1
|
Jiwa NA, Ketang’enyi E, Nganyanyuka K, Mbwanji R, Mwenisongole D, Masuka E, Brown M, Charles M, Mwasomola DL, Nyangalima T, Olomi W, Komba L, Gwimile J, Kasambala B, Mwita L. Factors associated with the acceptability of Lopinavir/Ritonavir formulations among children living with HIV/AIDS attending care and treatment clinics in Mbeya and Mwanza, Tanzania. PLoS One 2024; 19:e0292424. [PMID: 38165867 PMCID: PMC10760675 DOI: 10.1371/journal.pone.0292424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/20/2023] [Indexed: 01/04/2024] Open
Abstract
INTRODUCTION Children living with chronic illnesses are offered formulations based on manufacturer and distributor research. The aim of this study is to better understand the perspectives of children and their caregivers in accepting Lopinavir/ritonavir (LPV/r) formulations. METHODS 362 participants were recruited from two pediatric HIV/AIDS clinics in Mbeya and Mwanza, Tanzania, from December 2021 to May 2022. A translated questionnaire was piloted and validated at both clinics, followed by the implementation of a cross-sectional study. RESULTS 169 participants (47.1%) reported general difficulties in swallowing, regardless of formulation, while 34.3% and 38.5% reported vomiting tablets and syrups, respectively. Statistical significance is shown to support that children can swallow medications if they can eat stiffened porridge (Ugali). This correlated with the lower incidence of younger children being able to swallow compared to older children (above six years of age). Children older than six years preferred taking tablets (independent of daily dosage) better than other formulations. Significantly, older children who attend school were associated with high odds of swallowing medicine (AOR = 3.06, 95%CI; 1.32-7.05); however, age was not found to be statistically related to ease of administration for Lopinavir/Ritonavir in this study. CONCLUSIONS Lopinavir/Ritonavir tablets remain the most accepted formulation among children and adolescents with HIV/AIDS. This study highlights the impact of various factors affecting the acceptability of pediatric formulation, suggesting that children younger than six years, unable to eat Ugali and not attending schools may be most vulnerable regarding their ability to accept Lopinavir/Ritonavir formulations. Further studies are needed to assess the acceptability of other medications in chronically ill children.
Collapse
Affiliation(s)
| | | | | | - Ruth Mbwanji
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| | | | - Eutropia Masuka
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| | - Mary Brown
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| | - Mary Charles
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| | | | | | - Willyhelmina Olomi
- National Institute of Medical Research (NIMR)- Mbeya Medical Research Center (MMRC), Mbeya, Tanzania
| | - Lilian Komba
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| | - Judith Gwimile
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| | - Bertha Kasambala
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| | - Lumumba Mwita
- Baylor College of Medicine Children’s Foundation, Mwanza, Tanzania
| |
Collapse
|
2
|
Persaud D, Bryson Y, Nelson BS, Tierney C, Cotton MF, Coletti A, Jao J, Spector SA, Mirochnick M, Capparelli EV, Costello D, Szewczyk J, Nicodimus N, Stranix-Chibanda L, Kekitiinwa AR, Korutaro V, Reding C, Carrington MN, Majji S, Yin DE, Jean-Philippe P, Chadwick EG. HIV-1 reservoir size after neonatal antiretroviral therapy and the potential to evaluate antiretroviral-therapy-free remission (IMPAACT P1115): a phase 1/2 proof-of-concept study. Lancet HIV 2024; 11:e20-e30. [PMID: 38061376 PMCID: PMC11094801 DOI: 10.1016/s2352-3018(23)00236-9] [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: 04/20/2023] [Revised: 08/23/2023] [Accepted: 09/11/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Infants born with HIV-1 require lifelong antiretroviral therapy (ART). We aimed to assess whether very early ART in neonates might restrict HIV-1 reservoirs, an important step towards ART-free remission. METHODS IMPAACT P1115 is an ongoing, phase 1/2, proof-of-concept study in which infants were enrolled at 30 research clinics in 11 countries (Brazil, Haiti, Kenya, Malawi, South Africa, Tanzania, Thailand, Uganda, the USA, Zambia, and Zimbabwe) into two cohorts. Infants at least 34 weeks' gestational age at high risk for in-utero HIV-1 with either untreated maternal HIV-1 (cohort 1) or who were receiving pre-emptive triple antiretroviral prophylaxis outside of the study (maternal ART permissible; cohort 2) were included. All infants initiated treatment within 48 h of life. Cohort 1 initiated three-drug nevirapine-based ART, and cohort 2 initiated three-drug nevirapine-based prophylaxis then three-drug nevirapine-based ART following HIV diagnosis by age 10 days. We added twice-daily coformulated oral ritonavir 75 mg/m2 and lopinavir 300 mg/m2 from 14 days of life and 42 weeks postmenstrual age. We discontinued nevirapine 12 weeks after two consecutive plasma HIV-1 RNA levels below limit of detection. We tracked virological suppression, safety outcomes, and meeting a predetermined biomarker profile at age 2 years (undetectable RNA since week 48, HIV-1 antibody-negative, HIV-1 DNA not detected, and normal CD4 count and CD4 percentage) to assess qualification for analytical treatment interruption. This study is registered with ClinicalTrials.gov, NCT02140255. FINDINGS Between Jan 23, 2015, and Dec 14, 2017, 440 infants were included in cohort 1 and 20 were included in cohort 2. 54 of these infants (34 from cohort 1 and 20 from cohort 2) had confirmed in-utero HIV-1 and were enrolled to receive study ART. 33 (61%) of 54 infants were female and 21 (39%) were male. The estimated probability of maintaining undetectable plasma RNA through to 2 years was 33% (95% CI 17-49) in cohort 1 and 57% (28-78) in cohort 2. Among infants maintaining protocol-defined virological control criteria through to study week 108, seven of 11 (64%, 95% CI 31-89) in cohort 1 and five of seven (71%, 29-96) in cohort 2 had no detected HIV-1 DNA. Ten of 12 (83%, 52-100) in cohort 1 and all seven (100%, 59-100) in cohort 2 tested HIV-1 antibody-negative at week 108. Among 54 infants initiated on very early ART, ten (19%; six in cohort 1 and four in cohort 2) met all criteria for possible analytical treatment interruption. Reversible grade 3 or 4 adverse events occurred in 15 (44%) of 34 infants in cohort 1 and seven (35%) of 20 infants in cohort 2. INTERPRETATION Very early ART for in-utero HIV-1 can achieve sustained virological suppression in association with biomarkers indicating restricted HIV-1 reservoirs by age 2 years, which might enable potential ART-free remission. FUNDING National Institute of Allergy and Infectious Diseases, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, and the National Institute of Mental Health.
Collapse
Affiliation(s)
- Deborah Persaud
- Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yvonne Bryson
- University of California Los Angeles, Los Angeles, CA, USA
| | - Bryan S Nelson
- Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Camlin Tierney
- Harvard T H Chan School of Public Health, Boston, MA, USA
| | | | | | - Jennifer Jao
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stephen A Spector
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | | | | | - Diane Costello
- University of California Los Angeles, Los Angeles, CA, USA
| | - Joseph Szewczyk
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicol Nicodimus
- University of Zimbabwe, Clinical Trials Research Centre, Harare, Zimbabwe
| | | | | | - Violet Korutaro
- Baylor College of Medicine Children's Foundation, Kampala, Uganda
| | - Christina Reding
- Frontier Science and Technology Research Foundation, Amherst, NY, USA
| | - Mary N Carrington
- Frederick National Laboratory for Cancer Research and Laboratory of Integrative Cancer Immunology National Cancer Institute, MD, USA
| | - Sai Majji
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Dwight E Yin
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patrick Jean-Philippe
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ellen G Chadwick
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
3
|
Chabala C, Turkova A, Kapasa M, LeBeau K, Tembo CH, Zimba K, Weisner L, Zyambo K, Choo L, Chungu C, Lungu J, Mulenga V, Crook A, Gibb D, McIlleron H. Inadequate Lopinavir Concentrations With Modified 8-Hourly Lopinavir/Ritonavir 4:1 Dosing During Rifampicin-based Tuberculosis Treatment in Children Living With HIV. Pediatr Infect Dis J 2023; 42:899-904. [PMID: 37506295 PMCID: PMC10501348 DOI: 10.1097/inf.0000000000004047] [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: 06/25/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Lopinavir/ritonavir plasma concentrations are profoundly reduced when co-administered with rifampicin. Super-boosting of lopinavir/ritonavir is limited by nonavailability of single-entity ritonavir, while double-dosing of co-formulated lopinavir/ritonavir given twice-daily produces suboptimal lopinavir concentrations in young children. We evaluated whether increased daily dosing with modified 8-hourly lopinavir/ritonavir 4:1 would maintain therapeutic plasma concentrations of lopinavir in children living with HIV receiving rifampicin-based antituberculosis treatment. METHODS Children with HIV/tuberculosis coinfection weighing 3.0 to 19.9 kg, on rifampicin-based antituberculosis treatment were commenced or switched to 8-hourly liquid lopinavir/ritonavir 4:1 with increased daily dosing using weight-band dosing approach. A standard twice-daily dosing of lopinavir/ritonavir was resumed 2 weeks after completing antituberculosis treatment. Plasma sampling was conducted during and 4 weeks after completing antituberculosis treatment. RESULTS Of 20 children enrolled; 15, 1-7 years old, had pharmacokinetics sampling available for analysis. Lopinavir concentrations (median [range]) on 8-hourly lopinavir/ritonavir co-administered with rifampicin (n = 15; area under the curve 0-24 55.32 mg/h/L [0.30-398.7 mg/h/L]; C max 3.04 mg/L [0.03-18.6 mg/L]; C 8hr 0.90 mg/L [0.01-13.7 mg/L]) were lower than on standard dosing without rifampicin (n = 12; area under the curve 24 121.63 mg/h/L [2.56-487.3 mg/h/L]; C max 9.45 mg/L [0.39-26.4 mg/L]; C 12hr 3.03 mg/L [0.01-17.7 mg/L]). During and after rifampicin cotreatment, only 7 of 15 (44.7%) and 8 of 12 (66.7%) children, respectively, achieved targeted pre-dose lopinavir concentrations ≥1mg/L. CONCLUSIONS Modified 8-hourly dosing of lopinavir/ritonavir failed to achieve adequate lopinavir concentrations with concurrent antituberculosis treatment. The subtherapeutic lopinavir exposures on standard dosing after antituberculosis treatment are of concern and requires further evaluation.
Collapse
Affiliation(s)
- Chishala Chabala
- From the Department of Paediatrics, University of Zambia, School of Medicine, Lusaka, Zambia
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Anna Turkova
- Medical Research Council–Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, United Kingdom
| | - Monica Kapasa
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Kristen LeBeau
- Medical Research Council–Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, United Kingdom
| | - Chimuka H. Tembo
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Kevin Zimba
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Lubbe Weisner
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
| | - Khozya Zyambo
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Louise Choo
- Medical Research Council–Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, United Kingdom
| | - Chalilwe Chungu
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Joyce Lungu
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Veronica Mulenga
- University Teaching Hospital-Children’s Hospital, Lusaka, Zambia
| | - Angela Crook
- Medical Research Council–Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, United Kingdom
| | - Diana Gibb
- Medical Research Council–Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, United Kingdom
| | - Helen McIlleron
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Faculty of Health Sciences, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
4
|
Salerno SN, Capparelli EV, McIlleron H, Gerhart JG, Dumond JB, Kashuba AD, Denti P, Gonzalez D. Leveraging physiologically based pharmacokinetic modeling to optimize dosing for lopinavir/ritonavir with rifampin in pediatric patients. Pharmacotherapy 2023; 43:638-649. [PMID: 35607886 PMCID: PMC9684348 DOI: 10.1002/phar.2703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/28/2022] [Indexed: 11/11/2022]
Abstract
STUDY OBJECTIVE Treatment of HIV and tuberculosis co-infection leads to significant mortality in pediatric patients, and treatment can be challenging due to the clinically significant drug-drug interaction (DDI) between lopinavir/ritonavir (LPV/RTV) and rifampin. Doubling LPV/RTV results in insufficient lopinavir trough concentrations in pediatric patients. The objective of this study was to leverage physiologically based pharmacokinetic (PBPK) modeling to optimize the adjusted doses of LPV/RTV in children receiving the WHO-revised doses of rifampin (15 mg/kg daily). DESIGN Adult and pediatric PBPK models for LPV/RTV with rifampin were developed, including CYP3A and P-glycoprotein inhibition and induction. SETTING (OR DATA SOURCE) Data for LPV/RTV model development and evaluation were available from the pediatric AIDS Clinical Trials Group. PATIENTS Dosing simulations were next performed to optimize dosing in children (2 months to 8 years of age). INTERVENTION Exposure following super-boosted LPV/RTV with 10 and 15 mg/kg PO daily rifampin was simulated. MEASUREMENTS AND MAIN RESULTS Simulated parameters were within twofold observations for LPV, RTV, and rifampin in adults and children ≥2 weeks old. The model predicted that, in healthy adults receiving 400/100 mg oral LPV/RTV twice daily (BID), co-treatment with 600 mg oral rifampin daily decreased the steady-state area under the concentration vs. time curve of LPV by 79%, in line with the observed change of 75%. Simulated and observed concentration profiles were comparable for LPV/RTV (230/57.5 mg/m2 ) PO BID without rifampin and 230/230 mg/m2 LPV/RTV PO BID with 10 mg/kg PO daily rifampin in pediatric patients. Sixteen mg/kg of super-boosted LPV (LPV/RTV 1:1) PO BID with 15 mg/kg PO daily rifampin achieved simulated LPV troughs >1 mg/L in ≥93% of virtual children weighing 3.0-24.9 kg, which was comparable with 10 mg/kg PO daily rifampin. CONCLUSIONS Super-boosted LPV/RTV with 15 mg/kg rifampin achieves therapeutic LPV troughs in HIV/TB-infected simulated children.
Collapse
Affiliation(s)
- Sara N. Salerno
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edmund V. Capparelli
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Helen McIlleron
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Jacqueline G. Gerhart
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Julie B. Dumond
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angela D.M. Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paolo Denti
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Svedmyr A, Hack H, Anderson BJ. Interactions of the protease inhibitor, ritonavir, with common anesthesia drugs. Paediatr Anaesth 2022; 32:1091-1099. [PMID: 35842922 PMCID: PMC9543968 DOI: 10.1111/pan.14529] [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: 04/01/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/27/2022]
Abstract
The protease inhibitor, ritonavir, is a strong inhibitor of CYP 3A. The drug is used for management of the human immunovirus and is currently part of an oral antiviral drug combination (nirmatrelvir-ritonavir) for the early treatment of SARS-2 COVID-19-positive patients aged 12 years and over who have recognized comorbidities. The CYP 3A enzyme system is responsible for clearance of numerous drugs used in anesthesia (e.g., alfentanil, fentanyl, methadone, rocuronium, bupivacaine, midazolam, ketamine). Ritonavir will have an impact on drug clearances that are dependent on ritonavir concentration, anesthesia drug intrinsic hepatic clearance, metabolic pathways, concentration-response relationship, and route of administration. Drugs with a steep concentration-response relationship (ketamine, midazolam, rocuronium) are mostly affected because small changes in concentration have major changes in effect response. An increase in midazolam concentration is observed after oral administration because CYP 3A in the gastrointestinal wall is inhibited, causing a large increase in relative bioavailability. Fentanyl infusion may be associated with a modest increase in plasma concentration and effect, but the large between subject variability of pharmacokinetic and pharmacodynamic concentration changes suggests it will have little impact on an individual patient, especially when used with adverse effect monitoring. It has been proposed that drugs that have no or only a small metabolic pathway involving the CYP 3A enzyme be used during anesthesia, for example, propofol, atracurium, remifentanil, and the volatile agents. That anesthesia approach denies children of drugs with considerable value. It is better that the inhibitory changes in clearance of these drugs are understood so that rational drug choices can be made to tailor drug use to the individual patient. Altered drug dose, anticipation of duration of effect, timing of administration, use of reversal agents and perioperative monitoring would better behoove children undergoing anesthesia.
Collapse
Affiliation(s)
- Anders Svedmyr
- Dept AnaesthesiaStarship Children's HospitalAucklandNew Zealand
| | - Henrik Hack
- Dept AnaesthesiaStarship Children's HospitalAucklandNew Zealand
| | | |
Collapse
|
6
|
Ya S, Ding W, Li S, Du K, Zhang Y, Li C, Liu J, Li F, Li P, Luo T, He L, Xu A, Gao D, Qiu B. On-Chip Construction of Liver Lobules with Self-Assembled Perfusable Hepatic Sinusoid Networks. ACS APPLIED MATERIALS & INTERFACES 2021; 13:32640-32652. [PMID: 34225454 DOI: 10.1021/acsami.1c00794] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Although various liver chips have been developed using emerging organ-on-a-chip techniques, it remains an enormous challenge to replicate the liver lobules with self-assembled perfusable hepatic sinusoid networks. Herein we develop a lifelike bionic liver lobule chip (LLC), on which the perfusable hepatic sinusoid networks are achieved using a microflow-guided angiogenesis methodology; additionally, during and after self-assembly, oxygen concentration is regulated to mimic physiologically dissolved levels supplied by actual hepatic arterioles and venules. This liver lobule design thereby produces more bionic liver microstructures, higher metabolic abilities, and longer lasting hepatocyte function than other liver-on-a-chip techniques that are able to deliver. We found that the flow through the unique micropillar design in the cell coculture zone guides the radiating assembly of the hepatic sinusoid, the oxygen concentration affects the morphology of the sinusoid by proliferation, and the oxygen gradient plays a key role in prolonging hepatocyte function. The expected breadth of applications our LLC is suited to is demonstrated by means of preliminarily testing chronic and acute hepatotoxicity of drugs and replicating growth of tumors in situ. This work provides new insights into designing more extensive bionic vascularized liver chips, while achieving longer lasting ex-vivo hepatocyte function.
Collapse
Affiliation(s)
- Shengnan Ya
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weiping Ding
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Lab for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shibo Li
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Kun Du
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yuanyuan Zhang
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Chengpan Li
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jing Liu
- School of Biology, Food and Environment Engineering, Hefei University, Hefei, Anhui 230601, China
| | - Fenfen Li
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Lab for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ping Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Tianzhi Luo
- School of Engineering Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Liqun He
- School of Engineering Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ao Xu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Bensheng Qiu
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Lab for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
7
|
Yang C, Huang Y, Liu S. Therapeutic Development in COVID-19. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1318:435-448. [PMID: 33973193 DOI: 10.1007/978-3-030-63761-3_25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19) caused by the SARS-CoV-2, the disease has spread rapidly worldwide and developed into a global pandemic, causing a significant impact on the global health system and economic development. Scientists have been racing to find effective drugs and vaccines for the treatment and prevention of COVID-19. However, due to the diversity of clinical manifestations caused by COVID-19, no standard antiviral regimen beyond supportive therapy has been established. Ongoing clinical trials are underway to evaluate the efficacy of drugs that primarily act on the viral replication cycle or enhanced immunity of patients. This chapter will summarize the currently used antiviral and adjuvant therapies in clinical practice and provide a theoretical basis for the future treatment of COVID-19.
Collapse
Affiliation(s)
- Chan Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yuan Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Guangzhou, China.
| |
Collapse
|
8
|
von Hentig N, Angioni C, Königs C. Determination of lopinavir/ritonavir concentrations in four different oral solutions for the application of antiretroviral therapy in very young, HIV-1-infected children. South Afr J HIV Med 2021; 22:1222. [PMID: 34192069 PMCID: PMC8182455 DOI: 10.4102/sajhivmed.v22i1.1222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/30/2021] [Indexed: 12/03/2022] Open
Affiliation(s)
- Nils von Hentig
- Internal Medicine II, HIVCENTER, Goethe University Hospital, Frankfurt, Germany
| | - Carlo Angioni
- Institute of Clinical Pharmacology, Goethe University Hospital, Frankfurt, Germany
| | - Christoph Königs
- Department of Pediatrics and Adolescent Medicine, Goethe University Hospital, Frankfurt, Germany
| |
Collapse
|
9
|
Owor M, Tierney C, Ziemba L, Browning R, Moye J, Graham B, Reding C, Costello D, Norman J, Wiesner L, Hughes E, Whalen ME, Purdue L, Mmbaga BT, Kamthunzi P, Kawalazira R, Nathoo K, Bradford S, Coletti A, Aweeka F, Musoke P. Pharmacokinetics and Safety of Zidovudine, Lamivudine, and Lopinavir/Ritonavir in HIV-infected Children With Severe Acute Malnutrition in Sub-Saharan Africa: IMPAACT Protocol P1092. Pediatr Infect Dis J 2021; 40:446-452. [PMID: 33464021 PMCID: PMC8043511 DOI: 10.1097/inf.0000000000003055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Severe acute malnutrition (SAM) may alter the pharmacokinetics (PK), efficacy, and safety of antiretroviral therapy. The phase IV study, IMPAACT P1092, compared PK, safety, and tolerability of zidovudine (ZDV), lamivudine (3TC), and lopinavir/ritonavir (LPV/r) in children with and without SAM. MATERIALS AND METHODS Children living with HIV 6 to <36 months of age with or without World Health Organization (WHO)-defined SAM received ZDV, 3TC, and LPV/r syrup for 48 weeks according to WHO weight band dosing. Intensive PK sampling was performed at weeks 1, 12, and 24. Plasma drug concentrations were measured using liquid chromatography tandem mass spectrometry. Steady-state mean area under the curve (AUC0-12h) and clearance (CL/F) for each drug were compared. Grade ≥3 adverse events were compared between cohorts. RESULTS Fifty-two children were enrolled across 5 sites in Africa with 44% (23/52) female, median age 19 months (Q1, Q3: 13, 25). Twenty-five children had SAM with entry median weight-for-height Z-score (WHZ) -3.4 (IQR -4.0, -3.0) and 27 non-SAM had median WHZ -1.0 (IQR -1.8, -0.1). No significant differences in mean AUC0-12h or CL/F were observed (P ≥ 0.09) except for lower 3TC AUC0-12h (GMR, 0.60; 95% CI, 0.4-1.0; P = 0.047) at week 12, higher ZDV AUC0-12h (GMR, 1.52; 1.2-2.0; P = 0.003) at week 24 in the SAM cohort compared with non-SAM cohort. Treatment-related grade ≥3 events did not differ significantly between cohorts (24.0% vs. 25.9%). CONCLUSION PK and safety findings for ZDV, 3TC, and LPV/r support current WHO weight band dosing of syrup formulations in children with SAM.
Collapse
Affiliation(s)
- Maxensia Owor
- From the Makerere University—Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - Camlin Tierney
- Department of Biostatistics, Center for Biostatistics in AIDS Research, Harvard T.H Chan School of Public Health, Boston, MA
| | - Lauren Ziemba
- Department of Biostatistics, Center for Biostatistics in AIDS Research, Harvard T.H Chan School of Public Health, Boston, MA
| | - Renee Browning
- National Institute of Allergy and Infectious Diseases, Bethesda, MD
| | | | | | | | - Diane Costello
- IMPAACT Laboratory Center, University of California, Los Angeles, CA
| | - Jennifer Norman
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Emma Hughes
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA
| | - Meghan E. Whalen
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA
| | | | - Blandina Theophil Mmbaga
- Kilimanjaro Clinical Research Institute—Kilimanjaro Christian Medical Center and Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | | | - Rachel Kawalazira
- College of Medicine, Johns Hopkins Research Project, Blantyre, Malawi
| | - Kusum Nathoo
- College of Health Sciences, University of Zimbabwe, Avondale, Harare, Zimbabwe
| | | | | | - Francesca. Aweeka
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA
| | - Philippa Musoke
- From the Makerere University—Johns Hopkins University Research Collaboration, Kampala, Uganda
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
10
|
Waalewijn H, Turkova A, Rakhmanina N, Cressey TR, Penazzato M, Colbers A, Burger DM. Optimizing Pediatric Dosing Recommendations and Treatment Management of Antiretroviral Drugs Using Therapeutic Drug Monitoring Data in Children Living With HIV. Ther Drug Monit 2019; 41:431-443. [PMID: 31008997 PMCID: PMC6636807 DOI: 10.1097/ftd.0000000000000637] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/03/2019] [Indexed: 12/27/2022]
Abstract
INTRODUCTION This review summarizes the current dosing recommendations for antiretroviral (ARV) drugs in the international pediatric guidelines of the World Health Organization (WHO), US Department of Health and Human Services (DHHS), and Pediatric European Network for Treatment of AIDS (PENTA), and evaluates the research that informed these approaches. We further explore the role of data generated through therapeutic drug monitoring in optimizing the dosing of ARVs in children. METHODS A PubMed search was conducted for the literature on ARV dosing published in English. In addition, the registration documentation of European Medicines Agency and the US Food and Drug Administration for currently used ARVs and studies referenced by the WHO, DHHS, and EMA guidelines were screened. Resulting publications were screened for papers containing data on the area under the concentration-time curve, trough concentration, and peak concentration. Studies with enrolled participants with a median or mean age of ≥18 years were excluded. No restriction on publishing date was applied. DISCUSSION AND CONCLUSION Pediatric ARV dosing is frequently based on data obtained from small studies and is often simplified to facilitate dosing in the context of a public health approach. Pharmacokinetic parameters of pediatric ARVs are subject to high interpatient variation and this leads to a potential risk of underdosing or overdosing when drugs are used in real life. To ensure optimal use of ARVs and validate dosing recommendations for children, it is essential to monitor ARV dosing more thoroughly with larger sample sizes and to include diverse subpopulations. Therapeutic drug monitoring data generated in children, where available and affordable, have the potential to enhance our understanding of the appropriateness of simplified pediatric dosing strategies recommended using a public health approach and to uncover suboptimal dosing or other unanticipated issues postmarketing, further facilitating the ultimate goal of optimizing pediatric ARV treatment.
Collapse
Affiliation(s)
- Hylke Waalewijn
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Anna Turkova
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London
- Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Natella Rakhmanina
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University
- Division of Infectious Diseases, Children's National Medical Center
- Elizabeth Glaser Pediatric AIDS Foundation, Washington, District of Columbia
| | - Tim R. Cressey
- PHPT/IRD UMI 174, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Immunology and Infectious Diseases, Harvard T.H Chan School of Public Health, Boston, Massachusetts
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom; and
| | - Martina Penazzato
- Treatment and Care, Department of HIV/AIDS, World Health Organization, Geneva, Switzerland
| | - Angela Colbers
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, the Netherlands
| | - David M. Burger
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
11
|
Whalen ME, Kajubi R, Chamankhah N, Huang L, Orukan F, Wallender E, Kamya MR, Dorsey G, Jagannathan P, Rosenthal PJ, Mwebaza N, Aweeka FT. Reduced Exposure to Piperaquine, Compared to Adults, in Young Children Receiving Dihydroartemisinin-Piperaquine as Malaria Chemoprevention. Clin Pharmacol Ther 2019; 106:1310-1318. [PMID: 31173649 DOI: 10.1002/cpt.1534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/07/2019] [Indexed: 11/10/2022]
Abstract
Dihydroartemisinin (DHA)-piperaquine is being evaluated as intermittent preventive therapy for malaria, but dosing has not been optimized for children. We assessed exposure to DHA and piperaquine in Ugandan children at two ages during infancy. Intensive sampling was performed in 32 children at 32 weeks of age, 31 children at 104 weeks, and 30 female adult controls. Compared with adults, DHA area under the concentration-time curve (AUC0-8 hr ) was 52% higher at 32 weeks and comparable at 104 weeks. Compared with adults, piperaquine AUC0-21 d was 35% lower at 32 weeks and 53% lower at 104 weeks. Terminal piperaquine concentrations on days 7, 14, and 21 were lower in children compared with adults and lower at 104 compared with 32 weeks. Piperaquine exposure was lower in young children compared with adults, and lower at 104 compared with 32 weeks of age, suggesting a need for age-based DHA-piperaquine dose optimization for chemoprevention.
Collapse
Affiliation(s)
- Meghan E Whalen
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Richard Kajubi
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda.,Department of Pharmacology and Therapeutics, Makerere University College of Health Sciences, Kampala, Uganda
| | - Nona Chamankhah
- Department of Pharmacy, Rady Children's Hospital, San Diego, California, USA
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Francis Orukan
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Erika Wallender
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Moses R Kamya
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | | | - Philip J Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| | - Norah Mwebaza
- Infectious Disease Research Collaboration, Makerere University College of Health Sciences, Kampala, Uganda.,Department of Pharmacology and Therapeutics, Makerere University College of Health Sciences, Kampala, Uganda
| | - Francesca T Aweeka
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco General Hospital, San Francisco, California, USA
| |
Collapse
|