS toxicities between different CYP2B6 metabolizer categories. Patients with very extensive or extensive CYP2B6 metabolism more frequently reported severe CNS adverse experiences compared to intermediate, slow metabolizers and very slow metabolizers reported less toxicity despite their higher plasma EFV concentrations (p = 0.041). The association between composite metabolizer group and CNS toxicities disappeared at the six month time point (p=0.425, figure not shown).We did not observe an association between composite CYP2B6 metabolism and composite failure (Table 4). However, there was a tendency of loss to follow-up occurring more frequently in both the very extensive and slow/very slow metabolizer groups compared to intermediate metabolizers, OR 1.42 (0.84-2.36) and 1.50 (0.93-2.42) respectively. Similarly a mild protective effect was observed for the outcome of detectable viral load for both the extensive and slow/very slow metabolizer groups, although none of these associations met the traditional thresholds for statistical significance (Table 4).
Figure 2.
CNS toxicities by composite CYP P450 metabolizer group
Table 4.
Association Between Composite CYP2B6 Metabolism and Treatment Endpoints
Failure | Detectable Viral Load | Loss to Follow-up | |||||||
---|---|---|---|---|---|---|---|---|---|
OR | (95%CI) | P | OR | (95%CI) | P | OR | (95%CI) | P | |
Composite Metabolizer | |||||||||
Very Extensive | 0.99 | (0.65-1.50) | 0.68 | (0.34-1.29) | 1.42 | (0.84-2.36) | |||
Extensive | 0.84 | (0.57-1.23) | 0.58 | (0.31-1.04) | 1.00 | (0.61-1.64) | |||
Intermediate | 1.00 | (Ref) | 1.00 | (Ref) | 1.00 | (Ref) | |||
Slow/Very Slow | 1.18 | (0.80-1.72) | 0.74 | (0.40-1.33) | 1.50 | (0.93-2.42) |
Composite virologic endpoint was defined as death, HIV RNA >25 copies/ml, or loss to follow-up at 6 months
Discussion
The present study confirms the influence of two well-described cytochrome P450 polymorphisms, CYP2B6 983T>C and CYP2A6 -48T>G, which were both associated with increased plasma EFV exposure [10, 11, 34]. We additionally identified 4 cytochrome P450 variants (CYP2B6 -745A>G, 15817A>C, 18492T>C, and CYP2A6 -750T>C) associated with decreased plasma EFV exposure in HIV-infected individuals initiating ART treatment in Botswana. We observed an association between composite CYP2B6-mediated EFV metabolism and CNS toxicities, with extensive metabolizers reporting more and very slow metabolizers reporting less toxicities. These data suggest that CNS-related toxicities might not be a result of super-therapeutic EFV levels in this HIV-infected population of African ancestry.
While prior research has found EFV levels exceeding 4 mg/mL to be associated with neurotoxicity [8], Ribaudo et al. found the relation between EFV concentrations and CNS adverse experiences to be apparent among Caucasian, but not among African-American patients [18]. This discrepancy may have been due to a more complex relation between EFV metabolism and symptoms than previously recognized. Recent in vitro studies suggest a major neurotoxic role for the major phase I metabolite of EFV, 8-hydroxyefavirenz (8-OH-EFV) due to its ability to generate reactive oxygen species [18]. Efavirenz metabolites may contribute to drug effectiveness but also to toxicity and interactions. Thus, there is a need for extensive EFV metabolic profiling. In extensive metabolizers, even though parent EFV is low, 8-OH metabolite concentrations higher. But in slow metabolizers, despite high EFV concentrations, the concentration of 8-OH-EFV would be expected to be lower due to upregulated hydrolysis by the CYP2A6 enzyme into 7-OH-EFV.
P-glycoprotein, an efflux membrane transporter encoded by ABCB1 is predominantly found at the blood brain barrier that limits entry into the CNS for a large number of drugs. Several studies assessed EFV substrate specificity for the main ATP-dependent drug efflux transporters suggesting that EFV does not interact with ABCB1 [35–39] and is therefore unlikely to explain our findings. These data are in line with the observations of Winston et al. who showed central spine fluid (CSFG) 8-OH-EFV not to be differential between CYP2B6 516G>T genotypes [40]. They postulated an EFV dose-dependent up regulation of efflux transporters specifically affecting CNS 8-OH-EFV removal [41]. Similarly, Aouri et al. recently measured both phase I and II metabolites of EFV in CSF and found that 8-OH-EFV did not differ between CYP2B6 genotypes [42]. They did not observe a direct association between EFV or any metabolite and CNS toxicity, however they might have been underpowered for an association analysis with only 71 patients. While we did not measure plasma or CSF concentrations of 8-OH-EFV, our data are consistent with the hypothesis that EFV-related CNS toxicity might mediated via a neurotoxic 8-OH-EFV metabolite [43]. However, additional studies are essential to explain this phenomenon.
Several CYP2B6 polymorphisms evaluated other than G516T have been implicated in the inter-individual variability in plasma EFV levels previously. The current investigation validates several previously repeatedly identified SNPs in a cohort of African HIV patients treated with EFV in a clinical setting. Our population PK analyses on apparent EFV clearance support the previously observed association of the missense CYP2B6 983T>C (rs28399499) [9–11, 21, 34, 44–48] and intronic CYP2A6 -48T>G (rs28399433) SNPs with poor EFV clearance. We additionally identified 4 SNPs with substantially reduced MOFV and altered EFV clearance estimates by >20%, namely CYP2B6 -745A>G, 15817A>C, 18492T>C, and CYP2A6 -750T>C.
CYP2B6 18492T>C (rs2279345) has been previously associated with increased metabolism of EFV, resulting in lower EFV plasma levels as compared to patients with the TT genotype [49–53]. Similarly, rs4802101 (CYP2B6 −750T>C) is a promoter SNP part of CYP2B6 *28 and located within a transcription factor binding site. It has been associated with increased exposure to EFV as compared to CYP2B6 *1 haplotype in people with HIV [19]. It should be noted that since these polymorphism are relatively frequent in African populations, a substantial proportion of the patients might be at the risk of having sub-therapeutic EFV plasma concentrations and consequently higher rates of resistance when failing EFV [8].
To our knowledge, we identified two SNPs not described to date in relation to EFV pharmacokinetics. First, rs4803417 (CYP2B6 15817A>C) located in the UTR-5 region of CYP2B6 and has been associated with lowering circulating concentrations of a major brominated flame retardant, BDE-47 [54]. Since there is evidence that in Botswana atmospheric polybrominated diphenyl ethers (PBDEs) levels have been steadily increasing [55, 56], it is intriguing to consider that PBDE-induced CYP activation might be at play [57]. Second, rs61663607 (-745A>G) is a polymorphism located in the 5 prime flanking region of CYP2A6 that disrupts the core region of a CCAAT box to which the regulatory element NF-Y binds [58, 59] Genetic polymorphisms in the promoter region of CYP2A6 have been implicated for decreased promoter activity and subsequent transcriptional activity [58, 60]. The epigenomic signature of the SNP obtained from ChIP-seq data of the Roadmap Epigenomics Consortium reveals the presence of both a genetic enhancer and confirms the presence of poised (e.g. inactive) promoter [61]. Our study found that patients heterozygous for the minor allele have increased apparent EFV clearance, suggesting increased CYP2A6 activity. Therefore, the CYP2A6 -745A>G potential mechanisms of switching between promoter, enhancer, and transcriptional transition states of CYP2A6 chromatin and the effect on EFV pharmacokinetics requires further investigation.
Major strengths of our study include 1) a large sample size, 2) a racially homogenous group, and 3) the routine care setting. Several limitations apply. Although the association between genotype and CNS toxicities was substantial, the reporting of EFV-based toxicities in Africa was lower than in Western countries [22]. In our study in Botswana, 66.3% of patients reported at least one EFV-based adverse event, whereas prevalences of 9.4%, 30.9%, 42.7%, and 83.3% were reported in Ghana, Uganda, Zimbabwe, and South Africa respectively [15, 20, 21, 62]. These observations highlight the need for an adverse events assessment method tailored for and validated in African HIV-infected individuals. Furthermore, we lacked a precise measure of adherence, such as microelectronic monitors [63], and so the accuracy of adherence measures might be sub-optimal. Detectable viral load – one of the study endpoints – was defined at the low level of >25 copies/ml and therefore may not be associated with clinical failure or virologic transmission if individuals engage in unprotected sex [64]. Data on whether resistance emerged in these failures was unavailable. Moreover, a substantial part of the patient population was lost to care within the 6 month time frame, where others have demonstrated these individuals are more likely to have poor outcomes [33]. Also, the current study is restricted to an average follow-up time of 6 months, and so questions regarding the impact of these polymorphisms in the response to therapeutic drugs and the long-term virologic and immunological response in this population remain to be answered. However, in a follow-up study in Botswana we observed that the CYP2B6 516G>T polymorphism showed a protective association with regards to the maintenance of HIV suppression [14]. Finally, in this exploratory analysis evaluating a total of 21 SNPs we cannot exclude that these findings were not due to chance and therefore future replication is essential to establish the validity of these findings.
In conclusion, extensive SNPs in cytochrome P450 enzymes appear to contribute to variability of EFV pharmacokinetic parameters in HIV patients and appear to be associated with greater CNS toxicity. Additional investigation is required to replicate our seemingly paradoxical observation with regard to EFV-induced CNS toxicity, identify potential underlying causal variants, and characterize their biological mechanistic importance.
Figure 1.
LD Blocks and Allele Frequencies for SNPs
Table 3.
Clearance-Based Classification of CYP2B6 SNPs into a Composite Metabolizer Group.
516G>T | 983T>C | Extensive SNP | N | Composite Metabolizer |
---|---|---|---|---|
0 | 0 | 0 | 69 | Extensive |
0 | 0 | 1 | 141 | Very Extensive |
0 | 1 | 0 | 57 | Intermediate |
0 | 1 | 1 | 36 | Intermediate |
0 | 2 | 0 | 6 | Very Slow |
0 | 2 | 1 | 0 | Slow |
1 | 0 | 0 | 179 | Intermediate |
1 | 0 | 1 | 127 | Extensive |
1 | 1 | 0 | 66 | Slow |
1 | 1 | 1 | 3 | Intermediate |
1 | 2 | 0 | 0 | Very Slow |
1 | 2 | 1 | 0 | Slow |
2 | 0 | 0 | 112 | Slow |
2 | 0 | 1 | 2 | Intermediate |
2 | 1 | 0 | 1 | Very Slow |
2 | 1 | 1 | 0 | Slow |
2 | 2 | 0 | 0 | Very Slow |
2 | 2 | 1 | 0 | Very Slow |
Acknowledgments
We thank the medical staff at the Bontleng, BH3, Broadhurst Traditional Area, Morwa, Nkoyaphiri, Phase II, and Village Infectious Diseases Care Clinics for their assistance with carrying out this study. We also thank the Ministry of Health of Botswana for supporting the project and the patients who participated.
Footnotes
Conflict of Interest
All authors had full access to study data and agreed to submit for publication. The corresponding author had final responsibility for the decision to submit for publication. No medical writers were used and no agency made any payments for writing. Neither the funding agency nor pharmaceutical companies supporting the trial played any role in collection, analysis, interpretation, or reporting of these data.
Author Contributions
M.V. wrote the article; R.G., G.P.B., M.V, B.L.S., and M.M. designed the research; M.V., S.L.B, A.Z, M.G., B.R., X.H., A.P.S., M.M., B.L.S., G.P.B., A.R. and R.G. performed the research; M.V. analyzed the data. The final manuscript reviewed and approved by all authors.
References
- 1.UNAIDS. Global Report: UNAIDS Report on the Global AIDS Epidemic 2014. Geneva: UNAIDS; 2014. [Google Scholar]
- 2.Smith PF, DiCenzo R, Morse GD. Clinical pharmacokinetics of non-nucleoside reverse transcriptase inhibitors. Clin Pharmacokinet. 2001;40(12):893–905. doi: 10.2165/00003088-200140120-00002. [DOI] [PubMed] [Google Scholar]
- 3.Robertson K, Liner J, Meeker RB. Antiretroviral neurotoxicity. J Neurovirol. 2012;18(5):388–99. doi: 10.1007/s13365-012-0120-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.World Health Organization. Global update on the health sector response to HIV 2014. Geneva, Switzerland: 2014. [Google Scholar]
- 5.Leutscher PD, Stecher C, Storgaard M, Larsen CS. Discontinuation of efavirenz therapy in HIV patients due to neuropsychiatric adverse effects. Scand J Infect Dis. 2013;45(8):645–51. doi: 10.3109/00365548.2013.773067. [DOI] [PubMed] [Google Scholar]
- 6.Group ES. Efficacy of 400 mg efavirenz versus standard 600 mg dose in HIV-infected, antiretroviral-naive adults (ENCORE1): a randomised, double-blind, placebo-controlled, non-inferiority trial. Lancet. 2014;383(9927):1474–82. doi: 10.1016/S0140-6736(13)62187-X. Epub 2014/02/14. [DOI] [PubMed] [Google Scholar]
- 7.Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents [Internet] 2016 Available at http://aidsinfo.nih.gov/contentfiles/lvguidelines/AdultandAdolescentGL.pdf: Accessed July 6th, 2016.
- 8.Ward BA, Gorski JC, Jones DR, Hall SD, Flockhart DA, 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(1):287–300. doi: 10.1124/jpet.103.049601. [DOI] [PubMed] [Google Scholar]
- 9.Sinxadi PZ, Leger PD, McIlleron HM, Smith PJ, Dave JA, Levitt NS, et al. Pharmacogenetics of plasma efavirenz exposure in HIV-infected adults and children in South Africa. Br J Clin Pharmacol. 2015;80(1):146–56. doi: 10.1111/bcp.12590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Haas DW, Kwara A, Richardson DM, Baker P, Papageorgiou I, Acosta EP, et al. Secondary metabolism pathway polymorphisms and plasma efavirenz concentrations in HIV-infected adults with CYP2B6 slow metabolizer genotypes. J Antimicrob Chemother. 2014;69(8):2175–82. doi: 10.1093/jac/dku110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Holzinger ER, Grady B, Ritchie MD, Ribaudo HJ, Acosta EP, Morse GD, et al. Genome-wide association study of plasma efavirenz pharmacokinetics in AIDS Clinical Trials Group protocols implicates several CYP2B6 variants. Pharmacogenet Genomics. 2012;22(12):858–67. doi: 10.1097/FPC.0b013e32835a450b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gross R, Aplenc R, Tenhave T, Foulkes AS, Thakur R, Mosepele M, et al. Slow efavirenz metabolism genotype is common in Botswana. J Acquir Immune Defic Syndr. 2008;49(3):336–7. doi: 10.1097/QAI.0b013e31817c1ed0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Gross R, Bellamy SL, Ratshaa B, Han X, Vujkovic M, Aplenc R, et al. CYP2B6 genotypes and early efavirenz-based HIV treatment outcomes in Botswana. AIDS. 2017;31(15):2107–13. doi: 10.1097/QAD.0000000000001593. Epub 2017/07/12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Vujkovic M, Bellamy SL, Zuppa AF, Gastonguay M, Moorthy GS, Ratshaa BR, et al. Brief Report: CYP2B6 516G>T Minor Allele Protective of Late Virologic Failure in Efavirenz-Treated HIV-Infected Patients in Botswana. J Acquir Immune Defic Syndr. 2017;75(4):488–91. doi: 10.1097/QAI.0000000000001442. Epub 2017/05/10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gounden V, van Niekerk C, Snyman T, George JA. Presence of the CYP2B6 516G> T polymorphism, increased plasma Efavirenz concentrations and early neuropsychiatric side effects in South African HIV-infected patients. AIDS Res Ther. 2010;7:32. doi: 10.1186/1742-6405-7-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mukonzo JK, Owen JS, Ogwal-Okeng J, Kuteesa RB, Nanzigu S, Sewankambo N, et al. Pharmacogenetic-based efavirenz dose modification: suggestions for an African population and the different CYP2B6 genotypes. PLoS One. 2014;9(1):e86919. doi: 10.1371/journal.pone.0086919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Rotger M, Colombo S, Furrer H, Bleiber G, Buclin T, Lee BL, 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):1–5. doi: 10.1097/01213011-200501000-00001. Epub 2005/05/03. [DOI] [PubMed] [Google Scholar]
- 18.Ribaudo HJ, Liu H, Schwab M, Schaeffeler E, Eichelbaum M, Motsinger-Reif AA, 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(5):717–22. doi: 10.1086/655470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rotger M, Tegude H, Colombo S, Cavassini M, Furrer H, Decosterd L, et al. Predictive value of known and novel alleles of CYP2B6 for efavirenz plasma concentrations in HIV-infected individuals. Clin Pharmacol Ther. 2007;81(4):557–66. doi: 10.1038/sj.clpt.6100072. [DOI] [PubMed] [Google Scholar]
- 20.Mukonzo JK, Okwera A, Nakasujja N, Luzze H, Sebuwufu D, Ogwal-Okeng J, et al. Influence of efavirenz pharmacokinetics and pharmacogenetics on neuropsychological disorders in Ugandan HIV-positive patients with or without tuberculosis: a prospective cohort study. BMC Infect Dis. 2013;13:261. doi: 10.1186/1471-2334-13-261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sarfo FS, Zhang Y, Egan D, Tetteh LA, Phillips R, Bedu-Addo G, 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(2):491–9. doi: 10.1093/jac/dkt372. [DOI] [PubMed] [Google Scholar]
- 22.Clifford DB, Evans S, Yang Y, Acosta EP, Goodkin K, Tashima K, et al. Impact of efavirenz on neuropsychological performance and symptoms in HIV-infected individuals. Ann Intern Med. 2005;143(10):714–21. doi: 10.7326/0003-4819-143-10-200511150-00008. [DOI] [PubMed] [Google Scholar]
- 23.Takahashi M, Ibe S, Kudaka Y, Okumura N, Hirano A, Suzuki T, 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(8):983–7. doi: 10.1089/aid.2006.0193. [DOI] [PubMed] [Google Scholar]
- 24.Fumaz CR, Munoz-Moreno JA, Molto J, Negredo E, Ferrer MJ, Sirera G, et al. Long-term neuropsychiatric disorders on efavirenz-based approaches: quality of life, psychologic issues, and adherence. J Acquir Immune Defic Syndr. 2005;38(5):560–5. doi: 10.1097/01.qai.0000147523.41993.47. [DOI] [PubMed] [Google Scholar]
- 25.Stahle L, Moberg L, Svensson JO, Sonnerborg A. Efavirenz plasma concentrations in HIV-infected patients: inter- and intraindividual variability and clinical effects. Ther Drug Monit. 2004;26(3):267–70. doi: 10.1097/00007691-200406000-00008. [DOI] [PubMed] [Google Scholar]
- 26.van Luin M, Bannister WP, Mocroft A, Reiss P, Di Perri G, Peytavin G, et al. Absence of a relation between efavirenz plasma concentrations and toxicity-driven efavirenz discontinuations in the EuroSIDA study. Antivir Ther. 2009;14(1):75–83. [PubMed] [Google Scholar]
- 27.Kappelhoff BS, van Leth F, Robinson PA, MacGregor TR, Baraldi E, Montella F, et al. Are adverse events of nevirapine and efavirenz related to plasma concentrations? Antivir Ther. 2005;10(4):489–98. [PubMed] [Google Scholar]
- 28.Csajka C, Marzolini C, Fattinger K, Decosterd LA, Fellay J, Telenti A, et al. Population pharmacokinetics and effects of efavirenz in patients with human immunodeficiency virus infection. Clin Pharmacol Ther. 2003;73(1):20–30. doi: 10.1067/mcp.2003.22. [DOI] [PubMed] [Google Scholar]
- 29.Srivastava P, Moorthy GS, Gross R, Barrett JS. A sensitive and selective liquid chromatography/tandem mass spectrometry method for quantitative analysis of efavirenz in human plasma. PLoS One. 2013;8(6):e63305. doi: 10.1371/journal.pone.0063305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.International HapMap C. The International HapMap Project. Nature. 2003;426(6968):789–96. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
- 31.Grossberg R, Zhang Y, Gross R. A time-to-prescription-refill measure of antiretroviral adherence predicted changes in viral load in HIV. J Clin Epidemiol. 2004;57(10):1107–10. doi: 10.1016/j.jclinepi.2004.04.002. [DOI] [PubMed] [Google Scholar]
- 32.Clifford DB, Evans S, Yang Y, Acosta EP, Ribaudo H, Gulick RM, et al. Long-term impact of efavirenz on neuropsychological performance and symptoms in HIV-infected individuals (ACTG 5097s) HIV Clin Trials. 2009;10(6):343–55. doi: 10.1310/hct1006-343. Epub 2010/02/06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Brinkhof MW, Boulle A, Weigel R, Messou E, Mathers C, Orrell C, et al. Mortality of HIV-infected patients starting antiretroviral therapy in sub-Saharan Africa: comparison with HIV-unrelated mortality. PLoS Med. 2009;6(4):e1000066. doi: 10.1371/journal.pmed.1000066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Swart M, Evans J, Skelton M, Castel S, Wiesner L, Smith PJ, et al. An Expanded Analysis of Pharmacogenetics Determinants of Efavirenz Response that Includes 3′-UTR Single Nucleotide Polymorphisms among Black South African HIV/AIDS Patients. Front Genet. 2015;6:356. doi: 10.3389/fgene.2015.00356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Winzer R, Langmann P, Zilly M, Tollmann F, Schubert J, Klinker H, et al. No influence of the P-glycoprotein genotype (MDR1 C3435T) on plasma levels of lopinavir and efavirenz during antiretroviral treatment. Eur J Med Res. 2003;8(12):531–4. Epub 2004/01/09. [PubMed] [Google Scholar]
- 36.Dirson G, Fernandez C, Hindlet P, Roux F, German-Fattal M, Gimenez F, et al. Efavirenz does not interact with the ABCB1 transporter at the blood-brain barrier. Pharm Res. 2006;23(7):1525–32. doi: 10.1007/s11095-006-0279-5. Epub 2006/06/17. [DOI] [PubMed] [Google Scholar]
- 37.Stormer E, von Moltke LL, Perloff MD, Greenblatt DJ. Differential modulation of P-glycoprotein expression and activity by non-nucleoside HIV-1 reverse transcriptase inhibitors in cell culture. Pharm Res. 2002;19(7):1038–45. doi: 10.1023/a:1016430825740. Epub 2002/08/16. [DOI] [PubMed] [Google Scholar]
- 38.Janneh O, Chandler B, Hartkoorn R, Kwan WS, Jenkinson C, Evans S, et al. Intracellular accumulation of efavirenz and nevirapine is independent of P-glycoprotein activity in cultured CD4 T cells and primary human lymphocytes. J Antimicrob Chemother. 2009;64(5):1002–7. doi: 10.1093/jac/dkp335. Epub 2009/09/15. [DOI] [PubMed] [Google Scholar]
- 39.Burhenne J, Matthee AK, Pasakova I, Roder C, Heinrich T, Haefeli WE, et al. No evidence for induction of ABC transporters in peripheral blood mononuclear cells in humans after 14 days of efavirenz treatment. Antimicrob Agents Chemother. 2010;54(10):4185–91. doi: 10.1128/AAC.00283-10. Epub 2010/07/28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Winston A, Amin J, Clarke A, Else L, Amara A, Owen A, et al. Cerebrospinal fluid exposure of efavirenz and its major metabolites when dosed at 400 mg and 600 mg once daily: a randomized controlled trial. Clin Infect Dis. 2015;60(7):1026–32. doi: 10.1093/cid/ciu976. [DOI] [PubMed] [Google Scholar]
- 41.Ashraf T, Robillard K, Chan GN, Bendayan R. Role of CNS transporters in the pharmacotherapy of HIV-1 associated neurological disorders. Curr Pharm Des. 2014;20(10):1543–63. doi: 10.2174/13816128113199990464. [DOI] [PubMed] [Google Scholar]
- 42.Aouri M, Barcelo C, Ternon B, Cavassini M, Anagnostopoulos A, Yerly S, et al. In Vivo Profiling and Distribution of Known and Novel Phase I and Phase II Metabolites of Efavirenz in Plasma, Urine, and Cerebrospinal Fluid. Drug Metab Dispos. 2016;44(1):151–61. doi: 10.1124/dmd.115.065839. [DOI] [PubMed] [Google Scholar]
- 43.Abdelhady AM, Desta Z, Jiang F, Yeo CW, Shin JG, Overholser BR. Population pharmacogenetic-based pharmacokinetic modeling of efavirenz, 7-hydroxy- and 8-hydroxyefavirenz. J Clin Pharmacol. 2014;54(1):87–96. doi: 10.1002/jcph.208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ngaimisi E, Mugusi S, Minzi O, Sasi P, Riedel KD, Suda A, et al. Effect of rifampicin and CYP2B6 genotype on long-term efavirenz autoinduction and plasma exposure in HIV patients with or without tuberculosis. Clin Pharmacol Ther. 2011;90(3):406–13. doi: 10.1038/clpt.2011.129. [DOI] [PubMed] [Google Scholar]
- 45.Bienvenu E, Swart M, Dandara C, Ashton M. The role of genetic polymorphisms in cytochrome P450 and effects of tuberculosis co-treatment on the predictive value of CYP2B6 SNPs and on efavirenz plasma levels in adult HIV patients. Antiviral Res. 2014;102:44–53. doi: 10.1016/j.antiviral.2013.11.011. [DOI] [PubMed] [Google Scholar]
- 46.Swart M, Skelton M, Ren Y, Smith P, Takuva S, Dandara C. High predictive value of CYP2B6 SNPs for steady-state plasma efavirenz levels in South African HIV/AIDS patients. Pharmacogenet Genomics. 2013;23(8):415–27. doi: 10.1097/FPC.0b013e328363176f. [DOI] [PubMed] [Google Scholar]
- 47.Gandhi M, Greenblatt RM, Bacchetti P, Jin C, Huang Y, Anastos K, et al. A single-nucleotide polymorphism in CYP2B6 leads to >3-fold increases in efavirenz concentrations in plasma and hair among HIV-infected women. J Infect Dis. 2012;206(9):1453–61. doi: 10.1093/infdis/jis508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Leger P, Dillingham R, Beauharnais CA, Kashuba AD, Rezk NL, Fitzgerald DW, et al. CYP2B6 variants and plasma efavirenz concentrations during antiretroviral therapy in Port-au-Prince, Haiti. J Infect Dis. 2009;200(6):955–64. doi: 10.1086/605126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Carr DF, la Porte CJ, Pirmohamed M, Owen A, Cortes CP. Haplotype structure of CYP2B6 and association with plasma efavirenz concentrations in a Chilean HIV cohort. J Antimicrob Chemother. 2010;65(9):1889–93. doi: 10.1093/jac/dkq260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sukasem C, Cressey TR, Prapaithong P, Tawon Y, Pasomsub E, Srichunrusami C, et al. Pharmacogenetic markers of CYP2B6 associated with efavirenz plasma concentrations in HIV-1 infected Thai adults. Br J Clin Pharmacol. 2012;74(6):1005–12. doi: 10.1111/j.1365-2125.2012.04288.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sukasem C, Manosuthi W, Koomdee N, Santon S, Jantararoungtong T, Prommas S, et al. Low level of efavirenz in HIV-1-infected Thai adults is associated with the CYP2B6 polymorphism. Infection. 2014;42(3):469–74. doi: 10.1007/s15010-013-0560-6. [DOI] [PubMed] [Google Scholar]
- 52.Sukasem C, Chamnanphon M, Koomdee N, Santon S, Jantararoungtong T, Prommas S, et al. Pharmacogenetics and clinical biomarkers for subtherapeutic plasma efavirenz concentration in HIV-1 infected Thai adults. Drug Metab Pharmacokinet. 2014;29(4):289–95. doi: 10.2133/dmpk.dmpk-13-rg-077. [DOI] [PubMed] [Google Scholar]
- 53.Manosuthi W, Sukasem C, Thongyen S, Nilkamhang S, Manosuthi S, Sungkanuparph S. CYP2B6 18492T->C polymorphism compromises efavirenz concentration in coinfected HIV and tuberculosis patients carrying CYP2B6 haplotype *1/*1. Antimicrob Agents Chemother. 2014;58(4):2268–73. doi: 10.1128/AAC.02384-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Penell J, Lind L, Fall T, Syvanen AC, Axelsson T, Lundmark P, et al. Genetic variation in the CYP2B6 gene is related to circulating 2,2′,4,4′-tetrabromodiphenyl ether (BDE-47) concentrations: an observational population-based study. Environ Health. 2014;13(1):34. doi: 10.1186/1476-069X-13-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Pozo K, Harner T, Wania F, Muir DCG, Jones KC, Barrie LA. Toward a Global Network for Persistent Organic Pollutants in Air: Results from the GAPS Study. Environmental Science & Technology. 2006;40(16):4867–73. doi: 10.1021/es060447t.. [DOI] [PubMed] [Google Scholar]
- 56.Shunthirasingham C, Gouin T, Lei YD, Wania CO, Mmereki BT, Masamba W, et al. Year-round measurements of PBDEs in the atmosphere of tropical Costa Rica and subtropical Botswana. 5th International Symposium on Brominated Flame Retardants; April 7–9, 2010; Kyoto, Japan. 2010. [Google Scholar]
- 57.Sueyoshi T, Li L, Wang H, Moore R, Kodavanti PR, Lehmler HJ, et al. Flame retardant BDE-47 effectively activates nuclear receptor CAR in human primary hepatocytes. Toxicol Sci. 2014;137(2):292–302. doi: 10.1093/toxsci/kft243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.von Richter O, Pitarque M, Rodriguez-Antona C, Testa A, Mantovani R, Oscarson M, et al. Polymorphic NF-Y dependent regulation of human nicotine C-oxidase (CYP2A6) Pharmacogenetics. 2004;14(6):369–79. doi: 10.1097/00008571-200406000-00006. [DOI] [PubMed] [Google Scholar]
- 59.Kheradpour P, Kellis M. Systematic discovery and characterization of regulatory motifs in ENCODE TF binding experiments. Nucleic Acids Res. 2014;42(5):2976–87. doi: 10.1093/nar/gkt1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Al Koudsi N, Ahluwalia JS, Lin SK, Sellers EM, Tyndale RF. A novel CYP2A6 allele (CYP2A6*35) resulting in an amino-acid substitution (Asn438Tyr) is associated with lower CYP2A6 activity in vivo. Pharmacogenomics J. 2009;9(4):274–82. doi: 10.1038/tpj.2009.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Roadmap Epigenomics C, Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, et al. Integrative analysis of 111 reference human epigenomes. Nature. 2015;518(7539):317–30. doi: 10.1038/nature14248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Dhoro M, Zvada S, Ngara B, Nhachi C, Kadzirange G, Chonzi P, et al. CYP2B6*6, CYP2B6*18, Body weight and sex are predictors of efavirenz pharmacokinetics and treatment response: population pharmacokinetic modeling in an HIV/AIDS and TB cohort in Zimbabwe. BMC Pharmacol Toxicol. 2015;16:4. doi: 10.1186/s40360-015-0004-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gross R, Bilker WB, Friedman HM, Strom BL. Effect of adherence to newly initiated antiretroviral therapy on plasma viral load. AIDS. 2001;15(16):2109–17. doi: 10.1097/00002030-200111090-00006. [DOI] [PubMed] [Google Scholar]
- 64.Quinn TC, Brookmeyer R, Kline R, Shepherd M, Paranjape R, Mehendale S, et al. Feasibility of pooling sera for HIV-1 viral RNA to diagnose acute primary HIV-1 infection and estimate HIV incidence. AIDS. 2000;14(17):2751–7. doi: 10.1097/00002030-200012010-00015. [DOI] [PubMed] [Google Scholar]