1
|
Farhan N, Dahal UP, Wahlstrom J. Development and Evaluation of Ontogeny Functions of the Major UDP-Glucuronosyltransferase Enzymes to Underwrite Physiologically Based Pharmacokinetic Modeling in Pediatric Populations. J Clin Pharmacol 2024; 64:1222-1235. [PMID: 38898531 DOI: 10.1002/jcph.2484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024]
Abstract
Uridine 5'-diphospho-glucuronosyltransferases (UGTs) demonstrate variable expression in the pediatric population. Thus, understanding of age-dependent maturation of UGTs is critical for accurate pediatric pharmacokinetics (PK) prediction of drugs that are susceptible for glucuronidation. Ontogeny functions of major UGTs have been previously developed and reported. However, those ontogeny functions are based on in vitro data (i.e., enzyme abundance, in vitro substrate activity, and so on) and therefore, may not translate to in vivo maturation of UGTs in the clinical setting. This report describes meta-analysis of the literature to develop and compare ontogeny functions for 8 primary UGTs (UGT1A1, UGT1A4, UGT1A6, UGT1A9, UGT2B7, UGT2B10, UGT2B15, and UGT2B17) based on published in vitro and in vivo studies. Once integrated with physiologically based pharmacokinetics modeling models, in vivo activity-based ontogeny functions demonstrated somewhat greater prediction accuracy (mean squared error, MSE: 0.05) compared to in vitro activity (MSE: 0.104) and in vitro abundance-based ontogeny functions (MSE: 0.129).
Collapse
Affiliation(s)
- Nashid Farhan
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | - Upendra P Dahal
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | - Jan Wahlstrom
- Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California, USA
| |
Collapse
|
2
|
Fokam J, Bouba Y, Ajeh RA, Guebiapsi DT, Essamba S, Zeh Meka AF, Lifanda E, Ada RA, Yakouba L, Mbengono NB, Djomo ARD, Tetang SN, Sosso SM, Babodo JC, Ambomo OFN, Temgoua EM, Medouane C, Atsinkou SN, Mvogo JL, Onana RM, Anoubissi JDD, Ketchaji A, Nka AD, Gouissi DHA, Ka'e AC, Fainguem NN, Kamgaing RS, Takou D, Tchouaket MCT, Semengue ENJ, Atsama MA, Nwobegahay J, Vuchas C, Nsimen AN, Bille BE, Gatchuessi SK, Ateba FN, Kesseng D, Billong SC, Armenia D, Santoro MM, Ceccherini-Silberstein F, Koki PN, Hamsatou HC, Colizzi V, Ndjolo A, Perno CF, Zoung-Kanyi Bissek AC. Evaluation of Viral Suppression in Paediatric Populations: Implications for the Transition to Dolutegravir-Based Regimens in Cameroon: The CIPHER-ADOLA Study. Biomedicines 2024; 12:2083. [PMID: 39335597 PMCID: PMC11440115 DOI: 10.3390/biomedicines12092083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Mortality in children accounts for 15% of all AIDS-related deaths globally, with a higher burden among Cameroonian children (25%), likely driven by poor virological response. We sought to evaluate viral suppression (VS) and its determinants in a nationally representative paediatric and young adult population receiving antiretroviral therapy (ART). A cross-sectional and multicentric study was conducted among Cameroonian children (<10 years), adolescents (10-19 years) and young adults (20-24 years). Data were collected from the databases of nine reference laboratories from December 2023 to March 2024. A conditional backward stepwise regression model was built to assess the predictors of VS, defined as a viral load (VL) <1000 HIV-RNA copies/mL. Overall, 7558 individuals (females: 73.2%) were analysed. Regarding the ART regimen, 17% of children, 80% of adolescents and 83% of young adults transitioned to dolutegravir (DTG)-based regimens. Overall VS was 82.3%, with 67.3% (<10 years), 80.5% (10-19 years) and 86.5% (20-24 years), and p < 0.001. VS was 85.1% on a DTG-based regimen versus 80.0% on efavirenz/nevirapine and 65.6% on lopinavir/ritonavir or atazanavir/ritonavir. VS was higher in females versus males (85.8% versus 78.2%, p < 0.001). The VS rate remained stable around 85% at 12 and 24 months but dropped to about 80% at 36 months after ART initiation, p < 0.009. Independent predictors of non-VS were younger age, longer ART duration (>36 months), backbone drug (non-TDF/3TC) and anchor drug (non-DTG based). In this Cameroonian paediatric population with varying levels of transition to DTG, overall VS remains below the 95% targets. Predictors of non-VS are younger age, non-TDF/3TC- and non-DTG-based regimens. Thus, efforts toward eliminating paediatric AIDS should prioritise the transition to a DTG-based regimen in this new ART era.
Collapse
Affiliation(s)
- Joseph Fokam
- Faculty of Health Sciences, University of Buea, Buea P.O. Box 63, Cameroon
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Yagai Bouba
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
- Faculty of Medicine, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Rogers Awoh Ajeh
- Faculty of Health Sciences, University of Buea, Buea P.O. Box 63, Cameroon
- HIV, Tuberculosis and Malaria Global Funds Subvention Coordination Unit (UCS), Ministry of Public Health, Yaoundé P.O. Box 2459, Cameroon
| | - Dominik Tameza Guebiapsi
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Suzane Essamba
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Albert Franck Zeh Meka
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Ebiama Lifanda
- Health Office, United States Agency for International Development (USAID), Yaoundé P.O. Box 817, Cameroon
| | - Rose Armelle Ada
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Liman Yakouba
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Nancy Barbara Mbengono
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Audrey Raissa Dzaddi Djomo
- Health Office, United States Agency for International Development (USAID), Yaoundé P.O. Box 817, Cameroon
| | - Suzie Ndiang Tetang
- Essos Hospital (CHE), National Social Welfare Centre, Yaoundé P.O. Box 5777, Cameroon
| | - Samuel Martin Sosso
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
| | - Jocelyne Carmen Babodo
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | | | - Edith Michele Temgoua
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Caroline Medouane
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Sabine Ndejo Atsinkou
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Justin Leonel Mvogo
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Roger Martin Onana
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Jean de Dieu Anoubissi
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Alice Ketchaji
- Division of Disease, Epidemic and Pandemic Control, Ministry of Public Health, Yaoundé P.O. Box 3038, Cameroon
| | - Alex Durand Nka
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
| | - Davy-Hyacinthe Anguechia Gouissi
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé P.O. Box 1364, Cameroon
| | - Aude Christelle Ka'e
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Nadine Nguendjoung Fainguem
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
| | - Rachel Simo Kamgaing
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
| | - Désiré Takou
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
| | - Michel Carlos Tommo Tchouaket
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
| | | | - Marie Amougou Atsama
- Research Center on Emerging and Re-Emerging Diseases (CREMER), Yaoundé P.O. Box 13033, Cameroon
| | - Julius Nwobegahay
- Centre for Research and Military Health (CRESAR), Yaoundé P.O. Box 15939, Cameroon
| | - Comfort Vuchas
- The Bamenda Center for Health Promotion and Research, Bamenda P.O. Box 586, Cameroon
| | - Anna Nya Nsimen
- The Bamenda Center for Health Promotion and Research, Bamenda P.O. Box 586, Cameroon
| | | | | | | | - Daniel Kesseng
- Mother-Child Centre, Chantal BIYA Foundation, Yaoundé P.O. Box 1936, Cameroon
| | - Serge Clotaire Billong
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé P.O. Box 1364, Cameroon
| | - Daniele Armenia
- Faculty of Medicine, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Maria Mercedes Santoro
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | | | - Paul Ndombo Koki
- Mother-Child Centre, Chantal BIYA Foundation, Yaoundé P.O. Box 1936, Cameroon
| | - Hadja Cherif Hamsatou
- Central Technical Group, National AIDS Control Committee (NACC), Yaoundé P.O. Box 2459, Cameroon
| | - Vittorio Colizzi
- Faculty of Science and Technology, University of Bandjoun, Bandjoun P.O. Box 127, Cameroon
| | - Alexis Ndjolo
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé P.O. Box 3077, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé P.O. Box 1364, Cameroon
| | - Carlo-Federico Perno
- Multimodal Medicine Laboratory, Bambino Gesù Children Hospital, IRCCS, 00165 Rome, Italy
| | - Anne-Cecile Zoung-Kanyi Bissek
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé P.O. Box 1364, Cameroon
- Division of Health Operational Research, Ministry of Public Health, Yaoundé P.O. Box 1937, Cameroon
| |
Collapse
|
3
|
Rungsapphaiboon A, Wacharachaisurapol N, Anugulruengkitt S, Sirikutt P, Phasomsap C, Tawan M, Saisaengjan C, Na Nakorn Y, Paiboon N, Songtaweesin WN, Tawon Y, Cressey TR, Puthanakit T. Pharmacokinetics of Generic Pediatric Dolutegravir Dispersible Tablet in Thai Young Children Living With HIV Weighing Below Twenty Kilograms. Pediatr Infect Dis J 2024; 43:789-794. [PMID: 39018516 PMCID: PMC11250107 DOI: 10.1097/inf.0000000000004366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Dolutegravir (DTG) dispersible tablet (DTG-DT) is a pediatric-friendly formulation. We aimed to describe the pharmacokinetics and virologic responses of generic DTG-DT in children weighing <20 kg. METHODS Children living with HIV-1 and <7 years of age weighing 6 to <20 kg were eligible. A generic 10-mg scored DTG-DT was administered to children using 3 weight bands (WB): WB1 (6 to <10 kg), WB2 (10 to <14 kg) and WB3 (14 to <20 kg), at doses of 20 mg (higher than World Health Organization recommendation of 15 mg), 20 mg and 25 mg, respectively. Steady-state intensive pharmacokinetics (PK) was performed in fasting condition with blood sampling at predose and 1, 2, 3, 4, 6 and 24 hours postdose. DTG PK parameters were estimated using a noncompartmental analysis, and DTG trough concentrations (C 24 ) and 24-hour area under the concentration-time curve were calculated. Comparisons were made with ODYSSEY and IMPAACT 2019. And 90% effective concentration of 0.32 mg/L was used as a reference individual DTG C 24 concentration. RESULTS From August 2021 to March 2023, 29 Thai children with a median (interquartile range) age of 3.2 (1.5-4.8) years were enrolled; 8 in WB1, 9 in WB2 and 12 in WB3. All children were treatment experienced and 59% had HIV RNA <200 copies/mL. Overall geometric mean (coefficient of variation percentage) DTG C 24 was 1.0 (46%) mg/L [WB1, 0.9 (53%); WB2, 0.9 (27%); WB3, 1.2 (51%)]. Geometric mean (coefficient of variation percentage) 24-hour area under the concentration-time curve was 83.2 (24%) mg h/L [WB1, 84.3 (31%); WB2, 76.9 (16%); WB3, 87.6 (25%)]. At weeks 24 and 48, 90% and 92% of participants had plasma HIV RNA <200 copies/mL. CONCLUSIONS Generic DTG-DT provided adequate drug exposure in children weighing 6 to <20 kg. The exploratory dose of DTG 20 mg for children weighing 6 to <10 kg showed similar PK parameters to World Health Organization doses in the other WB.
Collapse
Affiliation(s)
- Athiporn Rungsapphaiboon
- From the Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pediatrics, Khon Kaen Hospital, Khon Kaen, Thailand
| | - Noppadol Wacharachaisurapol
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University
- Department of Pharmacology, Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Faculty of Medicine, Chulalongkorn University
| | - Suvaporn Anugulruengkitt
- From the Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University
| | - Pugpen Sirikutt
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Chayapa Phasomsap
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University
| | - Monta Tawan
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University
| | - Chutima Saisaengjan
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University
| | - Yossawadee Na Nakorn
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Nantika Paiboon
- From the Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wipaporn Natalie Songtaweesin
- From the Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University
| | - Yardpiroon Tawon
- PK Laboratory Department, AMS-PHPT Research Collaboration, Faculty of Associated Medical Science, Chiang Mai University, Chiang Mai, Thailand
| | - Tim R. Cressey
- PK Laboratory Department, AMS-PHPT Research Collaboration, Faculty of Associated Medical Science, Chiang Mai University, Chiang Mai, Thailand
| | - Thanyawee Puthanakit
- From the Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University
| |
Collapse
|
4
|
Chandasana H, van Dijkman SC, Mehta R, Bush M, Rabie H, Flynn P, Cressey TR, Acosta EP, Brooks KM. Population Pharmacokinetic Modeling of Abacavir/Dolutegravir/Lamivudine to Support a Fixed-Dose Combination in Children with HIV-1. Infect Dis Ther 2024; 13:1877-1891. [PMID: 38961048 PMCID: PMC11266315 DOI: 10.1007/s40121-024-01008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024] Open
Abstract
INTRODUCTION Once-daily fixed-dose combinations (FDC) containing abacavir (ABC), dolutegravir (DTG), and lamivudine (3TC) have been approved in the US for adults and children with HIV weighing ≥ 6 kg. This analysis assessed the ability of previously developed ABC, DTG, and 3TC pediatric population pharmacokinetic (PopPK) models using multiple formulations to describe and predict PK data in young children using dispersible tablet (DT) and tablet formulations of ABC/DTG/3TC FDC in the IMPAACT 2019 study. METHODS IMPAACT 2019 was a Phase I/II study assessing the PK, safety, tolerability, and efficacy of ABC/DTG/3TC FDC in children with HIV-1. Intensive and sparse PK samples were collected over 48 weeks. Existing drug-specific pediatric PopPK models for ABC (2-compartment), DTG (1-compartment), and 3TC (1-compartment) were applied to the IMPAACT 2019 drug concentration data without re-estimation (external validation) of PopPK parameters. Drug exposures were then simulated across World Health Organization weight bands for children weighing ≥ 6 to < 40 kg for each drug and compared with pre-defined exposure target ranges. RESULTS Goodness-of-fit and visual predictive check plots demonstrated that the previously developed pediatric PopPK models sufficiently described and predicted the data. Thus, new PopPK models describing the IMPAACT 2019 data were unnecessary. Across weight bands, the predicted geometric mean (GM) for ABC AUC0-24 ranged from 14.89 to 18.50 μg*h/ml, DTG C24 ranged from 0.74 to 0.95 μg/ml, and 3TC AUC0-24 ranged from 10.50 to 13.20 μg*h/ml. These exposures were well within the pre-defined target ranges set for each drug. CONCLUSION This model-based approach leveraged existing pediatric data and models to confirm dosing of ABC/DTG/3TC FDC formulations in children with HIV-1. This analysis supports ABC/DTG/3TC FDC dosing in children weighing ≥ 6 kg.
Collapse
Affiliation(s)
- Hardik Chandasana
- Clinical Pharmacology Modeling and Simulation, GSK, 1250 South Collegeville Road, Collegeville, PA, 19426, USA.
| | | | | | | | - Helena Rabie
- University of Stellenbosch, Cape Town, South Africa
| | | | - Tim R Cressey
- Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | | | | |
Collapse
|
5
|
Meque I, Herrera N, Nhangave A, Mandlate D, Guilaze R, Tambo A, Mussa A, Bhatt N, Gill MM. The rollout of paediatric dolutegravir and virological outcomes among children living with HIV in Mozambique. South Afr J HIV Med 2024; 25:1578. [PMID: 39113779 PMCID: PMC11304402 DOI: 10.4102/sajhivmed.v25i1.1578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/26/2024] [Indexed: 08/10/2024] Open
Abstract
Background In 2022, Mozambique introduced Dolutegravir 10mg (pDTG), as part of paediatric antiretroviral therapy for children weighing < 20 kg. Understanding real-world challenges during national rollout can strengthen health systems in resource-limited settings. Objectives We described the transition rate to, and new initiation of, pDTG, viral load suppression (VLS) post-pDTG, and factors associated with VLS among children living with HIV. Method We conducted a retrospective cohort study involving children aged < 9 years and abstracted data from clinical sources. We used logistic regression to assess VLS and pDTG initiation predictors. Results Of 1353 children, 1146 initiated pDTG; 196 (14.5%) had no recorded weight. Post-pDTG switch, 98.9% (950/961) of children maintained the same nucleoside reverse transcriptase inhibitor backbone. After initiating Abacavir/Lamivudine+pDTG, 834 (72.8%) children remained on the regimen, 156 (13.6%) switched off (majority to Dolutegravir 50mg), 22 (1.9%) had ≥ 2 anchor drug switches; 134 (11.7%) had no documented follow-up regimen. Factors associated with pDTG initiation or switch were younger age (adjusted odds ratio [AOR] = 0.71 [0.63-0.80]) and a recorded weight (AOR = 55.58 [33.88-91.18]). VLS among the 294 children with a viral load (VL) test after ≥ 5 months post-pDTG was 75.5% (n = 222/294). Pre-pDTG VLS rate among treatment-experienced children was 56.5% (n = 130/230). Factors associated with VLS were older age (AOR = 1.18 [1.03-1.34]) and previous VLS (AOR = 2.27 [1.27-4.06]). Conclusion Most eligible children initiated pDTG per guidelines, improving post-pDTG VLS. Challenges included unexplained switches off pDTG after initiation, low VL coverage and inadequate documentation in clinic records.
Collapse
Affiliation(s)
- Ivete Meque
- Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Nicole Herrera
- Elizabeth Glaser Pediatric AIDS Foundation, Washington DC, United States of America
| | - Amâncio Nhangave
- Núcleo Provincial de Pesquisa de Gaza, Direcção Provincial de Saúde de Gaza, Xai-Xai, Mozambique
| | - Dórcia Mandlate
- Núcleo Provincial de Pesquisa de Inhambane, Direcção Provincial de Saúde de Inhambane, Inhambane, Mozambique
| | - Rui Guilaze
- Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Ana Tambo
- Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Abdul Mussa
- Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Nilesh Bhatt
- Elizabeth Glaser Pediatric AIDS Foundation, Washington DC, United States of America
| | - Michelle M. Gill
- Elizabeth Glaser Pediatric AIDS Foundation, Washington DC, United States of America
| |
Collapse
|
6
|
Bamford A, Hamzah L, Turkova A. Paediatric antiretroviral therapy challenges with emerging integrase resistance. Curr Opin HIV AIDS 2024; 19:01222929-990000000-00104. [PMID: 38967797 PMCID: PMC11451947 DOI: 10.1097/coh.0000000000000876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
PURPOSE OF REVIEW Universal antiretroviral (ART) coverage and virological suppression are fundamental to ending AIDS in children by 2030. Availability of new paediatric dolutegravir (DTG)-based ART formulations is a major breakthrough and will undoubtedly help achieve this goal, but treatment challenges still remain. RECENT FINDINGS Paediatric formulations remain limited compared to those for adults, especially for young children, those unable to tolerate DTG or with DTG-based first-line ART failure. Tenofovir alafenamide is virologically superior to standard-of-care backbone drugs in second-line, but paediatric formulations are not widely available. The roles of resistance testing and recycling of backbone drugs following first-line ART failure remain to be determined. Results of trials of novel treatment strategies including dual therapy and long-acting agents are awaited. Although numbers are currently small, safe and effective ART options are urgently required for children developing DTG resistance. SUMMARY The antiretroviral treatment gap between adults and children persists. The potential benefits from rollout of new paediatric DTG-based fixed-dose combination ART for first-line treatment are considerable. However, children remain disadvantaged when DTG-based first-line ART fails or cannot be used. Research efforts to address this inequity require prioritisation in order to ensure health outcomes are optimised for all ages in all settings.
Collapse
Affiliation(s)
- Alasdair Bamford
- Great Ormond Street Hospital for Children NHS Foundation Trust
- UCL Great Ormond Street Institute of Child Health
- MRC Clinical Trials Unit at UCL
| | - Lisa Hamzah
- St George's University Hospital NHS Trust, London, UK
| | - Anna Turkova
- Great Ormond Street Hospital for Children NHS Foundation Trust
- MRC Clinical Trials Unit at UCL
| |
Collapse
|
7
|
Desmonde S, Dame J, Malateste K, David A, Amorissani-Folquet M, N'Gbeche S, Sylla M, Takassi E, Kouakou K, Tossa LB, Yonaba C, Leroy V. Disparities in access to Dolutegravir in West African children, adolescents and young adults aged 0-24 years living with HIV. A IeDEA Pediatric West African cohort analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.24.24307900. [PMID: 38826257 PMCID: PMC11142258 DOI: 10.1101/2024.05.24.24307900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Introduction We describe the 24-month incidence of Dolutegravir (DTG)-containing antiretroviral treatment (ART) initiation since its introduction in 2019 in the pediatric West African IeDEA cohorts. Methods We included all patients aged 0-24 years on ART, from nine clinics in Côte d'Ivoire (n=4), Ghana, Nigeria, Mali, Benin, and Burkina Faso. Baseline varied by clinic and was defined as date of first DTG prescription; patients were followed-up until database closure/death/loss to follow-up (LTFU, no visit ≥ 7 months), whichever came first. We computed the cumulative incidence function for DTG initiation; associated factors were explored in a shared frailty model, accounting for clinic heterogeneity. Results Since 2019, 3,350 patients were included; 49% were female;79% had been on ART ≥ 12 months. Median baseline age was 12.9 years (IQR: 9-17). Median follow-up was 14 months (IQR: 7-22). The overall cumulative incidence of DTG initiation reached 35.5% (95% CI: 33.7-37.2) and 56.4% (95% CI: 54.4-58.4) at 12 and 24 months, respectively. In univariate analyses, those aged <5 years and females were overall less likely to switch. Adjusted on ART line and available viral load (VL) at baseline, females >10 years were less likely to initiate DTG compared to males of the same age (aHR among 10-14 years: 0.62, 95% CI: 0.54-0.72; among ≥15 years: 0.43, 95% CI: 0.36-0.50), as were those with detectable VL (> 50 copies/mL) compared to those in viral suppression (aHR: 0.86, 95% CI: 0.77-0.97) and those on PIs compared to those on NNRTIs (aHR after 12 months of roll-out: 0.75, 95% CI: 0.65-0.86). Conclusion: Access to paediatric DTG was incomplete and unequitable in West African settings: children <5years, females ≥ 10 years and those with detectable viral load were least likely to access DTG. Maintained monitoring and support of treatment practices is required to better ensure universal and equal access. Key messages What is already known on this topic?: Dolutegravir (DTG)-based ART regimens are recommended as the preferred first-line ART regimens recommended by the World Health Organisation in all people living with HIV since 2018, with a note of caution for pregnant women, then confirmed in all children with approved DTG dosing and adolescents since 2019.Deployment of universal DTG access in adults in West Africa has faced challenges such as infrastructure challenges, and healthcare system disparities, and was hindered by initial perinatal safety concerns affecting greatly women of childbearing age.Specific data on access to DTG in children, adolescents and young adults in West Africa is limited.What this study adds ?: This study describes the dynamic of the DTG roll-out over the first 24 months and its correlates since 2019 in a large West African multicentric cohort of children, adolescents and youth.We observed a rapid scale-up of DTG among children, adolescents and young adults living with HIV in West Africa, despite the COVID-19 pandemic.However, DTG access after 24 months was incomplete and unequitable, with adolescent girls and young women being less likely to initiate DTG compared to males, as were those with a detectable viral load (> 50 copies/mL) compared to those in success.Younger children < 5 years were also less likely to initiate DTG, explained by the later approval of paediatric formulations and their low availability.How this study might affect research, practice or policy?: Maintained monitoring, training and updating guidance for healthcare workers is essential to ensure universal access to DTG, especially for females, for whom inequity begins age 10 years.Efforts to improve access to universal DTG in West Africa require multifaceted interventions including healthcare infrastructure improvement and facilitation of paediatric antiretroviral forecasting and planification.
Collapse
|
8
|
Jacobs TG, Mumbiro V, Cassia U, Zimba K, Nalwanga D, Ballesteros A, Domínguez-Rodríguez S, Tagarro A, Madrid L, Mutata C, Chitsamatanga M, Bwakura-Dangarembizi M, Passanduca A, Buck WC, Nduna B, Chabala C, Najjingo E, Musiime V, Moraleda C, Colbers A, Mujuru HA, Rojo P, Burger DM. Twice-Daily Dosing of Dolutegravir in Infants on Rifampicin Treatment: A Pharmacokinetic Substudy of the EMPIRICAL Trial. Clin Infect Dis 2024; 78:702-710. [PMID: 37882611 PMCID: PMC10954323 DOI: 10.1093/cid/ciad656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/29/2023] [Accepted: 10/25/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND We evaluated dolutegravir pharmacokinetics in infants with human immunodeficiency virus (HIV) receiving dolutegravir twice daily (BID) with rifampicin-based tuberculosis (TB) treatment compared with once daily (OD) without rifampicin. METHODS Infants with HIV aged 1-12 months, weighing ≥3 kg, and receiving dolutegravir BID with rifampicin or OD without rifampicin were eligible. Six blood samples were taken over 12 (BID) or 24 hours (OD). Dolutegravir pharmacokinetic parameters, HIV viral load (VL) data, and adverse events (AEs) were reported. RESULTS Twenty-seven of 30 enrolled infants had evaluable pharmacokinetic curves. The median (interquartile range) age was 7.1 months (6.1-9.9), weight was 6.3 kg (5.6-7.2), 21 (78%) received rifampicin, and 11 (41%) were female. Geometric mean ratios comparing dolutegravir BID with rifampicin versus OD without rifampicin were area under curve (AUC)0-24h 0.91 (95% confidence interval, .59-1.42), Ctrough 0.95 (0.57-1.59), Cmax 0.87 (0.57-1.33). One infant (5%) receiving rifampicin versus none without rifampicin had dolutegravir Ctrough <0.32 mg/L, and none had Ctrough <0.064 mg/L. The dolutegravir metabolic ratio (dolutegravir-glucuronide AUC/dolutegravir AUC) was 2.3-fold higher in combination with rifampicin versus without rifampicin. Five of 82 reported AEs were possibly related to rifampicin or dolutegravir and resolved without treatment discontinuation. Upon TB treatment completion, HIV viral load was <1000 copies/mL in 76% and 100% of infants and undetectable in 35% and 20% of infants with and without rifampicin, respectively. CONCLUSIONS Dolutegravir BID in infants receiving rifampicin resulted in adequate dolutegravir exposure, supporting this treatment approach for infants with HIV-TB coinfection.
Collapse
Affiliation(s)
- Tom G Jacobs
- Department of Pharmacy, Radboudumc Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vivian Mumbiro
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | - Uneisse Cassia
- Universidade Eduardo Mondlane Faculdade de Medicina, Maputo, Mozambique
| | - Kevin Zimba
- University Teaching Hospitals-Children’s Hospital, Lusaka, Zambia
| | - Damalie Nalwanga
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Alvaro Ballesteros
- Pediatric Unit for Research and Clinical Trials, Hospital 12 de Octubre Health Research Institute, Biomedical Foundation of Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Sara Domínguez-Rodríguez
- Pediatric Unit for Research and Clinical Trials, Hospital 12 de Octubre Health Research Institute, Biomedical Foundation of Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alfredo Tagarro
- Pediatric Unit for Research and Clinical Trials, Hospital 12 de Octubre Health Research Institute, Biomedical Foundation of Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Service, Infanta Sofia University Hospital, Servicio Madrileño de Salud, Madrid, Spain
- Universidad Europea de Madrid, Madrid, Spain
| | - Lola Madrid
- Pediatric Unit for Research and Clinical Trials, Hospital 12 de Octubre Health Research Institute, Biomedical Foundation of Hospital Universitario 12 de Octubre, Madrid, Spain
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | | | | | - Alfeu Passanduca
- Universidade Eduardo Mondlane Faculdade de Medicina, Maputo, Mozambique
| | - W Chris Buck
- Universidade Eduardo Mondlane Faculdade de Medicina, Maputo, Mozambique
- David Geffen School of Medicine, University of California–Los Angeles, Los Angeles, California, USA
| | - Bwendo Nduna
- Arthur Davidson Children’s Hospital, Ndola, Zambia
| | - Chishala Chabala
- University Teaching Hospitals-Children’s Hospital, Lusaka, Zambia
- School of Medicine, University of Zambia, Lusaka, Zambia
- HerpeZ, Lusaka, Zambia
| | | | - Victor Musiime
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
- Joint Clinical Research Centre, Kampala, Uganda
| | - Cinta Moraleda
- Pediatric Unit for Research and Clinical Trials, Hospital 12 de Octubre Health Research Institute, Biomedical Foundation of Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Service, Hospital Universitario 12 de Octubre, Servicio Madrileño de Salud, Madrid, Spain
| | - Angela Colbers
- Department of Pharmacy, Radboudumc Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hilda A Mujuru
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | - Pablo Rojo
- Pediatric Unit for Research and Clinical Trials, Hospital 12 de Octubre Health Research Institute, Biomedical Foundation of Hospital Universitario 12 de Octubre, Madrid, Spain
- Pediatric Service, Hospital Universitario 12 de Octubre, Servicio Madrileño de Salud, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | - David M Burger
- Department of Pharmacy, Radboudumc Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Makumbi S, Bajunirwe F, Ford D, Turkova A, South A, Lugemwa A, Musiime V, Gibb D, Tamwesigire IK. Voluntariness of consent in paediatric HIV clinical trials: a mixed-methods, cross-sectional study of participants in the CHAPAS-4 and ODYSSEY trials in Uganda. BMJ Open 2024; 14:e077546. [PMID: 38431301 PMCID: PMC10910635 DOI: 10.1136/bmjopen-2023-077546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVES To examine the voluntariness of consent in paediatric HIV clinical trials and the associated factors. DESIGN Mixed-methods, cross-sectional study combining a quantitative survey conducted concurrently with indepth interviews. SETTING AND PARTICIPANTS From January 2021 to April 2021, we interviewed parents of children on first-line or second-line Anti-retroviral therapy (ART) in two ongoing paediatric HIV clinical trials [CHAPAS-4 (ISRCTN22964075) and ODYSSEY (ISRCTN91737921)] at the Joint Clinical Research Centre Mbarara, Uganda. OUTCOME MEASURES The outcome measures were the proportion of parents with voluntary consent, factors affecting voluntariness and the sources of external influence. Parents rated the voluntariness of their consent on a voluntariness ladder. Indepth interviews described participants' lived experiences and were aimed at adding context. RESULTS All 151 parents randomly sampled for the survey participated (84% female, median age 40 years). Most (67%) gave a fully voluntary decision, with a score of 10 on the voluntariness ladder, whereas 8% scored 9, 9% scored 8, 6% scored 7, 8% scored 6 and 2.7% scored 4. Trust in medical researchers (adjusted OR 9.90, 95% CI 1.01 to 97.20, p=0.049) and male sex of the parent (adjusted OR 3.66, 95% CI 1.00 to 13.38, p=0.05) were positively associated with voluntariness of consent. Prior research experience (adjusted OR 0.31, 95% CI 0.12 to 0.78, p=0.014) and consulting (adjusted OR 0.25. 95% CI 0.10 to 0.60, p=0.002) were negatively associated with voluntariness. Consultation and advice came from referring health workers (36%), spouses (29%), other family members (27%), friends (15%) and researchers (7%). The indepth interviews (n=14) identified the health condition of the child, advice from referring health workers and the opportunity to access better care as factors affecting the voluntariness of consent. CONCLUSIONS This study demonstrated a high voluntariness of consent, which was enhanced among male parents and by parents' trust in medical researchers. Prior research experience of the child and advice from health workers and spouses were negatively associated with the voluntariness of parents' consent. Female parents and parents of children with prior research experience may benefit from additional interventions to support voluntary participation.
Collapse
Affiliation(s)
- Shafic Makumbi
- Joint Clinical Research Centre, Mbarara, Uganda
- Mbarara University of Science and Technology, Mbarara, Uganda
| | - Francis Bajunirwe
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Deborah Ford
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Anna Turkova
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Annabelle South
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | | | - Victor Musiime
- Joint Clinical Research Centre, Mbarara, Uganda
- Makerere University, Kampala, Uganda
| | - Diana Gibb
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Imelda K Tamwesigire
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| |
Collapse
|
10
|
Bevers LAH, Waalewijn H, Szubert AJ, Chabala C, Bwakura-Dangarembizi M, Makumbi S, Nangiya J, Mumbiro V, Mulenga V, Musiime V, Burger DM, Gibb DM, Colbers A. Pharmacokinetic Data of Dolutegravir in Second-line Treatment of Children With Human Immunodeficiency Virus: Results From the CHAPAS4 Trial. Clin Infect Dis 2023; 77:1312-1317. [PMID: 37280040 PMCID: PMC10640690 DOI: 10.1093/cid/ciad346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Dolutegravir (DTG), combined with a backbone of 2 nucleoside reverse transcriptase inhibitors, is currently the preferred first-line treatment for human immunodeficiency virus (HIV) in childhood. CHAPAS4 is an ongoing randomized controlled trial investigating second-line treatment options for children with HIV. We did a nested pharmacokinetic (PK) substudy within CHAPAS4 to evaluate the DTG exposure in children with HIV taking DTG with food as part of their second-line treatment. METHODS Additional consent was required for children on DTG enrolled in the CHAPAS4 trial to participate in this PK substudy. Children weighing 14-19.9 kg took 25 mg DTG as dispersible tablets and children ≥20 kg took 50 mg film-coated tablets. Steady-state 24-hour DTG plasma concentration-time PK profiling was done at t = 0 and 1, 2, 4, 6, 8, 12, and 24 hours after observed DTG intake with food. Reference adult PK data and pediatric data from the ODYSSEY trial were used primarily for comparison. The individual target trough concentration (Ctrough) was defined as 0.32 mg/L. RESULTS Thirty-nine children on DTG were included in this PK substudy. The geometric mean (GM) area under the concentration-time curve over the dosing interval (AUC0-24h) was 57.1 hours × mg/L (coefficient of variation [CV%], 38.4%), which was approximately 8% below the average AUC0-24h in children in the ODYSSEY trial with comparable dosages, but above the adult reference. The GM (CV%) Ctrough was 0.82 mg/L (63.8%), which was comparable to ODYSSEY and adult reference values. CONCLUSIONS This nested PK substudy shows that the exposure of DTG taken with food in children on second-line treatment is comparable with that of children in the ODYSSEY trial and adult references. Clinical Trials Registration.ISRCTN22964075.
Collapse
Affiliation(s)
- Lisanne A H Bevers
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hylke Waalewijn
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, South Africa
| | - Alexander J Szubert
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Chishala Chabala
- Children’s Hospital, University Teaching Hospital, Lusaka, Zambia
| | | | - Shafic Makumbi
- Joint Clinical Research Centre, Mbarara Regional Centre of Excellence, Mbarara, Uganda
| | - Joan Nangiya
- Joint Clinical Research Centre, Research Department, Kampala, Uganda
| | | | - Veronica Mulenga
- Children’s Hospital, University Teaching Hospital, Lusaka, Zambia
| | - Victor Musiime
- Joint Clinical Research Centre, Research Department, Kampala, Uganda
| | - David M Burger
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Diana M Gibb
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Angela Colbers
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Chandasana H, Thapar M, Hayes S, Baker M, Gibb DM, Turkova A, Ford D, Ruel T, Wiznia A, Fairlie L, Bwakura-Dangarembizi M, Mujuru H, Alvero C, Farhad M, Hazra R, Townley E, Buchanan A, Bollen P, Waalewijn H, Colbers A, Burger D, Acosta EP, Singh R. Population Pharmacokinetic Modeling of Dolutegravir to Optimize Pediatric Dosing in HIV-1-Infected Infants, Children, and Adolescents. Clin Pharmacokinet 2023; 62:1445-1459. [PMID: 37603217 PMCID: PMC10520196 DOI: 10.1007/s40262-023-01289-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND AND OBJECTIVE HIV treatment options remain limited in children. Dolutegravir is a potent and well-tolerated, once-daily HIV-1 integrase inhibitor recommended for HIV-1 infection in both adults and children down to 4 weeks of age. To support pediatric dosing of dolutegravir in children, we used a population pharmacokinetic model with dolutegravir data from the P1093 and ODYSSEY clinical trials. The relationship between dolutegravir exposure and selected safety endpoints was also evaluated. METHODS A population pharmacokinetic model was developed with data from P1093 and ODYSSEY to characterize the pharmacokinetics and associated variability and to evaluate the impact of pharmacokinetic covariates. The final population pharmacokinetic model simulated exposures across weight bands, doses, and formulations that were compared with established adult reference data. Exploratory exposure-safety analyses evaluated the relationship between dolutegravir pharmacokinetic parameters and selected clinical laboratory parameters and adverse events. RESULTS A total of N = 239 participants were included, baseline age ranged from 0.1 to 17.5 years, weight ranged from 3.9 to 91 kg, 50% were male, and 80% were black. The final population pharmacokinetic model was a one-compartment model with first-order absorption and elimination, enabling predictions of dolutegravir concentrations in the pediatric population across weight bands and doses/formulations. The predicted geometric mean trough concentration was comparable to the adult value following a 50-mg daily dose of dolutegravir for all weight bands at recommended doses. Body weight, age, and formulation were significant predictors of dolutegravir pharmacokinetics in pediatrics. Additionally, during an exploratory exposure-safety analysis, no correlation was found between dolutegravir exposure and selected safety endpoints or adverse events. CONCLUSIONS The dolutegravir dosing in children ≥ 4 weeks of age on an age/weight-band basis provides comparable exposures to those historically observed in adults. Observed pharmacokinetic variability was higher in this pediatric population and no additional safety concerns were observed. These results support the weight-banded dosing of dolutegravir in pediatric participants currently recommended by the World Health Organization.
Collapse
Affiliation(s)
- Hardik Chandasana
- Clinical Pharmacology, Modeling and Simulation, GSK, 1250 South Collegeville Road, Collegeville, PA, 19406, USA.
| | | | | | | | - Diana M Gibb
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Anna Turkova
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Deborah Ford
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Theodore Ruel
- University of California, San Francisco, San Francisco, CA, USA
| | - Andrew Wiznia
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lee Fairlie
- Faculty of Health Sciences, Wits RHI, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Hilda Mujuru
- Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | | | - Mona Farhad
- Frontier Science Foundation, Brookline, MA, USA
| | - Rohan Hazra
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Ellen Townley
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Pauline Bollen
- Department of Pharmacy, Radboudumc Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hylke Waalewijn
- Department of Pharmacy, Radboudumc Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Angela Colbers
- Department of Pharmacy, Radboudumc Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David Burger
- Department of Pharmacy, Radboudumc Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Rajendra Singh
- Clinical Pharmacology, Modeling and Simulation, GSK, 1250 South Collegeville Road, Collegeville, PA, 19406, USA
| |
Collapse
|
12
|
Turkova A, White E, Kekitiinwa AR, Mumbiro V, Kaudha E, Liberty A, Ahimbisibwe GM, Moloantoa T, Srirompotong U, Mosia NR, Puthanakit T, Kobbe R, Fortuny C, Kataike H, Bbuye D, Na-Rajsima S, Coelho A, Lugemwa A, Bwakura-Dangarembizi MF, Klein N, Mujuru HA, Kityo C, Cotton MF, Ferrand RA, Giaquinto C, Rojo P, Violari A, Gibb DM, Ford D. Neuropsychiatric manifestations and sleep disturbances with dolutegravir-based antiretroviral therapy versus standard of care in children and adolescents: a secondary analysis of the ODYSSEY trial. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:718-727. [PMID: 37562418 PMCID: PMC7616346 DOI: 10.1016/s2352-4642(23)00164-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/03/2023] [Accepted: 06/12/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Cohort studies in adults with HIV showed that dolutegravir was associated with neuropsychiatric adverse events and sleep problems, yet data are scarce in children and adolescents. We aimed to evaluate neuropsychiatric manifestations in children and adolescents treated with dolutegravir-based treatment versus alternative antiretroviral therapy. METHODS This is a secondary analysis of ODYSSEY, an open-label, multicentre, randomised, non-inferiority trial, in which adolescents and children initiating first-line or second-line antiretroviral therapy were randomly assigned 1:1 to dolutegravir-based treatment or standard-of-care treatment. We assessed neuropsychiatric adverse events (reported by clinicians) and responses to the mood and sleep questionnaires (reported by the participant or their carer) in both groups. We compared the proportions of patients with neuropsychiatric adverse events (neurological, psychiatric, and total), time to first neuropsychiatric adverse event, and participant-reported responses to questionnaires capturing issues with mood, suicidal thoughts, and sleep problems. FINDINGS Between Sept 20, 2016, and June 22, 2018, 707 participants were enrolled, of whom 345 (49%) were female and 362 (51%) were male, and 623 (88%) were Black-African. Of 707 participants, 350 (50%) were randomly assigned to dolutegravir-based antiretroviral therapy and 357 (50%) to non-dolutegravir-based standard-of-care. 311 (44%) of 707 participants started first-line antiretroviral therapy (ODYSSEY-A; 145 [92%] of 157 participants had efavirenz-based therapy in the standard-of-care group), and 396 (56%) of 707 started second-line therapy (ODYSSEY-B; 195 [98%] of 200 had protease inhibitor-based therapy in the standard-of-care group). During follow-up (median 142 weeks, IQR 124-159), 23 participants had 31 neuropsychiatric adverse events (15 in the dolutegravir group and eight in the standard-of-care group; difference in proportion of participants with ≥1 event p=0·13). 11 participants had one or more neurological events (six and five; p=0·74) and 14 participants had one or more psychiatric events (ten and four; p=0·097). Among 14 participants with psychiatric events, eight participants in the dolutegravir group and four in standard-of-care group had suicidal ideation or behaviour. More participants in the dolutegravir group than the standard-of-care group reported symptoms of self-harm (eight vs one; p=0·025), life not worth living (17 vs five; p=0·0091), or suicidal thoughts (13 vs none; p=0·0006) at one or more follow-up visits. Most reports were transient. There were no differences by treatment group in low mood or feeling sad, problems concentrating, feeling worried or feeling angry or aggressive, sleep problems, or sleep quality. INTERPRETATION The numbers of neuropsychiatric adverse events and reported neuropsychiatric symptoms were low. However, numerically more participants had psychiatric events and reported suicidality ideation in the dolutegravir group than the standard-of-care group. These differences should be interpreted with caution in an open-label trial. Clinicians and policy makers should consider including suicidality screening of children or adolescents receiving dolutegravir. FUNDING Penta Foundation, ViiV Healthcare, and UK Medical Research Council.
Collapse
Affiliation(s)
- Anna Turkova
- Medical Research Council Clinical Trials Unit at University College London, London, UK.
| | - Ellen White
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | | | - Vivian Mumbiro
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | | | - Afaaf Liberty
- Perinatal HIV Research Unit, University of the Witwarsrand, Johannesburg, South Africa
| | | | - Tumelo Moloantoa
- Perinatal HIV Research Unit, University of the Witwarsrand, Johannesburg, South Africa
| | | | - Nozibusiso Rejoice Mosia
- Department of Paediatrics and Children Health, King Edward VIII Hospital, Enhancing Care Foundation, University of KwaZulu-Natal, Durban, South Africa
| | - Thanyawee Puthanakit
- HIVNAT, Thai Red Cross AIDS Research Center, Bangkok, Thailand; Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Robin Kobbe
- Institute for Infection Research and Vaccine Development, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Clàudia Fortuny
- Infectious Diseases Department, Institut de Recerca Sant Joan de Déu, Sant Joan de Déu Children's Hospital, Barcelona, Spain; Department of Surgery and Medico-Surgical Specialties, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Hajira Kataike
- Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda
| | | | | | - Alexandra Coelho
- INSERM/ANRS SC10-US19, Essais Thérapeutiques et Maladies Infectieuses, Villejuif, France
| | | | | | - Nigel Klein
- Infection, Immunity & Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, UK; Africa Health Research Institute, Kwazulu-Natal, South Africa
| | - Hilda A Mujuru
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | - Cissy Kityo
- Joint Clinical Research Centre, Kampala, Uganda
| | - Mark F Cotton
- Children's Infectious Diseases Clinical Research Unit, Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, University of Stellenbosch, Cape Town, South Africa
| | - Rashida A Ferrand
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Carlo Giaquinto
- Department of Women and Child Health, Padova, University of Padova, Italy
| | - Pablo Rojo
- Pediatric Infectious Diseases Unit, Hospital 12 de Octubre, Madrid, Spain
| | - Avy Violari
- Perinatal HIV Research Unit, University of the Witwarsrand, Johannesburg, South Africa
| | - Diana M Gibb
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Deborah Ford
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| |
Collapse
|
13
|
Brooks KM, Kiser JJ, Ziemba L, Ward S, Rani Y, Cressey TR, Masheto GR, Cassim H, Deville JG, Ponatshego PL, Patel F, Aurpibul L, Barnabas SL, Mustich I, Coletti A, Heckman B, Krotje C, Lojacono M, Yin DE, Townley E, Moye J, Majji S, Acosta EP, Ryan K, Chandasana H, Brothers CH, Buchanan AM, Rabie H, Flynn PM. Pharmacokinetics, safety, and tolerability of dispersible and immediate-release abacavir, dolutegravir, and lamivudine tablets in children with HIV (IMPAACT 2019): week 24 results of an open-label, multicentre, phase 1-2 dose-confirmation study. Lancet HIV 2023; 10:e506-e517. [PMID: 37541705 PMCID: PMC10642428 DOI: 10.1016/s2352-3018(23)00107-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Child-friendly fixed-dose combination (FDC) antiretroviral therapy (ART) options are limited. We evaluated the pharmacokinetics, safety, and tolerability of dispersible and immediate-release FDC abacavir, dolutegravir, and lamivudine taken once per day in children younger than 12 years with HIV. METHODS IMPAACT 2019 was an international, phase 1-2, multisite, open-label, non-comparative dose-confirmation study of abacavir, dolutegravir, and lamivudine in children younger than 12 years. Participants were enrolled across five weight bands: those weighing 6 kg to less than 25 kg received abacavir (60 mg), dolutegravir (5 mg), and lamivudine (30 mg) dispersible tablets (three to six tablets depending on body weight), and those weighing 25 kg to less than 40 kg received abacavir (600 mg), dolutegravir (50 mg), and lamivudine (300 mg) in an immediate-release tablet. At entry, participants were ART naive or ART experienced and virologically suppressed on stable ART for 6 months or more. Dose confirmation was based on pharmacokinetic and safety criteria in the first five to seven participants in each weight band to week 4; all participants were followed up to week 48. We present the results for the primary objectives to assess pharmacokinetics, confirm dosing, and evaluate safety through 24 weeks across all weight bands. The trial is registered with ClinicalTrials.gov (NCT03760458). FINDINGS 57 children were enrolled and initiated study drug (26 [46%] female and 31 [54%] male; 37 [65%] Black, 18 [32%] Asian, and 1 [2%] had race reported as unknown). Within each weight band, 6 kg to less than 10 kg, 10 kg to less than 14 kg, 14 kg to less than 20 kg, 20 kg to less than 25 kg, and 25 kg or higher: the geometric mean dolutegravir area under the concentration time curve over the 24 h dosing interval (AUC0-24 h) was 75·9 h·μg/mL (33·7%), 91·0 h·μg/mL (36·5%), 71·4 h·μg/mL (23·5%), 84·4 h·μg/mL (26·3%), and 71·8 h·μg/mL (13·9%); dolutegravir concentrations 24 h after dosage (C24 h) were 0·91 μg/mL (67·6%), 1·22 μg/mL (77·5%), 0·79 μg/mL (44·2%), 1·35 μg/mL (95·5%), and 0·98 μg/mL (27·9%); abacavir AUC0-24 h was 17·7 h·μg/mL (38·8%), 19·8 h·μg/mL (50·6%), 15·1 h·μg/mL (40·3%), 17·4 h·μg/mL (19·4%), and 25·7 h·μg/mL (14·6%); lamivudine AUC0-24 h was 10·7 h·μg/mL (46·0%), 14·2 h·μg/mL (23·9%), 13·0 h·μg/mL (15·6%), 14·5 h·μg/mL (16·6%), and 21·7 h·μg/mL (26·2%), respectively. Pharmacokinetic targets and safety criteria were met within each weight band, and thus dosing of abacavir, dolutegravir, and lamivudine was confirmed at the originally selected doses. 54 (95%) of participants were treatment experienced and all who continued taking the study drug remained virologically suppressed (<200 copies per mL) through week 24. Virological suppression was achieved in two of three participants who were ART naive by week 24. There were no grade 3 or higher adverse events related to abacavir, dolutegravir, and lamivudine and no discontinuations because of toxicity to week 24. Both formulations were well tolerated. INTERPRETATION Dosing of abacavir, dolutegravir, and lamivudine was confirmed in children weighing 6 kg to less than 40 kg, and both FDC formulations were safe, well tolerated, and efficacious through 24 weeks of treatment. These findings support global efforts to expand the availability of FDC abacavir, dolutegravir, and lamivudine to children with HIV. FUNDING National Institute of Allergy and Infectious Diseases, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, the National Institute of Mental Health, ViiV Healthcare, and GlaxoSmithKline.
Collapse
Affiliation(s)
- Kristina M Brooks
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Jennifer J Kiser
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren Ziemba
- Centre for Biostatistics in AIDS Research, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Shawn Ward
- Frontier Science Foundation, Brookline, MA, USA
| | - Yasha Rani
- Frontier Science Foundation, Brookline, MA, USA
| | - Tim R Cressey
- PHPT-Chiangrai Prachanukroh Hospital, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | | | - Haseena Cassim
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | - Faeezah Patel
- Wits RHI, University of the Witwatersrand, Johannesburg, South Africa
| | - Linda Aurpibul
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand
| | | | | | | | | | | | | | - Dwight E Yin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ellen Townley
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jack Moye
- National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Sai Majji
- National Institute of Child Health and Human Development, Bethesda, MD, USA
| | | | - Kevin Ryan
- University of Alabama-Birmingham, Birmingham, AL, USA
| | | | | | | | - Helena Rabie
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Science, University of Stellenbosch, Cape Town, South Africa
| | | |
Collapse
|
14
|
Lain MG, Vaz P, Sanna M, Ismael N, Chicumbe S, Simione TB, Cantarutti A, Porcu G, Rinaldi S, de Armas L, Dinh V, Pallikkuth S, Pahwa R, Palma P, Cotugno N, Pahwa S. Viral Response among Early Treated HIV Perinatally Infected Infants: Description of a Cohort in Southern Mozambique. Healthcare (Basel) 2022; 10:2156. [PMID: 36360495 PMCID: PMC9691232 DOI: 10.3390/healthcare10112156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Early initiation of antiretroviral therapy and adherence to achieve viral load suppression (VLS) are crucial for reducing morbidity and mortality of perinatally HIV-infected infants. In this descriptive cohort study of 39 HIV perinatally infected infants, who started treatment at one month of life in Mozambique, we aimed to describe the viral response over 2 years of follow up. VLS ≤ 400 copies/mL, sustained VLS and viral rebound were described using a Kaplan-Meier estimator. Antiretroviral drug transmitted resistance was assessed for a sub-group of non-VLS infants. In total, 61% of infants reached VLS, and 50% had a rebound. Cumulative probability of VLS was 36%, 51%, and 69% at 6, 12 and 24 months of treatment, respectively. The median duration of VLS was 7.4 months (IQR 12.6) and the cumulative probability of rebound at 6 months was 30%. Two infants had resistance biomarkers to drugs included in their treatment regimen. Our findings point to a low rate of VLS and high rate of viral rebound. More frequent viral response monitoring is advisable to identify infants with rebound and offer timely adherence support. It is urgent to tailor the psychosocial support model of care to this specific age group and offer differentiated service delivery to mother-baby pairs.
Collapse
Affiliation(s)
- Maria Grazia Lain
- Fundação Ariel Glaser Contra o SIDA Pediátrico, Maputo P.O.Box 2822, Mozambique
| | - Paula Vaz
- Fundação Ariel Glaser Contra o SIDA Pediátrico, Maputo P.O.Box 2822, Mozambique
| | - Marco Sanna
- Research Unit of Clinical Immunology and Vaccinology, Children’s Hospital Bambino Gesù, IRCCS, 0165 Rome, Italy
| | - Nalia Ismael
- Technological Platforms Department, Instituto Nacional de Saúde, Marracuene, Maputo 1120, Mozambique
| | - Sérgio Chicumbe
- Health System and Policy Program, Instituto Nacional de Saúde, Marracuene, Maputo 1120, Mozambique
| | | | - Anna Cantarutti
- National Centre for Healthcare Research and Pharmaco-Epidemiology, Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy
| | - Gloria Porcu
- National Centre for Healthcare Research and Pharmaco-Epidemiology, Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy
| | - Stefano Rinaldi
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lesley de Armas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vinh Dinh
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rajendra Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Children’s Hospital Bambino Gesù, IRCCS, 0165 Rome, Italy
- Department of Systems Medicine, University of Rome “Tor Vergata”, 0133 Rome, Italy
| | - Nicola Cotugno
- Research Unit of Clinical Immunology and Vaccinology, Children’s Hospital Bambino Gesù, IRCCS, 0165 Rome, Italy
- Department of Systems Medicine, University of Rome “Tor Vergata”, 0133 Rome, Italy
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
15
|
Amuge P, Lugemwa A, Wynne B, Mujuru HA, Violari A, Kityo CM, Archary M, Variava E, White E, Turner RM, Shakeshaft C, Ali S, Nathoo KJ, Atwine L, Liberty A, Bbuye D, Kaudha E, Mngqibisa R, Mosala M, Mumbiro V, Nanduudu A, Ankunda R, Maseko L, Kekitiinwa AR, Giaquinto C, Rojo P, Gibb DM, Turkova A, Ford D. Once-daily dolutegravir-based antiretroviral therapy in infants and children living with HIV from age 4 weeks: results from the below 14 kg cohort in the randomised ODYSSEY trial. Lancet HIV 2022; 9:e638-e648. [PMID: 36055295 PMCID: PMC9646993 DOI: 10.1016/s2352-3018(22)00163-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Young children living with HIV have few treatment options. We aimed to assess the efficacy and safety of dolutegravir-based antiretroviral therapy (ART) in children weighing between 3 kg and less than 14 kg. METHODS ODYSSEY is an open-label, randomised, non-inferiority trial (10% margin) comparing dolutegravir-based ART with standard of care and comprises two cohorts (children weighing ≥14 kg and <14 kg). Children weighing less than 14 kg starting first-line or second-line ART were enrolled in seven HIV treatment centres in South Africa, Uganda, and Zimbabwe. Randomisation, which was computer generated by the trial statistician, was stratified by first-line or second-line ART and three weight bands. Dispersible 5 mg dolutegravir was dosed according to WHO weight bands. The primary outcome was the Kaplan-Meier estimated proportion of children with virological or clinical failure by 96 weeks, defined as: confirmed viral load of at least 400 copies per mL after week 36; absence of virological suppression by 24 weeks followed by a switch to second-line or third-line ART; all-cause death; or a new or recurrent WHO stage 4 or severe WHO stage 3 event. The primary outcome was assessed by intention to treat in all randomly assigned participants. A primary Bayesian analysis of the difference in the proportion of children meeting the primary outcome between treatment groups incorporated evidence from the higher weight cohort (≥14 kg) in a prior distribution. A frequentist analysis was also done of the lower weight cohort (<14 kg) alone. Safety analyses are presented for all randomly assigned children in this study (<14 kg cohort). ODYSSEY is registered with ClinicalTrials.gov, NCT02259127. FINDINGS Between July 5, 2018, and Aug 26, 2019, 85 children weighing less than 14 kg were randomly assigned to receive dolutegravir (n=42) or standard of care (n=43; 32 [74%] receiving protease inhibitor-based ART). Median age was 1·4 years (IQR 0·6-2·0) and median weight 8·1 kg (5·4-10·0). 72 (85%) children started first-line ART and 13 (15%) started second-line ART. Median follow-up was 124 weeks (112-137). By 96 weeks, treatment failure occurred in 12 children in the dolutegravir group (Kaplan-Meier estimated proportion 31%) versus 21 (48%) in the standard-of-care group. The Bayesian estimated difference in treatment failure (dolutegravir minus standard of care) was -10% (95% CI -19% to -2%; p=0·020), demonstrating superiority of dolutegravir. The frequentist estimated difference was -18% (-36% to 2%; p=0·057). 15 serious adverse events were reported in 11 (26%) children in the dolutegravir group, including two deaths, and 19 were reported in 11 (26%) children in the standard-of-care group, including four deaths (hazard ratio [HR] 1·08 [95% CI 0·47-2·49]; p=0·86). 36 adverse events of grade 3 or higher were reported in 19 (45%) children in the dolutegravir group, versus 34 events in 21 (49%) children in the standard-of-care group (HR 0·93 [0·50-1·74]; p=0·83). No events were considered related to dolutegravir. INTERPRETATION Dolutegravir-based ART was superior to standard of care (mainly protease inhibitor-based) with a lower risk of treatment failure in infants and young children, providing support for global dispersible dolutegravir roll-out for younger children and allowing alignment of adult and paediatric treatment. FUNDING Paediatric European Network for Treatment of AIDS Foundation, ViiV Healthcare, UK Medical Research Council.
Collapse
Affiliation(s)
- Pauline Amuge
- Baylor College of Medicine Children's Foundation-Uganda, Kampala, Uganda
| | | | - Ben Wynne
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Hilda A Mujuru
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | - Avy Violari
- Perinatal HIV Research Unit, University of the Witwatersrand, South Africa
| | | | - Moherndran Archary
- Department of Paediatrics and Children Health, King Edward VIII Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Ebrahim Variava
- Perinatal HIV Research Unit, University of the Witwatersrand, South Africa
| | - Ellen White
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Rebecca M Turner
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Clare Shakeshaft
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Shabinah Ali
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Kusum J Nathoo
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | | | - Afaaf Liberty
- Perinatal HIV Research Unit, University of the Witwatersrand, South Africa
| | - Dickson Bbuye
- Baylor College of Medicine Children's Foundation-Uganda, Kampala, Uganda
| | | | - Rosie Mngqibisa
- Department of Paediatrics and Children Health, King Edward VIII Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Modehei Mosala
- Perinatal HIV Research Unit, University of the Witwatersrand, South Africa
| | - Vivian Mumbiro
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | | | | | - Lindiwe Maseko
- Perinatal HIV Research Unit, University of the Witwatersrand, South Africa
| | | | - Carlo Giaquinto
- Department of Women and Child Health, University of Padova, Italy; Penta Foundation, Padova, Italy
| | - Pablo Rojo
- Pediatric Infectious Diseases Unit, Hospital 12 de Octubre, Madrid, Spain
| | - Diana M Gibb
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Anna Turkova
- Medical Research Council Clinical Trials Unit at University College London, London, UK
| | - Deborah Ford
- Medical Research Council Clinical Trials Unit at University College London, London, UK.
| |
Collapse
|
16
|
Blanche S. Young children still to treat, unfortunately. THE LANCET HIV 2022; 9:e600-e601. [DOI: 10.1016/s2352-3018(22)00199-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022]
|
17
|
Turkova A, Waalewijn H, Chan MK, Bollen PDJ, Bwakura-Dangarembizi MF, Kekitiinwa AR, Cotton MF, Lugemwa A, Variava E, Ahimbisibwe GM, Srirompotong U, Mumbiro V, Amuge P, Zuidewind P, Ali S, Kityo CM, Archary M, Ferrand RA, Violari A, Gibb DM, Burger DM, Ford D, Colbers A. Dolutegravir twice-daily dosing in children with HIV-associated tuberculosis: a pharmacokinetic and safety study within the open-label, multicentre, randomised, non-inferiority ODYSSEY trial. Lancet HIV 2022; 9:e627-e637. [PMID: 35868341 PMCID: PMC9630157 DOI: 10.1016/s2352-3018(22)00160-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Children with HIV-associated tuberculosis (TB) have few antiretroviral therapy (ART) options. We aimed to evaluate the safety and pharmacokinetics of dolutegravir twice-daily dosing in children receiving rifampicin for HIV-associated TB. METHODS We nested a two-period, fixed-order pharmacokinetic substudy within the open-label, multicentre, randomised, controlled, non-inferiority ODYSSEY trial at research centres in South Africa, Uganda, and Zimbabwe. Children (aged 4 weeks to <18 years) with HIV-associated TB who were receiving rifampicin and twice-daily dolutegravir were eligible for inclusion. We did a 12-h pharmacokinetic profile on rifampicin and twice-daily dolutegravir and a 24-h profile on once-daily dolutegravir. Geometric mean ratios for trough plasma concentration (Ctrough), area under the plasma concentration time curve from 0 h to 24 h after dosing (AUC0-24 h), and maximum plasma concentration (Cmax) were used to compare dolutegravir concentrations between substudy days. We assessed rifampicin Cmax on the first substudy day. All children within ODYSSEY with HIV-associated TB who received rifampicin and twice-daily dolutegravir were included in the safety analysis. We described adverse events reported from starting twice-daily dolutegravir to 30 days after returning to once-daily dolutegravir. This trial is registered with ClinicalTrials.gov (NCT02259127), EudraCT (2014-002632-14), and the ISRCTN registry (ISRCTN91737921). FINDINGS Between Sept 20, 2016, and June 28, 2021, 37 children with HIV-associated TB (median age 11·9 years [range 0·4-17·6], 19 [51%] were female and 18 [49%] were male, 36 [97%] in Africa and one [3%] in Thailand) received rifampicin with twice-daily dolutegravir and were included in the safety analysis. 20 (54%) of 37 children enrolled in the pharmacokinetic substudy, 14 of whom contributed at least one evaluable pharmacokinetic curve for dolutegravir, including 12 who had within-participant comparisons. Geometric mean ratios for rifampicin and twice-daily dolutegravir versus once-daily dolutegravir were 1·51 (90% CI 1·08-2·11) for Ctrough, 1·23 (0·99-1·53) for AUC0-24 h, and 0·94 (0·76-1·16) for Cmax. Individual dolutegravir Ctrough concentrations were higher than the 90% effective concentration (ie, 0·32 mg/L) in all children receiving rifampicin and twice-daily dolutegravir. Of 18 children with evaluable rifampicin concentrations, 15 (83%) had a Cmax of less than the optimal target concentration of 8 mg/L. Rifampicin geometric mean Cmax was 5·1 mg/L (coefficient of variation 71%). During a median follow-up of 31 weeks (IQR 30-40), 15 grade 3 or higher adverse events occurred among 11 (30%) of 37 children, ten serious adverse events occurred among eight (22%) children, including two deaths (one tuberculosis-related death, one death due to traumatic injury); no adverse events, including deaths, were considered related to dolutegravir. INTERPRETATION Twice-daily dolutegravir was shown to be safe and sufficient to overcome the rifampicin enzyme-inducing effect in children, and could provide a practical ART option for children with HIV-associated TB. FUNDING Penta Foundation, ViiV Healthcare, UK Medical Research Council.
Collapse
Affiliation(s)
- Anna Turkova
- Medical Research Council Clinical Trials Unit, University College London, London, UK.
| | - Hylke Waalewijn
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Man K Chan
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Pauline D J Bollen
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | - Mark F Cotton
- Children's Infectious Diseases Clinical Research Unit, Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, University of Stellenbosch, Cape Town, South Africa
| | | | - Ebrahim Variava
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | - Vivian Mumbiro
- University of Zimbabwe Clinical Research Centre, Harare, Zimbabwe
| | | | - Peter Zuidewind
- Children's Infectious Diseases Clinical Research Unit, Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, University of Stellenbosch, Cape Town, South Africa
| | - Shabinah Ali
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | | | - Moherndran Archary
- Department of Paediatrics and Child Health, King Edward VIII Hospital, Enhancing Care Foundation, University of KwaZulu-Natal, Durban, South Africa
| | | | - Avy Violari
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Diana M Gibb
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - David M Burger
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Deborah Ford
- Medical Research Council Clinical Trials Unit, University College London, London, UK
| | - Angela Colbers
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
18
|
Rabie H, Lishman J, Frigati LJ. Moving forward with dolutegravir in children weighing less than 20 kg. Lancet HIV 2022; 9:e301-e302. [PMID: 35489373 DOI: 10.1016/s2352-3018(22)00062-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Affiliation(s)
- Helena Rabie
- Department of Paediatrics and Child Health, Stellenbosch University and Tygerberg Hospital, Faculty of medicine and Health Sciences, Parrow, Cape Town, 7505, South Africa.
| | - Juanita Lishman
- Department of Paediatrics and Child Health, Stellenbosch University and Tygerberg Hospital, Faculty of medicine and Health Sciences, Parrow, Cape Town, 7505, South Africa
| | - Lisa Jane Frigati
- Department of Paediatrics and Child Health, Stellenbosch University and Tygerberg Hospital, Faculty of medicine and Health Sciences, Parrow, Cape Town, 7505, South Africa
| |
Collapse
|
19
|
Third-Line Antiretroviral Therapy: What Do We Do When the Appropriate Formulations Are Not Available? CHILDREN 2022; 9:children9040473. [PMID: 35455517 PMCID: PMC9032725 DOI: 10.3390/children9040473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022]
Abstract
Children on antiretroviral therapy have limited options, particularly if they are failing therapy and live in resource-poor settings. We describe three cases where children accessed third-line antiretroviral therapy off-label, or used them extemporaneously with successful outcomes. We then review the evidence for performing this measure. There is an urgent need for appropriate formulations to treat young children who require a third-line or salvage regimen.
Collapse
|