Skip to main content

Factors affecting antiretroviral pharmacokinetics in HIV-infected women with virologic suppression on combination antiretroviral therapy: a cross-sectional study

Abstract

Background

Although some studies show higher antiretroviral concentrations in women compared to men, data are limited. We conducted a cross-sectional study of HIV-positive women to determine if protease inhibitor (PI) and non-nucleoside reverse transcriptase inhibitor (NNRTI) Cmin and Cmax values were significantly different than historical general population (predominantly male) averages and to evaluate correlates of higher concentrations.

Methods

HIV-positive women with virologic suppression (viral load < 50copies/mL) on their first antiretroviral regimen were enrolled. Timed blood samples for Cmin and Cmax were drawn weekly for 3 weeks. The ratio of each individual’s median Cmin and Cmax to the published population mean values for their PI or NNRTI was calculated and assessed using Wilcoxon sign-rank. Intra- and inter-patient variability of antiretroviral drug levels was assessed using coefficient of variation and intra-class correlation. Linear regression was used to identify correlates of the square root-transformed Cmin and Cmax ratios.

Results

Data from 82 women were analyzed. Their median age was 41 years (IQR=36-48) and duration of antiretrovirals was 20 months (IQR=9-45). Median antiretroviral Cmin and Cmax ratios were 1.21 (IQR=0.72-1.89, p=0.003) (highest ratios for nevirapine and lopinavir) and 0.82 (IQR=0.59-1.14, p=0.004), respectively. Nevirapine and efavirenz showed the least and unboosted atazanavir showed the most intra- and inter-patient variability. Higher CD4+ count correlated with higher Cmin. No significant correlates for Cmax were found.

Conclusions

Compared to historical control data, Cmin in the women enrolled was significantly higher whereas Cmax was significantly lower. Antiretroviral Cmin ratios were highly variable within and between participants. There were no clinically relevant correlates of drug concentrations.

Trial registration

NCT00433979

Peer Review reports

Background

Since the advent of combination antiretroviral therapy (cART), there has been a dramatic decrease in the mortality of individuals infected with human immunodeficiency virus (HIV) [1]. Traditionally, this combination has included two drugs from the antiretroviral class of nucleoside reverse transcriptase inhibitors (NRTIs) and either one from the class of protease inhibitors (PIs) or one from the class of non-nucleoside reverse transcriptase inhibitors (NNRTIs) [2]. If the third agent is from the PI class, it is often combined with a low dose of ritonavir in order to boost its drug levels [2]. More recently, raltegavir has been added as an option for the third agent, but remains rarely used in Canada due to cost [3].

Despite these tremendous advances in HIV management, there remain several important complications related to antiretroviral drug use, one of the most notable being drug-related adverse events (AEs) and toxicities [4], which can negatively impact patients’ quality of life, contributing to non-adherence and drug resistance and ultimately effectiveness. Adverse drug reactions to antiretrovirals are a major reason for discontinuing or changing therapy [5, 6]. Furthermore, these drug-related complications can significantly contribute to morbidity, hospitalizations, and mortality in this population [710].

Women constitute one of the fastest-rising population groups at risk for infection with HIV, representing over 50% of cases worldwide, and approximately 25% of new cases in the United States (U.S.) and 28% of new cases in Canada [13]. Surprisingly, little is known about the differential efficacy and toxicity of various antiretroviral drugs in women compared to men [11]. This gap in knowledge is a result of the initial exclusion and continued underrepresentation of women in antiretroviral clinical trials [12]. This circumstance has slowly started to change, and there are now more longitudinal studies examining women-specific issues [13, 14]. Many studies in the general population have shown that AEs are more common in women than in men [15]. In the HIV-infected population, higher incidence rates of increased systemic symptoms (such as nausea, vomiting and diarrhea), as well as organ toxicity (including anemia, hepatotoxicity, pancreatitis, lactic acidosis, peripheral neuropathy, and notable lipodystrophy), have been observed in women compared to men [11, 16, 17]. For nevirapine, female gender and higher CD4+ cell counts were risk factors for fatal hepatitis, and this observation has led regulatory authorities to release warnings on its use in certain female populations [18].

Most of these studies assessing sex differences in antiretroviral AEs rates are limited in that they only identify the issue of increased toxicity in women and do not try to elucidate the cause or management [16, 19, 20]. The potential causes of these gender differences in antiretroviral toxicities may have a sound biologic basis possibly related to differences in physiology and/or the influence of sex hormones on drug metabolism. All aspects of drug handling and exposure may be different in women versus men, including bioavailability (with lower gastric emptying time due to hormonal contraception use or pregnancy) and distribution (lower body weight, smaller organ size, higher body fat content, altered gastric motility, greater organ blood flow, and altered protein binding secondary to endogenous or exogenous estrogens). Metabolism and elimination of drugs have also exhibited gender differences related to differences in expression and activity of various drug transporters and metabolizing enzymes [20, 21]. Of particular interest, a better understanding of antiretroviral pharmacokinetics (PK) in women and how these drug levels impact AEs and toxicities in women is crucial, and will lead to more effective methods of treatment, reduced discomfort, possible enhanced adherence and improved morbidity and/or mortality.

Although a few investigations have considered PK differences that occur between men and women, most of these studies involved the use of older antiretroviral agents and were assessed in small sample sizes [2129]. Furthermore, a number of potentially confounding variables such as race, age, weight, menstruation, and hepatitis co-infection have not been explored as determinants impacting drug levels in a female population. We conducted a cross-sectional study of HIV-positive women taking their first combination ART to determine if drug levels (Cmin and Cmax) of currently used PIs and NNRTIs were significantly higher in this population as compared to the historical general (predominantly male) population and to evaluate correlates of higher concentrations.

Methods

Ethics statement

The study was reviewed and received ethics approval by the Full Institutional Research Ethics Board (REB) of main coordinating research centre, Women’s College Research Institute, Toronto, Canada (REB# 2006–003). Additional Full Institutional REB approval was obtained from each research site prior to commencement. All study personnel were trained in and practiced under the principles of the Declaration of Helsinki. All study candidates were informed about this study and written informed consent was obtained from every participant prior enrollment.

Study population

We carried out a cross-sectional study with participants who met the following inclusion criteria: 1) HIV-positive, 2) biologically female, 3) 18 years of age or older, 4) taking their first cART regimen containing either a PI or an NNRTI with a backbone of NRTI as per common practice for at least 3 months (but could have had prior switches that were not due to virologic failure), 5) taking either a PI or an NNRTI but not both, 6) if taking a PI, must have been taking only one PI excluding low dose ritonavir used as boosting and 7) had evidence of full virologic suppression (HIV-1 RNA VL < 50 copies/mL) on at least two occasions at least one month apart. The patient population was limited to women who were on their first cART regimen in order to ensure a more homogeneous population. The current analysis was limited to candidates who were taking the following antiretroviral drugs (atazanavir, atazanavir boosted with ritonavir, lopinavir boosted with ritonavir, efavirenz and nevirapine) [3, 30]. Participants could not take both a PI and NNRTI or two PIs excluding ritonavir as there are multiple drug interactions between these agents which would lead to uninterpretable results. Further, participating women had to have full virologic suppression to avoid inclusion of women who experience difficulty with drug adherence. The 3 month requirement for being on a cART regimen was mandated to 1) ensure stabilization of adherence as there may be an adjustment period as patients get accustomed to taking their new drug regimen, 2) eliminate early discontinuations due to drug toxicities which often happen during the first 3 months of therapy, and to a lesser extend 3) to ensure drug steady state as there is some variation between drugs [5].

A planned sample size of 80 was calculated in order to estimate if the mean ratio of the Cmin values to historical values for the general population was significantly different from 1.0, assuming an alternative hypothesis of 1.2 with a standard deviation of 0.64, 80% power and a significance level of 0.05. Recruitment was conducted from February, 2007 to November, 2008 from 14 primary care and specialty HIV clinics from across Canada. Recruitment and study qualification determination was carried out by the site investigator and research staff. The recruitment was carried out in a non-random consecutive manner as research staff were instructed to invite every consecutive qualifying woman who received services in their clinic on all days that care was provided.

Data collection

Study visits occurred at weekly intervals for a three-week period. During the baseline visit, demographic and HIV and other medical history data was collected including concurrent medications, validated questionnaires pertaining to antiretroviral adherence [31] and symptom distress [32] were completed, weight was measured and blood work to assess laboratory values was drawn.

Pharmacokinetic analysis

At each of the three visits, patients had a pre-dose (Cmin) and a maximum (Cmax) PI or NNRTI drug level drawn. Patients were asked to fast from midnight of the previous night. One hour before their scheduled morning dose the pre-dose blood collection was drawn (Cmin t) (alternative arrangements were made if the patient took her drugs in the evening). Following the pre-dose blood collection, patients consumed a standard breakfast (consisting of 50% carbohydrate, 30% fat, 20% protein) after which time the morning antiretroviral dose was administered. Cmax levels were drawn at 2 hours post-dose for atazanavir 400 mg QD [33], 3 hours post-dose for atazanavir/ritonavir 300/100 mg QD [33], 4 hours post-dose for lopinavir/ritonavir 400/100 mg BID [34], 5 hours post-dose for efavirenz 600 mg QD [35], and 2 hours post-dose for nevirapine (both 200 mg BID and 400 mg QD [36]). The plasma drug level samples were all stored in cryovials at −20°C or lower and shipped for concurrent assessment at the end of the study. Concentrations of the PIs and the NNRTIs in plasma were measured simultaneously by sensitive and selective, validated high-performance liquid chromatography coupled to tandem mass-spectroscopy (LC-MS/MS) [37]. All samples were analyzed at the pharmacokinetic laboratory at the Ottawa Hospital Research Institute in Ottawa, Canada.

The PK endpoints for each patient were determined by taking the median of the three weekly values for the Cmin and Cmax. The rationale for measuring drug levels at three separate time points was that there is potentially intra-individual variability in drug levels, especially for PIs [38, 39]. Using the median value for the endpoints eliminated outliers. The three drug level measurements allowed for the calculation of intra-patient variability.

Statistical analyses

Baseline characteristics of the study population were summarized using medians and interquartile ranges (IQR) for continuous variables and frequencies and proportions for categorical variables.

The mean Cmin and Cmax drug levels in the historical HIV population were taken from the most recent product monograph when possible, or from a published study if the product monograph did not report these values [3336]. Next, for each subject the ratio of their median Cmin and Cmax for their main PI or NNRTI to this published drug level was calculated and used as the primary outcome as previously reported by Burger et al. [40] Due to the lack of published population PK data for ritonavir when included as part of regimen in a “boosting” role, we only calculated the ratios for the main PI in a participant’s regimen. Differences between the Cmin ratio and Cmax ratio to the population mean were conducted using a Wilcoxon sign-rank test for a median of 1. For comparison purposes, PK levels were dichotomized into high and low levels with a high level defined as ≥ 1.5 X arithmetic population mean of the Cmin and Cmax for each drug. Inter-patient variability of Cmin and Cmax for each antiretroviral drug was assessed by calculating the coefficient of variation (CV) using each individual’s median Cmin and Cmax. An intra-patient CV for each participant was calculated from the Cmin and Cmax values obtained at each of the 3 visits. These values are summarized using median and IQR of the individual CVs. The inter-patient CV is a measure of variation among individuals whereas intra-patient CV measures variation within an individual The inter-patient CV is calculated as the ratio of the standard deviation to the mean X 100; the higher the value, the more variability exists. Finally, linear regression models were fit to assess correlates of the square root-transformed Cmin and Cmax ratios. The square-root transformation was used because the untransformed Cmin and Cmax ratios were both right-skewed. The regression models included indicator variables to account for the different antiretroviral medications and each potential correlate was included separately in an adjusted model. Statistical analyses were performed using SAS Version 9.2 (SAS Institute, Cary, North Carolina, USA).

Results

Study population

Ninety women were enrolled from 14 sites across Canada between 2/2007 and 11/2008. Eight women were excluded for the following reasons: one due to missing data, six were not on a current cART regimen deemed eligible to be included in this analysis, and one because she was not on standard dosing schedule. The data is summarized for the remaining 82 patients. Median age of the study population was 41 years (IQR 36–48) and 56% identified as Black. The median times since their HIV diagnosis and start of their current cART regimen were 7 years (IQR 3–11) and 20 months (IQR 9–45), respectively. Fifty-seven percent were taking a PI-containing regimen (81% being ritonavir-boosted and 19% unboosted) and 43% a NNRTI-containing regimen. All participants had an undetectable viral load with a median CD4+ count at the time of enrolment of 487 cells/μL (IQR 380–621). Additional demographic and clinical variables of the study population are summarized in Table 1. No relevant concurrent medications were being taken by the participants.

Table 1 Demographic characteristics of study participants

Drug level ratios by antiretroviral drug

The Cmin and Cmax summaries and historical values used to calculate the ratios are reported in Table 2[3336]. Paired values for the Cmin and Cmax data for each dosing regime are shown in Figure 1. Overall, the median ratio of the participants’ Cmin to historical mean values was 1.21 (IQR 0.72-1.89, p=0.003) and the median ratio of Cmax to historical mean values was 0.82 (IQR 0.59-1.14, p=0.004). Twenty-eight participants (34%) had Cmin ≥ 1.5 X arithmetic population mean of the Cmin for each drug and seven participants (9%) had Cmax ≥ 1.5 X arithmetic population mean of the Cmax for each drug; all on an NNRTI. The median ratios of Cmin and Cmax to historical mean values for each specific antiretroviral drug overall and for each dose are presented in Table 3.

Table 2 Reference population mean and Study Participants C min and C max by antiretroviral drug
Figure 1
figure 1

Paired C min and C max values for each participant by antiretroviral drug and dose. The Cmin values are identified by circles and the Cmax values by triangles. The reported population mean Cmin and Cmax values for each antiretroviral drug and dose are presented with the hatched lines (−−−).

Table 3 Ratio of C min and C max values to Historical Population Means by antiretroviral drug and dose

Intra-patient and inter-patient variability of antiretroviral drug levels

The intra- and inter-patient variability of each antiretroviral drug’s Cmin and Cmax was assessed using CV; data are presented in Table 4. Data from 2 participants for whom PK samples were only obtained at one visit were excluded from the intra-patient variability calculations. Nevirapine BID and lopinavir/ritonavir BID showed the least inter-patient variability with the lowest CVs for both Cmin and Cmax. Nevirapine BID and efavirenz had the lowest intra-patient variability variability for Cmin, while nevirapine BID, lopinavir/ritonavir BID, and efavirenz demonstrated lowest intra-patient variability for Cmax. Inter- and intra- patient variability was high for atazanavir, particularly when not boosted with ritonavir.

Table 4 Inter-patient and Intra-patient variability of C min and C max for each antiretroviral agent

Linear regression models for antiretroviral Cmin and Cmax

Linear regression models to assess the relationship between the square root-transformed Cmin ratio and demographic and clinical variables of the population were carried out (Table 5). Higher CD4+ cell count was the only variable significantly associated with higher Cmin ratios (Beta coefficient=0.04/ 100 cells per μL increase, 95% CI=0.003-0.080; p=0.03). Similar analyses were carried out for the assessment of the correlates of the square root-transformed Cmax ratios and only injection drug use showed a trend to be negatively associated with Cmax ratio (Beta coefficient=−0.10, 95% CI=−0.23-0.02; p=0.10).

Table 5 Linear regression models for square-root transformed C min and C max

Discussion

In this cross-sectional study of 82 HIV-positive women taking cART with full virologic suppression, median pre-dose drug levels were found to be significantly higher than historical controls. Approximately one- third of women had a Cmin value more than 1.5 times higher than historical control values. Since the majority of historical controls consist of men, this finding supports the growing literature indicating that drug levels are higher in women than in men.

Our findings also support the increasing body of literature on the gender differences in drug disposition and PK of all drugs. However, in the HIV population, these differences may in part be due to race as many of the historical controlled participants were White men, and as in our study, many of the females infected with HIV globally are Black. Several studies have investigated the relationship of antiretroviral PK with genetically determined factors that might differ by individual ancestral history [4145]. Racial differences have been found in P-glycoprotein (PGP) activity, an efflux protein which pumps its substrates, including the PIs, out of cells, away from their site of action. For example, Africans are four times more likely than white Americans or Japanese individuals to have the CC genotype at position 3435 of the gene coding for PGP [45]. Fellay and colleagues [41] demonstrated that persons with the CC genotype have higher PGP-activity, higher serum drug levels. Similarly, there are racial differences in the prevalence of slow-metabolizers of specific pathways, for example 2D6 and 2B6, of the cytochrome P-450 system of drug metabolism in the liver [43]. Thus, there may be genetically determined heterogeneity in the in vivo transport and metabolism of antiretroviral agents, resulting in variation in serum and intracellular drug levels.

A number of other studies have assessed the gender differences in antiretroviral PK, particularly of the older PIs. In a study of 186 patients (15.6% female), Fletcher and colleagues [23] demonstrated that serum levels of saquinavir were significantly higher in women than in men, independent of body size. Another study sought to characterize the PK of saquinavir (1000 mg BID), lopinavir (400 mg BID), and ritonavir (100 mg BID) in a multidrug rescue therapy study [24]. Twenty-five patients (28% women) were included in the study group and fifteen (20% women) were included in the comparison group that did not receive saquinavir. Area under the curve (AUC), Cmax, and Cmin values for saquinavir and ritonavir were significantly higher in women than in men, though there were no significant differences in weight or body mass indexes between genders. Work by Dickinson and colleagues, who looked at the PK of saquinavir and ritonavir in 34 patients on this combination, showed that in women a higher exposure to saquinavir might, at least in part, be driven by higher exposure to ritonavir [25]. Pharmacokinetic studies of newer PIs have only been reported in product monographs limited to data showing that women have modest increases in AUC of approximately 20% for lopinavir, atazanavir and darunavir [33, 34, 46]. In terms of NNRTIs, investigators have demonstrated higher serum efavirenz and nevirapine levels in women [2629]. A previous report using a full 12 hour PK of nevirapine showed a gender difference of an 18.9% lower AUC in males when corrected for body weight [26]. In the same study, pregnant women had lower nevirapine exposure and this effect did not seem to be driven by body weight.

Our data also adds to the literature on gender differences in Cmax of current antiretroviral agents. These observations should be interpreted with caution as the collection of Cmax samples in this study was done at a single standardized timepoint; therefore, if the Cmax sampling time was slightly off from the real Tmax then the resulting Cmax observations would be interpreted as lower. In our study, atazanavir had the lowest ratios of both Cmin and Cmax versus historical reference values, but also the greatest observed inter-patient variability. In general, a higher degree of variability was noted for Cmin as compared to Cmax values.

Few studies have assessed intra-patient variability of antiretroviral drug levels for either gender. Over our 3-week sampling period, we found higher intra-patient variability in the PI-based regimens compared to the NNRTIs; a similar finding to the retrospective summary provided by Fabbiani and colleagues [47]. More specifically, unboosted atazanavir had the highest intra- and inter-patient CVs for Cmin and Cmax, whereas efavirenz and nevirapine had the lowest CVs for intra-patient variability. This most likely reflects the longer half-life for both nevirapine and efavirenz, compared to PIs. Despite ritonavir boosting for both, the intra-patient variability for lopinavir Cmax was substantially lower than that of atazanavir, which may reflect the absence of a food and gastric pH effects on lopinavir absorption as compared to atazanavir absorption. In addition, lopinavir is co-formulated with ritonavir, thus ensuring simultaneous coadministration of both drugs, whereas atazanavir absorption may be impacted if ritonavir is not taken at the same time for reasons such as patient choice (e.g., concerns of selective side effects of ritonavir) or forgetfulness since ritonavir capsules require refrigeration. In our study, inter-patient CV for Cmin and Cmax of boosted and unboosted atazanavir appeared to be lower than historical values from predominantly male populations, while intrapatient CV for Cmin of unboosted atazanavir appeared to be higher than historical data. For NNRTIs, observed intra- and inter-patient Cmin CVs of efavirenz and nevirapine appeared to be lower than historical controls. As such, the existence of sex-based differences in antiretroviral variability cannot be ruled out.

Correlates of antiretroviral drug levels in women have only been investigated by few studies. Gibbons and colleagues examined the potential for age-dependent changes in lopinavir and efavirenz levels in female subjects in a retrospective analysis of therapeutic drug monitoring on non-pregnant women receiving either lopinavir/ritonavir 400 mg /100 mg twice daily or efavirenz 600 mg daily [48]. They found that women > 50 years of age had significantly higher 8–16 hour efavirenz levels when compared to women < 40 years of age (p=0.046). In our study, we found that Cmin ratio increased with CD4+ cell count and there was some indication that it was lower among injection drug users. There were no statistically significant correlates of Cmax. Of note, there was no correlation between drug levels and body weight or body mass index. The lack of statistical significance may be related to the attempts to make our population homogenous.

Our study has a number of limitations, most notably the lack of a concurrent male control group which would have allowed real-time assessment of sex differences in drug levels. The fact that historical control data was used results in the inability to report on the demographics of the controls including the true proportion of cases that were male, ethnicity, weight, and co-infection status. The restriction of our study to women on their first cART regimen with virologic suppression to ensure homogeneity likely contributed to the lack of range amongst our covariates, decreasing our ability to detect differences and associations. If higher concentrations were associated with toxicity, then the women may have switched off the regimen or been inconsistently adherent and experienced viral failure and would not have been eligible for this data set. The demand and time commitment for the participants was high and likely led to some degree of selection bias of women who are committed to therapy and research. Also, there were small sample sizes for each drug dosing (e.g. only 4 participations taking lopinavir/ritonavir 800 mg/100 mg OD and 8 taking nevirapine 400 mg OD) and for this reason it was difficult to make conclusions regarding specific drugs and drug dosing.

Conclusion

In summary, our data adds to the growing literature on the gender differences of antiretroviral drug levels. Our study showed that the PIs and NNRTIs overall Cmin ratios were significantly higher in our HIV-positive female participants as compared to men (historical controls). In particular, we observed the highest Cmin for nevirapine and lopinavir. These latter observations add to the literature by providing data on newer antiretroviral agents, and could explain nevirapine’s important gender-specific drug toxicity. Our study was also able to study intra- and inter-patient variability for PIs and NNRTIs Cmin ratios and found significant variability particularly for unboosted and boosted atazanavir. This intra-patient variability could have clinical implications with respect to toxicity and efficacy. These findings require further study to elucidate the mechanism and clinical consequences of these differences and results. It also emphasizes the importance of gender-specific analyses when investigating antiretroviral efficacy and toxicity.

Abbreviations

AEs:

Adverse events

AIDS:

Acquired immune deficiency syndrome

ART:

Antiretroviral therapy

AUC:

Area under the curve

BID:

Twice daily

BMI:

Body mass index

cART:

Combination antiretroviral therapy

CHUL:

Centre Hospitalier Universitaire de Quebec

CI:

Confidence interval

CIHR:

Canadian Institutes of Health Research

Cmax:

Maximum plasma concentration of the drug

Cmin:

Minimum plasma concentration of the drug

CV:

Coefficient variation

CVs:

Coefficient variations

FRSQ:

Fonds de recherche en santé du Québec

HIV:

Human immunodeficiency virus

IDU:

Injection drug user

IQR:

Interquartile range

NNRTI:

Non nucleoside reverse transcriptase inhibitor

NNRTIs:

Non nucleoside reverse transcriptase inhibitors

PGP:

P-glycoprotein

PI:

Protease inhibitor

PIs:

Protease inhibitors

PK:

Pharmacokinetics

QD:

Once daily

REB:

Research ethics board

TDM:

Therapeutic drug monitoring

VL:

Viral load

References

  1. Palella FJ, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg SD: Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med. 1998, 338 (13): 853-860. 10.1056/NEJM199803263381301.

    Article  PubMed  Google Scholar 

  2. Zeldin RK, Petruschke RA: Pharmacologic and therapeutic properties of ritonavir-boosted protease inhibitor therapy in HIV-infected patients. J Antimicrob Chemother. 2004, 53 (1): 4-9.

    Article  CAS  PubMed  Google Scholar 

  3. Panel on Antiretroviral Guidelines for Adults and Adolescents: Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services. 2011, 1-167. [http://www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf]

    Google Scholar 

  4. Montessori V, Press N, Harris M, Akagi L, Montaner JS: Adverse effects of antiretroviral therapy for HIV infection. Can Med Assoc J. 2004, 170 (2): 229-238.

    Google Scholar 

  5. Elzi L, Marzolini C, Furrer H, Ledergerber B, Cavassini M, Hirschel B, Vernazza P, Bernasconi E, Weber R, Battegay M: Swiss HIV Cohort Study. Treatment modification in human immunodeficiency virus-infected individuals starting combination antiretroviral therapy between 2005–2008. Arch Int Med. 2010, 170 (1): 57-65. 10.1001/archinternmed.2009.432.

    Article  CAS  Google Scholar 

  6. Cicconi P, Cozzi-Lepri A, Castagna A, Trecarichi EM, Antinori A, Gatti F, Cassola G, Sighinolfi L, Castelli P, d’Arminio Monforte A, ICoNA Foundation Study Group: Insights into reasons for discontinuation according to year of starting first regimen of highly active antiretroviral therapy in a cohort of antiretroviral-naïve patients. HIV Med. 2010, 11 (2): 104-13. 10.1111/j.1468-1293.2009.00750.x.

    Article  CAS  PubMed  Google Scholar 

  7. Foisy MM, Gough K, Quan C, Harris K, Ibanez D, Phillips A: Hospitalization due to adverse drug reactions and drug interactions before and after HAART. Can J Inf Dis. 2000, 11 (4): 193-201.

    CAS  Google Scholar 

  8. Mehta U, Durrheim DN, Blockman M, Kredo T, Gounden R, Barnes KI: Adverse drug reactions in adult medical inpatients in a South African hospital serving a community with a high HIV/AIDS prevalence:prospective observational study. Br J Clin Pharmacol. 2008, 65 (3): 396-406. 10.1111/j.1365-2125.2007.03034.x.

    Article  PubMed  Google Scholar 

  9. Núñez MJ, Martín-Carbonero L, Moreno V, Valencia E, Garcia-Samaniego J, Castillo JG, Barreiro P, González-Lahoz J, Soriano V: Impact of antiretroviral treatment-related toxicities on hospital admissions in HIV-infected patients. AIDS Res Hum Retroviruses. 2006, 22 (9): 825-829. 10.1089/aid.2006.22.825.

    Article  PubMed  Google Scholar 

  10. Núñez M: Clinical syndromes and consequences of antiretroviral-related hepatotoxicity. Hepatology. 2010, 52 (3): 1143-55. 10.1002/hep.23716.

    Article  PubMed  Google Scholar 

  11. Clark R: Sex differences in antiretroviral therapy-associated intolerance and adverse events. Drug Safe. 2005, 28 (12): 1075-1083. 10.2165/00002018-200528120-00003.

    Article  CAS  Google Scholar 

  12. d’Arminio Monforte A, González L, Haberl A, Sherr L, Ssanyu-Sseruma W, Walmsley SL, Women for Positive Action: Better mind the gap:addressing the shortage of HIV-positive women in clinical trials. AIDS. 2010, 24 (8): 1091-1094. 10.1097/QAD.0b013e3283390db3.

    Article  PubMed  Google Scholar 

  13. Barkan SE, Melnick SL, Preston-Martin S, Weber K, Kalish LA, Miotti P, Young M, Greenblatt R, Sacks H, Feldman J, The Women’s Interagency HIV Study: WIHS collaborative study group. Epidemiology. 1998, 9 (2): 117-125. 10.1097/00001648-199803000-00004.

    Article  CAS  PubMed  Google Scholar 

  14. Schoeni-Affolter F, Ledergerber B, Rickenbach M, Rudin C, Günthard HF, Telenti A, Furrer H, Yerly S, Francioli P, Swiss HIV Cohort Study: Cohort: profile:the Swiss HIV cohort study. Int J Epidemiol. 2010, 39 (5): 1179-1189.

    Article  PubMed  Google Scholar 

  15. Tran C, Knowles SR, Liu BA, Shear NH: Gender differences in adverse drug reactions. J Clin Pharmacol. 1998, 38 (11): 1003-1009. 10.1177/009127009803801103.

    Article  CAS  PubMed  Google Scholar 

  16. Nicastri E, Leone S, Angeletti C, Palmisano L, Sarmati L, Chiesi A, Geraci A, Vella S, Narciso P, Corpolongo A, Andreoni M: Sex issues in HIV-1-infected persons during highly active antiretroviral therapy:a systematic review. J Antimicrob Chemother. 2007, 60: 724-732. 10.1093/jac/dkm302.

    Article  CAS  PubMed  Google Scholar 

  17. Emery J, Pick N, Mills EJ, Cooper CL: Gender differences in clinical, immunological, and virological outcomes in highly active antiretroviral-treated HIV-HCV coinfected patients. Patient Prefer Adherence. 2010, 4: 97-103.

    PubMed  PubMed Central  Google Scholar 

  18. Viramune (nevirapine) Package insert. 2010, Ridgefield, CT, USA: Boehringer Ingelheim Pharmaceuticals, Inc

  19. Floridia M, Giuliano M, Palmisano L, Vella S: Gender differences in the treatment of HIV infection. Pharmacol Res. 2008, 58 (3–4): 173-182.

    Article  CAS  PubMed  Google Scholar 

  20. Umeh OC, Currier JS: Sex differences in pharmacokinetics and toxicity of antiretroviral therapy. Exper Opin Drug Metab Toxicol. 2006, 2 (2): 273-283. 10.1517/17425255.2.2.273.

    Article  CAS  Google Scholar 

  21. Gandhi M, Aweeka F, Greenblatt RM, Blashke TF: Sex differences in pharmacokinetics and pharmacodynamics. Annu Rev Pharmacol Toxicol. 2004, 44: 499-523. 10.1146/annurev.pharmtox.44.101802.121453.

    Article  CAS  PubMed  Google Scholar 

  22. Ofotokun I, Chuck SK, Hitti JE: Antiretroviral pharmacokinetic profile: a review of sex differences. Gend Med. 2007, 4 (2): 106-119. 10.1016/S1550-8579(07)80025-8.

    Article  PubMed  Google Scholar 

  23. Fletcher CV, Jiang H, Brundage RC, Acosta EP, Haubrich R, Katzenstein D, Gulick RM: Sex-based differences in saquinavir pharmacology and virologic response in AIDS Clinical Trials Group Study 359. J Infect Dis. 2004, 189 (7): 1176-1184. 10.1086/382754.

    Article  CAS  PubMed  Google Scholar 

  24. Ribera E, Lopez RM, Diaz M, Pou L, Ruiz L, Falcó V, Crespo M, Azuaje C, Ruiz I, Ocaña I, Clotet B, Pahissa A: Steady-state pharmacokinetics of a double-boosting regimen of saquinavir soft gel plus lopinavir plus minidose ritonavir in human immunodeficiency virus-infected adults. Antimicrob Agents Chemother. 2004, 48 (11): 4256-4262. 10.1128/AAC.48.11.4256-4262.2004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dickinson L, Boffito M, Back DJ, Khoo SH, Pozniak AL, Mugyenyi P, Merry C, Autar RS, Burger DM, Aarons LJ: Population pharmacokinetics of ritonavir-boosted saquinavir regimens in HIV-infected individuals. J Antimicrob Chemother. 2008, 62 (6): 1344-1355. 10.1093/jac/dkn399.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. von Hentig N, Carlebach A, Gute P, Knecht G, Klauke S, Rohrbacher M, Stocker H, Kurowski M, Harder S, Staszewski S, Haberl A: A comparison of the steady-stage pharmacokinetics of nevirapine in men, nonpregnant women and women in late pregnancy. Br J Clin Pharmacol. 2006, 62 (5): 552-559. 10.1111/j.1365-2125.2006.02664.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Regazzi M, Villani P, Seminari E, Ravasi G, Cusato M, Marubbi F, Meneghetti G, Maserati R: Sex differences in nevirapine disposition in HIV-infected patients. AIDS. 2003, 17 (16): 2399-2400. 10.1097/00002030-200311070-00018.

    Article  PubMed  Google Scholar 

  28. Kappelhoff BS, van Leth F, MacGregor TR, Lange J, Beijenen JH, Huitema AD, 2NN study group: Nevirapine and efavirenz pharmacokinetics and covariate analysis in the 2NN study. Antivi Ther. 2005, 10 (1): 145-55.

    CAS  Google Scholar 

  29. Currier J, Averitt Bridge D, Hagins D, Zorrilla CD, Feinberg J, Ryan R, Falcon R, Tennenberg A, Mrus J, Squires K, GRACE (Gender, Race, And Clinical Experience) Study Group: Sex-based outcomes of darunavir-ritonavir therapy:a single-group trial. Ann Intern Med. 2010, 153 (6): 349-357. 10.7326/0003-4819-153-6-201009210-00002.

    Article  PubMed  PubMed Central  Google Scholar 

  30. European AIDS Clinical Society. EACS Guidelines Version 6.0. 2011, [http://www.europeanaidsclinicalsociety.org/images/stories/EACS-Pdf/EACSGuidelines-v6.0-English.pdf], October

  31. Chesney MA, Ickovics JR, Chambers DB, Gifford AL, Neidig J, Zwickl B, Wu AW: Self-reported adherence to antiretroviral medications among participants in HIV clinical trials: the AACTG adherence instruments. Patient Care Committee & Adherence Working Group of the Outcomes Committee of the Adult AIDS Clinical Trials Group (AACTG). AIDS Care. 2000, 12 (3): 255-266. 10.1080/09540120050042891.

    Article  CAS  PubMed  Google Scholar 

  32. Justice AC, Holmes W, Gifford AL, Rabeneck L, Zackin R, Sinclair G, Weissman S, Neidig J, Marcus C, Chesney M, Cohn SE, Wu AW, Adult AIDS Clinical Trials Unit Outcomes Committee: Development and validation of a self-completed HIV symptom index. J Clin Epidemiol. 2001, 54: S77-S90. 10.1016/S0895-4356(01)00449-8.

    Article  PubMed  Google Scholar 

  33. Reyataz (atazanavir sulfate) Package insert. 2010, Princeton, NJ, USA: Bristol-Myers Squibb Company

  34. Kaletra (lopinavir/ritonavir) Package insert. 2010, North Chicago, IL, USA: Abbott Laboratories

  35. Sustiva (efavirenz) Package insert. 2010, Princeton, NJ, USA: Bristol-Myers Squibb Company

  36. van Heeswijk RP, Veldkamp AI, Mulder JW, Meenhorst PL, Wit FW, Lange JM, Danner SA, Foudraine NA, Kwakkelstein MO, Reiss P, Beijnen JH, Hoetelmans RM: The steady-state pharmacokinetics of nevirapine during once daily and twice daily dosing in HIV-1-infected individuals. AIDS. 2000, 14 (8): F77-82. 10.1097/00002030-200005260-00001.

    Article  CAS  PubMed  Google Scholar 

  37. Lorello G, la Porte C, Pilon R, Zhang G, Karnauchow T, MacPherson P: Discordance in HIV-1 viral loads and antiretroviral drug concentrations comparing semen and blood plasma. HIV Med. 2009, 10 (9): 548-554. 10.1111/j.1468-1293.2009.00725.x.

    Article  CAS  PubMed  Google Scholar 

  38. Nettles RE, Kieffer TL, Parsons T, Johnson J, Cofrancesco J, Gallant JE, Carson KA, Siliciano RF, Flexner C: Marked intraindividual variability in antiretroviral concentrations may limit the utility of therapeutic drug monitoring. Clin Infect Dis. 2006, 42 (8): 1189-1196. 10.1086/501458.

    Article  CAS  PubMed  Google Scholar 

  39. Moltó J, Blanco A, Miranda C, Miranda J, Puig J, Valle M, Delavarga M, Fumaz CR, Barbanoj MJ, Clotet B: Variability in non-nucleoside reverse transcriptase and protease inhibitors concentrations among HIV-infected adults in routine clinical practice. Brit J Clin Pharmoco. 2007, 63 (6): 715-721. 10.1111/j.1365-2125.2006.02834.x.

    Article  Google Scholar 

  40. Burger D, Hugen P, Reiss P, Gyssens I, Schneider M, Kroon F, Schreij G, Brinkman K, Richter C, Prins J, Aarnoutse R, Lange J, ATHENA Cohort Study Group: Therapeutic drug monitoring of nelfinavir and indinavir in treatment-naïve-HIV-1-infected individuals. AIDS. 2003, 17 (8): 1157-1165. 10.1097/00002030-200305230-00007.

    Article  CAS  PubMed  Google Scholar 

  41. Fellay J, Marzolini C, Meaden ER, Back DJ, Buclin T, Chave JP, Decosterd LA, Furrer H, Opravil M, Pantaleo G, Retelska D, Ruiz L, Schinkel AH, Vernazza P, Eap CB, Telenti A, Swiss HIV Cohort Study: Response to antiretroviral treatment in HIV-1-infected individuals with allelic variants of the multidrug resistance transporter 1:a pharmacogenetics study. Lancet. 2002, 359 (9300): 30-36. 10.1016/S0140-6736(02)07276-8.

    Article  CAS  PubMed  Google Scholar 

  42. Wegner S, Vahey M, Dolan M, Wallace M, Aronson N, Barile A, Emmons W, Frazier S, Stephan K, Nau M, Piscitelli S, Harrigan R, Larder B: Racial Differences in Clinical Efficacy of Efavirenz-Based Antiretroviral Therapy [abstract]. 2002, Seattle, WA, USA: 9th Conference on Retroviruses and Opportunistic Infections, 24-28. Abstract 429-W

    Google Scholar 

  43. Baede-van Dijk PA, Hugen PW, Verweij-van Wissen CP, Koopmans PP, Burger DM, Hekster YA: Analysis of variation in plasma concentrations of nelfinavir and its active metabolite M8 in HIV-positive patients. AIDS. 2001, 15 (8): 991-998. 10.1097/00002030-200105250-00007.

    Article  CAS  PubMed  Google Scholar 

  44. Ameyaw MM, Regateiro F, Li T, Liu X, Tariq M, Mobarek A, Thornton N, Folayan GO, Githang’a J, Indalo A, Ofori-Adjei D, Price-Evans DA, McLeod HL: MDR1 pharmacogenetics:frequency of the C3435T mutation in exon 26 is significantly influenced by ethnicity. Pharmacogenetics. 2001, 11: 217-221. 10.1097/00008571-200104000-00005.

    Article  CAS  PubMed  Google Scholar 

  45. Schaeffeler E, Eichelbaum M, Brinkmann U, Penger A, Asante-Poku S, Zanger UM, Schwab M: Frequency of C3435T polymorphism of MDR1 gene in African people. Lancet. 2001, 358 (9279): 383-384. 10.1016/S0140-6736(01)05579-9.

    Article  CAS  PubMed  Google Scholar 

  46. Prezista (darunavir). Package insert (2010). Toronto, ON, Canada: Janssen Inc

  47. Fabbiani M, Di Giambenedetto S, Bracciale L, Bacarelli A, Ragazzoni E, Cauda R, Navarra P, De Luca A: Pharmacokinetic variability of antiretroviral drugs and correlation with virological outcome: 2 years of experience in routine clinical practice. J Antimicrob Chemother. 2009, 64 (1): 109-117. 10.1093/jac/dkp132.

    Article  CAS  PubMed  Google Scholar 

  48. Gibbons SE, Back DJ, Khoo SH: Is there evidence of age-dependent variation in lopinavir or efavirenz levels in female subjects in the clinical setting? [abstract]. 2005, Quebec City, QC, Canada: 6th International Workshop on Clinical Pharmacology of HIV Therapy, 28-30. Abstract 54

    Google Scholar 

Pre-publication history

Download references

Acknowledgements

We would like to thank the research coordinators, members of the Steering Committee and the participants of the study. This work was funded by the Canadian Institutes of Health Research (CIHR) (Grant # HBF – 90187) and the CIHR Canadian HIV Trials Network. Five investigators are also the recipients of salary support from the Canadian Institutes of Health Research (MRL), the Ontario HIV Treatment Network (JMR, SLW, JBA), Fonds de la Recherche en Santé du Québec (MBK), and the Skate the Dream Fund from the Toronto Western Hospital Foundation (JMR).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mona Rafik Loutfy.

Additional information

Competing interests

There are no financial or non-financial competing interests related to this paper and project.

Authors’ contributions

MRL was the senior most responsible investigator for the project who had study idea, wrote the protocol, sought funding and coordinated the project. MRL wrote the first and final drafts of the manuscript and is the corresponding author. CJlP and ALT provided pharmacokinetic expertise to the project and provided substantive edits to the manuscript. CJlP carried out the PK testing of all the samples. SLB conducted the statistical analyses under the direction of JMR, MRL and CJlP. SLB and JMR provided substantive edits to the manuscript. SB assisted with referencing. SW, MBK, NP, BC, JBA, ARR, KG, JC, DH, DB, FMS, AdP, HL, ST contributed to the study idea and protocol, acted as site investigators and enrolled participants for the study, and contributed to editing the manuscript. All authors reviewed the manuscript during preparation, provided critical feedback and approved the final manuscript.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Loutfy, M.R., Walmsley, S.L., Klein, M.B. et al. Factors affecting antiretroviral pharmacokinetics in HIV-infected women with virologic suppression on combination antiretroviral therapy: a cross-sectional study. BMC Infect Dis 13, 256 (2013). https://doi.org/10.1186/1471-2334-13-256

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2334-13-256

Keywords