1
|
Bekker A, Yang J, Wang J, Cotton MF, Cababasay M, Wiesner L, Moye J, Browning R, Nakwa FL, Rabie H, Violari A, Mirochnick M, Cressey TR, Capparelli EV. Safety and Pharmacokinetics of Lopinavir/Ritonavir Oral Solution in Preterm and Term Infants Starting Before 3 Months of Age. Pediatr Infect Dis J 2024; 43:355-360. [PMID: 38190642 PMCID: PMC10939833 DOI: 10.1097/inf.0000000000004243] [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] [Indexed: 01/10/2024]
Abstract
BACKGROUND Study of liquid lopinavir/ritonavir (LPV/r) in young infants has been limited by concerns for its safety in neonates. METHODS International Maternal Pediatric Adolescent AIDS Clinical Trials Network P1106 was a phase IV, prospective, trial evaluating the safety and pharmacokinetics of antiretroviral medications administered according to local guidelines to South African preterm and term infants <3 months of age. Safety evaluation through 24-week follow-up included clinical, cardiac and laboratory assessments. Pharmacokinetic data from P1106 were combined with data from International Maternal Pediatric Adolescent AIDS Clinical Trials Network studies P1030 and P1083 in a population pharmacokinetics model used to simulate LPV exposures with a weight-band dosing regimen in infants through age 6 months. RESULTS Safety and pharmacokinetics results were similar in 13/28 (46%) infants initiating LPV/r <42 weeks postmenstrual age (PMA) and in those starting ≥42 weeks PMA. LPV/r was started at a median (range) age of 47 (13-121) days. No grade 3 or higher adverse events were considered treatment related. Modeling and simulation predicted that for infants with gestational age ≥27 weeks who receive the weight-band dosing regimen, 82.6% will achieve LPV trough concentration above the target trough concentration of 1.0 µg/mL and 56.6% would exceed the observed adult lower limit of LPV exposure of 55.9 µg·h/mL through age 6 months. CONCLUSIONS LPV/r oral solution was safely initiated in a relatively small sample size of infants ≥34 weeks PMA and >2 weeks of life. No serious drug-related safety signal was observed; however, adrenal function assessments were not performed. Weight-band dosing regimen in infants with gestational age ≥27 weeks is predicted to result in LPV exposures equivalent to those observed in other pediatric studies.
Collapse
Affiliation(s)
- Adrie Bekker
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Jincheng Yang
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, USA
- Clinical Pharmacology and Quantitative pharmacology, CPSS, AstraZeneca R&D, Waltham, MA, USA
| | - Jiajia Wang
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mark F. Cotton
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Mae Cababasay
- Clinical Pharmacology and Quantitative pharmacology, CPSS, AstraZeneca R&D, Waltham, MA, USA
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, University of Cape Town, Cape Town, South Africa
| | - Jack Moye
- Division of Extramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development
| | - Renee Browning
- Division of AIDS, National Institute of Allergy and Infectious Diseases
| | - Firdose L. Nakwa
- Department of Pediatrics and Child Health, Faculty of Health Sciences, School of Clinical Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Helena Rabie
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Avy Violari
- Perinatal HIV research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Mark Mirochnick
- Division of Neonatology, Department of Pediatrics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Tim. R Cressey
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Molecular & Clinical Pharmacology, University of Liverpool, UK
| | - Edmund V. Capparelli
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, USA
| |
Collapse
|
2
|
Bekker A, Salvadori N, Rabie H, du Toit S, Than-In-At K, Groenewald M, Cressey R, Nielsen J, Capparelli EV, Lallemant M, Cotton MF, Cressey TR. Paediatric abacavir-lamivudine fixed-dose dispersible tablets and ritonavir-boosted lopinavir granules in neonates exposed to HIV (PETITE study): an open-label, two-stage, single-arm, phase 1/2, pharmacokinetic and safety trial. Lancet HIV 2024; 11:e86-e95. [PMID: 38296364 DOI: 10.1016/s2352-3018(23)00289-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Existing solid antiretroviral fixed-dose combination formulations are preferred over liquid formulations in children, but their suitability for neonates is unknown. We evaluated the pharmacokinetics and safety of paediatric abacavir-lamivudine fixed-dose dispersible tablets and ritonavir-boosted lopinavir granules in neonates. METHODS In this open-label, two-stage, single-arm, phase 1/2, pharmacokinetic and safety trial, generic abacavir- lamivudine (120:60 mg) double-scored dispersible tablets and lopinavir boosted with ritonavir (40:10 mg) granules were studied. Neonates exposed to HIV (≥37 weeks gestational age) of no more than 3 days of age with birthweights of 2000-4000 g were identified through routine care in a tertiary hospital in Cape Town, South Africa. In stage 1, the pharmacokinetics and safety of two single doses were assessed to select the multidose strategy for stage 2. Neonates received a single dose of abacavir-lamivudine (30:15 mg, a quarter of a tablet) and lopinavir boosted with ritonavir (40:10 mg - one sachet) orally between 3 days and 14 days of age, and a second dose of a quarter tablet of abacavir-lamivudine and lopinavir boosted with ritonavir (80:20 mg, two sachets) 10-14 days later in stage 1. The multidose strategy selected in stage 2 was a quarter of the abacavir-lamivudine (30:15 mg) fixed-dose dispersible tablet once per day and two sachets of the lopinavir boosted with ritonavir (80:20 mg) granules twice per day from birth to age 28 days. In both stages two intensive pharmacokinetic visits were done, one at less than 14 days of life (pharmacokinetics 1) and another 10-14 days later (pharmacokinetics 2). Safety visits were done 1-2 weeks after each pharmacokinetic visit. Primary objectives were to assess pharmacokinetics and safety of abacavir, lamivudine, and lopinavir. Pharmacokinetic endpoints were area under the concentration time curve (AUC), maximum concentration, and concentration at end of dosing interval in all participants with at least one evaluable pharmacokinetic visit. Safety endpoints included grade 3 or worse adverse events, and grade 3 or worse treatment-related adverse events, occurring between study drug initiation and end of study. This completed trial is registered with the Pan African Clinical Trials Registry (PACTR202007806554538). FINDINGS Between Aug 18, 2021, and Aug 18, 2022, 24 neonates were enrolled into the trial and received study drugs. Eight neonates completed stage 1, meeting interim pharmacokinetic and safety criteria. In stage 2, 16 neonates received study drugs. Geometric mean abacavir and lamivudine exposures (AUC0-24) were higher at 6-14 days (51·7 mg × h/L for abacavir and 17·2 mg × h/L for lamivudine) than at 19-24 days of age (25·0 mg × h/L and 11·3 mg × h/L), whereas they were similar for lopinavir over this period (AUC 0-12 58·5 mg × h/L vs 46·4 mg × h/L). Abacavir geometric mean AUC0-24 crossed the upper reference range at pharmacokinetics 1, but rapidly decreased. Lamivudine and lopinavir AUC0-tau were within range. No grade 2 or worse adverse events were related to study drugs. One neonate had a grade 1 prolonged corrected QT interval using the Fridericia method that spontaneously resolved. INTERPRETATION Abacavir-lamivudine dispersible tablets and ritonavir-boosted lopinavir granules in neonates were safe and provided drug exposures similar to those in young infants. Although further safety data are needed, this regimen presents a new option for HIV prevention and treatment from birth. Accelerating neonatal pharmacokinetic studies of novel antiretroviral therapies is essential for neonates to also benefit from state-of-the-art treatments. FUNDING Unitaid.
Collapse
Affiliation(s)
- Adrie Bekker
- Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Nicolas Salvadori
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand; Department of Statistics, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| | - Helena Rabie
- Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Samantha du Toit
- Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Kanchana Than-In-At
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Marisa Groenewald
- Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Ratchada Cressey
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - James Nielsen
- Hassenfield Children's Hospital at NYU Langone, New York, NY, USA
| | - Edmund V Capparelli
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Science, University of California San Diego, San Diego, CA, USA
| | - Marc Lallemant
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Mark F Cotton
- Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Tim R Cressey
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand; Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
3
|
Mohammed O, Alemayehu E, Bisetegn H, Tilahun M, Gedefie A, Ebrahim E, Fiseha M, Necho M, Fiseha T. Prevalence of hepatotoxicity among HIV-infected patients in Ethiopia: a systematic review and meta-analysis. BMC Infect Dis 2022; 22:826. [DOI: 10.1186/s12879-022-07838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Abstract
Background
Globally, the human immunodeficiency virus has been recognized as a major public health concern. The direct toxicity of antiretroviral medicines or their active metabolites causes liver cell destruction by different mechanisms, inducing immune-mediated inflammation, oxidative stress, and other mechanisms. On the other hand, the virus itself also produces hepatotoxicity. Therefore, this systematic review and meta-analysis aimed to assess the pooled prevalence of hepatotoxicity among HIV-infected patients in Ethiopia.
Methods
PubMed, Science Direct, Cochrane Library, Web of Science, and ResearchGate databases were used to find relevant articles. As well, various professional associations were searched to retrieve grey literature. The Newcastle–Ottawa Quality Assessment Scale was used to assess the quality of recruited studies. The data were extracted using Microsoft Excel, and the meta-analysis was carried out using STATA 14 software. I2 and Cochran’s Q test were employed to assess the presence of heterogeneity between studies. A random effect model was used. The funnel plot and Egger’s statistics were used to assess publication bias. Moreover, subgroup analysis and sensitivity analysis were also done.
Results
The pooled prevalence of hepatotoxicity among HIV patients in Ethiopia was 25.45% (95% CI = 20.06–30.84%). There was high heterogeneity, with an I2 value of 93.7%. Subgroup analysis by HAART status showed a higher pooled prevalence of hepatotoxicity among HIV patients taking HAART (23.63%) than among HAART naive patients (7.29%). In subgroup analysis, the pooled prevalence of hepatotoxicity among HIV/Tb co-infected and HIV mono-infected patients was 26.3% and 17.94%, respectively.
Conclusion
The current systematic review and meta-analysis showed a high prevalence of hepatotoxicity among HIV-infected patients. Therefore, regular monitoring of hepatotoxicity among HIV-infected patients is required in order to avoid liver damage and other complications.
Systematic review registration PROSPERO (2022:CRD42022334704)
Collapse
|
4
|
Tam J, Lau E, Read S, Bitnun A. Is Routine Therapeutic Drug Monitoring of Anti-Retroviral Agents Warranted in Children Living with HIV? J Pediatr Pharmacol Ther 2022; 27:551-557. [DOI: 10.5863/1551-6776-27.6.551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 11/29/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVE
The utility of routine therapeutic drug monitoring (TDM) in children living with HIV has not been extensively studied. The purpose of this study was to assess this strategy.
METHODS
This was a single-center, prospective observational study of routine TDM for protease inhibitors (PIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and integrase strand transfer inhibitors (INSTIs) in children living with HIV who were receiving antiretroviral therapy (ART) between February and December 2014. Outcome measures included the proportion of serum antiretroviral (ARV) medication concentrations in the therapeutic range (target values extrapolated from adult data) and the effect of serum concentrations on virologic control, medication adherence, and toxicity.
RESULTS
Forty-eight children with a median age of 13 years (interquartile range, 3–18) were included. Median viral load (VL) and CD4% were <40 copies/mL (range, <40–124) and 37.4% (range, 8.4–47.9), respectively. Adherence was considered excellent in 95.8% of patients. Of the 50 serum trough concentrations (PI n = 19 [38%]; NNRTI n = 27 [54%]; INSTI n = 4 [8%]), 66% (n = 33) were in the therapeutic range, 12% (n = 6) were subtherapeutic, and 22% (n = 11) were supratherapeutic. There was no statistically significant correlation between serum ARV concentrations and patient demographics, VL, CD4%, or adherence. No clinically significant adverse events were noted. One dose adjustment was made for a subtherapeutic serum raltegravir concentration, likely attributable to interaction with ritonavir.
CONCLUSIONS
This study does not support routine TDM in healthy children living with HIV who are well controlled on antiretroviral medication regimens. A more targeted strategy, such as when adherence is questioned or when there are suspected drug interactions, may be more appropriate.
Collapse
Affiliation(s)
- Jennifer Tam
- Division of Infectious Diseases, Department of Pediatrics (JT, SR, AB), The Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto (JT, EL, SR, AB), Toronto, ON, Canada
- Division of Infectious Diseases, Department of Pediatrics (JT), BC Children's Hospital, Vancouver, BC, Canada
| | - Elaine Lau
- University of Toronto (JT, EL, SR, AB), Toronto, ON, Canada
- Department of Pharmacy (EL), The Hospital for Sick Children, Toronto, ON, Canada
| | - Stanley Read
- Division of Infectious Diseases, Department of Pediatrics (JT, SR, AB), The Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto (JT, EL, SR, AB), Toronto, ON, Canada
| | - Ari Bitnun
- Division of Infectious Diseases, Department of Pediatrics (JT, SR, AB), The Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto (JT, EL, SR, AB), Toronto, ON, Canada
| |
Collapse
|
5
|
Bekker A, Rabie H, Salvadori N, du Toit S, Than-in-at K, Groenewald M, Andrieux-Meyer I, Kumar M, Cressey R, Nielsen J, Capparelli E, Lallemant M, Cotton MF, Cressey TR. Pharmacokinetics and Safety of the Abacavir/Lamivudine/Lopinavir/Ritonavir Fixed-Dose Granule Formulation (4-in-1) in Neonates: PETITE Study. J Acquir Immune Defic Syndr 2022; 89:324-331. [PMID: 34855626 PMCID: PMC8826609 DOI: 10.1097/qai.0000000000002871] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/15/2021] [Indexed: 12/03/2022]
Abstract
BACKGROUND Antiretroviral options for neonates (younger than 28 days) should be expanded. We evaluated the pharmacokinetics, safety, and acceptability of the "4-in-1" fixed-dose pediatric granule formulation of abacavir/lamivudine/lopinavir/ritonavir (30/15/40/10 mg) in neonates. METHODS The PETITE study is an ongoing phase I/II, open-label, single-arm, 2-stage trial conducted in South Africa. In stage 1, term neonates exposed to HIV on standard antiretroviral prophylaxis (nevirapine ± zidovudine) received single dose(s) of the 4-in-1 formulation, followed by intensive pharmacokinetic sampling and safety assessments. At each PK visit, blood was drawn after an observed dose at 1, 2, 4, 8, and 12 hours postdose. In this study, we have reported the planned interim pharmacokinetic and safety analysis after completion of the single-dose administration. RESULTS Sixteen neonates, with a median (range) birth weight of 3130 g (2790-3590 g), completed 24 pharmacokinetic visits. The 4-in-1 formulation imposed relatively high doses of abacavir [8.6 mg/kg (6.6-11.4)] and lamivudine [4.3 mg/kg (3.3-5.7)] but lower doses of lopinavir [11.5 mg/kg (8.8-15.2)]. The geometric means (GM, 90% CI) AUC0-12 of abacavir, lamivudine, and lopinavir were 29.87 (26.29-33.93), 12.61 (10.72-14.83), and 3.49 (2.13-5.72) µg.h/mL, respectively. Lopinavir GM AUC0-12 was below the predefined target (20-100 µg.h/mL), and ritonavir concentrations were only detectable in 4 of the 120 (3%) samples. No adverse events were related to study drugs. No neonate had difficulty swallowing the 4-in-1 formulation. CONCLUSIONS The high doses of abacavir and lamivudine (in mg/kg) and AUCs were safe, and the formulation was well tolerated; however, lopinavir/ritonavir exposures were extremely low, preventing its use in neonates use in neonates. Alternative pediatric solid antiretroviral formulations must be studied in neonates.
Collapse
Affiliation(s)
- Adrie Bekker
- Department of Pediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Helena Rabie
- Department of Pediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Nicolas Salvadori
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
| | - Samantha du Toit
- Department of Pediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Kanchana Than-in-at
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Marisa Groenewald
- Department of Pediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | | | | | - Ratchada Cressey
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - James Nielsen
- Hassenfield Children's Hospital at NYU Langone, New York, NY
| | | | - Marc Lallemant
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Mark F. Cotton
- Department of Pediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | - Tim R. Cressey
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
6
|
Rosebush JC, Best BM, Chadwick EG, Butler K, Moye J, Smith E, Bradford S, Reding CA, Mathiba SR, Hanley S, Aziz M, Homans J, Acosta EP, Murtaugh W, Vourvahis M, Mcfadyen L, Hayward K, Mirochnick M, Samson P. Pharmacokinetics and safety of maraviroc in neonates. AIDS 2021; 35:419-427. [PMID: 33252481 PMCID: PMC7856036 DOI: 10.1097/qad.0000000000002762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate safety and pharmacokinetics of maraviroc administered with standard antiretroviral prophylaxis to HIV-1 exposed infants and to determine the appropriate dose of maraviroc during the first 6 weeks of life. DESIGN A phase I, multicentre, open-label study enrolling two sequential cohorts. METHODS IMPAACT 2007 participants enrolled by day 3 of life and were stratified by exposure to maternal efavirenz. Cohort 1 participants received two single 8 mg/kg maraviroc doses 1 week apart with pharmacokinetic sampling after each dose. Cohort 2 participants received 8 mg/kg maraviroc twice daily through 6 weeks of life with pharmacokinetic sampling at weeks 1 and 4. Maraviroc exposure target was Cavg at least 75 ng/ml. Laboratory and clinical evaluations assessed safety. RESULTS Fifteen Cohort 1 and 32 Cohort 2 HIV-exposed neonates were enrolled (median gestational age 39 weeks, 51% male). All 13 evaluable Cohort 1 infants met the pharmacokinetic target. Median exposure for the 25 evaluable Cohort 2 infants met the pharmacokinetic target but variability was high, with 17-33% of infants below target at Weeks 1 and 4. Pharmacokinetic target achievement was similar between efavirenz exposure strata. No Grade 3+ toxicities, early study or treatment discontinuations due to maraviroc occurred. CONCLUSION Median maraviroc exposure met the Cavg target in neonates receiving 8 mg/kg twice daily, although exposures were variable. Maternal efavirenz use did not impact maraviroc exposure and no discontinuations were due to maraviroc toxicity/intolerance. No infants acquired HIV-1 infection during follow-up. Maraviroc 8 mg/kg twice daily appears well tolerated during the first 6 weeks of life.
Collapse
Affiliation(s)
| | - Brookie M Best
- University of California San Diego, La Jolla, California
| | - Ellen G Chadwick
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Kevin Butler
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - John Moye
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda
| | - Elizabeth Smith
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | | | - Christina A Reding
- Frontier Science & Technology Research Foundation, Inc., Amherst, New York, USA
| | | | - Sherika Hanley
- Centre for the AIDS Programme in South Africa, University of KwaZulu-Natal, Durban, South Africa
| | - Mariam Aziz
- Rush University Medical Center, Chicago, Illinois
| | - James Homans
- University of Southern California, Los Angeles, California
| | - Edward P Acosta
- Deparment of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama
| | - William Murtaugh
- IMPAACT Laboratory Center, Children's Hospital of Los Angeles, Los Angeles, California
| | - Manoli Vourvahis
- Clinical Pharmacology, Pfizer Global Research and Development, New York, New York, USA
| | - Lynn Mcfadyen
- Pharmacometrics, Pfizer Global Research and Development, Sandwich, UK
| | - Katy Hayward
- ViiV HealthCare, Research Triangle Park, North Carolina
| | | | - Pearl Samson
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Frontier Science & Technology Research Foundation, Inc., Amherst, New York, USA
| |
Collapse
|
7
|
Gilleece DY, Tariq DS, Bamford DA, Bhagani DS, Byrne DL, Clarke DE, Clayden MP, Lyall DH, Metcalfe DR, Palfreeman DA, Rubinstein DL, Sonecha MS, Thorley DL, Tookey DP, Tosswill MJ, Utting MD, Welch DS, Wright MA. British HIV Association guidelines for the management of HIV in pregnancy and postpartum 2018. HIV Med 2020; 20 Suppl 3:s2-s85. [PMID: 30869192 DOI: 10.1111/hiv.12720] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Dr Yvonne Gilleece
- Honorary Clinical Senior Lecturer and Consultant Physician in HIV and Genitourinary Medicine, Brighton and Sussex University Hospitals NHS Trust
| | - Dr Shema Tariq
- Postdoctoral Clinical Research Fellow, University College London, and Honorary Consultant Physician in HIV, Central and North West London NHS Foundation Trust
| | - Dr Alasdair Bamford
- Consultant in Paediatric Infectious Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London
| | - Dr Sanjay Bhagani
- Consultant Physician in Infectious Diseases, Royal Free Hospital NHS Trust, London
| | - Dr Laura Byrne
- Locum Consultant in HIV Medicine, St George's University Hospitals NHS Foundation Trust, London
| | - Dr Emily Clarke
- Consultant in Genitourinary Medicine, Royal Liverpool and Broadgreen University Hospitals NHS Trust
| | - Ms Polly Clayden
- UK Community Advisory Board representative/HIV treatment advocates network
| | - Dr Hermione Lyall
- Clinical Director for Children's Services and Consultant Paediatrician in Infectious Diseases, Imperial College Healthcare NHS Trust, London
| | | | - Dr Adrian Palfreeman
- Consultant in Genitourinary Medicine, University Hospitals of Leicester NHS Trust
| | - Dr Luciana Rubinstein
- Consultant in Genitourinary Medicine, London North West Healthcare University NHS Trust, London
| | - Ms Sonali Sonecha
- Lead Directorate Pharmacist HIV/GUM, Chelsea and Westminster Healthcare NHS Foundation Trust, London
| | | | - Dr Pat Tookey
- Honorary Senior Lecturer and Co-Investigator National Study of HIV in Pregnancy and Childhood, UCL Great Ormond Street Institute of Child Health, London
| | | | - Mr David Utting
- Consultant Obstetrician and Gynaecologist, Brighton and Sussex University Hospitals NHS Trust
| | - Dr Steven Welch
- Consultant in Paediatric Infectious Diseases, Heart of England NHS Foundation Trust, Birmingham
| | - Ms Alison Wright
- Consultant Obstetrician and Gynaecologist, Royal Free Hospitals NHS Foundation Trust, London
| |
Collapse
|
8
|
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
|
9
|
Yang J, Nikanjam M, Best BM, Pinto J, Chadwick EG, Daar ES, Havens PL, Rakhmanina N, Capparelli EV. Population Pharmacokinetics of Lopinavir/Ritonavir: Changes Across Formulations and Human Development From Infancy Through Adulthood. J Clin Pharmacol 2018; 58:1604-1617. [PMID: 30252146 PMCID: PMC6604805 DOI: 10.1002/jcph.1293] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/06/2018] [Indexed: 11/05/2022]
Abstract
Lopinavir/ritonavir (LPV/r) is recommended by the World Health Organization as first-line treatment for HIV-infected infants and young children. We performed a composite population pharmacokinetic (PK) analysis on LPV plasma concentration data from 6 pediatric and adult studies to determine maturation and formulation effects from infancy to adulthood. Intensive PK data were available for infants, children, adolescents, and adults (297 intensive profiles/1662 LPV concentrations). LPV PK data included 1 adult, 1 combined pediatric-adult, and 4 pediatric studies (age 6 weeks to 63 years) with 3 formulations (gel-capsule, liquid, melt-extrusion tablets). LPV concentrations were modeled using nonlinear mixed effects modeling (NONMEM v. 7.3; GloboMax, Hanover, Maryland) with a one compartment semiphysiologic model. LPV clearance was described by hepatic plasma flow (QHP ) times hepatic extraction (EH ), with EH estimated from the PK data. Volume was scaled by linear weight (WT/70)1.0 . Bioavailability was assessed separately as a function of hepatic extraction and the fraction absorbed from the gastrointestinal tract. The absorption component of bioavailability increased with age and tablet formulation. Monte Carlo simulations of the final model using current World Health Organization weight-band dosing recommendations demonstrated that participants younger than 6 months of age had a lower area under the drug concentration-time curve (94.8 vs >107.4 μg hr/mL) and minimum observed concentration of drug in blood plasma (5.0 vs > 7.1 μg/mL) values compared to older children and adults. Although World Health Organization dosing recommendations include a larger dosage (mg/m2 ) in infants to account for higher apparent clearance, they still result in low LPV concentrations in many infants younger than 6 months of age receiving the liquid formulation.
Collapse
Affiliation(s)
- Jincheng Yang
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, California
| | - Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, California
| | - Brookie M. Best
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, California
- Division of Host-Microbe Systems and Therapeutics, University of California San Diego School of Medicine, San Diego, California
| | - Jorge Pinto
- Division of Immunology, School of Medicine, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Ellen G. Chadwick
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Eric S. Daar
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Los Angeles, California
| | - Peter L. Havens
- Division of Pediatric Infectious Diseases, Medical College of Wisconsin and Children’s Hospital of Wisconsin, Milwauke, CA
| | - Natella Rakhmanina
- Division of Pediatric Infectious Diseases, Children’s National Medical Center; The George Washington University, Washington DC
- Elizabeth Glaser Pediatric AIDS Foundation, Washington DC
| | - Edmund V. Capparelli
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, California
- Division of Host-Microbe Systems and Therapeutics, University of California San Diego School of Medicine, San Diego, California
| |
Collapse
|
10
|
Mavigner M, Habib J, Deleage C, Rosen E, Mattingly C, Bricker K, Kashuba A, Amblard F, Schinazi RF, Lawson B, Vanderford TH, Jean S, Cohen J, McGary C, Paiardini M, Wood MP, Sodora DL, Silvestri G, Estes J, Chahroudi A. Simian Immunodeficiency Virus Persistence in Cellular and Anatomic Reservoirs in Antiretroviral Therapy-Suppressed Infant Rhesus Macaques. J Virol 2018; 92:e00562-18. [PMID: 29997216 PMCID: PMC6146711 DOI: 10.1128/jvi.00562-18] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/29/2018] [Indexed: 11/20/2022] Open
Abstract
Worldwide, nearly two million children are infected with human immunodeficiency virus (HIV), with breastfeeding accounting for the majority of contemporary HIV transmissions. Antiretroviral therapy (ART) has reduced HIV-related morbidity and mortality but is not curative. The main barrier to a cure is persistence of latent HIV in long-lived reservoirs. However, our understanding of the cellular and anatomic sources of the HIV reservoir during infancy and childhood is limited. Here, we developed a pediatric model of ART suppression in orally simian immunodeficiency virus (SIV)-infected rhesus macaque (RM) infants, with measurement of virus persistence in blood and tissues after 6 to 9 months of ART. Cross-sectional analyses were conducted to compare SIV RNA and DNA levels in adult and infant RMs naive to treatment and on ART. We demonstrate efficient viral suppression following ART initiation in SIV-infected RM infants with sustained undetectable plasma viral loads in the setting of heterogeneous penetration of ART into lymphoid and gastrointestinal tissues and low drug levels in the brain. We further show reduction in SIV RNA and DNA on ART in lymphoid tissues of both infant and adult RMs but stable (albeit low) levels of SIV RNA and DNA in the brains of viremic and ART-suppressed infants. Finally, we report a large contribution of naive CD4+ T cells to the total CD4 reservoir of SIV in blood and lymph nodes of ART-suppressed RM infants that differs from what we show in adults. These results reveal important aspects of HIV/SIV persistence in infants and provide insight into strategic targets for cure interventions in a pediatric population.IMPORTANCE While antiretroviral therapy (ART) can reduce HIV replication, the virus cannot be eradicated from an infected individual, and our incomplete understanding of HIV persistence in reservoirs greatly complicates the generation of a cure for HIV infection. Given the immaturity of the infant immune system, it is critically important to study HIV reservoirs specifically in this population. Here, we established a pediatric animal model to simulate breastfeeding transmission and study SIV reservoirs in rhesus macaque (RM) infants. Our study demonstrates that ART can be safely administered to infant RMs for prolonged periods and that it efficiently controls viral replication in this model. SIV persistence was shown in blood and tissues, with similar anatomic distributions of SIV reservoirs in infant and adult RMs. However, in the peripheral blood and lymph nodes, a greater contribution of the naive CD4+ T cells to the SIV reservoir was observed in infants than in adults.
Collapse
Affiliation(s)
- Maud Mavigner
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jakob Habib
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Elias Rosen
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Cameron Mattingly
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Katherine Bricker
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Angela Kashuba
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Franck Amblard
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Raymond F Schinazi
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Benton Lawson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Thomas H Vanderford
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sherrie Jean
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Joyce Cohen
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Colleen McGary
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Mirko Paiardini
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Matthew P Wood
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Donald L Sodora
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Jacob Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Ann Chahroudi
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Emory+Children's Center for Childhood Infections and Vaccines, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Schultheiß M, Kling S, Lenker U, von Bibra M, Rosenkranz B, Klinker H. Lopinavir serum concentrations of critically ill infants: a pharmacokinetic investigation in South Africa. Med Microbiol Immunol 2018; 207:339-343. [PMID: 29974233 DOI: 10.1007/s00430-018-0550-5] [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: 04/11/2018] [Accepted: 06/29/2018] [Indexed: 10/28/2022]
Abstract
The role of therapeutic drug monitoring in pediatric antiretroviral therapy is unclear. A little pharmacokinetic datum from clinical practice exists beyond controlled approval studies including clinically stable children. The aim of this study is to quantify LPV exposure of critically ill infants in an ICU and-by identifying risk factors for inadequate exposure-to define sensible indications for TDM in pediatric HIV care; in addition, assume total drug adherence in ICU to compare LPV exposure with a setting of unknown adherence. In this prospective investigation, 15 blood samples from critically ill infants in the pediatric ICU at Tygerberg Hospital were analyzed for LPV-serum concentrations. They were then compared to those of 22 blood samples from out-patient children. Serum-level measurements were performed with an established high-performance liquid chromatography method. All LPV-serum levels of ICU patients were higher than a recommended Ctrough (= 1.000 ng/ml), 60% of levels were higher than Cmax (8.200 ng/ml). Partly, serum levels reached were extremely high (Maximum: 28.778 ng/ml). Low bodyweight and age correlated significantly with high LPV concentrations and were risk factors for serum levels higher than Cmax. Significantly fewer serum levels from infants in ICU care (mean: 11.552 ng/ml ± SD 7760 ng/ml) than from out-patient children (mean: 6.756 ng/ml ± SD 6.003 ng/ml) were subtherapeutic (0 vs. 28%, p = 0.008). Under total adherence in the ICU group, there were no subtherapeutic serum levels, while, in out-patient, children with unknown adherence 28% of serum levels were found subtherapeutic. Low bodyweight and age are risk factors for reaching potentially toxic LPV levels in this extremely fragile population. TDM can be a reasonable tool to secure sufficient and safe drug exposure in pediatric cART.
Collapse
Affiliation(s)
- Michael Schultheiß
- Medizinische Klinik und Poliklinik II, Schwerpunkt Infektiologie, Universitätsklinikum Würzburg, ZIM Haus A3/A4, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.
| | - Sharon Kling
- Department of Pediatrics and Child Health, Ward A9, Tygerberg Hospital, University of Stellenbosch, Francie van Zijl Drive, Tygerberg, 7505, South Africa
| | - Ulrike Lenker
- Medizinische Klinik und Poliklinik II, Schwerpunkt Infektiologie, Universitätsklinikum Würzburg, ZIM Haus A3/A4, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Miriam von Bibra
- Medizinische Klinik und Poliklinik II, Schwerpunkt Infektiologie, Universitätsklinikum Würzburg, ZIM Haus A3/A4, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Bernd Rosenkranz
- Division of Clinical Pharmacology, Department of Medicine, Tygerberg Hospital, University of Stellenbosch, Francie van Zijl Drive, Tygerberg, 7505, South Africa
| | - Hartwig Klinker
- Medizinische Klinik und Poliklinik II, Schwerpunkt Infektiologie, Universitätsklinikum Würzburg, ZIM Haus A3/A4, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| |
Collapse
|
12
|
Shiau S, Abrams EJ, Arpadi SM, Kuhn L. Early antiretroviral therapy in HIV-infected infants: can it lead to HIV remission? Lancet HIV 2018; 5:e250-e258. [PMID: 29739699 PMCID: PMC7487171 DOI: 10.1016/s2352-3018(18)30012-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/19/2018] [Indexed: 12/27/2022]
Abstract
Interventions to prevent mother-to-child HIV transmission have been extremely successful, but new HIV infections continue to occur in infants. Strong evidence indicates that combination antiretroviral therapy (ART) for treatment should be started in HIV-infected infants to prevent early morbidity and mortality. In 2013, the report of the Mississippi baby, who was started on ART within 30 h of life and maintained off-treatment remission for 27 months before HIV was once again detectable, generated renewed interest in very early ART initiation. The case stimulated interest in the possibility of HIV remission, which we define as maintenance of plasma viraemia below the threshold of detection in the absence of ART, after early treatment initiation. The possibility of HIV remission elicits much hope, given that children with HIV infection currently face a lifetime of treatment. The potential for early ART to lead to HIV remission in infants can be thought of in terms of six factors: rapidity of viral suppression with very early ART; initial viral suppression rate with early ART; later virological control after early treatment; the effect of early treatment on the viral reservoir size; outcomes of randomised trials of structured treatment interruption; and the likelihood of viral rebound in neonates after ART cessation. Review of existing data suggests that achieving long-term remission off treatment remains elusive, and concentrated attention and commitment of the scientific community is needed to investigate the factors that might help to reach this goal.
Collapse
Affiliation(s)
- Stephanie Shiau
- Gertrude H. Sergievsky Center, Mailman School of Public Health, Columbia University, New York, NY, USA; College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA; Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Elaine J Abrams
- Department of Pediatrics, Mailman School of Public Health, Columbia University, New York, NY, USA; College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA; ICAP, Mailman School of Public Health, Columbia University, New York, NY, USA; Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephen M Arpadi
- Gertrude H. Sergievsky Center, Mailman School of Public Health, Columbia University, New York, NY, USA; Department of Pediatrics, Mailman School of Public Health, Columbia University, New York, NY, USA; College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA; ICAP, Mailman School of Public Health, Columbia University, New York, NY, USA; Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, Mailman School of Public Health, Columbia University, New York, NY, USA; College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA; Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
13
|
Beghin JC, Yombi JC, Ruelle J, Van der Linden D. Moving forward with treatment options for HIV-infected children. Expert Opin Pharmacother 2017; 19:27-37. [PMID: 28879787 DOI: 10.1080/14656566.2017.1377181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Current international guidelines recommend to treat all HIV-1 infected patients regardless of CD4 cell count. Despite the remarkable worldwide progress for universal access to antiretroviral during the last decade, the pediatric population remains fragile due to lack of randomized studies, inappropriate antiretroviral formulations, adherence difficulties, drug toxicity and development of resistance. AREAS COVERED This review summarizes the latest recommendations and advances for the treatment of HIV-infected children and highlights the potential complications of a lifelong antiretroviral treatment initiated early in life. EXPERT OPINION International guidelines recommend to start combination antiretroviral therapy (cART) as fast as possible in all children diagnosed with HIV-1. The principal goal is to improve survival and reduce mortality as well as rapidly decrease HIV reservoirs. This remains a challenge in resource-limited settings were diagnostic tools and treatment access may be limited. Different new strategies are in the pipeline such as immunotherapy in combination with very early cART initiation to seek remission or functional cure. For the time being and awaiting for long term remission or cure, there is a need for further pharmacokinetics studies, more pediatric formulations with improved palatability and implementation of randomized trials for the newer antiretroviral drugs.
Collapse
Affiliation(s)
- Jean-Christophe Beghin
- a Hôpital Universitaire des Enfants Reine Fabiola , Université Libre de Bruxelles (ULB) , Brussels , Belgium.,b AIDS Reference Laboratory , Université Catholique de Louvain , Brussels , Belgium.,c Institute of Experimental and Clinical Research (IREC) , Université Catholique de Louvain , Brussels , Belgium
| | - Jean Cyr Yombi
- c Institute of Experimental and Clinical Research (IREC) , Université Catholique de Louvain , Brussels , Belgium.,d Department of Internal Medicine, Infectious Diseases and Tropical Medicine Unit , Cliniques Universitaires St Luc , Brussels , Belgium
| | - Jean Ruelle
- c Institute of Experimental and Clinical Research (IREC) , Université Catholique de Louvain , Brussels , Belgium.,e Medical Molecular Biology Unit , Cliniques Universitaires St Luc , Brussels , Belgium
| | - Dimitri Van der Linden
- c Institute of Experimental and Clinical Research (IREC) , Université Catholique de Louvain , Brussels , Belgium.,f Pediatric Infectious Diseases, General Pediatrics, Pediatric Department , Cliniques Universitaires St Luc , Brussels , Belgium
| |
Collapse
|
14
|
Jiménez de Ory S, González-Tomé MI, Fortuny C, Mellado MJ, Soler-Palacin P, Bustillo M, Ramos JT, Muñoz-Fernández MA, Navarro ML. New diagnoses of human immunodeficiency virus infection in the Spanish pediatric HIV Cohort (CoRISpe) from 2004 to 2013. Medicine (Baltimore) 2017; 96:e7858. [PMID: 28953612 PMCID: PMC5626255 DOI: 10.1097/md.0000000000007858] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Vertical human immunodeficiency virus (HIV) infection has decreased in industrialized countries in recent decades, but there are no studies on the mechanisms of HIV transmission among infected children in Spain. Our aim was to study the characteristics and trends of diagnoses of vertically HIV-infected children in Spain from 2004 to 2013.Vertically HIV-infected children were selected if they were diagnosed from 2004 to 2013, were aged 0 to 18 years old, and were included in the Cohort of the Spanish Pediatric HIV Network (CoRISpe). Demographic, clinical, immunological, and virological data at diagnosis were obtained. The rate of diagnoses of vertically HIV-infected children was calculated as the number of cases per 100,000 inhabitants. Obstetric data of mothers of Spanish children and prophylaxis at childbirth and postpartum were obtained.A total of 218 HIV-infected children were included in the study. Of this sample, 182 children (83.5%) were perinatally HIV infected, and 125 out of those 182 children (68.7%) were born in Spain. The vertically HIV-infected Spanish children were diagnosed earlier and were in better clinical and immunological condition at diagnosis than were foreign children. The rate of vertically HIV-infected children declined from 0.09 in 2004 to 0.03 in 2013 due to the decrease in the rate of children born in Spain (0.08 in 2004 vs 0.01 in 2013). A total of 60 out of 107 mothers (56.1%) of Spanish children were diagnosed at or after childbirth. However, this number declined between 2004 and 2013.The rate of new HIV diagnoses of vertically HIV-infected children decreased significantly between 2004 and 2013 from 0.09 to 0.03 per 100,000 inhabitants.
Collapse
Affiliation(s)
- Santiago Jiménez de Ory
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón
- Spanish Human Immunodeficiency Virus Hospital Gregorio Marañón BioBank (Spanish HIV HGM BioBank)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN)
- Servicio de Pediatría, Hospital Clínico San Carlos
| | - María Isabel González-Tomé
- Servicio de Infecciosas Pediátricas, Hospital Universitario Doce de Octubre, Instituto de Investigación Hospital 12 de Octubre
| | - Claudia Fortuny
- Unidad de Enfermedades Infecciosas, Servicio de Pediatría, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues del Llobregat, Barcelona
| | - Maria Jose Mellado
- Servicio de Pediatría Hospitalaria y Enfermedades Infecciosas y Tropicales Pediátricas, Hospital Universitario Infantil La Paz and Hospital Carlos III
| | - Pere Soler-Palacin
- Unitat de Patologia Infecciosa i Immunodeficiències de Pediatria, Hospital Universitari Vall d’Hebron, Institut de Recerca Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona
| | - Matilde Bustillo
- Servicio de Pediatría, Unidad Infectología, Hospital Infantil Universitario Miguel Servet, Zaragoza
| | | | - Maria Angeles Muñoz-Fernández
- Sección Inmunología, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón
- Spanish Human Immunodeficiency Virus Hospital Gregorio Marañón BioBank (Spanish HIV HGM BioBank)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN)
| | - Maria Luisa Navarro
- Sección de Enfermedades Infecciosas, Servicio de Pediatría, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
15
|
Srinivas NR. Prediction of area under the concentration-time curve for lopinavir from peak or trough lopinavir concentrations in patients receiving lopinavir-ritonavir therapy. Am J Health Syst Pharm 2016; 73:376-85. [PMID: 26953282 DOI: 10.2146/ajhp150417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE A "single time point" strategy for predicting the area under the concentration-time curve (AUC) for lopinavir in patients receiving ritonavir-boosted lopinavir therapy was investigated. METHODS Linear regression equations describing the relationships of lopinavir peak and trough concentrations to lopinavir AUC values were established using pharmacokinetic data from published studies of patients or healthy subjects receiving lopinavir and ritonavir at standard dosages. The resulting "trough-AUC model" and "peak-AUC model" were used to predict lopinavir AUC values in the evaluated study populations (total n = 479); those values were then compared with reported AUC values. RESULTS Lopinavir peak or trough concentrations were strongly correlated with lopinavir AUC values (r = 0.9947 and r = 0.9541, respectively). For about 94% of calculations using the peak-AUC model and 87% of calculations using the trough-AUC model, differences between predicted and observed AUC values were in the range of 0.76-1.5 fold; the associated r values were 0.9514 (p < 0.001) and 0.9345 (p < 0.001), respectively. The mean absolute predictive error was less than 6% with the use of either the peak-AUC model or the trough-AUC model, with corresponding values for root-mean-square error of 17.6% and 23.5%, respectively. CONCLUSION Equations incorporating lopinavir peak and trough concentrations were found to satisfactorily predict lopinavir AUC values in data sets describing patients receiving lopinavir with ritonavir boosting. Variability in predictions was higher with use of the trough-AUC model.
Collapse
|
16
|
Safety of combination antiretroviral prophylaxis in high-risk HIV-exposed newborns: a retrospective review of the Canadian experience. J Int AIDS Soc 2016; 19:20520. [PMID: 26880241 PMCID: PMC4753845 DOI: 10.7448/ias.19.1.20520] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 12/10/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022] Open
Abstract
Introduction The optimal management of infants born to HIV-positive mothers who are untreated or have detectable viral load prior to delivery remains controversial. Despite the increasing use of combination antiretroviral therapy (cART) for post-exposure prophylaxis (PEP) of neonates at high risk of HIV infection, there is little safety and pharmacokinetic data to support this approach. The objective of this study was to evaluate the safety and tolerability of cART for PEP in HIV-exposed neonates. Methods Retrospective study on 148 cART and 145 Zidovudine (ZDV) monotherapy-exposed infants identified from four Canadian centres where cART for PEP has routinely been prescribed in high-risk situations. Physician-reported adverse events and clinical outcomes were extracted by chart review. Haematological and growth parameters at birth, one and six months of age were compared between cART and ZDV-exposed infants using multivariate mixed effects modelling. Results Non-specific signs and symptoms were reported in 10.2% of cART recipients versus none of the ZDV recipients. Treatment was discontinued prematurely in 9.5% of cART recipients versus 2.1% of ZDV recipients (p=0.01). In the multivariate model, cART recipients had lower mean haemoglobin (decrease of 2.07 g/L) over the 6-month period compared with ZDV recipients (p=0.04), but no effect was seen on absolute neutrophil count. cART recipients had lower weight and smaller head circumference at birth and one month of age compared with ZDV-exposed infants; these differences were no longer significant at six months of age. Conclusions cART administered at treatment doses for PEP in neonates was generally well tolerated, though a higher incidence of non-specific signs and symptoms and early treatment discontinuation occurred among cART recipients.
Collapse
|
17
|
Nagot N, Kankasa C, Tumwine JK, Meda N, Hofmeyr GJ, Vallo R, Mwiya M, Kwagala M, Traore H, Sunday A, Singata M, Siuluta C, Some E, Rutagwera D, Neboua D, Ndeezi G, Jackson D, Maréchal V, Neveu D, Engebretsen IMS, Lombard C, Blanche S, Sommerfelt H, Rekacewicz C, Tylleskär T, Van de Perre P. Extended pre-exposure prophylaxis with lopinavir-ritonavir versus lamivudine to prevent HIV-1 transmission through breastfeeding up to 50 weeks in infants in Africa (ANRS 12174): a randomised controlled trial. Lancet 2016; 387:566-573. [PMID: 26603917 DOI: 10.1016/s0140-6736(15)00984-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Strategies to prevent postnatal mother-to-child transmission of HIV-1 in Africa, including infant prophylaxis, have never been assessed past 6 months of breastfeeding, despite breastfeeding being recommended up to 12 months after birth. We aimed to compare the efficacy and safety of infant prophylaxis with the two drug regimens (lamivudine or lopinavir-ritonavir) to prevent postnatal HIV-1 transmission up to 50 weeks of breastfeeding. METHODS We did a randomised controlled trial in four sites in Burkina Faso, South Africa, Uganda, and Zambia in children born to HIV-1-infected mothers not eligible for antiretroviral therapy (CD4 count >350 cells per μL). An independent researcher electronically generated a randomisation schedule; we then used sequentially numbered envelopes to randomly assign (1:1) HIV-1-uninfected breastfed infants aged 7 days to either lopinavir-ritonavir or lamivudine (paediatric liquid formulations, twice a day) up to 1 week after complete cessation of breastfeeding or at the final visit at week 50. We stratified the randomisation by country and used permuted blocks of four and six. We used a study label on drug bottles to mask participants, study physicians, and assessors to the treatment allocation. The primary outcome was infant HIV-1 infection between age 7 days and 50 weeks, diagnosed every 3 months with HIV-1 DNA PCR, in the modified intention-to-treat population (all who attended at least one follow-up visit). This trial is registered with ClinicalTrials.gov, number NCT00640263. FINDINGS Between Nov 16, 2009, and May 7, 2012, we enrolled and randomised 1273 infants and analysed 1236; 615 assigned to lopinavir-ritonavir or 621 assigned to lamivudine. 17 HIV-1 infections were diagnosed in the study period (eight in the lopinavir-ritonavir group and nine in the lamivudine group), resulting in cumulative HIV-1 infection of 1.4% (95% CI 0.4-2.5) and 1.5% (0.7-2.5), respectively. Infection rates did not differ between the two drug regimens (hazard ratio [HR] of lopinavir-ritonavir versus lamivudine of 0.90, 95% CI 0.35-2.34; p=0.83). Clinical and biological severe adverse events did not differ between groups; 251 (51%) infants had a grade 3-4 event in the lopinavir-ritonavir group compared with 246 (50%) in the lamivudine group. INTERPRETATION Infant HIV-1 prophylaxis with lopinavir-ritonavir was not superior to lamivudine and both drugs led to very low rates of HIV-1 postnatal transmission for up to 50 weeks of breastfeeding. Infant pre-exposure prophylaxis should be extended until the end of HIV-1 exposure and mothers should be informed about the persistent risk of transmission throughout breastfeeding. FUNDING INSERM/National Agency for Research on AIDS and Viral Hepatitis (including funds from the Total Foundation), European Developing Countries Clinical Trials Partnership, Research Council of Norway.
Collapse
Affiliation(s)
- Nicolas Nagot
- INSERM U 1058, Montpellier, France; Université Montpellier, Montpellier, France; CHU Montpellier, Montpellier, France
| | - Chipepo Kankasa
- University of Zambia, School of Medicine, Department of Paediatrics and Child Health, University Teaching Hospital, Lusaka, Zambia
| | - James K Tumwine
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Nicolas Meda
- Centre of International Research for Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | - G Justus Hofmeyr
- Effective Care Research Unit, Cecilia Makiwane Hospital, East London Hospital Complex, East London, South Africa; School of Public Health, University of the Western Cape, South Africa
| | - Roselyne Vallo
- INSERM U 1058, Montpellier, France; Université Montpellier, Montpellier, France
| | - Mwiya Mwiya
- University of Zambia, School of Medicine, Department of Paediatrics and Child Health, University Teaching Hospital, Lusaka, Zambia
| | - Mary Kwagala
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Hugues Traore
- Centre of International Research for Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | - Amwe Sunday
- Effective Care Research Unit, Cecilia Makiwane Hospital, East London Hospital Complex, East London, South Africa; School of Public Health, University of the Western Cape, South Africa
| | - Mandisa Singata
- Effective Care Research Unit, Cecilia Makiwane Hospital, East London Hospital Complex, East London, South Africa; School of Public Health, University of the Western Cape, South Africa
| | - Chafye Siuluta
- University of Zambia, School of Medicine, Department of Paediatrics and Child Health, University Teaching Hospital, Lusaka, Zambia
| | - Eric Some
- Centre of International Research for Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | - David Rutagwera
- University of Zambia, School of Medicine, Department of Paediatrics and Child Health, University Teaching Hospital, Lusaka, Zambia
| | - Desire Neboua
- Centre of International Research for Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | - Grace Ndeezi
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Debra Jackson
- School of Public Health, University of the Western Cape, South Africa
| | - Valérie Maréchal
- INSERM U 1058, Montpellier, France; Université Montpellier, Montpellier, France
| | - Dorine Neveu
- INSERM U 1058, Montpellier, France; Université Montpellier, Montpellier, France
| | | | - Carl Lombard
- Biostatistics Unit, Medical Research Council of South Africa, Cape Town, South Africa
| | - Stéphane Blanche
- University of Paris V, Department of Paediatrics, URC/CIC Necker-Cochin, Hôpital Necker, Paris, France
| | - Halvor Sommerfelt
- Centre for International Health, University of Bergen, Bergen, Norway; Centre for Intervention Science in Maternal and Child Health, Centre for International Health, University of Bergen, Bergen, Norway; Department of International Public Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Claire Rekacewicz
- French National Institute of Health and Medical Research-National Agency for Research on AIDS and Viral Hepatitis (INSERM-ANRS), Paris, France
| | | | - Philippe Van de Perre
- INSERM U 1058, Montpellier, France; Université Montpellier, Montpellier, France; CHU Montpellier, Montpellier, France.
| |
Collapse
|
18
|
Uprety P, Chadwick EG, Rainwater-Lovett K, Ziemniak C, Luzuriaga K, Capparelli EV, Yenokyan G, Persaud D. Cell-Associated HIV-1 DNA and RNA Decay Dynamics During Early Combination Antiretroviral Therapy in HIV-1-Infected Infants. Clin Infect Dis 2015; 61:1862-70. [PMID: 26270687 DOI: 10.1093/cid/civ688] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/30/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The decay of human immunodeficiency virus type 1 (HIV-1)-infected cells during early combination antiretroviral therapy (cART) in infected infants is not defined. METHODS HIV-1 DNA, including 2-long terminal repeat (2-LTR) circles, and multiply spliced (ms-) and unspliced (us-) HIV-1 RNA concentrations were measured at 0, 24, 48, and 96 weeks of cART in infants from the IMPAACT P1030 trial receiving lopinavir-ritonavir-based cART. The ratio of HIV-1 DNA concentrations to replication-competent genomes was also estimated. Linear mixed effects models with random intercept and linear splines were used to estimate patient-specific decay kinetics of HIV-1 DNA. RESULTS The median HIV-1 DNA concentration before cART at a median age of 2 months was 3.2 log10 copies per million PBMC. With cART, the average estimated patient-specific change in HIV-1 DNA concentrations was -0.040 log10/week (95% confidence interval [CI], -.05, -.03) between 0 and 24 weeks and -0.017 log10/week between 24 and 48 weeks (95% CI, -.024, -.01). 2-LTR circles decreased with cART but remained detectable through 96 weeks. Pre-cART HIV-1 DNA concentration was correlated with time to undetectable plasma viral load and post-cART HIV-1 DNA at 96 weeks; although HIV-1 DNA concentrations exceeded replication-competent HIV-1 genomes by 148-fold. Almost all infants had ms- and usRNA detected pre-cART, with 75% having usRNA through 96 weeks of cART. CONCLUSIONS By 2 months of age, a large pool of HIV-1-infected cells is established in perinatal infection, which influences time to undetectable viral load and reservoir size. This has implications for informing novel approaches aimed at early restriction of HIV-1 reservoirs to enable virologic remission and cure.
Collapse
Affiliation(s)
- Priyanka Uprety
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ellen G Chadwick
- Department of Pediatrics, Northwestern University School of Medicine, Chicago, Illinois
| | | | - Carrie Ziemniak
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Katherine Luzuriaga
- Department of Pediatrics, University of Massachusetts Medical School, Worcester
| | | | - Gayane Yenokyan
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Deborah Persaud
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Giandhari J, Basson AE, Coovadia A, Kuhn L, Abrams EJ, Strehlau R, Morris L, Hunt GM. Genetic Changes in HIV-1 Gag-Protease Associated with Protease Inhibitor-Based Therapy Failure in Pediatric Patients. AIDS Res Hum Retroviruses 2015; 31:776-82. [PMID: 25919760 DOI: 10.1089/aid.2014.0349] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Studies have shown a low frequency of HIV-1 protease drug resistance mutations in patients failing protease inhibitor (PI)-based therapy. Recent studies have identified mutations in Gag as an alternate pathway for PI drug resistance in subtype B viruses. We therefore genotyped the Gag and protease genes from 20 HIV-1 subtype C-infected pediatric patients failing a PI-based regimen. Major protease resistance mutations (M46I, I54V, and V82A) were identified in eight (40%) patients, as well as Gag cleavage site (CS) mutations (at codons 373, 374, 378, 428, 431, 449, 451, and 453) in nine (45%) patients. Four of these Gag CS mutations occurred in the absence of major protease mutations at PI failure. In addition, amino acid changes were noted at Gag non-CS with some predicted to be under HLA/KIR immune-mediated pressure and/or drug selection pressure. Changes in Gag during PI failure therefore warrant further investigation of the Gag gene and its role in PI failure in HIV-1 subtype C infection.
Collapse
Affiliation(s)
- Jennifer Giandhari
- Centre for HIV and STIs, National Institute for Communicable Diseases (NICD), Johannesburg, South Africa
- Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Adriaan E. Basson
- Centre for HIV and STIs, National Institute for Communicable Diseases (NICD), Johannesburg, South Africa
- Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Ashraf Coovadia
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Elaine J. Abrams
- International Center for AIDS Programs, Mailman School of Public Health, Columbia University, New York, New York
| | - Renate Strehlau
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases (NICD), Johannesburg, South Africa
- Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Gillian M. Hunt
- Centre for HIV and STIs, National Institute for Communicable Diseases (NICD), Johannesburg, South Africa
- Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
20
|
de Ruiter A, Taylor GP, Clayden P, Dhar J, Gandhi K, Gilleece Y, Harding K, Hay P, Kennedy J, Low-Beer N, Lyall H, Palfreeman A, O'Shea S, Tookey P, Tosswill J, Welch S, Wilkins E. British HIV Association guidelines for the management of HIV infection in pregnant women 2012 (2014 interim review). HIV Med 2015; 15 Suppl 4:1-77. [PMID: 25604045 DOI: 10.1111/hiv.12185] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Smith C, Forster JE, Levin MJ, Davies J, Pappas J, Kinzie K, Barr E, Paul S, McFarland EJ, Weinberg A. Serious adverse events are uncommon with combination neonatal antiretroviral prophylaxis: a retrospective case review. PLoS One 2015; 10:e0127062. [PMID: 26000984 PMCID: PMC4441417 DOI: 10.1371/journal.pone.0127062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/10/2015] [Indexed: 12/12/2022] Open
Abstract
Six weeks of zidovudine (ZDV) is recommended for postnatal prophylaxis of HIV-exposed infants, but combination antiretrovirals are indicated if HIV transmission risk is increased. We investigated the frequency and severity of adverse events (AE) in infants receiving multiple drug prophylaxis compared to ZDV alone. In this retrospective review of 148 HIV-exposed uninfected infants born between 1997–2009, we determined clinical and laboratory AE that occurred between days of life 8–42. Thirty-six infants received combination prophylaxis; among those, a three-drug regimen containing ZDV, lamivudine, and nevirapine was most common (53%). Rates of laboratory AE grade ≥1 were as follows for the combination prophylaxis and ZDV alone groups, respectively: neutropenia 55% and 39%; anemia 50% and 39%; thrombocytopenia 0 and 3%; elevated aspartate aminotransferase 3% and 3%; elevated alanine aminotransferase 0 and 1%; hyperbilirubinemia 19% and 42%. Anemia occurred more frequently in infants who received three-drug prophylaxis compared to infants who received ZDV alone (63% vs. 39%, p = 0.04); all anemia AE were grade 1 or 2 in the three-drug prophylaxis group. Overall, 75% of infants on combination prophylaxis and 66% of infants on ZDV alone developed grade ≥1 AE (p = 0.32), and 17% of infants in either group developed grade ≥3 AE. Stavudine was substituted for ZDV in 23 infants due to anemia or neutropenia. After this antiretroviral change, 50% of evaluable infants demonstrated improvement in AE grade, and 25% had no change. In conclusion, low grade anemia, neutropenia, and hyperbilirubinemia occurred frequently regardless of the prophylactic regimen, but serious AE were uncommon. Although most AE were typical of ZDV toxicity, the combination of ZDV with lamivudine and nevirapine resulted in an increased frequency of low-grade anemia. Further studies are needed to identify prophylactic regimens with less toxicity for infants born to HIV-infected mothers.
Collapse
Affiliation(s)
- Christiana Smith
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, United States of America
- * E-mail:
| | - Jeri E. Forster
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, United States of America
| | - Myron J. Levin
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, United States of America
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Jill Davies
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Obstetrics and Gynecology, Denver Health Medical Center, Denver, Colorado, United States of America
| | - Jennifer Pappas
- Children's Hospital Colorado, Aurora, Colorado, United States of America
| | - Kay Kinzie
- Children's Hospital Colorado, Aurora, Colorado, United States of America
| | - Emily Barr
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, United States of America
| | - Suzanne Paul
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, United States of America
| | - Elizabeth J. McFarland
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, United States of America
| | - Adriana Weinberg
- Department of Pediatric Infectious Diseases, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, United States of America
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
22
|
Davies MA. Research gaps in neonatal HIV-related care. South Afr J HIV Med 2015; 16:375. [PMID: 29568592 PMCID: PMC5843028 DOI: 10.4102/sajhivmed.v16i1.375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/16/2015] [Indexed: 12/19/2022] Open
Abstract
The South African prevention of mother to child transmission programme has made excellent progress in reducing vertical HIV transmission, and paediatric antiretroviral therapy programmes have demonstrated good outcomes with increasing treatment initiation in younger children and infants. However, both in South Africa and across sub-Saharan African, lack of boosted peri-partum prophylaxis for high-risk vertical transmission, loss to follow-up, and failure to initiate HIV-infected infants on antiretroviral therapy (ART) before disease progression are key remaining gaps in neonatal HIV-related care. In this issue of the Southern African Journal of HIV Medicine, experts provide valuable recommendations for addressing these gaps. The present article highlights a number of areas where evidence is lacking to inform guidelines and programme development for optimal neonatal HIV-related care.
Collapse
Affiliation(s)
- Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, South Africa
| |
Collapse
|
23
|
Nuttall JJC. Antiretroviral therapy during the neonatal period. South Afr J HIV Med 2015; 16:361. [PMID: 29568586 PMCID: PMC5843175 DOI: 10.4102/sajhivmed.v16i1.361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/10/2015] [Indexed: 01/05/2023] Open
Abstract
No abstract available
Collapse
Affiliation(s)
- James J C Nuttall
- Red Cross War Memorial Children's Hospital, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, South Africa
| |
Collapse
|
24
|
von Bibra M, Rosenkranz B, Pretorius E, Rabie H, Edson C, Lenker U, Cotton M, Klinker H. Are lopinavir and efavirenz serum concentrations in HIV-infected children in the therapeutic range in clinical practice? Paediatr Int Child Health 2014; 34:138-41. [PMID: 24225343 DOI: 10.1179/2046905513y.0000000090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND In antiretroviral treatment the role of therapeutic drug monitoring via measurement of serum levels remains unclear, especially in children. AIM To quantify exposure to LPV and EFV in children receiving therapy in a routine clinical setting in order to identify risk factors associated with inadequate drug exposure. METHOD A prospective study was conducted in a routine clinical setting in Tygerberg Children's Hospital, South Africa. A total of 53 random serum levels were analyzed. Serum concentrations were determined by an established high-performance liquid chromatography method. RESULTS Of 53 HIV-infected children treated with lopinavir (n = 29, median age 1·83 y) or efavirenz (n = 24, median age 9·3 years), 12 showed serum levels outside the therapeutic range (efavirenz) or below Cmin (lopinavir). Low bodyweight, rifampicin co-treatment, and significant comorbidity were potential risk factors for inadequate drug exposure. CONCLUSION These findings, together with previous studies, indicate that therapeutic drug monitoring can improve the management of antiretroviral therapy in children at risk.
Collapse
|
25
|
Moholisa RR, Schomaker M, Kuhn L, Meredith S, Wiesner L, Coovadia A, Strehlau R, Martens L, Abrams EJ, Maartens G, McIlleron H. Plasma lopinavir concentrations predict virological failure in a cohort of South African children initiating a protease-inhibitor-based regimen. Antivir Ther 2014; 19:399-406. [PMID: 24518130 DOI: 10.3851/imp2749] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2013] [Indexed: 01/16/2023]
Abstract
BACKGROUND Poor adherence to antiretroviral therapy contributes to pharmacokinetic variability and is the major determinant of virological failure. However, measuring treatment adherence is difficult, especially in children. We investigated the relationship between plasma lopinavir concentrations, pretreatment characteristics and viral load >400 copies/ml. METHODS A total of 237 HIV-infected children aged 4-42 months on lopinavir/ritonavir oral solution were studied prospectively and followed for up to 52 weeks. Viral load and lopinavir concentration were measured at clinic visits 12, 24, 36 and 52 weeks after starting treatment. Cox multiple failure events models were used to estimate the crude and adjusted effect of lopinavir concentrations on the hazard of viral load >400 copies/ml. RESULTS The median (IQR) pretreatment CD4(+) T-lymphocyte percentage was 18.80% (12.70-25.35) and 53% of children had a pretreatment viral load >750,000 copies/ml. The median (IQR) weight-for-age and height-for-age z-scores were -2.17 (-3.35--2.84) and -3.34 (-4.57--3.41), respectively. Median (IQR) lopinavir concentrations were 8.00 mg/l (4.11-12.42) at median (IQR) 3.50 h (2.67-4.25) after the dose. The hazard of viral load >400 copies/ml was increased with lopinavir concentrations <1 mg/l versus ≥1 mg/l (adjusted hazard ratio 2.3 [95% CI 1.63, 3.26]) and lower height-for-age z-scores. CONCLUSIONS Low lopinavir concentrations (<1 mg/l) are associated with viraemia in children. This measure could be used as a proxy for adherence and to determine which children are more likely to fail.
Collapse
Affiliation(s)
- Retsilisitsoe R Moholisa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shiau S, Kuhn L. Antiretroviral treatment in HIV-infected infants and young children: novel issues raised by the Mississippi baby. Expert Rev Anti Infect Ther 2014; 12:307-18. [PMID: 24506199 DOI: 10.1586/14787210.2014.888311] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The recent case report of an HIV-infected child in Mississippi with viral control post-antiretroviral therapy (ART) interruption has sparked interest in the possibility of 'functional cure' in infants if they initiate ART very soon after birth. The 'Mississippi baby' also raises many new questions around the clinical care of HIV-infected infants and young children, including when treatment should be initiated, why treatment should be initiated, what treatment should be initiated, and how to identify infants early enough to treat them adequately. Here, we review research conducted before the report of the 'Mississippi baby' highlighting the important new issues that now need to be taken into consideration.
Collapse
Affiliation(s)
- Stephanie Shiau
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | |
Collapse
|
27
|
Laughon MM, Avant D, Tripathi N, Hornik CP, Cohen-Wolkowiez M, Clark RH, Smith PB, Rodriguez W. Drug labeling and exposure in neonates. JAMA Pediatr 2014; 168:130-6. [PMID: 24322269 PMCID: PMC3927948 DOI: 10.1001/jamapediatrics.2013.4208] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IMPORTANCE Federal legislation has led to a notable increase in pediatric studies submitted to the Food and Drug Administration (FDA), resulting in new pediatric information in product labeling. However, approximately 50% of drug labels still have insufficient information on safety, efficacy, or dosing in children. Neonatal information in labeling is even scarcer because neonates comprise a vulnerable subpopulation for which end-point development is lagging and studies are more challenging. OBJECTIVE To quantify progress made in neonatal studies and neonatal information in product labeling as a result of recent legislation. DESIGN, SETTING, AND PARTICIPANTS We identified a cohort of drug studies between 1997 and 2010 that included neonates as a result of pediatric legislation using information available on the FDA website. We determined what studies were published in the medical literature, the legislation responsible for the studies, and the resulting neonatal labeling changes. We then examined the use of these drugs in a cohort of neonates admitted to 290 neonatal intensive care units (NICUs) (the Pediatrix Data Warehouse) in the United States from 2005 to 2010. EXPOSURE Infants exposed to a drug studied in neonates as identified by the FDA website. MAIN OUTCOMES AND MEASURES Number of drug studies with neonates and rate of exposure per 1000 admissions among infants admitted to an NICU. RESULTS In a review of the FDA databases, we identified 28 drugs studied in neonates and 24 related labeling changes. Forty-one studies encompassed the 28 drugs, and 31 (76%) of these were published. Eleven (46%) of the 24 neonatal labeling changes established safety and effectiveness. In a review of a cohort of 446,335 hospitalized infants, we identified 399 drugs used and 1,525,739 drug exposures in the first 28 postnatal days. Thirteen (46%) of the 28 drugs studied in neonates were not used in NICUs; 8 (29%) were used in fewer than 60 neonates. Of the drugs studied, ranitidine was used most often (15,627 neonates, 35 exposures per 1000 admissions). CONCLUSIONS AND RELEVANCE Few drug labeling changes made under pediatric legislation include neonates. Most drugs studied are either not used or rarely used in US NICUs. Strategies to increase the study of safe and effective drugs for neonates are needed.
Collapse
Affiliation(s)
- Matthew M. Laughon
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill
| | - Debbie Avant
- Food and Drug Administration, Rockville, Maryland
| | - Nidhi Tripathi
- Duke Clinical Research Institute and Duke University Medical Center, Durham, North Carolina
| | - Christoph P. Hornik
- Duke Clinical Research Institute and Duke University Medical Center, Durham, North Carolina
| | | | - Reese H. Clark
- Pediatrix Center for Research and Education, Sunrise, Florida
| | - P. Brian Smith
- Duke Clinical Research Institute and Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
28
|
Abstract
BACKGROUND There is scant data on young children receiving protease inhibitor-based therapy in real-life resource-limited settings and on the optimal timing of therapy among children who survive infancy. Our aim was to evaluate outcomes at the Hospital del Niño, Panama, where children have been routinely treated with lopinavir/ritonavir (LPV/r)-based therapy since 2002. METHODS Retrospective cohort analysis of all HIV-infected children enrolled in care between January 1, 1991, and June 1, 2011. Kaplan-Meier method and Cox proportional hazards regression were used to evaluate death, virologic suppression and virologic rebound. RESULTS Of 399 children contributing 1944 person-years of follow-up, 254 (63.7%) were treated with LPV/r and 94 (23.6%) were never treated with antiretrovirals (ARVs). Among infants, improved survival was associated with male gender (hazard rate of death[HRdeath] 0.54, 95% confidence interval [CI]: 0.32-0.92) and treatment with highly active antiretroviral therapy (HRdeath 0.32, 95% CI: 0.12-0.83), whereas residence outside of Panama City was associated with poorer survival (HRdeath 1.72, 95% CI: 1.01-2.94). Among children who survived to 1 year of age without exposure to ARVs, LPV/r-based therapy improved survival (HRdeath 0.07, 95% CI: 0.01-0.33). Virologic suppression was achieved in 42.1%, 70.5% and 85.1% by 12, 24 and 60 months of follow-up among children treated with LPV/r. Virologic suppression was not associated with prior ARV exposure or age at initiation of therapy but was associated with residence outside of Panama City (HR suppression 1.93, 95% CI: 1.19-3.14). Patients with a baseline viral load >100,000 copies/mL were less likely to achieve suppression (HR suppression 0.37, 95% CI: 0.21-0.66). No children who achieved virologic suppression after initiating LPV/r died. CONCLUSIONS LPV/r-based therapy improved survival not only in infants but also in children over 1 year of age. Age at initiation of LPV/r-based therapy or prior ARVs did not impact virologic outcomes.
Collapse
|
29
|
Nagot N, Kankasa C, Meda N, Hofmeyr J, Nikodem C, Tumwine JK, Karamagi C, Sommerfelt H, Neveu D, Tylleskär T, Van de Perre P. Lopinavir/Ritonavir versus Lamivudine peri-exposure prophylaxis to prevent HIV-1 transmission by breastfeeding: the PROMISE-PEP trial Protocol ANRS 12174. BMC Infect Dis 2012; 12:246. [PMID: 23039034 PMCID: PMC3482558 DOI: 10.1186/1471-2334-12-246] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 08/30/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Postnatal transmission of HIV-1 through breast milk remains an unsolved challenge in many resource-poor settings where replacement feeding is not a safe alternative. WHO now recommends breastfeeding of infants born to HIV-infected mothers until 12 months of age, with either maternal highly active antiretroviral therapy (HAART) or peri-exposure prophylaxis (PEP) in infants using nevirapine. As PEP, lamivudine showed a similar efficacy and safety as nevirapine, but with an expected lower rate of resistant HIV strains emerging in infants who fail PEP, and lower restrictions for future HIV treatment. Lopinavir/ritonavir (LPV/r) is an attractive PEP candidate with presumably higher efficacy against HIV than nevirapine or lamivudine, and a higher genetic barrier to resistance selection. It showed an acceptable safety profile for the treatment of very young HIV-infected infants. The ANRS 12174 study aims to compare the risk of HIV-1 transmission during and safety of prolonged infant PEP with LPV/r (40/10 mg twice daily if 2-4 kg and 80/20 mg twice daily if >4 kg) versus Lamivudine (7,5 mg twice daily if 2-4 kg, 25 mg twice daily if 4-8 kg and 50 mg twice daily if >8 kg) from day 7 until one week after cessation of BF (maximum 50 weeks of prophylaxis) to prevent postnatal HIV-1 acquisition between 7 days and 50 weeks of age. METHODS The ANRS 12174 study is a multinational, randomised controlled clinical trial conducted on 1,500 mother-infant pairs in Burkina Faso, South Africa, Uganda and Zambia. We will recommend exclusive breastfeeding (EBF) until 26th week of life and cessation of breastfeeding at a maximum of 49 weeks in both trial arms.HIV-uninfected infants at day 7 (± 2 days) born to HIV-1 infected mothers not eligible for HAART who choose to breastfeed their infants.The primary endpoint is the acquisition of HIV-1 (as assessed by HIV-1 DNA PCR) between day 7 and 50 weeks of age. Secondary endpoints are safety (including resistance, adverse events and growth) until 50 weeks and HIV-1-free survival until 50 weeks. DISCUSSION This study will provide a new evidence-based intervention to support HIV-1-infected women not eligible for HAART to safely breastfeed their babies.
Collapse
|
30
|
Anderson EJ, Yogev R. The glory of guidelines and the twilight of reality: controversies and challenges in the prevention and treatment of HIV in children. Expert Rev Anti Infect Ther 2012; 10:761-74. [PMID: 22943400 DOI: 10.1586/eri.12.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Since the discovery of HIV 30 years ago, we have learned much about HIV in children and adolescents. Dramatic declines have occurred in mother-to-child transmission of HIV in resource-rich countries. Resource-poor countries struggle with improving prevention of mother-to-child transmission due to the lack of universal antiretroviral treatment for pregnant and nursing mothers. In children infected with HIV, pharmacokinetic, safety and efficacy data have been determined for many of the older drugs. Data are lacking for the newer, safer and more effective currently available drugs, resulting in the pediatric guidelines lagging behind adult recommendations. Although guidelines for prevention and treatment are helpful, the way they are created causes them to lag behind new scientific evidence, and in some situations they will be confusing or only based on expert opinion. Improving prevention of HIV infection in adolescents and young adults and in treating those who become HIV infected is crucially important. The next 10 years hold tremendous opportunities for improvements in prevention and treatment of HIV in children, adolescents and young adults.
Collapse
Affiliation(s)
- Evan J Anderson
- Department of Pediatrics and Medicine, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA, USA.
| | | |
Collapse
|
31
|
11.0 References. HIV Med 2012. [DOI: 10.1111/j.1468-1293.2012.1030_12.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
32
|
Burckart GJ, Estes KE, Leong R, Mulugeta Y, Tandon V, Wang J, Abernethy DR, Jadhav PR. Methodological Issues in the Design of Paediatric Pharmacokinetic Studies. Pharmaceut Med 2012. [DOI: 10.1007/bf03256888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
33
|
Dynamics of the resting CD4(+) T-cell latent HIV reservoir in infants initiating HAART less than 6 months of age. AIDS 2012; 26:1483-90. [PMID: 22555165 DOI: 10.1097/qad.0b013e3283553638] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Identification of HIV infection in exposed infants facilitates early therapy, which may limit viral reservoirs that maintain HIV infection under HAART. METHODS The dynamics of the resting CD4 T-cell latent HIV reservoir was determined over the first 2 years of life in 17 HIV-infected infants initiating lopinavir/ritonavir-based HAART at a median age of 8.1 weeks and achieving adequate suppression of plasma viral load by 24 weeks. RESULTS The resting CD4 T-cell latent HIV reservoir was detected in 12 of 14 (86%) infants tested at 24 weeks of HAART [median frequency 1.88 infectious units per million (IUPM); range <0.22 to 81.7), and remained measurable (median IUPM = 0.32; range <0.22 to 3.25) in six of 10 (60%) children retested at 96 weeks. The reservoir declined, from 24 to 96 weeks of HAART, at an estimated mean rate of 0.028 log10 IUPM/month, corresponding to a half-life of 11 months (95% confidence interval 6-30 months]. A strong relationship was found between the frequency of latently infected CD4 T cells at 96 weeks of HAART and time to first undetectable plasma viral load (Spearman r = 0.91, P < 0.001). CONCLUSION Although the resting CD4 T-cell latent reservoir remains detectable over the first 2 years of HAART in a substantial proportion of infants, its size is associated with time to first undetectable viral load. To minimize HIV reservoirs in infants, rapid curtailment of viremia may limit HIV reservoirs and should be a therapeutic goal of early HAART in infants.
Collapse
|
34
|
8.0 Neonatal management. HIV Med 2012. [DOI: 10.1111/j.1468-1293.2012.1030_9.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
35
|
Taylor GP, Clayden P, Dhar J, Gandhi K, Gilleece Y, Harding K, Hay P, Kennedy J, Low-Beer N, Lyall H, Palfreeman A, Tookey P, Welch S, Wilkins E, de Ruiter A. British HIV Association guidelines for the management of HIV infection in pregnant women 2012. HIV Med 2012. [DOI: 10.1111/j.1468-1293.2012.01030.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- GP Taylor
- Communicable Diseases; Section of Infectious Diseases; Imperial College London; UK
| | - P Clayden
- UK Community Advisory Board representative/HIV treatment advocates network; London; UK
| | - J Dhar
- Genitourinary Medicine; University Hospitals of Leicester NHS Trust; Leicester; UK
| | - K Gandhi
- Heart of England NHS Foundation Trust; Birmingham; UK
| | | | - K Harding
- Guy's and St Thomas′ Hospital NHS Foundation Trust; London; UK
| | - P Hay
- St George's Healthcare NHS Trust; London; UK
| | - J Kennedy
- Homerton University Hospital NHS Foundation Trust; London; UK
| | - N Low-Beer
- Chelsea and Westminster Hospital NHS Foundation Trust; London; UK
| | - H Lyall
- Imperial College Healthcare NHS Trust; London; UK
| | - A Palfreeman
- Genitourinary Medicine; University Hospitals of Leicester NHS Trust; Leicester; UK
| | - P Tookey
- UCL Institute of Child Health; London; UK
| | - S Welch
- Paediatric Infectious Diseases; Heart of England NHS Foundation Trust; Birmingham; UK
| | - E Wilkins
- Infectious Diseases and Director of the HIV Research Unit; North Manchester General Hospital; Manchester; UK
| | - A de Ruiter
- Genitourinary Medicine; Guy's and St Thomas' NHS Foundation Trust; London; UK
| |
Collapse
|
36
|
Abstract
Because of serious adverse effects, the Food and Drug Administration warns against using lopinavir in infants younger than 42 weeks postconception. However, there is an imperative for early treatment. We report on our use of LPV in 8 premature HIV-infected infants. The median age at initiation was 27 days. Trough values guided dosing. Five infants needed doses above 300 mg/m. Although no adverse events were noted, lopinavir usage requires caution and careful monitoring.
Collapse
|
37
|
Wegzyn CM, Fredrick LM, Stubbs RO, Woodward WC, Norton M. Diarrhea associated with lopinavir/ritonavir-based therapy: results of a meta-analysis of 1469 HIV-1-infected participants. ACTA ACUST UNITED AC 2012; 11:252-9. [PMID: 22544446 DOI: 10.1177/1545109712442984] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Antiretroviral therapy is associated with adverse events (AEs). The most frequently reported AE associated with lopinavir/ritonavir (LPV/r) containing regimens is diarrhea. The objective of this meta-analysis is to describe the incidence, prevalence, and duration of diarrhea in individuals taking LPV/r. METHODS This is a meta-analysis of Abbott-conducted clinical trials. Inclusion criteria included prospective randomized clinical trials with the LPV/r tablet formulation and had AE data (moderate/severe diarrhea) available through 48 weeks of treatment. RESULTS Three trials (total 1469 participants) met the inclusion criteria. In all, 11.2% of participants reported moderate/severe diarrhea by week 8, with median time to resolution of 7.4 weeks. The overall 48-week incidence of moderate/severe diarrhea was 15.5%. The discontinuation rate due to moderate/severe diarrhea was 1.3%. CONCLUSIONS Moderate/severe diarrhea occurred in less than 1 in 6 participants taking LPV/r, typically started in the first 8 weeks of treatment and infrequently resulted in premature discontinuation.
Collapse
|
38
|
Tseng A, Foisy M, Hughes CA, Kelly D, Chan S, Dayneka N, Giguère P, Higgins N, Hills-Nieminen C, Kapler J, la Porte CJL, Nickel P, Park-Wyllie L, Quaia C, Robinson L, Sheehan N, Stone S, Sulz L, Yoong D. Role of the Pharmacist in Caring for Patients with HIV/AIDS: Clinical Practice Guidelines. Can J Hosp Pharm 2012; 65:125-45. [PMID: 22529405 PMCID: PMC3329905 DOI: 10.4212/cjhp.v65i2.1120] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Alice Tseng
- , BScPhm, PharmD, FSCHP, AAHIVP, is with the Immunodeficiency Clinic of the Toronto General Hospital, Toronto, Ontario
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Assessment of lopinavir pharmacokinetics with respect to developmental changes in infants and the impact on weight band-based dosing. Clin Pharmacol Ther 2011; 91:243-9. [PMID: 22190064 DOI: 10.1038/clpt.2011.218] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Improved antiretroviral therapies are needed for the treatment of HIV-infected infants, given the rapid progression of the disease and drug resistance resulting from perinatal exposure to antiretrovirals. We examined longitudinal pharmacokinetics (PK) data from a clinical trial of lopinavir/ritonavir (LPV/r) in HIV-infected infants in whom therapy was initiated at less than 6 months of age. A population PK analysis was performed using NONMEM to characterize changes in lopinavir (LPV) PK relating to maturational changes in infants, and to assess dosing requirements in this population. We also investigated the relationship between LPV PK and viral dynamic response. Age and ritonavir concentrations were the only covariates found to be significant. Population PK of LPV was characterized by high apparent clearance (CL/F) in young infants, which decreased with increasing age. Although younger infants had lower LPV concentrations, the viral dynamics did not correlate with initial LPV exposure. Monte Carlo simulations demonstrated that WHO weight band-based dosing recommendations predicted therapeutic LPV concentrations and provided drug exposure levels comparable to those resulting from US Food and Drug Administration (FDA)-suggested dosing regimens.
Collapse
|
40
|
Chadwick EG, Yogev R, Alvero CG, Hughes MD, Hazra R, Pinto JA, Robbins BL, Heckman BE, Palumbo PE, Capparelli EV. Long-term outcomes for HIV-infected infants less than 6 months of age at initiation of lopinavir/ritonavir combination antiretroviral therapy. AIDS 2011; 25:643-9. [PMID: 21297419 PMCID: PMC3068474 DOI: 10.1097/qad.0b013e32834403f6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the longitudinal pharmacokinetics, safety and efficacy of lopinavir/ritonavir (LPV/r) in HIV-infected infants initiating combination antiretroviral therapy (cART) between 2 weeks and 6 months of age. METHOD A prospective, open-label, multicenter Phase I/II study of LPV/r-based cART at a dose of 300/75 mg/m(2)/dose LPV/r twice daily. Intensive pharmacokinetic sampling at 12 months of age and quarterly predose LPV concentrations were collected and safety, virologic and immunologic responses were monitored every 4-12 weeks up to 252 weeks. RESULTS Thirty-one HIV-infected infants enrolled into two age cohorts, 14 days to <6 weeks and 6 weeks to <6 months; 29 completed ≥48 weeks of follow-up (median = 123 weeks, range 4-252). At 12 months of age, median LPV area under the curve was comparable for both age cohorts and similar to older children and adults. At week 48, 22 of 31 patients (71%) had HIV-1 RNA <400 copies/ml and 11 of 15 (73%) had <50 copies/ml; 29 of 31 achieved HIV-1 RNA <400 copies/ml on study treatment and 19 (66%) remained durably suppressed until the end of study; viral suppression correlated with a higher percentage of predose time points exceeding the LPV target of 1 μg/ml (92 vs. 71%, P = 0.002). CONCLUSION LPV/r at 300/75 mg/m(2)/dose as part of a cART regimen resulted in viral suppression through 96 weeks of treatment in >65% of young infants. Due to initially low LPV exposure in infants <6 weeks of age, frequent dose adjustment for weight gain is advisable and consideration should be given to studying a higher dose for very young infants.
Collapse
Affiliation(s)
- Ellen G Chadwick
- Division of Infectious Diseases, Children's Memorial Hospital, Northwestern University's Feinberg School of Medicine, Chicago, Illinois, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Klinklom A, Puthanakit T, Gorowara M, Phasomsap C, Kerr S, Sriheara C, Ananworanich J, Burger D, Ruxrungtham K, Pancharoen C. Low dose lopinavir/ritonavir tablet achieves adequate pharmacokinetic parameters in HIV-infected Thai adolescents. Antivir Ther 2011; 17:283-9. [DOI: 10.3851/imp1958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2011] [Indexed: 10/15/2022]
|
42
|
McKeegan K, Rutstein R, Lowenthal E. Postnatal infant HIV prophylaxis: a survey of U.S. practice. AIDS Patient Care STDS 2011; 25:1-4. [PMID: 21162689 DOI: 10.1089/apc.2010.0255] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
For neonates identified as at increased risk of acquiring HIV perinatally, the optimal postnatal prophylaxis regimen is not known. Current United States Public Health Service guidelines recognize that combination postnatal prophylaxis may be considered in some situations but that there are little data regarding the effectiveness and safety of any postnatal regimen besides zidovudine. The actual use of combination postnatal regimens in the United States has not previously been described. We conducted a national, Web-based survey between December 2009 and January 2010 to describe the percent of providers who prescribe combination postnatal prophylaxis, the antiretroviral combinations they used, and the risk factors that might elicit combination postnatal prophylaxis. 472 known or possible perinatal HIV providers were queried; 42% (n = 197) responded and 68% of respondents (134) were eligible to complete the survey. Sixty-two percent (n = 83) of participating providers reported use or recommendation of combination postnatal prophylaxis in the last year. Three drugs, zidovudine, lamivudine and nevirapine, comprised 77% of first-choice combination regimens. Lopinivir-ritonivir (LPV/RTV) was included in 16% of all reported regimens. Combination postnatal prophylaxis was strongly preferred in patient-based scenarios with additional risk factors for perinatal HIV transmission.
Collapse
Affiliation(s)
- Kathleen McKeegan
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Richard Rutstein
- Divisions of General Pediatrics and Special Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth Lowenthal
- Divisions of General Pediatrics and Special Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Martinez BL, Riordan FAI. Novel strategies in the use of lopinavir/ritonavir for the treatment of HIV infection in children. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2010; 2:59-67. [PMID: 22096385 PMCID: PMC3218684 DOI: 10.2147/hiv.s6616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Lopinavir/ritonavir (LPV/r) is considered by many as the first choice protease inhibitor (PI) for children. This co-formulation avoids the need for children to take ritonavir separately to “boost” the levels of lopinavir. LPV/r has high virologic potency, an excellent toxicity profile and a high barrier to the development of viral resistance. However, LPV/r has poor tolerability of the oral suspension (due to the poor taste of ritonavir), difficult dosing requirements and metabolic side effects, especially hyperlipidemia. The new tablet low-dose formulation (100/25 mg) may allow more convenient antiretroviral treatment in children. Novel strategies of LPV/r in childhood could maximize its advantages. For example, infants infected with HIV despite single dose Nevirapine after birth need effective combination antiretroviral treatment. This can be given using a higher dose of LPV/r with therapeutic drug monitoring. Other novel uses include once daily LPV/r regimens in older children and adolescents and lower doses of LPV/r in certain populations, which may decrease hyperlipidemia. Heavily pre-treated children might benefit from a double PI/r regimen which includes LPV/r. The high potency of LPV/r needs to be balanced with convenient regimens, to enhance adherence and decrease toxicity whenever possible. The aim of this review is to discuss the rationale behind these novel strategies of LPV/r use in pediatric antiretroviral treatment as well as their results and limitations.
Collapse
Affiliation(s)
- Beatriz Larru Martinez
- Laboratorio Inmuno-Biología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
44
|
Abstract
We report twin neonates who were born prematurely at 32 weeks of gestation to a mother with human immunodeficiency virus infection. One of the twins developed complete heart block and dilated cardiomyopathy related to lopinavir/ritonavir therapy, a boosted protease-inhibitor agent, while the other twin developed mild bradycardia. We recommend caution in the use of lopinavir/ritonavir in the immediate neonatal period.
Collapse
|
45
|
Van der Linden D, Callens S, Brichard B, Colebunders R. Pediatric HIV: new opportunities to treat children. Expert Opin Pharmacother 2009; 10:1783-91. [PMID: 19558340 DOI: 10.1517/14656560903012377] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Treating HIV-infected children remains a challenge due to a lack of treatment options, appropriate drug formulations and, in countries with limited resources, insufficient access to diagnostic tests and treatment. OBJECTIVE To summarize current data concerning new opportunities to improve the treatment of HIV-infected children. METHODS This review includes data from the most recently published peer-reviewed publications, guidelines or presentations at international meetings concerning new ways to treat HIV-infected children. RESULTS/CONCLUSIONS New WHO guidelines recommend starting combination antiretroviral treatment in all infants aged < 1 year. Although this is common practice in some high-income countries, implementation of these recommendations in countries with limited resources is still a challenge. There is still an important gap between the availability of licensed drugs in children compared with adults. There remains a need for further pharmacokinetic studies, and for more pediatric formulations of antiretroviral drugs with improved palatability.
Collapse
Affiliation(s)
- Dimitri Van der Linden
- Cliniques Universitaires UCL St Luc, Pediatrics Department, 10 Avenue Hippocrate, 1200 Brussels, Belgium.
| | | | | | | |
Collapse
|
46
|
Current awareness: Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2009. [DOI: 10.1002/pds.1652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|