Correspondence to Dr Christine Sekaggya-Wiltshire; [email protected]
STRENGTHS AND LIMITATIONS OF THIS STUDY
The study’s comprehensive scope includes multiple healthcare facilities in Uganda, with a long-term follow-up to robustly evaluate outcomes and adverse events.
This study will critically evaluate population pharmacokinetics (PK) of adverse events, and the genetic analysis will enhance understanding of individual responses to 3-month isoniazid and rifapentine in Uganda.
Continuous directly observed therapy is not done in the programmatic setting, potentially causing reporting bias.
Introduction
23% of the people who developed tuberculosis (TB) in 2022 were from the WHO Africa region.1 In 2019, Africa recorded 2.46 million new TB cases, a quarter of the global cases, with the highest incidence rate worldwide of 231 cases per 100 000.2 HIV-TB rates are five times higher in Africa than the worldwide average. The incidence rate of TB in Uganda is among the highest in the world at 200 cases per 100 000 and like the rest of Africa, the rates of HIV-TB are one of the highest (81 cases per 100 000).1
TB’s global burden extends beyond active cases, with a quarter of the global population harbouring latent TB infection (LTBI).3 Approximately 5%–10% of persons with LTBI progress to active TB disease during their lifetime4–6; however, TB preventive therapy (TPT) reduces TB incidence by 40%,7 8 which is crucial in order to achieve WHO’s goal of ending TB by 2030.9
WHO recommends TPT for all persons living with HIV (PLWH) and TB contacts with latent TB.4 Uganda recently rolled out TPT using the 3 months of weekly isoniazid and rifapentine (3HP) in 2022.10 3HP has better completion rates, as shown by a study in Pakistan where household contacts of all ages were more likely to complete TPT with shorter weekly regimens, in a programmatic setting in a high-burden country.11 3HP is also non-inferior to the 6 months of isoniazid.12–16 However, there are limited data on 3HP safety and drug pharmacokinetics in routine clinical settings from Africa, especially in the paediatric and adolescent population.14
A recent systematic review and meta-analysis that included 175 publications describing TPT-related adverse events in 277 cohorts found that 3HP had the highest incidence of any AE and AE-related treatment discontinuation, although it also had a low incidence of hepatotoxic AEs.17 Several studies have also demonstrated pharmacokinetic variability of isoniazid and that of rifamycins in people receiving treatment for active TB disease.18–20 In a recent study evaluating the pharmacokinetics of isoniazid in 30 people living with HIV, high isoniazid exposures were found, however, the adverse drug reactions (ADRs) were not linked to the 3HP. Polymorphisms in NAT-2 are associated with variability in isoniazid concentrations and may contribute to the occurrence of ADRs.21 In Taiwan, 22.8% of participants were slow metabolisers of isoniazid and NAT2 rs1041983 polymorphisms were associated with severe ADRs.22 The mechanism underlying some ADRs including isoniazid-induced hepatotoxicity has been referred to as a ‘metabolic idiosyncrasy’ because it is incompletely understood.23 24 The reasons for interindividual variability of isoniazid concentrations, especially in LTBI patients, are still poorly described, and how those variable exposures might contribute to ADRs is still unknown.18 23–26 Isoniazid and its metabolites may be directly cytotoxic, particularly in the presence of rifampin or similar drugs that induce cytochrome P450. Other molecular mechanisms might contribute to ADRs including immune-mediated processes influenced by HLA immunogenetic markers.27 The effect of recently described HLA genotypes and other drug metabolising enzymes, such as cytochrome 2E1 (CYP2E1) and uridine 5'-diphospho-glucuronosyltransferase, on relative concentrations of isoniazid and rifamycins hepatotoxicity risk in slow vs fast acetylators needs to be elucidated further.
The majority of the data available on 3HP is from clinical trials which we may not be able to entirely extrapolate to the ‘real-life’ settings. This study will review the rate of adverse events resulting from 3HP in Uganda compared with other settings where evaluation and management of adverse events may differ. The study will also generate and compare pharmacokinetic, pharmacogenomic and immunogenetic data from patients in a routine clinical setting experiencing ADR and matched controls without ADR to determine the mechanism underlying interindividual variability of ADR in LTBI patient populations. We, therefore, aimed to describe the safety profile of 3HP among people receiving TPT and the effect of ADRs on TPT completion rates. We also sought to describe the pharmacokinetic and pharmacogenomic determinants of the ADRs. We hypothesise that 3HP-related ADR will be minimal and those who do experience ADRs are more likely to discontinue treatment.
Methods and analysis
Study site
The study is being conducted at the Infectious Diseases Clinic (IDI) at Mulago National Referral Hospital, Kampala, Uganda, where over 8000 HIV-infected patients are managed annually. The study also takes place in Kampala City Council Authority health facilities.
Study population
Patients are enrolled if they meet the following inclusion criteria: (1) individuals greater or equal to 2 years who have been initiated on TPT using the isoniazid/rifapentine regimen according to standard of care, (2) both PLWHIV and HIV-uninfected individuals are eligible and (3) subjects who are willing and are able to comply with study procedures.
Patients are excluded from the study if they meet any one of the following criteria: (1) women who are pregnant at the time of screening and (2) individuals who have already received 3HP for more than 1 month.
Study design
This is a cohort study including 651 participants with a nested case–control study (150 cases and 150 controls). Over 18 months, we enrolled consecutive patients who were already initiated on TPT by the facility clinician as per standard of care. At enrolment, baseline clinical and demographic information are collected including age, sex, HIV status, type of Antiretriviral Therapy (ART) where applicable, weight/body mass index (BMI), concomitant medications and comorbidities, prior history of TB treatment and history of substance abuse. Blood samples for baseline alanine aminotransferase (ALT) and total bilirubin are also drawn as shown in table 1.
Table 1Study procedures
Screening | Baseline | Week 2 | Month 1 | Month 2 | Month 3 | Months 6–27 (3 monthly) | |
Patient history and physical examination | x | x | x | x | x | x | |
Clinical adverse event assessment | x | x | x | x | |||
Pregnancy test | x | x | x | x | |||
ALT/bilirubin | x | x | x | x | |||
Assessment for TB/TB reactivation | x | x | x | X | |||
Witnessed ingestion of 3HP | x | x | x | ||||
PK for rifapentine and isoniazid | x * | x * | x * | x * | |||
Genetic analysis (cases and controls) | x ** | x ** | x ** | x ** | |||
Adherence counselling | x | x | x | x | x |
ALT, alanine aminotransferase; 3HP, 3-month isoniazid and rifapentine; PK, pharmacokinetics; TB, tuberculosis.
The 3HP is provided through standard of care and administered to the patient as a weekly dose of rifapentine and isoniazid for up to 12 doses (3 months). Treatment is self-administered at home except on the study visit where treatment will be given as directly observed therapy and in order to conduct the pharmacokinetic analysis. The majority of the participants living with HIV were on dolutegravir-based regimens and participants who experience any antiretroviral drug interactions with 3HP are managed accordingly in addition to discontinuation of the 3HP. We assess adherence by self-report and review of the TPT card. A number of doses missed are recorded.
Nested case–control study
Cases are selected from patients within the cohort study who have developed adverse events that are related to the drug based on the Naranjo score. Controls are selected by incidence density sampling from those within the cohort study who do not develop any ADR related to drug. Cases and controls are matched by duration on 3HP, sex and age.
Follow-up
All enrolled participants are followed up to evaluate for ADRs after 2 weeks from enrolment and then monthly during the 3 months of 3HP treatment. TPT cards are given to every participant to record each dose that has been taken. During every follow-up visit, participants are also required to come with empty sachets of the drugs they have taken before they can be given drugs for the following month. Following completion of 3HP, 3 monthly visits are conducted for 2 years, where one visit will be conducted every 3 months for a total period of 2 years.
Primary outcomes
Our primary outcomes are ADRs and TPT completion.
Adverse drug reactions
ADRs are assessed using clinical history, physical examination and laboratory tests (ALT and total bilirubin). The ALT and bilirubin measurements are performed at the IDI Core laboratory, which is a College of American Pathologists certified laboratory.
Participants who present with symptoms suspected to be ADRs are further evaluated and additional laboratory tests may be performed. ADRs that may have been experienced between visits are also evaluated and recorded. ADRs are assessed for relationship to the drug using the Naranjo scale.28 Follow-up is conducted to determine the outcome of the ADR and whether subsequent doses of TPT are taken or not. Division of AIDS (DAIDS) V.2.229 is used to grade the severity of ADRs.
TPT completion rates
TPT completion is defined as taking all 12 doses of HP within 16 weeks of starting treatment. For those who undergo interruption of treatment for any reason, the total duration of their TPT is recorded. Interruption and restarting of TPT are in accordance with national treatment guidelines which state that If interruption is within 3 days stick to same day of the week and go back to one’s normal routine. If interruption >3 days; take the next dose on your usual day: This means one has skipped a week and will need to continue the medication for an additional week. 3HP should completed within 4 months or else TPT should be restarted all over again.10
Secondary outcome
Our secondary outcome is the efficacy of 3HP in terms of TB reactivation or a TB episode following TPT within 2 years of treatment completion
Evaluation for TB after TPT
All participants in the cohort are followed up for 2 years following 3HP initiation. Screening for signs and symptoms of TB is conducted at the monthly and 3-monthly visits using the intensified case finding form.
A gene expert will be done for all presumptive TB patients, and if MTB is not detected, further clinical evaluation including a chest X-ray will be done.
HIV positive adults in whom TB is not picked up by gene expert and are very sick (CD4 less than 100) will be tested for TB using Urine TB LAM test.
Pharmacokinetic analysis
Blood sampling for PK analysis is done for patients (cases and controls) within the nested case–control study to measure rifapentine and isoniazid concentrations. The first PK sampling occurs within 2 weeks of being selected as a case or control. The second PK sampling is performed before or at the month 3 visit. A meal is provided prior to drug intake. Blood samples are drawn predose, at 4 to 6 hours and 24 hours postdose. Time and date of the last dose are noted on these occasions.
Drug concentrations are measured using ultraperformance liquid chromatographic-tandem mass spectrometry or high-performance liquid chromatography.
Pharmacogenomic analysis
Blood sampling is performed for polymorphisms in AADAC and CYP2E1 (rs2070673). In addition, NAT2 acetylator status is determined using a three-single-nucleotide polymorphism panel of 191G>A (rs1801279), 341T>C (rs1041983) and 857G>A (rs1799931).
DNA extraction is performed using commercially available kits according to the manufacturer’s instructions. Genotyping is conducted using real-time PCR allelic discrimination using the TaqMan assay (Applied Biosystems, Warrington, U.K.) at the IDI Translational laboratory.
HLA genotyping for class 1 will be performed using PCR. Allele-specific PCR, using sequence-specific primers will be performed according to the protocol and recommendations of the manufacturer.
Patient and public involvement
In the exploration of adverse events associated with 3HP within Uganda’s routine clinical setting, the patient and public involvement section stands as a testament to our dedication to inclusivity and collaboration. At the outset, patients and the public were intimately involved in shaping the study’s direction, with their insights guiding the formulation of research questions. Drawing from their lived experiences, preferences and priorities, our study aims to resonate deeply within the community it serves. Throughout the design and execution phases, patients and the public have been indispensable partners. Their input has steered decisions concerning study design, the selection of outcome measures and the crafting of recruitment strategies. Furthermore, their ongoing engagement extends to critical decisions regarding the plans for disseminating study findings. By fostering this level of meaningful engagement, our study not only amplifies the voices of those affected by TB but also ensures that our findings are both accessible and actionable within Uganda’s healthcare landscape, contributing to more effective TB control strategies in resource-limited settings.
Sample size determination for cohort study
In this study, the primary endpoint for primary objective 2 is the proportion of patients who complete treatment. We aim to compare the completion rates among patients who experience ADR versus those who do not experience ADR. Using a difference in proportions, TPT completion rates of 87.3% among patients who do not experience ADR and assuming 70% completion among patients with ADR, type I error of 5% and 90% power, design effect of 2.0 to account for between cluster correlation, we will enrol a total of 614 study participants.
Subjects may withdraw from the study at any time at their own request, or they may be withdrawn at any time at the discretion of the investigator or sponsor for safety or behavioural reasons, or the inability of the participant to comply with the protocol required schedule of study visits or procedures. Participants who are withdrawn from the study can be replaced.
Sample size calculation for case–control study
In a study by Meng Rui et al, the mean drug concentration among patients who experienced ADR was 0.25 (SD 0.10). In our study, we assume that patients who do not experience ADR will have a 15% lower drug concentration. Using a difference in means as the primary endpoint, assuming a 5% type 1 error, 80% power, we will need to enrol 129 cases and 129 controls. The sample sizes for both groups have been adjusted for missing data and incomplete observations and a final sample size of 150 cases and 150 controls will be enrolled.
Analysis of outcomes
Description of safety profile
Safety profile of 3HP will be described using descriptive statistics including proportions. We will determine the proportion of patients who had any ADR and those with grade 3 and above events with their relatedness to the treatment.
Effect of ADRs on TPT completion
We will use χ2 tests to compare the proportion of patients with ADR who completed 3HP versus those who did not experience ADR. Additional comparisons of TPT completions will be made based on differences in demographic characteristics and risk categories including (HIV+, children <5 years and gender), behavioural risk factors like substance abuse, comorbid conditions and health facility. We will use univariable and generalised linear models to determine the factors associated with 3HP completion and these characteristics and calculate adjusted risk ratios. We will use robust SEs to account for correlation within health facilities. In a sensitivity analysis and given the availability and completeness of all participants’ visits, we will conduct a Kaplan-Meier survival analysis to estimate the time to drop out from 3HP treatment and compare across different variables, for example, sex, HIV status, BMI category and others using log-rank tests. All analyses will be performed by using STATA V.14.0.
Pharmacokinetic-pharmacodynamic modelling
Non-linear mixed effects modelling technique will be used develop a model to establish the population parameters for rifapentine and isoniazid and the variability around these parameters. Using this approach, we will then determine the PK parameters: area under the concentration-time curve and maximum concentrations of rifapentine and isoniazid and the factors that affect these PK parameters including age, sex and pharmacogenomic variants in NAT-2, AADAC and CYP2E1.
Ethics and dissemination
Ethical approval for this study was sought and approvals were received from the Infectious Diseases Institute Research and Ethics Committee, IREC Ref: 001/2021 and the Uganda National Council of Science and Technology (UNCST), UNCST Folio Number: HS1582ES.
Informed consent is obtained from all participants. Children between 8 and 17 years are requested for assent, in addition to consent from their parents or legally authorised representative. For children less than 8 years, consent is sought from their parents/legally authorised representative. The study is conducted in accordance with good clinical practice and Declaration of Helsinki.
Study status
The study began enrolment in February 2022 and completed recruitment in August 2023. All participants have completed TPT and are undergoing follow-up for TB reactivation. Follow-up will be completed in September 2025.
Ethics statements
Patient consent for publication
Not applicable.
Contributors This study was conceptualised by CS-W and SZ-M. The study design was conceived by CS-W, SZ-M, EAOL, JM, KED, JP, BC, TS and MPS. IM, RRN, LA, FA, BO, RO, JB and JN assisted with data collection while JP and GB assisted with data analysis. IM, RN and CS-W wrote the manuscript. All authors were involved in drafting the work or reviewing it critically for important intellectual content and all authors have agreed to this publication. All named authors adhere to the 302 authorship guidelines of BMJ Open. CS-W is the guarantor.
Funding The National Institute of Allergy And Infectious Diseases of the National Institutes of Health under Award Number R01AI160434 supported research reported in this publication. KED is supported by NIAID K24AI150349.
Disclaimer The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Competing interests None declared.
Patient and public involvement Patients and/or the public were involved in the design, or conduct, or reporting, or dissemination plans of this research. Refer to the Methods section for further details.
Provenance and peer review Not commissioned; externally peer reviewed.
1 Organization WH. Global tuberculosis report 2023. Geneva, 2023.
2 Organization WH. Global tuberculosis report 2013. World Health Organization; 2013.
3 Houben RMGJ, Dodd PJ. The Global Burden of Latent Tuberculosis Infection: A Re-estimation Using Mathematical Modelling. PLoS Med 2016; 13: e1002152. doi:10.1371/journal.pmed.1002152
4 Getahun H, Matteelli A, Abubakar I, et al. Management of latent Mycobacterium tuberculosis infection: WHO guidelines for low tuberculosis burden countries. Eur Respir J 2015; 46: 1563–76. doi:10.1183/13993003.01245-2015
5 Sutherland I. Recent studies in the epidemiology of tuberculosis, based on the risk of being infected with tubercle bacilli. Adv Tuberc Res 1976; 19: 1–63.
6 Whalen CC, Johnson JL, Okwera A, et al. A Trial of Three Regimens to Prevent Tuberculosis in Ugandan Adults Infected with the Human Immunodeficiency Virus. N Engl J Med 1997; 337: 801–8. doi:10.1056/NEJM199709183371201
7 Sterling TR, Njie G, Zenner D, et al. Guidelines for the treatment of latent tuberculosis infection: Recommendations from the National Tuberculosis Controllers Association and CDC, 2020. Am J Transplant 2020; 20: 1196–206. doi:10.1111/ajt.15841
8 Zenner D, Beer N, Harris RJ, et al. Treatment of Latent Tuberculosis Infection: An Updated Network Meta-analysis. Ann Intern Med 2017; 167: 248–55. doi:10.7326/M17-0609
9 Millington KA. Ending tuberculosis. 2018.
10 Government of uganda ministry of health. Guidelines for programmatic management of latent tb infection in uganda 'Closing the tb tap' a health worker guide. 2021.
11 Jaswal M, Farooq S, Madhani F, et al. Implementing 3HP vs. IPT as TB preventive treatment in Pakistan. Int Tuberc Lung Dis 2022; 26: 741–6. doi:10.5588/ijtld.21.0676
12 Belknap R, Holland D, Feng P-J, et al. Self-administered Versus Directly Observed Once-Weekly Isoniazid and Rifapentine Treatment of Latent Tuberculosis Infection: A Randomized Trial. Ann Intern Med 2017; 167: 689–97. doi:10.7326/M17-1150
13 Horsburgh CR, Goldberg S, Bethel J, et al. Latent TB Infection Treatment Acceptance and Completion in the United States and Canada. Chest 2010; 137: 401–9. doi:10.1378/chest.09-0394
14 Villarino ME, Scott NA, Weis SE, et al. Treatment for preventing tuberculosis in children and adolescents: a randomized clinical trial of a 3-month, 12-dose regimen of a combination of rifapentine and isoniazid. JAMA Pediatr 2015; 169: 247–55. doi:10.1001/jamapediatrics.2014.3158
15 Sandul AL, Nwana N, Holcombe JM, et al. High Rate of Treatment Completion in Program Settings With 12-Dose Weekly Isoniazid and Rifapentine for Latent Mycobacterium tuberculosis Infection. Clin Infect Dis 2017; 65: 1085–93. doi:10.1093/cid/cix505
16 Sun H-Y, Huang Y-W, Huang W-C, et al. Twelve-dose weekly rifapentine plus isoniazid for latent tuberculosis infection: A multicentre randomised controlled trial in Taiwan. Tuberculosis (Edinb) 2018; 111: 121–6. doi:10.1016/j.tube.2018.05.013
17 Melnychuk L, Perlman-Arrow S, Lisboa Bastos M, et al. A Systematic Review and Meta-Analysis of Tuberculous Preventative Therapy Adverse Events. Clin Infect Dis 2023; 77: 287–94. doi:10.1093/cid/ciad246
18 Sekaggya-Wiltshire C, von Braun A, Scherrer AU, et al. Anti-TB drug concentrations and drug-associated toxicities among TB/HIV-coinfected patients. J Antimicrob Chemother 2017; 72: 1172–7. doi:10.1093/jac/dkw534
19 Sekaggya-Wiltshire C, von Braun A, Lamorde M, et al. Delayed Sputum Culture Conversion in Tuberculosis–Human Immunodeficiency Virus–Coinfected Patients With Low Isoniazid and Rifampicin Concentrations. Clin Infect Dis 2018; 67: 708–16. doi:10.1093/cid/ciy179
20 Sekaggya-Wiltshire C, Chirehwa M, Musaazi J, et al. Low Antituberculosis Drug Concentrations in HIV-Tuberculosis-Coinfected Adults with Low Body Weight: Is It Time To Update Dosing Guidelines? Antimicrob Agents Chemother 2019; 63: 02174–18: e02174-18. doi:10.1128/AAC.02174-18
21 Jarrett RT, van der Heijden Y, Shotwell MS, et al. High Isoniazid Exposures When Administered with Rifapentine Once Weekly for Latent Tuberculosis in Individuals with Human Immunodeficiency Virus. Antimicrob Agents Chemother 2023; 67: e01297-22. doi:10.1128/aac.01297-22
22 Lee M-R, Huang H-L, Lin S-W, et al. Isoniazid Concentration and NAT2 Genotype Predict Risk of Systemic Drug Reactions during 3HP for LTBI. JCM 2019; 8: 812. doi:10.3390/jcm8060812
23 Metushi IG, Cai P, Zhu X, et al. A Fresh Look at the Mechanism of Isoniazid-Induced Hepatotoxicity. Clin Pharmacol Ther 2011; 89: 911–4. doi:10.1038/clpt.2010.355
24 Metushi I, Uetrecht J, Phillips E. Mechanism of isoniazid-induced hepatotoxicity: then and now. Br J Clin Pharmacol 2016; 81: 1030–6. doi:10.1111/bcp.12885
25 Sharma SK, Balamurugan A, Saha PK, et al. Evaluation of Clinical and Immunogenetic Risk Factors for the Development of Hepatotoxicity during Antituberculosis Treatment. Am J Respir Crit Care Med 2002; 166: 916–9. doi:10.1164/rccm.2108091
26 Rogers Z, Hiruy H, Pasipanodya JG, et al. The Non-Linear Child: Ontogeny, Isoniazid Concentration, and NAT2 Genotype Modulate Enzyme Reaction Kinetics and Metabolism. EBioMedicine 2016; 11: 118–26. doi:10.1016/j.ebiom.2016.07.031
27 Watkins PB. Idiosyncratic Liver Injury: Challenges and Approaches. Toxicol Pathol 2005; 33: 1–5. doi:10.1080/01926230590888306
28 Murayama H, Sakuma M, Takahashi Y, et al. Improving the assessment of adverse drug reactions using the Naranjo Algorithm in daily practice: The Japan Adverse Drug Events Study. Pharmacology Res & Perspec 2018; 6: e00373. doi:10.1002/prp2.373
29 The division of aids (daids) table for grading the severity of adult and pediatric adverse events. 2017.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024 Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ Group. http://creativecommons.org/licenses/by-nc/4.0/ This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/ . Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Introduction
Tuberculosis (TB) is the leading infectious cause of death globally. Despite WHO recommendations for TB preventive therapy (TPT), challenges persist, including incompletion of treatment and adverse drug reactions (ADRs). There is limited data on the 3-month isoniazid and rifapentine (3HP) pharmacokinetics, pharmacogenomics and their relation with ADRs. Our study aims to describe the pharmacokinetic and pharmacogenomics of 3HP used for TPT, the ADRs and their association with completion rates, and TPT outcomes, providing vital insights for TB control strategies in resource-limited settings.
Methods
This is an observational cohort study with a nested case–control study. We enrolled consecutive patients who had been initiated on TPT using the 3HP regimen. These are followed up biweekly and then monthly during the active phase of treatment and 3 monthly for 2 years following completion of TPT. ADR evaluation includes clinical assessment and liver function tests. Cases are selected from those who experience ADRs and controls from those who do not. Serum isoniazid and rifapentine concentrations are measured and pharmacogenomic analysis for NAT2, AADAC and CYP2E1 polymorphisms are done. Participants are followed up for 2 years to determine TPT outcomes.
Analysis
The safety profile of 3HP will be assessed using descriptive statistics, including proportions of patients experiencing ADRs and grade 3 or above events related to treatment. χ2 tests and regression models will determine predictors of ADRs and their impact on treatment completion. Pharmacokinetic-pharmacodynamic modelling will establish population parameters and factors influencing rifapentine and isoniazid concentrations.
Ethics and dissemination
Ethical approval of this study inclusive of all the appropriate documents was obtained from the Infectious Diseases Institute Research and Ethics Committee and the Uganda National Council of Science and Technology. The study adheres to legal, ethical and Good Clinical Practice (GCP) guidelines. Deidentified genotype data from 300 patients will be shared after publication. The protocol and phenotype data will be publicly accessible. Abstracts will be submitted to conferences, and a manuscript will be published poststudy.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details




1 Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Central, Uganda
2 National TB and Leprosy Division, Republic of Uganda Ministry of Health, Kampala, Uganda
3 Vanderbilt University Medical Center, Nashville, Tennessee, USA
4 National Tuberculosis and Leprosy Program, Republic of Uganda Ministry of Health, Kampala, Uganda