1
|
Santos JR, Domingo P, Portilla J, Gutiérrez F, Imaz A, Vilchez H, Curran A, Valcarce-Pardeiro N, Payeras A, Bernal E, Montero-Alonso M, Yzusqui M, Clotet B, Videla S, Moltó J, Paredes R. A Randomized Trial of Dolutegravir Plus Darunavir/Cobicistat as a Switch Strategy in HIV-1-Infected Patients With Resistance to at Least 2 Antiretroviral Classes. Open Forum Infect Dis 2023; 10:ofad542. [PMID: 38023553 PMCID: PMC10661076 DOI: 10.1093/ofid/ofad542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Background Suppressed patients with drug-resistant HIV-1 require effective and simple antiretroviral therapy to maintain treatment adherence and viral suppression. Methods This randomized, open-label, noninferiority, multicenter pilot study involved HIV-infected adults who met the following criteria: confirmed HIV-1 RNA <50 copies/mL for ≥6 months preceding the study randomization, treatment with at least 3 antiretroviral drugs, and a history of drug resistance mutations against at least 2 antiretroviral classes but remaining fully susceptible to darunavir (DRV) and integrase inhibitors. Participants were randomized 1:1 to switch to dolutegravir (DTG; 50 mg once per day) plus DRV boosted with cobicistat (DRV/c; 800/150 mg once per day; 2D group) or continue with their baseline regimen (standard-of-care [SOC] group). The primary endpoint was the proportion of patients with HIV-1 RNA <50 copies/mL at week 48 relative to time to loss of virologic response, with a noninferiority margin set at -12.5%. Virologic failure was defined as confirmed HIV-1 RNA ≥50 copies/mL or a single determination of HIV-1 RNA >50 copies/mL followed by antiretroviral therapy discontinuation. Results Forty-five participants were assigned to the 2D group and 44 to the SOC group. Time to loss of virologic response showed no difference in the proportion maintaining HIV-1 RNA <50 copies/mL at week 48: 39 of 45 (86.7%; 95% CI, 73.21%-94.95%) in the 2D group vs 42 of 44 (95.4%; 95% CI, 84.53%-99.44%) in the SOC group (log-rank P = .159) with an estimated difference of -8.7 (95% CI, -22.72 to 5.14). Only 2 (4.5%) in the SOC group experienced virologic failure, and 3 participants from the 2D group experienced adverse events leading to treatment discontinuation. Conclusions In suppressed patients with at least 2 resistant antiretroviral classes, noninferiority could not be demonstrated by fully active DRV/c plus DTG. Nevertheless, there were no unexpected adverse events or virologic failure. DRV/c plus DTG may be considered a once-daily therapy option only for well-selected patients. Clinical Trials Registration. ClinicalTrials.gov (NCT03683524).
Collapse
Affiliation(s)
- José R Santos
- Infectious Diseases Department and Fundació Lluita contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Pere Domingo
- HIV Unit, Service of Internal Medicine, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Félix Gutiérrez
- Infectious Diseases Unit, Department of Internal Medicine, Hospital General Universitario de Elche, Elche, Spain
- University Miguel Hernández, Alicante, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
| | - Arkaitz Imaz
- HIV Unit, Infectious Diseases Service, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Helem Vilchez
- Infectious Diseases Unit, Internal Medicine Department, Hospital Universitari Son Espases, Fundació Institut d’Investigació Sanitària Illes Balears, Palma de Mallorca, Spain
| | - Adrià Curran
- HIV Unit, Service of Infectious Diseases, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | | | - Antoni Payeras
- Service of Internal Medicine, Hospital Son Llàtzer, Palma de Mallorca, Spain
| | - Enrique Bernal
- Infectious Diseases Unit, Hospital General Universitario Reina Sofía de Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
| | - Marta Montero-Alonso
- Infectious Diseases Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Miguel Yzusqui
- Department of Internal Medicine, Hospital General Nuestra Señora del Prado, Talavera de la Reina, Spain
| | - Bonaventura Clotet
- Infectious Diseases Department and Fundació Lluita contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
- IrsiCaixa AIDS Research Institute, Badalona, Spain
| | - Sebastià Videla
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, IDIBELL, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain
- Lluita contra les Infeccions Foundation, Badalona, Spain
| | - José Moltó
- Infectious Diseases Department and Fundació Lluita contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
- CIBERINFEC, ISCIII, Madrid, Spain
| | - Roger Paredes
- Infectious Diseases Department and Fundació Lluita contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Center for Global Health and Diseases, Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Prins HAB, Zino L, Svensson EM, Verbon A, de Bree GJ, Prins JM, Reiss P, Burger DM, Rokx C, Colbers A. Exposure and virologic outcomes of dolutegravir combined with ritonavir boosted darunavir in treatment-naïve individuals enrolled in the Netherlands Cohort Study on Acute HIV infection (NOVA). Int J Antimicrob Agents 2023; 61:106697. [PMID: 36470510 DOI: 10.1016/j.ijantimicag.2022.106697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 10/08/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022]
Abstract
To the authors' knowledge, there is currently no literature or guidance recommendation regarding whether the dose of dolutegravir (DTG) should be increased when co-administered with darunavir/ritonavir (DRV/r) in patients with acute human immunodeficiency virus infection (AHI). This study assessed the pharmacokinetics (PK) of twice-daily (BID) DTG and once-daily (QD) DRV/r, and compared this with DTG QD without DRV/r in patients with AHI. Forty-six participants initiated antiretroviral therapy within <24 h of enrolment: DTG 50 mg BID, DRV/r 800/100 mg QD, and two nucleoside reverse transcriptase inhibitors (NRTIs) for 4 weeks (Phase I); and DTG 50 mg QD with two NRTIs thereafter (Phase II: reference). Total DTG trough concentration (Ctrough) and area under the concentration-time profile of 0-24 h (AUC0-24h) were predicted using a population PK model. DTG glucuronidation metabolic ratio (MR) and DTG free fraction were determined and compared per treatment phase using geometric mean ratio (GMR) and 90% confidence interval (CI). Participants had a predicted geometric mean steady-state DTG Ctrough of 2.83 [coefficient of variation (CV%) 30.3%] mg/L (Phase I) and 1.28 (CV% 52.4%) mg/L (Phase II), with GMR of 2.20 (90% CI 1.90-2.55). Total exposure during DTG BID increased but did not double [AUC0-24h GMR 1.65 (90% CI 1.50-1.81) h.mg/L]. DTG glucuronidation MR increased by approximately 29% during Phase I. DTG Ctrough was above in-vivo EC90 (0.32 mg/L) during both phases, except in one participant during Phase I. At Week 8, 84% of participants had viral loads ≤40 copies/mL. The drug-drug interaction between DTG (BID) and DRV/r (QD) was due to induced glucuronidation, and is not clinically relevant in patients with AHI.
Collapse
Affiliation(s)
- H A B Prins
- Erasmus Medical Centre, Department of Medical Microbiology and Infectious Diseases and Department of Internal Medicine, Rotterdam, The Netherlands
| | - L Zino
- Radboud University Medical Centre, Department of Pharmacy and Radboud Institute for Health Sciences, Nijmegen, The Netherlands.
| | - E M Svensson
- Radboud University Medical Centre, Department of Pharmacy and Radboud Institute for Health Sciences, Nijmegen, The Netherlands; Uppsala University, Department of Pharmacy, Uppsala, Sweden
| | - A Verbon
- Erasmus Medical Centre, Department of Medical Microbiology and Infectious Diseases and Department of Internal Medicine, Rotterdam, The Netherlands
| | - G J de Bree
- Amsterdam University Medical Centre, University of Amsterdam, Department of Internal Medicine, Division of Infectious Diseases, and Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - J M Prins
- Amsterdam University Medical Centre, University of Amsterdam, Department of Internal Medicine, Division of Infectious Diseases, and Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - P Reiss
- Amsterdam University Medical Centre, University of Amsterdam, Department of Internal Medicine, Division of Infectious Diseases, and Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands; Amsterdam University Medical Centre, University of Amsterdam, Department of Global Health, and Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - D M Burger
- Radboud University Medical Centre, Department of Pharmacy and Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - C Rokx
- Erasmus Medical Centre, Department of Medical Microbiology and Infectious Diseases and Department of Internal Medicine, Rotterdam, The Netherlands
| | - A Colbers
- Radboud University Medical Centre, Department of Pharmacy and Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | | |
Collapse
|
3
|
Banda CG, Nkosi D, Allen E, Workman L, Madanitsa M, Chirwa M, Kapulula M, Muyaya S, Munharo S, Wiesner L, Phiri KS, Mwapasa V, Ter Kuile FO, Maartens G, Barnes KI. Effect of dihydroartemisinin/piperaquine for malaria intermittent preventive treatment on dolutegravir exposure in pregnant women living with HIV. J Antimicrob Chemother 2022; 77:1733-1737. [PMID: 35288747 PMCID: PMC9155593 DOI: 10.1093/jac/dkac081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/18/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND In sub-Saharan Africa, the burdens of malaria and HIV infections overlap. In settings with moderate-to-high malaria transmission intensity, pregnant women living with HIV (PLWH) require both ART and malaria intermittent preventive treatment (IPTp). Dihydroartemisinin/piperaquine has been identified as a promising alternative to sulfadoxine/pyrimethamine for IPTp. However, another antimalarial drug, artesunate/amodiaquine, similar to dihydroartemisinin/piperaquine, was previously shown to reduce dolutegravir exposure in non-pregnant adults. OBJECTIVES To investigate the effect of dihydroartemisinin/piperaquine on dolutegravir plasma exposure in pregnant women on dolutegravir-based ART. METHODS We conducted an open-label, non-randomized, fixed-sequence, pharmacokinetic study in PLWH in Malawi. Dolutegravir concentrations were measured over a 24 h period, before and after the recommended 3 day treatment dose of dihydroartemisinin/piperaquine in 12 pregnant women in their second or third trimester. Non-compartmental analysis was performed, and geometric mean ratios (GMRs) and 90% CIs were generated to compare dolutegravir pharmacokinetic parameters between the two treatment periods. RESULTS Co-administration of dihydroartemisinin/piperaquine and dolutegravir increased dolutegravir's overall exposure (AUC0-24) and Cmax by 30% (GMR 1.30; 90% CI 1.11-1.52) and 31% (GMR 1.31; 90% CI 1.13-1.51), respectively. The dolutegravir trough (C24) concentration increased by 42% (GMR 1.42; 90% CI 1.09-1.85). The combined treatments were well tolerated with no serious adverse events observed. CONCLUSIONS Dihydroartemisinin/piperaquine may be administered with dolutegravir-based ART in pregnant women as the modest increase in dolutegravir exposure, similar to pharmacokinetic parameter values published previously, ensures its efficacy without any clinically significant adverse events observed in this small study.
Collapse
Affiliation(s)
- Clifford G. Banda
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- Kamuzu University of Health Sciences, Blantyre, Malawi (formerly College of Medicine and Kamuzu College of Nursing, University of Malawi)
| | - Dumisile Nkosi
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Elizabeth Allen
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- WorldWide Antimalarial Resistance Network (WWARN), Pharmacology Scientific Group, University of Cape Town, Cape Town, South Africa
| | - Lesley Workman
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- WorldWide Antimalarial Resistance Network (WWARN), Pharmacology Scientific Group, University of Cape Town, Cape Town, South Africa
| | - Mwayiwawo Madanitsa
- Training and Research Unit of Excellence, Blantyre, Malawi
- Department of Clinical Sciences, Malawi University of Science and Technology, Limbe, Malawi
| | - Marumbo Chirwa
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | | | - Sharon Muyaya
- Training and Research Unit of Excellence, Blantyre, Malawi
| | - Steven Munharo
- Training and Research Unit of Excellence, Blantyre, Malawi
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Kamija S. Phiri
- Kamuzu University of Health Sciences, Blantyre, Malawi (formerly College of Medicine and Kamuzu College of Nursing, University of Malawi)
- Training and Research Unit of Excellence, Blantyre, Malawi
| | - Victor Mwapasa
- Kamuzu University of Health Sciences, Blantyre, Malawi (formerly College of Medicine and Kamuzu College of Nursing, University of Malawi)
| | - Feiko O. Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Gary Maartens
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Karen I. Barnes
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- WorldWide Antimalarial Resistance Network (WWARN), Pharmacology Scientific Group, University of Cape Town, Cape Town, South Africa
- Corresponding author. E-mail:
| |
Collapse
|
4
|
High-content analysis and Kinetic Image Cytometry identify toxicity and epigenetic effects of HIV antiretrovirals on human iPSC-neurons and primary neural precursor cells. J Pharmacol Toxicol Methods 2022; 114:107157. [PMID: 35143957 PMCID: PMC9103414 DOI: 10.1016/j.vascn.2022.107157] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Despite viral suppression due to combination antiretroviral therapy (cART), HIV-associated neurocognitive disorders (HAND) continue to affect half of people with HIV, suggesting that certain antiretrovirals (ARVs) may contribute to HAND. METHODS We examined the effects of nucleoside/nucleotide reverse transcriptase inhibitors tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) and the integrase inhibitors dolutegravir (DTG) and elvitegravir (EVG) on viability, structure, and function of glutamatergic neurons (a subtype of CNS neuron involved in cognition) derived from human induced pluripotent stem cells (hiPSC-neurons), and primary human neural precursor cells (hNPCs), which are responsible for neurogenesis. RESULTS Using automated digital microscopy and image analysis (high content analysis, HCA), we found that DTG, EVG, and TDF decreased hiPSC-neuron viability, neurites, and synapses after 7 days of treatment. Analysis of hiPSC-neuron calcium activity using Kinetic Image Cytometry (KIC) demonstrated that DTG and EVG also decreased the frequency and magnitude of intracellular calcium transients. Longer ARV exposures and simultaneous exposure to multiple ARVs increased the magnitude of these neurotoxic effects. Using the Microscopic Imaging of Epigenetic Landscapes (MIEL) assay, we found that TDF decreased hNPC viability and changed the distribution of histone modifications that regulate chromatin packing, suggesting that TDF may reduce neuroprogenitor pools important for CNS development and maintenance of cognition in adults. CONCLUSION This study establishes human preclinical assays that can screen potential ARVs for CNS toxicity to develop safer cART regimens and HAND therapeutics.
Collapse
|
5
|
Wolf E, Boesecke C, Balogh A, Bidner H, Cordes C, Heiken H, Krznaric I, Kümmerle T, Stellbrink HJ, Schneider J, Spinner CD. Virologic outcomes of switching to boosted darunavir plus dolutegravir with respect to history of drug resistance. AIDS Res Ther 2021; 18:58. [PMID: 34496848 PMCID: PMC8425038 DOI: 10.1186/s12981-021-00384-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/23/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE The DUALIS study showed that switching to boosted darunavir (bDRV) plus dolutegravir (DTG; 2DR) was non-inferior to continuous bDRV plus 2 nucleoside/nucleotide reverse-transcriptase inhibitors (NRTIs; 3DR) in treatment-experienced virologically suppressed people living with HIV (PLWH). We analyzed virologic outcomes with respect to treatment history and HIV drug resistance. DESIGN Post hoc analysis of a randomized trial. METHODS Main inclusion criteria were an HIV RNA level < 50 copies/mL for ≥ 24 weeks and no resistance to integrase strand transfer inhibitors or bDRV. Resistance-associated mutations (RAMs) were interpreted using the Stanford HIVdb mutation list. Outcomes measures were 48-week virologic response (HIV RNA < 50 copies/mL, FDA snapshot) and HIV RNA ≥ 50 copies/mL (including discontinuation due to a lack of efficacy or reasons other than adverse events and HIV RNA ≥ 50 copies/mL, referred to as snapshot non-response). RESULTS The analysis population included 263 patients (2DR: 131, 3DR: 132): 90.1% males; median age, 48 years; CD4 + T-cell nadir < 200/µl, 47.0%; ≥ 2 treatment changes, 27.4%; NRTI, non-NRTI (NNRTI), and major protease inhibitor (PI) RAMs in 9.5%, 14.4%, and 3.4%, respectively. In patients with RAMs in the 2DR and 3DR groups, virologic response rates were 87.8% and 96.0%, respectively; the corresponding rates in those without RAMs were 85.7% and 81.8%. RAMs were unrelated to virologic non-response in either group. No treatment-emergent RAMs were observed. CONCLUSIONS DTG + bDRV is an effective treatment option without the risk of treatment-emergent resistance for PLWH on suppressive first- or further-line treatment with or without evidence of pre-existing NRTI, NNRTI, or PI RAMs. TRIAL REGISTRATION EUDRA-CT Number 2015-000360-34; registered 07 April 2015; https://www.clinicaltrialsregister.eu/ctr-search/trial/2015-000360-34/DE .
Collapse
|
6
|
Shenkoya B, Atoyebi S, Eniayewu I, Akinloye A, Olagunju A. Mechanistic Modeling of Maternal Lymphoid and Fetal Plasma Antiretroviral Exposure During the Third Trimester. Front Pediatr 2021; 9:734122. [PMID: 34616699 PMCID: PMC8488224 DOI: 10.3389/fped.2021.734122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Pregnancy-induced changes in plasma pharmacokinetics of many antiretrovirals (ARV) are well-established. Current knowledge about the extent of ARV exposure in lymphoid tissues of pregnant women and within the fetal compartment is limited due to their inaccessibility. Subtherapeutic ARV concentrations in HIV reservoirs like lymphoid tissues during pregnancy may constitute a barrier to adequate virological suppression and increase the risk of mother-to-child transmission (MTCT). The present study describes the pharmacokinetics of three ARVs (efavirenz, dolutegravir, and rilpivirine) in lymphoid tissues and fetal plasma during pregnancy using materno-fetal physiologically-based pharmacokinetic models (m-f-PBPK). Lymphatic and fetal compartments were integrated into our previously validated adult PBPK model. Physiological and drug disposition processes were described using ordinary differential equations. For each drug, virtual pregnant women (n = 50 per simulation) received the standard dose during the third trimester. Essential pharmacokinetic parameters, including Cmax, Cmin, and AUC (0-24), were computed from the concentration-time data at steady state for lymph and fetal plasma. Models were qualified by comparison of predictions with published clinical data, the acceptance threshold being an absolute average fold-error (AAFE) within 2.0. AAFE for all model predictions was within 1.08-1.99 for all three drugs. Maternal lymph concentration 24 h after dose exceeded the reported minimum effective concentration (MEC) for efavirenz (11,514 vs. 800 ng/ml) and rilpivirine (118.8 vs. 50 ng/ml), but was substantially lower for dolutegravir (16.96 vs. 300 ng/ml). In addition, predicted maternal lymph-to-plasma AUC ratios vary considerably (6.431-efavirenz, 0.016-dolutegravir, 1.717-rilpivirine). Furthermore, fetal plasma-to-maternal plasma AUC ratios were 0.59 for efavirenz, 0.78 for dolutegravir, and 0.57 for rilpivirine. Compared with rilpivirine (0 h), longer dose forgiveness was observed for dolutegravir in fetal plasma (42 h), and for efavirenz in maternal lymph (12 h). The predicted low lymphoid tissue penetration of dolutegravir appears to be significantly offset by its extended dose forgiveness and adequate fetal compartment exposure. Hence, it is unlikely to be a predictor of maternal virological failure or MTCT risks. Predictions from our m-f-PBPK models align with recommendations of no dose adjustment despite moderate changes in exposure during pregnancy for these drugs. This is an important new application of PBPK modeling to evaluate the adequacy of drug exposure in otherwise inaccessible compartments.
Collapse
Affiliation(s)
- Babajide Shenkoya
- Department of Pharmaceutical Chemistry, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Shakir Atoyebi
- Department of Pharmaceutical Chemistry, Obafemi Awolowo University, Ile-Ife, Nigeria.,Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Ibrahim Eniayewu
- Department of Pharmaceutical Chemistry, Obafemi Awolowo University, Ile-Ife, Nigeria.,Department of Pharmaceutical and Medicinal Chemistry, University of Ilorin, Ilorin, Nigeria
| | - Abdulafeez Akinloye
- Department of Pharmaceutical Chemistry, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Adeniyi Olagunju
- Department of Pharmaceutical Chemistry, Obafemi Awolowo University, Ile-Ife, Nigeria.,Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
7
|
Barcelo C, Aouri M, Courlet P, Guidi M, Braun DL, Günthard HF, Piso RJ, Cavassini M, Buclin T, Decosterd LA, Csajka C. Population pharmacokinetics of dolutegravir: influence of drug-drug interactions in a real-life setting. J Antimicrob Chemother 2020; 74:2690-2697. [PMID: 31119275 DOI: 10.1093/jac/dkz217] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/04/2019] [Accepted: 04/23/2019] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES Dolutegravir is widely prescribed owing to its potent antiviral activity, high genetic barrier and good tolerability. The aim of this study was to characterize dolutegravir's pharmacokinetic profile and variability in a real-life setting and to identify individual factors and co-medications affecting dolutegravir disposition. METHODS A population pharmacokinetic model was developed using NONMEM®. Relevant demographic factors, clinical factors and co-medications were tested as potential covariates. Simulations based on the final model served to compare expected dolutegravir concentrations under standard and alternative dosage regimens in the case of drug-drug interactions. RESULTS A total of 620 dolutegravir plasma concentrations were collected from 521 HIV-infected individuals under steady-state conditions. A one-compartment model with first-order absorption and elimination best characterized dolutegravir pharmacokinetics. Typical dolutegravir apparent clearance (CL/F) was 0.93 L/h with 32% between-subject variability, the apparent volume of distribution was 20.2 L and the absorption rate constant was fixed to 2.24 h-1. Older age, higher body weight and current smoking were associated with higher CL/F. Atazanavir co-administration decreased dolutegravir CL/F by 38%, while darunavir modestly increased CL/F by 14%. Rifampicin co-administration showed the largest impact on CL/F. Simulations suggest that average dolutegravir trough concentrations are 63% lower after 50 mg/12h with rifampicin compared with a standard dosage of 50 mg/24h without rifampicin. Average trough concentrations after 100 mg/24h and 100 mg/12h with rifampicin are 92% and 25% lower than the standard dosage without rifampicin, respectively. CONCLUSIONS Patients co-treated with dolutegravir and rifampicin might benefit from therapeutic drug monitoring and individualized dosage increase, up to 100 mg/12 h in some cases.
Collapse
Affiliation(s)
- Catalina Barcelo
- Service of Clinical Pharmacology, University Hospital Centre and University of Lausanne, Bugnon 17, Lausanne, Switzerland
| | - Manel Aouri
- Service of Clinical Chemistry, University Hospital Centre and University of Lausanne, Bugnon 46, Lausanne, Switzerland
| | - Perrine Courlet
- Service of Clinical Pharmacology, University Hospital Centre and University of Lausanne, Bugnon 17, Lausanne, Switzerland
| | - Monia Guidi
- Service of Clinical Pharmacology, University Hospital Centre and University of Lausanne, Bugnon 17, Lausanne, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Michel-Servet 1, Geneva, Switzerland
| | - Dominique L Braun
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Rämistrasse 100, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Rämistrasse 100, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Rein J Piso
- Division of Infectious Diseases, Department of Medicine, Cantonal Hospital of Olten, Baslerstrasse 150, Olten, Switzerland
| | - Matthias Cavassini
- Service of Infectious Diseases, University Hospital Centre and University of Lausanne, Bugnon 46, Lausanne, Switzerland
| | - Thierry Buclin
- Service of Clinical Pharmacology, University Hospital Centre and University of Lausanne, Bugnon 17, Lausanne, Switzerland
| | - Laurent A Decosterd
- Service of Clinical Pharmacology, University Hospital Centre and University of Lausanne, Bugnon 17, Lausanne, Switzerland
| | - Chantal Csajka
- Service of Clinical Pharmacology, University Hospital Centre and University of Lausanne, Bugnon 17, Lausanne, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Michel-Servet 1, Geneva, Switzerland
| | | |
Collapse
|
8
|
Mboumba Bouassa RS, Mossoro-Kpinde CD, Gody JC, Veyer D, Péré H, Matta M, Robin L, Grésenguet G, Charpentier C, Bélec L. High predictive efficacy of integrase strand transfer inhibitors in perinatally HIV-1-infected African children in therapeutic failure of first- and second-line antiretroviral drug regimens recommended by the WHO. J Antimicrob Chemother 2020; 74:2030-2038. [PMID: 30891603 PMCID: PMC6587428 DOI: 10.1093/jac/dkz099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 12/23/2022] Open
Abstract
Objectives The predictive efficacy of integrase (IN) strand transfer inhibitors (INSTIs) was investigated in HIV-infected children born to HIV-infected mothers in Africa. Methods Plasma was collected at the Complexe Pédiatrique of Bangui, Central African Republic, from INSTI-naive children (n = 8) and adolescents (n = 10) in virological failure (viral load >1000 copies/mL) after 5 years of first- and/or second-line combination ART (cART). IN, reverse transcriptase (RT) and protease (P) genes were genotyped and drug resistance mutations (DRMs) to INSTIs, NRTIs, NNRTIs and PIs were interpreted using the Stanford algorithm. Results Successful IN, RT and P genotypes were obtained for 18, 13 and 15 children (median age 11 years, range 5–18; 8 were female), respectively. Two (2/18; 11.1%) viruses from children treated with a first-line regimen had INSTI DRMs at codon 138 (E138K and E138T), which is known to harbour major resistance mutations, and also had the accessory mutations L74I, G140K, G140R and G163R. The majority (16/18; 88.9%) of HIV-1 IN sequences demonstrated full susceptibility to all major INSTIs with a high frequency of natural polymorphic mutations. Most (12/15; 80%) genotyped viruses harboured at least one major DRM conferring resistance to at least one of the WHO-recommended antiretroviral drugs (NNRTIs, NRTIs and PIs) prescribed in first- and second-line regimens. Conclusions INSTIs could be proposed in first-line regimens in the majority of African children or adolescents and may constitute relevant therapeutic alternatives as second- and third-line cART regimens in HIV-infected children and adolescents living in sub-Saharan Africa.
Collapse
Affiliation(s)
- Ralph-Sydney Mboumba Bouassa
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France.,Ecole Doctorale (ED562) Bio Sorbonne Paris Cité (BioSPC), Université Paris Descartes, Paris, France.,Ecole Doctorale Régionale en Infectiologie Tropicale, Franceville, Gabon
| | - Christian Diamant Mossoro-Kpinde
- Laboratoire National de Biologie Clinique et de Santé Publique, Bangui, République Centrafricaine.,Faculté des Sciences de la Santé, Université de Bangui, Bangui, République Centrafricaine
| | - Jean-Chrysostome Gody
- Faculté des Sciences de la Santé, Université de Bangui, Bangui, République Centrafricaine.,Complexe Pédiatrique de Bangui, Bangui, République Centrafricaine
| | - David Veyer
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Hélène Péré
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Mathieu Matta
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Leman Robin
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Gérard Grésenguet
- Faculté des Sciences de la Santé, Université de Bangui, Bangui, République Centrafricaine
| | - Charlotte Charpentier
- INSERM, IAME, UMR 1137, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP-HP, Hôpital Bichat-Claude Bernard, Laboratoire de Virologie, Paris, France
| | - Laurent Bélec
- Laboratoire de Virologie, Hôpital Européen Georges Pompidou, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|