Skip to main content

Comparison of efavirenz levels in blood and hair with pharmacy refills as measures of adherence and predictors of viral suppression among people living with HIV in Nigeria

Abstract

Background

Strategies to support adherence are constrained by the lack of tools to objectively monitor medication intake in low-resource settings. Pharmacologic measures are objective, but pharmacy refill data is more accessible and cost-efficient. This study compared short-term and long-term efavirenz (EFV) drug levels with pharmacy refill adherence data (PRA) and evaluated their ability to predict viral suppression among people living with HIV in Nigeria.

Methods

Paired hair and dried blood spot (DBS) samples were obtained from 91 adults living with HIV receiving 600 mg EFV-based antiretroviral therapy (ART) and EFV concentrations were measured via validated methods using liquid-chromatography-mass-spectrometry. PRA was estimated from pharmacy records, based on the number of days a patient collected medication before or after the scheduled pick-up date. PRA was categorized into ≤ 74%, 75–94% and ≥ 95%, defined as poor, medium and high adherence, respectively. HIV viral loads closest to the hair sampling time (within 6 months) were also abstracted. Receiver Operating Characteristics (ROC) curve analyses compared the ability of adherence metrics to predict viral suppression.

Results

Based on PRA, 81% of participants had high adherence while 11% and 8% had medium and poor adherence, respectively. The median (IQR) EFV concentrations were 6.85 ng/mg (4.56–10.93) for hair and 1495.6 ng/ml (1050.7–2365.8) for DBS. Of the three measures of adherence, hair EFV concentration had the highest Area Under Curve (AUC) to predict viral suppression. Correlations between EFV concentrations in DBS and hair with PRA were positive (r = 0.12, P = 0.27 and r = 0.21, P = 0.05, respectively) but not strong.

Conclusions

EFV concentrations in hair were the strongest predictor of viral suppression and only weakly correlated with pharmacy refill adherence data in Nigeria. This study suggests that resource-limited settings may benefit from objective adherence metrics to monitor and support adherence.

Background

The efforts to end global HIV epidemics across every region of the world have yielded commendable results. Management of HIV disease involves several components including, early diagnosis, early initiation of antiretroviral therapy (ART), and ongoing adherence to medications. ART adherence is difficult to maintain, yet good adherence to ART is critical, particularly in resource-limited settings where viral load and resistance testing may not be routinely available to monitor ART outcomes. In addition, strategies to support adherence are constrained by the lack of tools to objectively monitor medication intake in low-resource settings. Although different ART regimens have different levels of adherence “forgiveness”, even recent studies have shown that an adherence rate ≥ 95% is associated with the most favorable treatment outcomes [1, 2]. Poor adherence to ART has been associated with lower rates of virologic suppression [3,4,5,6] and inflammation [7].

There is currently no gold standard method to accurately and independently measure medication adherence since each available method has inherent limitations. Pharmacologic measures are objective but pharmacy refill data is more accessible and cost-efficient. These methods also tend to be poorly correlated, including self-report, pill counts, electronic adherence monitors (e.g., medication event monitoring system), pharmacologic monitoring (e.g., drug levels in hair, plasma, dried blood spots (DBS), urine) and pharmacy refill adherence methods. Illustratively, self-reported adherence has shown poor agreement with pharmacologic metrics in many studies [6, 8,9,10], and, while medication event monitoring systems correlated well with pharmacologic methods [10], an inverse relationship between pharmacy refill data and DBS drug levels has been reported [11]. The advantages and disadvantages of each method have been well documented in previous reports [12,13,14,15,16].

Of the methods available to measure adherence, pharmacologic metrics are the most objective for monitoring actual drug ingestion, even though they are also imperfect. One major limitation of using antiretroviral drug levels in plasma (or levels in DBS for drugs not processed intracellularly, such as efavirenz) to measure adherence is that these metrics assess short-term exposure (within few days) and are thereby subject to “white coat compliance” [15], where patients increase their adherence to medication prior to clinic visits. Consequently, high adherence may be falsely recorded from short-term metrics. Measurements of drug concentrations in hair (or in DBS for intracellularly processed medications such as tenofovir) provides evidence of adherence for longer periods, up to weeks. Given that no single metric of adherence serves as a gold standard, a combination of methods has been suggested [2, 12, 16] in order to allow for a more robust assessment. For example, a combination of plasma (or DBS for most drugs) and hair drug levels can be used to determine short-term and long-term adherence, respectively. High drug concentrations in both matrices would indicate good adherence but discordant results with low levels in hair and high levels in plasma/DBS would provide evidence for white coat compliance.

In some resource-limited settings like Nigeria, pharmacy refill data or self-report are the most commonly-used methods to estimate patients’ adherence, although no previous study has confirmed their reliability in this population. To address this gap in the literature, our study compared pharmacy refill adherence data with drug levels in hair and dried blood spots among people living with HIV on efavirenz-based regimens in Nigeria.

Methods

Participants’ recruitment and sample collection

A cross-sectional study was conducted in the HIV clinic in a tertiary institution in South West Nigeria. Consenting patients receiving 600 mg efavirenz-based ART for at least 2 months were randomly selected in the waiting room of the HIV clinic at each clinic day until the required sample size was reached. A sample size of 91 was estimated with the single proportion formula considering that PRA adherence was 27% from an African study [17]; this sample size provides a 5% degree of precision and 5% type 1 error in a population less than 10,000 [18]. Hair and DBS samples were collected from the ninety-one (91) adult participants who gave informed consent.

Participants’ demographics and HIV viral loads closest to the hair sampling time (within 6 months) were abstracted from medical records. Pharmacy refill adherence (PRA) was estimated as a percentage from the pharmacy records, based on the number of days a patient collected medications before or after the scheduled medication pick-up date in line with the method used in estimating adherence at the Infectious Disease Institute, College of Medicine, University of Ibadan.

PRA was categorized into ≤ 74%, 75–94% and ≥ 95% [19], defined as poor, medium and high adherence, respectively. However, based on the receiver operator curve analysis, ≤ 75% or ≤ 85% or ≤ 95 or ≤ 100% of pills as estimated by the PRA were examined as threshold levels to predict viral suppression [20]. Ethical approval (NHREC/0/01/2008a) for this study was obtained from the joint UI/UCH ethical review board, University of Ibadan, Nigeria.

Hair and dried blood sample collections

The procedure for hair sample collection has been described elsewhere [21]. In brief, a small thatch of hair (20–30 strands) was cut with scissors as close as possible to the scalp and properly sealed as required. The hair samples were stored at room temperature until shipment to the Hair Analytical Laboratory (HAL) at the University of California San Francisco (UCSF), USA for quantification of efavirenz levels. For DBS, venous blood was collected from each participant and spotted on to Whatmann 903 protein saver cards (50 µl per spot). The procedure used has been previously reported [22].

Analyses of Efavirenz concentrations in hair and DBS

Efavirenz was quantified in hair using previously described and validated LC–MS/MS-based methods. Details of this procedure at the Hair Analytical Laboratory at UCSF has been described elsewhere [23] and the validated methods approved by Clinical Pharmacology and Quality Assurance Program of the National Institutes of Health [24]. Analysis of efavirenz concentrations in DBS was carried out at the Liverpool Bio-analytical Facility (BAF), Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool using validated liquid chromatography tandem mass spectrometry (LC–MS/MS)-based methods [25]. Details of the laboratory methods used in this study have been reported previously [22].

Statistical analysis

The association between hair or DBS (blood) efavirenz levels (which represents a short-term metric of exposure since efavirenz is not processed intracellularly) with PRA were assessed. The relationships between PRA and age, gender, and viral load were also assessed. Statistical analysis was carried out using IBM SPSS statistics version 25. Spearman rank correlation was used to assess the relationship between PRA, pharmacologic adherence metrics (efavirenz levels in hair and DBS), and the relevant covariates such as age, gender and viral load. Chi square statistics assessed differences in adherence by gender. Normality of continuous variables was tested using Shapiro–Wilk tests, Q-Q plots and histogram plots. Mean (standard deviation) or median (interquartile range) were used to summarize continuous variables while categorical variables were summarized by frequency and percentages in a descriptive analysis.

A receiver operator characteristic (ROC) curve of the association between specificity and sensitivity was generated to evaluate the predictive value of PRA, hair EFV concentrations and DBS EFV concentrations in forecasting viral suppression. The ROC curve was constructed by plotting the false positive rate (1-specificity) against the true positive rate (sensitivity). Detectable viral load was defined as greater than 50 copies/ml or greater than 1000 copies/ml [26, 27]. The adherence measure with the greatest total area was interpreted as having the best prediction of viral load suppression. Positive predictive value (PPV) and negative predictive value were also calculated. All analyses were performed at a significance level of 0.05.

Results

The respondents were 64 females (70.3%) and 27 males (29.7%), with a mean ± SD age of 44.5 ± 11.2 years and median (IQR) weight of 63 (64.0–73.0) Kg. The median (IQR) duration of EFV use was 45 (23.5–70.0) months. The concomitant antiretroviral agents were tenofovir disoproxil fumarate/lamivudine (TDF/3TC) (92.3%), abacavir/lamivudine (ABC/3TC) (5.5%) and TDF /emtricitabine (TDF/FTC) (2.2%). The PRA data revealed that 8%, 11% and 81% had poor, medium and high adherence, respectively. PRA was higher among females than males though not statistically significant (P = 0.472). Paired hair and dried blood spot (DBS) samples were analyzed successfully from all 91 participants.

The median (IQR) EFV concentration was 6.85 ng/mg (4.56–10.93) in hair while the median EFV concentration in blood was 1495.6 ng/ml (1050.7–2365.8). There was a strong correlation between EFV levels in hair and blood (r = 0.61, P < 0.0001) as shown in Fig. 1. The correlations between EFV concentrations in blood and hair with PRA were positive (r = 0.12, P = 0.27 and r = 0.21, P = 0.05, respectively) but weaker for blood levels. Age showed a trend with PRA, where younger participants tended to have better PRA compared to older participants (r = − 0.18, P = 0.08). Higher PRA and efavirenz DBS (blood) levels were not associated with lower HIV viral loads (r = − 0.12, P = 0.34; r = 0.13, P = 0.30, respectively). Higher hair concentrations also did not show significant association with HIV viral loads (r = − 0.14, P = 0.27). There was no significant correlation between DBS and hair EFV concentration with PRA (P > 0.05). However, the median DBS and hair EFV concentrations increased with increasing level of PRA (Table 1).

Fig. 1
figure 1

Correlation plot of hair EFV concentrations vs plasma EFV concentrations. Spearman r = 0.60 (CI 0.001425, 0.002286)

Table 1 Summary of PRA by DBS and hair concentration

The area under the Receiver Operating Characteristics Curve (AUROC) for predicting detectable viral loads using DBS and hair EFV concentrations ranged from 0.34 to 0.69 and were not significantly different across the two measures of detectable viral loads (P > 0.05) (Table 2). However, hair EFV concentrations had a higher AUROC to predict viral loads than DBS EFV concentrations at viral loads > 50 copies/mL (0.54 for hair vs 0.34 for DBS) and viral loads > 1000 copies/mL (0.69 for hair vs 0.47 for DBS) (Fig. 2 and Table 2).

Table 2 Area under the curve for the relationship between adherence (DBS EFV & Hair EFV concentration) and viral load (> 50 copies/ml and > 1000 copies/ml)
Fig. 2
figure 2

Receiver operating characteristic curve showing the relationship between adherence (DBS EFV & Hair EFV concentration) and viral load (> 50 copies/ml and > 1000 copies/ml). Blue line hair conc.; Green Line DBS conc.; Red diagonal reference

Sensitivity, specificity, PPV, NPV of using PRA adherence to predict detectable viral load are presented in Table 3. At viral loads > 50 copies/mL, all four measures of PRA had high sensitivity while at viral load > 1000 copies/mL, the four levels of PRA had lower sensitivity. There was no significant difference in sensitivity, specificity, PPV and NPV at the different levels of PRA in terms of predicting viral loads (P > 0.05) (Table 3).

Table 3 Pharmacy Refill Adherence (PRA) prediction of detectable viral load

Discussion

Accurate assessment of antiretroviral treatment adherence is necessary to tailor adherence support. Although pharmacy refill data is often used as an objective measure of adherence in Nigeria, our study showed a poor correlation between pharmacy refill data and pharmacologic methods of measuring adherence. Pharmacologic metrics, in which drug concentrations are measured in biological matrices such as hair, DBS, plasma, and urine, provide real evidence of medication ingestion. For drugs that are not processed intracellularly (like efavirenz), plasma, urine and DBS levels provide short-term metrics of exposure whereas hair levels provide long-term information on adherence. In our study, pharmacy refill data correlated poorly with short-and-long term objective metrics of efavirenz ingestion (Table 2). Previous reports have shown similar results where pharmacy refill data showed inverse correlations with tenofovir diphosphate (TFV-DP) drug levels in DBS [11].

Multiple studies have shown hair antiretroviral concentrations to be a strong predictor of virologic outcome [19, 28,29,30,31], with a systematic review and meta-analysis of these studies verifying this predictive utility [32]. Similarly, our study (the first to be performed in Nigeria) showed that hair concentrations of ART were predictive of virologic failure, verifying their utility as a tool to objectively monitor antiretroviral medication adherence. Traditionally, adherence >  = 95% was the preferred threshold for maintaining viral suppression in people living with HIV [33, 34], but our study and others [35] suggest that lower levels (> 80%) of adherence can also be associated with relatively high rates of viral suppression. In ROC analyses in this study, the AUC for hair concentrations in predicting virologic suppression (at either ≥ 50 copies/mL and ≥ 1000 copies/mL) was higher than either DBS levels or PRA.

In terms of correlation analyses, only weak positive correlations were found between pharmacy refill adherence and EFV concentrations in blood (r = 0.12, P = 0.27) and hair (r = 0.21, P = 0.05). Among study participants, 81% had high adherence (> 95%) estimated by PRA, but this high level of adherence was not verified by objective pharmacologic measures. This may mean that some participants consistently pick up their medication but may not consume them as prescribed. These results indicate that relying on pharmacy refill data as a sole method of assessing adherence in our population is not sufficient. The scale-up of pharmacologic methods to measure and support adherence in resource-limited settings could lead to better therapeutic outcomes.

Hair concentrations of antiretrovirals have several advantages when compared to other methods of assessing adherence. Self-reported adherence is usually associated with recall bias (patients may not be able to recall all missed doses) or social desirability bias, whereby patients respond to questions to please the researcher/clinicians even when they had poor adherence. In a study among Ugandan breast-feeding women on antiretroviral therapy, hair drug concentration was the strongest predictor of viral suppression while self-reported adherence did not show significant association with virologic outcomes [29]. Other methods of assessing adherence such as pill counts, medication events monitoring systems (MEMS), or pharmacy refill data, have similar shortfalls. For instance, not all MEMS cap openings amount to medication consumption [36]; moreover, network failures can affect the accuracy and reliability of MEMS as a sole method for adherence monitoring, especially in resource constrained settings. The accuracy of pill counts is also based on the assumption that all missing pills have been consumed. Finally, hair is easy to collect, has no biohazardous precautions, can be stored at room temperature, and hair measurements can be performed in resource-constrained settings [8], providing some advantages over blood-based methods. While socio-cultural factors such as fear of rituals may limit the willingness of some Nigerians to accept hair sampling [37], acceptance is generally high [38].

The major limitation of our study is that viral load measurements were not carried out at the same time as hair and DBS sample collection. Viral loads were determined from pharmacy records within a six-month period before sample collection. Actual viral loads at the time of sample collection may have varied from the viral load obtained from records for some participants. However, the majority of the participants (51 out of 63) already achieved sustained viral suppression, with viral loads less than 50 copies/mL. Therefore, variation in viral load may be very minimal within the specified period.

Conclusion

In this Nigeria-based study, pharmacy refill adherence data was not well correlated with objective adherence metrics (efavirenz levels in hair and blood), suggesting that pharmacy refill data may not accurately quantify ART ingestion in our population. Non-adherent patients may collect medicines but not take daily doses. Moreover, PRA and blood efavirenz levels were not significantly associated with HIV viral loads. The metric most associated with virologic suppression in receiver operating characteristic (ROC) analyses was hair ART levels. Resource-limited settings require easy-to-perform objective adherence metrics in order to monitor and support adherence.

Data availability

The datasets generated and/or analysed during the current study are not publicly available due to the fact that consent to make the data publicly available was not included in the informed consent process but the data are available from the corresponding author on reasonable request.

References

  1. Gachara G, Mavhandu LG, Rogawski ET, Manhaeve C, Bessong PO. Evaluating adherence to antiretroviral therapy using pharmacy refill records in a rural treatment site in South Africa. AIDS Res Treat. 2017;2017:5456219.

    PubMed  PubMed Central  Google Scholar 

  2. Mudhune V, Gvetadze R, Girde S, Ndivo R, Angira F, Zeh C, et al. Correlation of adherence by pill count, self-report, mems and plasma drug levels to treatment response among women receiving ARV therapy for PMTCT in Kenya. AIDS Behav. 2018;22(3):918–28.

    Article  Google Scholar 

  3. Oku AO, Owoaje ET, Ige OK, Oyo-Ita A. Prevalence and determinants of adherence to HAART amongst PLHIV in a tertiary health facility in South-South Nigeria. BMC Infect Dis. 2013;13:401.

    Article  Google Scholar 

  4. Adewuya AO, Afolabi MO, Ola BA, Ogundele OA, Ajibare AO, Oladipo BF, et al. The effect of psychological distress on medication adherence in persons with HIV infection in Nigeria. Psychosomatics. 2010;51(1):68–73.

    Article  Google Scholar 

  5. Marrazzo JM, Ramjee G, Richardson BA, Gomez K, Mgodi N, Nair G, et al. Tenofovir-based preexposure prophylaxis for HIV infection among African women. N Engl J Med. 2015;372(6):509–18.

    Article  Google Scholar 

  6. Dai JY, Hendrix CW, Richardson BA, Kelly C, Marzinke M, Chirenje ZM, et al. Pharmacological measures of treatment adherence and risk of HIV infection in the VOICE study. J Infect Dis. 2016;213(3):335–42.

    Article  Google Scholar 

  7. Musinguzi N, Castillo-Mancilla J, Morrow M, Byakwaga H, Mawhinney S, Burdo TH, et al. Antiretroviral therapy adherence interruptions are associated with systemic inflammation among Ugandans who achieved viral suppression. J Acquir Immune Defic Syndr. 2019;82(4):386–91.

    Article  CAS  Google Scholar 

  8. Gandhi M, Devi S, Bacchetti P, Chandy S, Heylen E, Phung N, et al. Measuring adherence to antiretroviral therapy via hair concentrations in India. J Acquir Immune Defic Syndr. 2019;81(2):202–6.

    Article  CAS  Google Scholar 

  9. Alcaide ML, Ramlagan S, Rodriguez VJ, Cook R, Peltzer K, Weiss SM, et al. Self-report and dry blood spot measurement of antiretroviral medications as markers of adherence in pregnant women in rural South Africa. AIDS Behav. 2017;21(7):2135–40.

    Article  Google Scholar 

  10. Baxi SM, Liu A, Bacchetti P, Mutua G, Sanders EJ, Kibengo FM, et al. Comparing the novel method of assessing PrEP adherence/exposure using hair samples to other pharmacologic and traditional measures. J Acquir Immune Defic Syndr. 2015;68(1):13–20.

    Article  CAS  Google Scholar 

  11. Castillo-Mancilla JR, Searls K, Caraway P, Zheng JH, Gardner EM, Predhomme J, et al. Short communication: Tenofovir diphosphate in dried blood spots as an objective measure of adherence in HIV-infected women. AIDS Res Hum Retrovir. 2015;31(4):428–32.

    Article  CAS  Google Scholar 

  12. Castillo-Mancilla JR, Haberer JE. Adherence measurements in HIV: new advancements in pharmacologic methods and real-time monitoring. Curr HIV/AIDS Rep. 2018;15(1):49–59.

    Article  Google Scholar 

  13. Kagee A, Nel A. Assessing the association between self-report items for HIV pill adherence and biological measures. AIDS Care. 2012;24(11):1448–52.

    Article  Google Scholar 

  14. Wendel CS, Mohler MJ, Kroesen K, Ampel NM, Gifford AL, Coons SJ. Barriers to use of electronic adherence monitoring in an HIV clinic. Ann Pharmacother. 2001;35(9):1010–5.

    Article  CAS  Google Scholar 

  15. Podsadecki TJ, Vrijens BC, Tousset EP, Rode RA, Hanna GJ. “White coat compliance” limits the reliability of therapeutic drug monitoring in HIV-1-infected patients. HIV Clin Trials. 2008;9(4):238–46.

    Article  Google Scholar 

  16. Spinelli MA, Haberer JE, Chai PR, Castillo-Mancilla J, Anderson PL, Gandhi M. Approaches to objectively measure antiretroviral medication adherence and drive adherence interventions. Curr HIV/AIDS Rep. 2020;17(4):301–14.

    Article  Google Scholar 

  17. Mekuria LA, Prins JM, Yalew AW, Sprangers MA, Nieuwkerk PT. Sub-optimal adherence to combination anti-retroviral therapy and its associated factors according to self-report, clinician-recorded and pharmacy-refill assessment methods among HIV-infected adults in Addis Ababa. AIDS Care. 2017;4:428–35.

    Article  Google Scholar 

  18. Israel G. Determining sample size. Univ Florida Facts Sheet PEOD-6. 1992;6.

  19. Gandhi M, Ameli N, Bacchetti P, Anastos K, Gange SJ, Minkoff H, et al. Atazanavir concentration in hair is the strongest predictor of outcomes on antiretroviral therapy. Clin Infect Dis. 2011;52(10):1267–75.

    Article  CAS  Google Scholar 

  20. Been SK, Yildiz E, Nieuwkerk PT, Pogány K, Van De Vijver DA, Verbon A. Self-reported adherence and pharmacy refill adherence are both predictive for an undetectable viral load among HIV-infected migrants receiving cART. PLoS ONE. 2017;12(11):e0186912.

    Article  Google Scholar 

  21. 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.

    Article  CAS  Google Scholar 

  22. Nwogu JN, Gandhi M, Owen A, Khoo S, Taiwo B, Olagunju A, et al. Associations between efavirenz concentrations, pharmacogenetics, and neurocognitive performance in people living with HIV in Nigeria. AIDS. 2021;35(12):1919–27.

    Article  CAS  Google Scholar 

  23. Huang Y, Gandhi M, Greenblatt RM, Gee W, Lin ET, Messenkoff N. Sensitive analysis of anti-HIV drugs, efavirenz, lopinavir and ritonavir, in human hair by liquid chromatography coupled with tandem mass spectrometry. Rapid Commun Mass Spectrom. 2008;22(21):3401–9.

    Article  CAS  Google Scholar 

  24. Browne RW, Rosenkranz SL, Wang Y, Taylor CR, DiFrancesco R, Morse GD. Sources of variability and accuracy of performance assessment in the clinical pharmacology quality assurance proficiency testing program for antiretrovirals. Ther Drug Monit. 2019;41(4):452–8.

    Article  CAS  Google Scholar 

  25. Amara AB, Else LJ, Tjia J, Olagunju A, Puls RL, Khoo S, et al. A validated method for quantification of efavirenz in dried blood spots using high-performance liquid chromatography-mass spectrometry. Ther Drug Monit. 2015;37(2):220–8.

    Article  CAS  Google Scholar 

  26. Da W, Li X, Qiao S, Zhou Y, Shen Z. Evaluation of self-report adherence measures and their associations with detectable viral load among people living with HIV (PLHIV) in China. PLoS ONE. 2018;13(8):e0203032.

    Article  Google Scholar 

  27. San Lio MM, Carbini R, Germano P, Guidotti G, Mancinelli S, Magid NA, et al. Evaluating adherence to highly active antiretroviral therapy with use of pill counts and viral load measurement in the drug resources enhancement against AIDS and malnutrition program in Mozambique. Clin Infect Dis. 2008;46(10):1609–16.

    Article  Google Scholar 

  28. Baxi SM, Greenblatt RM, Bacchetti P, Jin C, French AL, Keller MJ, et al. Nevirapine concentration in hair samples is a strong predictor of virologic suppression in a prospective cohort of HIV-infected patients. PLoS ONE. 2015;10(6):e0129100.

    Article  Google Scholar 

  29. Koss CA, Natureeba P, Mwesigwa J, Cohan D, Nzarubara B, Bacchetti P, et al. Hair concentrations of antiretrovirals predict viral suppression in HIV-infected pregnant and breastfeeding Ugandan women. AIDS. 2015;29(7):825–30.

    Article  CAS  Google Scholar 

  30. Gandhi M, Ameli N, Bacchetti P, Gange SJ, Anastos K, Levine A, et al. Protease inhibitor levels in hair strongly predict virologic response to treatment. AIDS. 2009;23(4):471–8.

    Article  CAS  Google Scholar 

  31. Yan J, Liu J, Su B, Pan X, Wang Z, Wu J, et al. Lamivudine concentration in hair and prediction of virologic failure and drug resistance among HIV patients receiving free ART in China. PLoS ONE. 2016;11(4):e0154421.

    Article  Google Scholar 

  32. Tamuzi JL, Bulabula AH, Milambo JP, Tshimwanga JL, Kazadi VT. Antiretroviral concentrations in hair as a tool for monitoring antiretroviral therapy adherence: systematic review and meta-analysis. Int J Innov Res Adv Stud. 2017;4(12):135–141.

    Google Scholar 

  33. Paterson DL, Swindells S, Mohr J, Brester M, Vergis EN, Squier C, et al. Adherence to protease inhibitor therapy and outcomes in patients with HIV infection. Ann Intern Med. 2000;133(1):21–30.

    Article  CAS  Google Scholar 

  34. Kobin AB, Sheth NU. Levels of adherence required for virologic suppression among newer antiretroviral medications. Ann Pharmacother. 2011;45(3):372–9.

    Article  CAS  Google Scholar 

  35. Leach OHE, Lu H, Caballero J, Thomas JE, Spencer EC, Cook RL. Defining the optimal cut-point of self-reported ART adherence to achieve viral suppression in the era of contemporary HIV therapy: a cross-sectional study. AIDS Res Ther. 2021;18(1):36.

    Article  Google Scholar 

  36. Deschamps AE, Van Wijngaerden E, Denhaerynck K, De Geest S, Vandamme AM. Use of electronic monitoring induces a 40-day intervention effect in HIV patients. J Acquir Immune Defic Syndr. 2006;43(2):247–8.

    Article  Google Scholar 

  37. Nwogu JN, Babalola CP, Ngene SO, Taiwo BO, Berzins B, Gandhi M. Willingness to donate hair samples for research among people living with HIV/AIDS attending a tertiary health facility in Ibadan. Niger AIDS Res Hum Retrovir. 2019;35(7):642–8.

    Article  Google Scholar 

  38. Herbertson EC, Lahiri CD, Nwogu JN, Soremekun RO, Olugbake OA, Ezechi OC, et al. High acceptability of donating hair and other biological samples for research among people living with HIV in an outpatient clinic in Lagos. Niger AIDS Res Hum Retrovir. 2021;37(9):676–82.

    Article  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the technical support of the staff of BAF, University of Liverpool, UK.

Funding

Research training for this publication was supported by the Fogarty International Center and National Institute of Mental Health (NIMH) of the National Institutes of Health (NIH) under award number D43TW009608, PI—Professor Babafemi Taiwo. This work is also partly supported by NIAID/NIH (2RO1AI098472 and R03 AI152773, PI Gandhi). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

JNN was involved in the conceptualization of the research, sample collection and analysis, data analysis and drafting of the manuscript. SON was involved in data analysis, interpretation and manuscript drafting. CPB was involved in the conceptualization of the research and drafting of the manuscript. AO was involved in sample analysis, data analysis and revising of the manuscript, AO; sample analysis and revising of manuscript, SHK; sample analysis and data interpretation, OAK; data interpretation and revising of the manuscript., BB; conceptualization of the research and revising of the manuscript, HO; sample analysis and revising of the manuscript, RT was involved in sample analysis and revising of the manuscript, MG; conceptualization of the research, drafting of the manuscript and revising of the manuscript, BT was involved in the conceptualization of the research and revising of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jacinta N. Nwogu or Chinedum P. Babalola.

Ethics declarations

Competing interests

The authors declare no competing interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nwogu, J.N., Ngene, S.O., Babalola, C.P. et al. Comparison of efavirenz levels in blood and hair with pharmacy refills as measures of adherence and predictors of viral suppression among people living with HIV in Nigeria. AIDS Res Ther 19, 35 (2022). https://doi.org/10.1186/s12981-022-00462-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12981-022-00462-3

Keywords