101
|
Quincardete D, Sidat M, Seehra G, Mitha K, Cassia U, Passanduca A, Langa S, Martins S, Elias B, Buck WC. Rapid neurodevelopmental recovery after ART initiation in an infant with HIV encephalopathy. SAGE Open Med Case Rep 2022; 10:2050313X221142236. [DOI: 10.1177/2050313x221142236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022] Open
Abstract
While there is ample evidence that antiretroviral therapy (ART) can improve cognitive outcomes in older children living with HIV, encephalopathy in infants has historically been considered an advanced disease presentation with less likelihood of neurodevelopmental recovery on treatment. More recent studies suggest that timely ART can halt encephalopathic disease progression and even lead to symptom resolution. Here we present a case of an HIV-positive infant diagnosed with encephalopathy who experienced impressive and rapid improvement with a multi-disciplinary care approach that included physical and occupational therapy and ART.
Collapse
Affiliation(s)
| | | | - Gurpreet Seehra
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kiran Mitha
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | - W Chris Buck
- Universidade Eduardo Mondlane, Maputo, Mozambique
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
102
|
Attia EF, Moraa H, Maleche-Obimbo E, Wamalwa D, Gómez LA, Rylance S, Vundla R, Ferrand RA, Karr CJ, John-Stewart GC, Benki-Nugent SF. Most Early-Treated Children With Perinatally Acquired HIV Have Preserved Lung Function at School Age. J Acquir Immune Defic Syndr 2022; 89:69-76. [PMID: 34878436 PMCID: PMC8665226 DOI: 10.1097/qai.0000000000002823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Impaired lung function is common among older children with perinatally acquired HIV (PHIV) who initiated antiretroviral therapy (ART) late in childhood. We determined the prevalence of abnormal spirometry and cofactors for impaired lung function among school-age children with PHIV who initiated ART when aged 12 months or younger. SETTING Children who received early ART in the Optimizing Pediatric HIV-1 Therapy study in Kenya and underwent spirometry at school age. METHODS Forced expiratory volume in 1 second (FEV1) and forced vital capacity (FVC) were measured. Abnormal spirometry was defined as follows: obstructive (FEV1/FVC <1.64 z score [zFEV1/FVC]) and restricted (zFVC <1.64 with zFEV1/FVC ≥1.64). Characteristics, including anthropometric and HIV-related data, were ascertained in infancy and at school age. Caregiver carbon monoxide exposure served as a proxy for school-age child exposure. Linear regression determined associations of cofactors with lung function. RESULTS Among 40 children, the median age was 5 months at ART initiation and 8.5 years at spirometry. The mean zFEV1, zFVC, and zFEV1/FVC (SD) were 0.21 (1.35), 0.31 (1.22), and -0.24 (0.82), respectively. Five (13%) children had abnormal spirometry. Spirometry z scores were significantly lower among children with pre-ART pneumonia, WHO HIV stage 3/4, higher HIV RNA at 6 months after ART initiation, low anthropometric z scores, and higher carbon monoxide exposure. CONCLUSIONS Most of the children with PHIV who initiated ART at age 12 months or younger had normal spirometry, suggesting that ART in infancy preserved lung function. However, 13% had abnormal spirometry despite early ART. Modifiable factors were associated with impaired lung function, providing potential targets for interventions to prevent chronic lung disease.
Collapse
Affiliation(s)
- Engi F. Attia
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Hellen Moraa
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | | | - Dalton Wamalwa
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Laurén A. Gómez
- Department of Global Health, University of Washington, Seattle, WA
| | - Sarah Rylance
- Department of Global Health, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Rumbidzayi Vundla
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Rashida A. Ferrand
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Catherine J. Karr
- Department of Pediatrics and Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
103
|
Gie A, Morrison J, Maree D, Laughton B, Browne SH, Cotton MF, Goussard P, Innes S. Childhood lung function following perinatal HIV infection and early antiretroviral therapy initiation: a cross-sectional study. ERJ Open Res 2022; 8:00691-2021. [PMID: 35350281 PMCID: PMC8943286 DOI: 10.1183/23120541.00691-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/02/2022] [Indexed: 11/09/2022] Open
Abstract
Despite the introduction of antiretroviral therapy (ART), HIV-associated pulmonary complications remain prevalent in children following perinatal HIV infection. In the post-ART era the incidence of opportunistic infections has decreased; however, non-infectious complications including diminished lung function are common. It is unclear whether early initiation of ART influences lung function later in life. We performed a cross-sectional study examining pulmonary function tests (PFT) (spirometry, plethysmography, carbon monoxide diffusing capacity) in HIV-unexposed (HU), HIV-exposed-uninfected (HEU) and perinatally HIV-infected children on early ART (HIV+) recruited from the Cape Town arms of the CHER and IMPAACT 1060 trials. PFT was performed once children could participate (October 2013 to January 2020). Global Lung Initiative reference software was used for Z-standardisation of lung function by sex, age and height. In total 394 children (HU n=90, HEU n=162, HIV+ n=142) underwent PFT, median age 8.7 (IQR 7.7-9.8) years. HIV+ had ART initiated at a median age of 17.6 (8.0-36.7) weeks. Forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and FEV1/FVC Z-scores were similar in all groups. Plethysmography demonstrated air-trapping with increased total lung capacity (TLC), functional residual capacity, residual volume (RV) and RV/TLC Z-scores in HIV+. There were no differences in alveolar volume; however, diffusing capacity was increased in HIV+. Our findings indicate that following perinatal HIV infection, early ART may attenuate HIV-associated lung disease and is associated with normal childhood spirometry. However plethysmography demonstrates that small airway dysfunction is more pronounced in HIV+. Longitudinal follow-up is required to assess if these children are at risk of obstructive airway disease later in life.
Collapse
Affiliation(s)
- Andre Gie
- Dept of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Julie Morrison
- Dept of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - David Maree
- Dept of Medicine, Division of Pulmonology, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Barbara Laughton
- Dept of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
- Family Center for Research with Ubuntu (FAMCRU), Stellenbosch University, Cape Town, South Africa
| | - Sara H. Browne
- Dept of Medicine, University of California San Diego, San Diego, CA, USA
| | - Mark F. Cotton
- Dept of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
- Family Center for Research with Ubuntu (FAMCRU), Stellenbosch University, Cape Town, South Africa
| | - Pierre Goussard
- Dept of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
| | - Steve Innes
- Dept of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Tygerberg Hospital, Stellenbosch University, Cape Town, South Africa
- Family Center for Research with Ubuntu (FAMCRU), Stellenbosch University, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
104
|
Dube-Pule A, Zanoni BC, Connolly C, Shabangu M, Archary M. Evaluation of an SMS-based mHealth intervention to enhance early infant diagnosis follow-up testing and assessment of postnatal prophylaxis. South Afr J HIV Med 2021; 22:1301. [PMID: 34917408 PMCID: PMC8661286 DOI: 10.4102/sajhivmed.v22i1.1301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/03/2021] [Indexed: 11/18/2022] Open
Abstract
Background Adherence to infant antiretroviral (ARV) postnatal prophylaxis and early infant diagnosis (EID) uptake is low in Africa. Promoting EID and adherence are necessary for this age group. Objectives We evaluated an SMS-based mobile health (mHealth) intervention to enhance adherence to ARV prophylaxis and knowledge of EID and prevention of mother-to-child transmission (PMTCT) among high-risk and low-risk mother–infant pairs. Method Two hundred and fifty-one mothers were recruited from King Edward VIII Hospital between December 2018 and October 2019. Participant information was captured, and SMS reminders were sent postnatally to promote immunisation attendance. Follow-up HIV polymerase chain reaction (PCR) test results were reviewed, and telephonic interviews were utilised for qualitative data. Results In all, 73.3% of infants had HIV PCR tests performed at 10 weeks. This high rate could be attributed to the mHealth intervention as this is considerably higher than other national studies, though not statistically significant compared to rates reported in the district at the same time. Factors that have impacted follow-up EID rates include poor maternal knowledge of EID time points and inadequate implementation of national PMTCT protocols. High-risk mothers were younger, commenced antenatal clinic visit later, were less knowledgeable on prophylaxis and have lower-birthweight infants than lower-risk mothers. Conclusion mHealth can play an important role in improving EID by increasing maternal knowledge. Further studies should focus on whether maternal education over an mHealth platform can increase knowledge on PMTCT and subsequently increase EID.
Collapse
Affiliation(s)
- Anele Dube-Pule
- Department of Paediatrics and Child Health, Faculty of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Brian C Zanoni
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, United States of America
| | - Cathy Connolly
- School of Public Health, University of KwaZulu-Natal, Durban, South Africa
| | - Majahonkhe Shabangu
- Sawubona Health Inc., Malden, Massachusetts, United States of America.,Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
| | - Moherndran Archary
- Department of Paediatrics and Child Health, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Department of Paediatrics, King Edward VIII Hospital, Durban, South Africa
| |
Collapse
|
105
|
Mellqvist H, Saggers RT, Elfvin A, Hentz E, Ballot DE. The effects of exposure to HIV in neonates at a referral hospital in South Africa. BMC Pediatr 2021; 21:485. [PMID: 34727920 PMCID: PMC8565056 DOI: 10.1186/s12887-021-02969-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
Background Fewer infants are infected with HIV through mother-to-child transmission, making HIV-exposed but uninfected (HEU) infants a growing population. HIV-exposure seems to affect immunology, early growth and development, and is associated with higher morbidity and mortality rates. Currently, there is a lack of information regarding the clinical effects of HIV-exposure during the neonatal period. Objectives To identify a possible difference in mortality and common neonatal morbidities in HEU neonates compared to HIV-unexposed neonates. Methods This was a retrospective, descriptive study of all neonates admitted to the neonatal unit at Charlotte Maxeke Johannesburg Academic Hospital between 1 January 2017 and 31 December 2018. HEU neonates were compared to HIV-unexposed neonates. Results There were 3236 neonates included, where 855 neonates were HEU. The HEU neonates had significantly lower birth weight and gestational age. The HEU neonates had higher rates of neonatal sepsis (19.8% vs 14.2%, OR 1.49, p < 0.001), specifically for late onset sepsis, and required more respiratory support. NCPAP and invasive ventilation was more common in the HEU group (36.3% vs 31.3% required NCPAP, p = 0.008, and 20.1% vs 15,0% required invasive ventilation, p < 0.001). Chronic lung disease was more common among HIV-exposed neonates (12.2% vs 8.7%, OR 1.46, p = 0.003). The difference in mortality rates between the study groups was not significant (10.8% of HEU neonates and 13.3% of HIV-unexposed). Conclusions HEU neonates had higher rates of neonatal sepsis, particularly late-onset sepsis, required more respiratory support and had higher rates of chronic lung disease. Mortality of HEU neonates was not different HIV-unexposed neonates.
Collapse
Affiliation(s)
- Helena Mellqvist
- Futurum, County Hospital Ryhov, Jonkoping, Sweden.,Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Robin T Saggers
- School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. .,Department of Paediatrics and Child Health, Charlotte Maxeke Johannesburg Academic Hospital, Jubilee Road, Parktown, Johannesburg, South Africa.
| | - Anders Elfvin
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Department of Pediatrics, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Elisabet Hentz
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Department of Pediatrics, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Daynia E Ballot
- School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
106
|
Amin O, Powers J, Bricker KM, Chahroudi A. Understanding Viral and Immune Interplay During Vertical Transmission of HIV: Implications for Cure. Front Immunol 2021; 12:757400. [PMID: 34745130 PMCID: PMC8566974 DOI: 10.3389/fimmu.2021.757400] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Despite the significant progress that has been made to eliminate vertical HIV infection, more than 150,000 children were infected with HIV in 2019, emphasizing the continued need for sustainable HIV treatment strategies and ideally a cure for children. Mother-to-child-transmission (MTCT) remains the most important route of pediatric HIV acquisition and, in absence of prevention measures, transmission rates range from 15% to 45% via three distinct routes: in utero, intrapartum, and in the postnatal period through breastfeeding. The exact mechanisms and biological basis of these different routes of transmission are not yet fully understood. Some infants escape infection despite significant virus exposure, while others do not, suggesting possible maternal or fetal immune protective factors including the presence of HIV-specific antibodies. Here we summarize the unique aspects of HIV MTCT including the immunopathogenesis of the different routes of transmission, and how transmission in the antenatal or postnatal periods may affect early life immune responses and HIV persistence. A more refined understanding of the complex interaction between viral, maternal, and fetal/infant factors may enhance the pursuit of strategies to achieve an HIV cure for pediatric populations.
Collapse
Affiliation(s)
- Omayma Amin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Jenna Powers
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine M. Bricker
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| |
Collapse
|
107
|
Dobe IS, Mocumbi AO, Majid N, Ayele B, Browne SH, Innes S. Earlier antiretroviral initiation is independently associated with better arterial stiffness in children living with perinatally acquired HIV with sustained viral suppression in Mozambique. South Afr J HIV Med 2021; 22:1282. [PMID: 34858652 PMCID: PMC8603154 DOI: 10.4102/sajhivmed.v22i1.1282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cardiovascular disease is a major driver of morbidity and mortality in adults living with HIV. The drivers of cardiovascular disease in children living with perinatally acquired HIV (PHIV) with sustained HIV viral suppression are unclear. OBJECTIVES We explored the contribution of HIV-specific risk factors to arterial stiffness independently of traditional risk factors (metabolic syndrome [MetS]) in prepubertal children with PHIV with sustained viral suppression in a low-income country in Africa. METHOD For this cross-sectional analysis, arterial stiffness was assessed by pulse wave velocity z-score (PWVz), measured using a Vicorder device. Metabolic syndrome components were measured. We retrospectively collected the antiretroviral therapy (ART) exposures, HIV stage, CD4 count and HIV viral load. A multivariate linear regression model was constructed for MetS components, retaining age and gender as obligatory variables. We then added HIV-related metrics to assess whether these had an independent or additive effect. RESULTS We studied 77 virally suppressed children with PHIV without evidence of cardiovascular disease (from medical history and physical examination). In the initial model, the PWVz was independently associated with each MetS component. The PWVz was higher in participants with proportionally greater visceral fat (waist/height ratio), elevated lipids (triglyceride/high-density lipoprotein ratio) and insulin resistance (log homeostatic model assessment [HOMA]). The addition of age at ART initiation increased the model R 2 value from 0.36 to 0.43. In the resulting model, younger age at ART initiation was independently associated with a better PWVz (P < 0.001). CONCLUSION Earlier ART initiation was independently associated with lower large artery stiffness. This effect was independent of the effect of elevated lipids, visceral fat and insulin resistance.
Collapse
Affiliation(s)
- Igor S. Dobe
- Division Non-Communicable Diseases, Instituto Nacional de Saúde, Maputo, Mozambique
| | - Ana O. Mocumbi
- Division Non-Communicable Diseases, Instituto Nacional de Saúde, Maputo, Mozambique
- Department of Internal Medicine, Faculty of Medicine, University Eduardo Mondlane, Maputo, Mozambique
| | | | - Birhanu Ayele
- Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Science, Stellenbosch University, Stellenbosch, South Africa
| | - Sara H. Browne
- Department of Medicine, Division of Infectious Diseases, University of California San Diego, San Diego, United States of America
| | - Steve Innes
- Department of Paediatrics and Child Health, Family Centre for Research with Ubuntu (FAMCRU), Stellenbosch University, Cape Town, South Africa
- Desmond Tutu HIV Centre, University of Cape Town, South Africa
| |
Collapse
|
108
|
Tarancón-Diez L, Rull A, Herrero P, Vazquez-Alejo E, Peraire J, Guillén S, Navarro-Gomez ML, Viladés C, Muñoz-Fernandez MÁ, Vidal F. Early antiretroviral therapy initiation effect on metabolic profile in vertically HIV-1-infected children. J Antimicrob Chemother 2021; 76:2993-3001. [PMID: 34463735 DOI: 10.1093/jac/dkab277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Early combined antiretroviral treatment (cART) in perinatally acquired HIV-1 children has been associated with a rapid viral suppression, small HIV-1 reservoir size and reduced mortality and morbidity. Immunometabolism has emerged as an important field in HIV-1 infection offering both relevant knowledge regarding immunopathogenesis and potential targets for therapies against HIV-1. OBJECTIVES To characterize the proteomic, lipidomic and metabolomic profile of HIV-1-infected children depending on their age at cART initiation. PATIENTS AND METHODS Plasma samples from perinatally HIV-1-infected children under suppressive cART who initiated an early cART (first 12 weeks after birth, EARLY, n = 10) and late cART (12-50 weeks after birth, LATE, n = 10) were analysed. Comparative plasma proteomics, lipidomics and metabolomics analyses were performed by nanoLC-Orbitrap, UHPLC-qTOF and GC-qTOF, respectively. RESULTS Seven of the 188 proteins identified exhibited differences comparing EARLY and LATE groups of HIV-1-infected children. Despite no differences in the lipidomic (n = 115) and metabolomic (n = 81) profiles, strong correlations were found between proteins and lipid levels as well as metabolites, including glucidic components and amino acids, with clinical parameters. The ratio among different proteins showed high discriminatory power of EARLY and LATE groups. CONCLUSIONS Protein signature show a different proinflammatory state associated with a late cART introduction. Its associations with lipid levels and the relationships found between metabolites and clinical parameters may potentially trigger premature non-AIDS events in this HIV-1 population, including atherosclerotic diseases and metabolic disorders. Antiretroviral treatment should be started as soon as possible in perinatally acquired HIV-1-infected children to prevent them from future long-life complications.
Collapse
Affiliation(s)
- Laura Tarancón-Diez
- Molecular Immunology Laboratory, Hospital General Universitario Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Anna Rull
- Universitat Rovira i Virgili, Tarragona, Spain.,Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.,Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Pol Herrero
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Ciències Òmiques (Unitat Mixta de Eurecat-Universitat Rovira i Virgili), Infraestructura Científico-Tècnica Singular (ICTS), Reus, Spain
| | - Elena Vazquez-Alejo
- Molecular Immunology Laboratory, Hospital General Universitario Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Joaquim Peraire
- Universitat Rovira i Virgili, Tarragona, Spain.,Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.,Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Sara Guillén
- Department of Pediatrics, Hospital Universitario de Getafe, Madrid, Spain
| | - Maria Luisa Navarro-Gomez
- Hospital General Universitario Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Consuelo Viladés
- Universitat Rovira i Virgili, Tarragona, Spain.,Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.,Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Mª Ángeles Muñoz-Fernandez
- Molecular Immunology Laboratory, Hospital General Universitario Gregorio Marañón, Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain.,Spanish HIV-HGM BioBank, Madrid, Spain
| | - Francesc Vidal
- Universitat Rovira i Virgili, Tarragona, Spain.,Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.,Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| |
Collapse
|
109
|
Payne H, Chan MK, Watters SA, Otwombe K, Hsiao NY, Babiker A, Violari A, Cotton MF, Gibb DM, Klein NJ. Early ART-initiation and longer ART duration reduces HIV-1 proviral DNA levels in children from the CHER trial. AIDS Res Ther 2021; 18:63. [PMID: 34587974 PMCID: PMC8482761 DOI: 10.1186/s12981-021-00389-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/16/2021] [Indexed: 11/21/2022] Open
Abstract
Background Reduction of the reservoir of latent HIV-infected cells might increase the possibility of long-term remission in individuals living with HIV. We investigated factors associated with HIV-1 proviral DNA levels in children receiving different antiretroviral therapy (ART) strategies in the children with HIV early antiretroviral therapy (CHER) trial. Methods Infants with HIV < 12 weeks old with CD4% ≥ 25% were randomized in the CHER trial to early limited ART for 40 or 96 weeks (ART-40 W, ART-96 W), or deferred ART (ART-Def). For ART-Def infants or following ART interruption in ART-40 W/ART-96 W, ART was started/re-started for clinical progression or CD4% < 25%. In 229 participants, HIV-1 proviral DNA was quantified by PCR from stored peripheral blood mononuclear cells from children who had received ≥ 24 weeks ART and two consecutive undetectable HIV-1 RNA 12–24 weeks apart. HIV-1 proviral DNA was compared between ART-Def and ART-96 W at week 96, and in all arms at week 248. Factors associated with HIV-1 proviral DNA levels were evaluated using linear regression. Findings Longer duration of ART was significantly associated with lower HIV-1 proviral DNA at both 96 (p = 0.0003) and 248 weeks (p = 0.0011). Higher total CD8 count at ART initiation was associated with lower HIV-1 proviral DNA at both 96 (p = 0.0225) and 248 weeks (p = 0.0398). Week 248 HIV-1 proviral DNA was significantly higher in those with positive HIV-1 serology at week 84 than those with negative serology (p = 0.0042). Intepretation Longer ART duration is key to HIV-1 proviral DNA reduction. Further understanding is needed of the effects of “immune-attenuation” through early HIV-1 exposure. Funding Wellcome Trust, National Institutes of Health, Medical Research Council.
Collapse
|
110
|
Lisboa A, Thorp M, Lambo L, Chaúque S, Elias B, Domingos C, Macassa E, Buck WC. Negative Rapid Serological Tests in an HIV-Infected Infant: A Call for Improved Inpatient Provider-Initiated Testing and Counseling Beginning With Breastfeeding Mothers. J Pediatric Infect Dis Soc 2021; 10:883-885. [PMID: 34037237 DOI: 10.1093/jpids/piab021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/16/2021] [Indexed: 11/14/2022]
Abstract
Rapid serological tests are unreliable for the diagnosis of HIV exposure and infection in infants. This case reports an HIV-infected infant with a delayed diagnosis due to multiple negative serological tests, highlighting the importance of maternal testing for provider-initiated testing and counseling in hospitalized infants.
Collapse
Affiliation(s)
- Anselmo Lisboa
- Department for Pediatrics, Hospital Central de Maputo, Maputo, Mozambique
| | - Marguerite Thorp
- Department for Pediatrics, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, USA
| | - Luisa Lambo
- Department for Pediatrics, Hospital Central de Maputo, Maputo, Mozambique
| | - Sílvia Chaúque
- Department for Pediatrics, Hospital Central de Maputo, Maputo, Mozambique
| | - Beatriz Elias
- Department for Pediatrics, Hospital Central de Maputo, Maputo, Mozambique
| | | | - Eugénia Macassa
- Department for Pediatrics, Hospital Central de Maputo, Maputo, Mozambique
| | - W Chris Buck
- Department for Pediatrics, Hospital Central de Maputo, Maputo, Mozambique.,Department for Pediatrics, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, USA
| |
Collapse
|
111
|
Acharya S, Palkar A, Sayed AP, Setia MS. Retrospective cohort analysis of survival of children living with HIV/AIDS in Mumbai, India. BMJ Open 2021; 11:e050534. [PMID: 34518268 PMCID: PMC8438856 DOI: 10.1136/bmjopen-2021-050534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES To understand the survival in a cohort of children living with HIV/AIDS (CLHAs) and to study the factors associated with survival in CLHAs in government antiretroviral therapy (ART) centres in Mumbai, India. DESIGN This is a retrospective cohort analysis. SETTING Data from electronic ART records of children from 15 ART centres in Mumbai, Maharashtra, India. PARTICIPANTS 2224 CLHAs registered in one of these ART centres from 2004 until October 2019. CLHAs up to the age of 18 at the time of registration were considered for these analyses. PRIMARY AND SECONDARY OUTCOMES We accessed the following data: date of test, date of initiation of ART, date of last follow-up, age at the time of registration, gender, potential route of infection, baseline CD4 counts, ART regimen, adherence and presence of co-infection (TB). We estimated the survival probabilities, plotted the Kaplan-Meier survival graphs and estimated HRs for mortality. RESULTS The mortality rate in our population was 22.75 (95% CI 20.02 to 25.85) per 1000 person-years. The 1-year survival was 0.92 (95% CI 0.91 to 0.93), 0.89 (95% CI 0.88 to 0.91) at 5 years and 0.85 (95% CI 0.83 to 0.87) at 10 years after initiation of ART. Children with adherence less than 80% had lowest survival in the first year (0.54, 95% CI 0.46 to 0.61). It reduced drastically at 5 and 10 years. After adjusting for demographic and clinical parameters, mortality was associated with poor adherence (<80%) (HR 11.70, 95% CI 8.82 to 15.53; p<0.001). However, CD4 counts of greater than 200 and age more than 1 year were protective. CONCLUSIONS Poor adherence to ART and low CD4 counts were significantly associated with higher mortality. Adherence counselling should be an important component of CLHA monitoring in all ART centres. It is also important to identify children early in the infection and start ART medications appropriately.
Collapse
Affiliation(s)
| | - Amol Palkar
- Mumbai Districts AIDS Control Society, Mumbai, India
| | - Anwar Parvez Sayed
- UW International Training and Education Center for Health, New Delhi, India
| | | |
Collapse
|
112
|
Zhang J, Atkins DL, Wagner AD, Njuguna IN, Neary J, Omondi VO, Otieno VA, Atieno WO, Odhiambo M, Wamalwa DC, John-Stewart G, Slyker JA, Weiner BJ, Beima-Sofie K. Financial Incentives for Pediatric HIV Testing (FIT): Caregiver Insights on Incentive Mechanisms, Focus Populations, and Acceptability for Programmatic Scale Up. AIDS Behav 2021; 25:2661-2668. [PMID: 34170433 DOI: 10.1007/s10461-021-03356-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 11/30/2022]
Abstract
Children living with HIV experience gaps in HIV testing globally; scaling up evidence-based testing strategies is critical for preventing HIV-related mortality. Financial incentives (FI) were recently demonstrated to increase uptake of pediatric HIV testing. As part of this qualitative follow-up study to the FIT trial (NCT03049917) conducted in Kenya, 54 caregivers participated in individual interviews. Interview transcripts were analyzed to identify considerations for scaling up FI for pediatric testing. Caregivers reported that FI function by directly offsetting costs or nudging caregivers to take action sooner. Caregivers found FI to be feasible and acceptable for broader programmatic implementation, and supported use for a variety of populations. Some concerns were raised about unintended consequences of FI, including caregivers bringing ineligible children to collect incentives and fears about the impact on linkage to care and retention if caregivers become dependent on FI.
Collapse
Affiliation(s)
- Junyi Zhang
- Department of Health Services, University of Washington, Seattle, WA, 98195, USA.
| | - Dana L Atkins
- Department of Global Health, University of Washington, UW Box #351620, Seattle, WA, 98195, USA
| | - Anjuli D Wagner
- Department of Global Health, University of Washington, UW Box #351620, Seattle, WA, 98195, USA
| | - Irene N Njuguna
- Department of Global Health, University of Washington, UW Box #351620, Seattle, WA, 98195, USA
- Research and Programs, Kenyatta National Hospital, Ngong Road, Nairobi, 00202, Kenya
| | - Jillian Neary
- Department of Epidemiology, University of Washington, Seattle, WA, 98104, USA
| | - Vincent O Omondi
- Pediatric Research Consortium, Kenya Pediatric Association, Nairobi, Kenya
| | - Verlinda A Otieno
- Pediatric Research Consortium, Kenya Pediatric Association, Nairobi, Kenya
| | - Winnie O Atieno
- Pediatric Research Consortium, Kenya Pediatric Association, Nairobi, Kenya
| | - Merceline Odhiambo
- Pediatric Research Consortium, Kenya Pediatric Association, Nairobi, Kenya
| | - Dalton C Wamalwa
- Department of Pediatrics, University of Nairobi, Nairobi, 00202, Kenya
| | - Grace John-Stewart
- Department of Global Health, University of Washington, UW Box #351620, Seattle, WA, 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA, 98104, USA
- Department of Medicine, University of Washington, Seattle, WA, 98104, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98104, USA
| | - Jennifer A Slyker
- Department of Global Health, University of Washington, UW Box #351620, Seattle, WA, 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA, 98104, USA
| | - Bryan J Weiner
- Department of Health Services, University of Washington, Seattle, WA, 98195, USA
- Department of Global Health, University of Washington, UW Box #351620, Seattle, WA, 98195, USA
| | - Kristin Beima-Sofie
- Department of Global Health, University of Washington, UW Box #351620, Seattle, WA, 98195, USA
| |
Collapse
|
113
|
Sutcliffe CG, Moyo N, Schue JL, Mutanga JN, Hamahuwa M, Munachoonga P, Maunga S, Thuma PE, Moss WJ. The NSEBA Demonstration Project: implementation of a point-of-care platform for early infant diagnosis of HIV in rural Zambia. Trop Med Int Health 2021; 26:1036-1046. [PMID: 33999480 PMCID: PMC8416694 DOI: 10.1111/tmi.13627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To describe the experience and resource requirements of implementing point-of-care testing for early infant diagnosis of HIV in rural Zambia. METHODS A demonstration project was conducted using a hub-and-spoke model in 2018-2019 at five clinics in rural Zambia. Two testing hubs were established, and all HIV-exposed infants were tested with the GeneXpert system. Data on costs, turnaround times and test results were collected. RESULTS Seven hundred and eighty six tests were conducted. At the hubs, results were available a median of 2.4 (IQR: 2.1, 2.8) hours after sample collection and most mothers (84%) received same-day results. At the spoke facilities, results were available a median of 9 days (IQR: 7, 12) after sample collection and provided to the mother a median of 16 days (IQR: 10, 28) after sample collection. Eleven children tested positive, and 9 (82%) started treatment a median of 13 days (IQR: 7, 21) after sample collection and on the day mothers received results. In contrast, results from matching samples sent for routine testing were available a median of 38 days (IQR: 27, 61) after sample collection and provided to the mother a median of 91 days (IQR: 47, 135) after sample collection. CONCLUSIONS Implementing point-of-care testing in a network of rural health centres in Zambia required significant initial and ongoing investment in infrastructure, training and supervision. However, point-of-care testing can rapidly diagnose HIV-infected infants, so they can benefit from early treatment.
Collapse
Affiliation(s)
| | | | - Jessica L. Schue
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | | | | | | | - Philip E. Thuma
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Macha Research Trust, Choma, Zambia
| | - William J. Moss
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
114
|
Insights from Clonal Expansion and HIV Persistence in Perinatal Infections. mBio 2021; 12:e0098321. [PMID: 34425702 PMCID: PMC8406253 DOI: 10.1128/mbio.00983-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The latent HIV reservoir forms early in the course of infection and is maintained for life despite effective antiretroviral treatment (ART), including early treatment. Perinatal HIV infection presents a unique opportunity to limit seeding of the reservoir through early ART. However, a greater understanding of the persistence of the integrated proviruses is needed for targeting the residual proviruses that form barriers to cure. A study was performed by Bale and Katusiime et al. (M. J. Bale, M. G. Katusiime, D. Wells, X. Wu, et al., mBio 12:e00568-21, 2021, https://doi.org/10.1128/mBio.00568-21) using in-depth integration site analysis in 11 children before ART and after up to nine years of ART. They have identified early development of long-lived proviruses, although the replication competence is unknown. A small fraction of cells bearing integrated proviruses clonally expand early during infection and persist. Integration in the oncogenes STAT5B and BACH2 were also found; these findings confirm the early development of clonal proliferation in perinatal HIV infection despite early effective ART, with a propensity for oncogenes.
Collapse
|
115
|
A new highly sensitive single-tube nested real-time PCR assay: Clinical utility in perinatal HIV-1 diagnosis. J Virol Methods 2021; 297:114273. [PMID: 34454987 DOI: 10.1016/j.jviromet.2021.114273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
Real time PCR is one of the major tools for molecular diagnosis, however not always reaches the required sensitivity, especially in detecting early infectious disease. To overcome this problem, nested PCR is commonly performed, since it is highly sensitive, but it is time-consuming, prone to cross-contamination and difficult to standardize. Therefore, we developed a sensitive and specific single-tube nested real-time PCR (STN-real-time PCR) assay and evaluated its clinical utility on early infant HIV-1 diagnosis (EID). The STN-real-time PCR enables the simultaneous amplification of four HIV-1 specific amplicons by the use of an internal and external pair of primers targeting ltr/gag region, and another one corresponding to human albumin as an internal control. Thermocycling had different annealing temperatures to favor the sequential use of each pair of primers, and included an initial touchdown step to broaden specificity and increase sensitivity. Finally, HIV-1 was detected by melting curve analysis. A total of 234 samples collected retrospectively and prospectively from HIV-1 exposed infants aged <18 months were used to evaluate the performance of the assay and compare it with a routine diagnostic nested-multiplex PCR. The developed assay had a limit of detection of 3 copies of HIV-1 DNA per reaction and had a sensitivity of 31 % more than routine diagnostic nested-multiplex PCR when testing samples near delivery. In conclusion, we developed a new assay by turning a conventional nested-PCR into a faster, more sensitive and feasible STN-real-time PCR assay for EID and potentially useful for detection of pathogens with variable genomes and present in low copy numbers.
Collapse
|
116
|
Madzime J, Cotton MF, Laughton B, van der Kouwe AJW, Meintjes EM, Jankiewicz M. Altered White Matter Tracts in the Somatosensory, Salience, Motor, and Default Mode Networks in 7-Year-Old Children Living with Human Immunodeficiency Virus: A Tractographic Analysis. Brain Connect 2021; 12:302-319. [PMID: 34107770 PMCID: PMC9131360 DOI: 10.1089/brain.2020.0948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction: Even with the increased access and early initiation of combination antiretroviral therapy, children with perinatally acquired human immunodeficiency virus (CPHIV) continue to demonstrate white matter alterations. Children perinatally HIV-exposed, but uninfected (CHEU) alike show differences in white matter integrity compared with children who are HIV-unexposed and uninfected (CHUU). Objectives: Mapping white matter connections that link gray matter regions that form resting-state (RS) functional networks may demonstrate whether structural and functional connectivity alterations in HIV infection and exposure may be related. We hypothesized reduced structural connectivity in CPHIV within the default mode network (DMN), visual, ventral DMN (vDMN), somatosensory, salience, auditory, motor, executive, basal ganglia, and posterior DMN (pDMN). We also hypothesized that CHEU will have increased structural connectivity compared with CHUU in the vDMN, somatosensory, pDMN, dorsal attention, salience, auditory, motor and basal ganglia. Methods: Study participants were 61 seven-year-old CPHIV and 46 age-matched children who are HIV uninfected (CHU) (19 CHEU). We used diffusion tensor imaging-based tractography to investigate white matter connections that link gray matter regions within RS functional networks. Results: We found altered white matter integrity in the somatosensory, salience, default mode, and motor networks of CPHIV compared with CHU. The superior temporal cortex, superior frontal cortex, and putamen were affected in all four networks and have also been reported to demonstrate morphological alterations in the same cohort. In CHEU, white matter integrity was higher in the visual network, pDMN, and motor network compared with CHUU. Conclusion: Our results suggest that altered white matter integrity may influence gray matter morphology and functional network alterations. Impact statement The long-term effects of human immunodeficiency virus (HIV) and exposure on the developing brain in the combination antiretroviral therapy era are still not well known. We use diffusion tensor imaging-based tractography to explore these effects on white matter connections that link gray matter regions within functional networks. Our findings provide a context for HIV-associated white matter and connectivity abnormalities.
Collapse
Affiliation(s)
- Joanah Madzime
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,Neurosciences Institute, University of Cape Town, Cape Town, South Africa
| | - Mark F Cotton
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,Neurosciences Institute, University of Cape Town, Cape Town, South Africa
| | - Barbara Laughton
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Stellenbosch, South Africa
| | - Andre J W van der Kouwe
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Ernesta M Meintjes
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,Neurosciences Institute, University of Cape Town, Cape Town, South Africa.,Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa
| | - Marcin Jankiewicz
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,Neurosciences Institute, University of Cape Town, Cape Town, South Africa.,Cape Universities Body Imaging Centre, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
117
|
Yee WL, Htay H, Mohamed Y, Nightingale CE, Tin HH, Thein W, Kyaw LL, Yee WW, Aye MM, Badman SG, Vallely AJ, Anderson D, Kelly-Hanku A, Luchters S. Operational experiences associated with the implementation of near point-of-care early infant diagnosis of HIV in Myanmar: a qualitative study. BMC Health Serv Res 2021; 21:863. [PMID: 34425814 PMCID: PMC8383426 DOI: 10.1186/s12913-021-06797-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 07/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background Timely diagnosis and early initiation of life-saving antiretroviral therapy are critical factors in preventing mortality among HIV-infected infants. However, resource-limited settings experience numerous challenges associated with centralised laboratory-based testing, including low rates of testing, complex sample referral pathways and unacceptably long turnaround times for results. Point-of-care (POC) HIV testing for HIV-exposed infants can enable same-day communication of results and early treatment initiation for HIV-infected infants. However, complex operational issues and service integration can limit utility and must be well understood prior to implementation. We explored and documented the challenges and enabling factors in implementing the POC Xpert® HIV-1 Qual test (Cepheid, Sunnyvale, CA, USA) for early infant diagnosis (EID) as part of routine services in four public hospitals in Myanmar. Methods This sub-study was part of a randomised controlled stepped-wedge trial (Australian and New Zealand Clinical Trials Registry, number 12616000734460) designed to investigate the impact of POC testing for EID in Myanmar and Papua New Guinea. Infants recruited during the intervention phase underwent POC testing at the participating hospitals as part of routine care. Semi-structured interviews with 23 caregivers, 12 healthcare providers and 10 key informants were used to explore experiences of POC-EID testing. The research team and hospital staff documented and discussed implementation challenges throughout the study. Results Overall, caregivers and healthcare workers were satisfied with the short turnaround time of the POC test. Occasional delays in POC testing were mostly attributable to late receipt of samples by laboratory technicians and communication constraints among healthcare staff. Hospital staff valued technical assistance from the research group and the National Health Laboratory. Despite staff shortages and infrastructure challenges such as unreliable electricity supply and cramped space, healthcare workers and caregivers found the implementation of the POC test to be feasible at pilot sites. Conclusions As plans for national scale-up evolve, there needs to be a continual focus on staff training, communication pathways and infrastructure. Other models of care, such as allowing non-laboratory-trained personnel to perform POC testing, and cost effectiveness should also be evaluated. Supplementary Information The online version contains supplementary material available at 10.1186/s12913-021-06797-3.
Collapse
Affiliation(s)
| | - Hla Htay
- Burnet Institute, Yangon, Myanmar
| | - Yasmin Mohamed
- Burnet Institute, Melbourne, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Claire E Nightingale
- Burnet Institute, Melbourne, Australia.,Monash Rural Health, Monash University, Bendigo, Australia
| | | | - Win Thein
- National Health Laboratory, Yangon, Myanmar
| | | | | | | | - Steven G Badman
- The Kirby Institute for Infection and Immunity in Society, UNSW Sydney, Sydney, Australia
| | - Andrew J Vallely
- The Kirby Institute for Infection and Immunity in Society, UNSW Sydney, Sydney, Australia
| | | | - Angela Kelly-Hanku
- The Kirby Institute for Infection and Immunity in Society, UNSW Sydney, Sydney, Australia.,Sexual and Reproductive Health Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Stanley Luchters
- Burnet Institute, Melbourne, Australia. .,School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia. .,Department of Population Health, Medical College, Aga Khan University, Nairobi, Kenya. .,Department of Public Health and Primary Care, Ghent University, Ghent, Belgium.
| |
Collapse
|
118
|
Hans L, von Allmen N, Edelmann A, Hofmann J, Nilsson AY, Simon CO, Seiverth B, Gohl P, Carmona S. Early Diagnosis of HIV-1 and HIV-2 Using Cobas HIV-1/HIV-2 Qualitative Test: A Novel Qualitative Nucleic Acid Amplification Test for Plasma, Serum, and Dried Blood Spot Specimens. J Acquir Immune Defic Syndr 2021; 87:1187-1195. [PMID: 33883470 PMCID: PMC8263138 DOI: 10.1097/qai.0000000000002713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/25/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Nucleic acid amplification tests (NATs) minimize the time from HIV infection to diagnosis, reducing transmission during acute HIV. NATs are especially useful for diagnosing HIV in children younger than 18 months and discriminating between HIV-1 and HIV-2. METHODS We evaluated the performance of the cobas HIV-1/HIV-2 qualitative (cobas HIV-1/2 Qual) test for use on cobas 6800/8800 Systems. The results of adult plasma and serum samples and pediatric dried blood spots were compared with those of the recomLine HIV-1 & HIV-2 Immunoglobulin G serological test and COBAS AmpliPrep/COBAS TaqMan HIV-1 qualitative test, v2.0. Genotype inclusivity and limits of detection were determined, and sensitivity on seroconversion panels was compared with that in the Bio-Rad Geenius HIV 1/2 Confirmatory Assay, Abbott ARCHITECT HIV Ag/Ab Combo serological test, and cobas TaqScreen MPX, v2.0. RESULTS Concordance of cobas HIV-1/2 Qual test with the comparator serological test and COBAS AmpliPrep/COBAS TaqMan test was ≥99.6% with all sample types. Reactivity with all HIV genotypes was 100%. LOD in plasma samples was 14.8, 12.6, and 27.9 copies/mL for HIV-1 group M, HIV-1 group O, and HIV-2, respectively, with similar results for serum samples. LOD in dried blood spots was 255 copies/mL for HIV-1 and 984 copies/mL for HIV-2. HIV infection was detected 18.9 days and 8.5 days earlier than the confirmatory and serological assays, respectively, and at a similar time to the NAT. CONCLUSIONS The cobas HIV-1/2 Qual test enables early and accurate diagnoses of HIV-1 and HIV-2 in adults and children across sample types. The assay could help avert transmission during acute HIV, simplify HIV diagnostic algorithms, and promote the survival of HIV-infected children.
Collapse
Affiliation(s)
- Lucia Hans
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa;
| | | | - Anke Edelmann
- Department of Virology, Labor Berlin—Charité Vivantes Services GmbH
| | - Jörg Hofmann
- Department of Virology, Labor Berlin—Charité Vivantes Services GmbH
| | - Alex Y. Nilsson
- Global Development, Roche Diagnostics International AG, Rotkreuz, Switzerland;
| | | | - Britta Seiverth
- Global Clinical Operations, Roche Diagnostics International AG, Rotkreuz, Switzerland; and
| | - Peter Gohl
- Bioscientia Institut für Medizinische Diagnostik, Ingelheim, Germany.
| | - Sergio Carmona
- Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Health Laboratory Services, Johannesburg, South Africa;
| |
Collapse
|
119
|
van Biljon N, Robertson F, Holmes M, Cotton MF, Laughton B, van der Kouwe A, Meintjes E, Little F. Multivariate approach for longitudinal analysis of brain metabolite levels from ages 5-11 years in children with perinatal HIV infection. Neuroimage 2021; 237:118101. [PMID: 33961998 PMCID: PMC8295244 DOI: 10.1016/j.neuroimage.2021.118101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/16/2021] [Accepted: 04/19/2021] [Indexed: 12/03/2022] Open
Abstract
Treatment guidelines recommend that children with perinatal HIV infection (PHIV) initiate antiretroviral therapy (ART) early in life and remain on it lifelong. As part of a longitudinal study examining the long-term consequences of PHIV and early ART on the developing brain, 89 PHIV children and a control group of 85 HIV uninfected children (HIV-) received neuroimaging at ages 5, 7, 9 and 11 years, including single voxel magnetic resonance spectroscopy (MRS) in three brain regions, namely the basal ganglia (BG), midfrontal gray matter (MFGM) and peritrigonal white matter (PWM). We analysed age-related changes in absolute metabolite concentrations using a multivariate approach traditionally applied to ecological data, the Correlated Response Model (CRM) and compared these to results obtained from a multilevel mixed effect modelling (MMEM) approach. Both approaches produce similar outcomes in relation to HIV status and age effects on longitudinal trajectories. Both methods found similar age-related increases in both PHIV and HIV- children in almost all metabolites across regions. We found significantly elevated GPC+PCh across regions (95% CI=[0.033; 0.105] in BG; 95% CI=[0.021; 0.099] in PWM; 95% CI=[0.059; 0.137] in MFGM) and elevated mI in MFGM (95% CI=[0.131; 0.407]) among children living with PHIV compared to HIV- children; additionally the CRM model also indicated elevated mI in BG (95% CI=[0.008; 0.248]). These findings suggest persistent inflammation across the brain in young children living with HIV despite early ART initiation.
Collapse
Affiliation(s)
- Noëlle van Biljon
- Department of Statistical Sciences, University of Cape Town, Private Bag X3, Rhodes Gift, 7707 Cape Town, South Africa; Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, South Africa
| | - Frances Robertson
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, South Africa; Cape Universities Body Imaging Centre, Cape Town, South Africa; Neuroscience Institute, University of Cape Town, South Africa
| | - Martha Holmes
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, South Africa; Neuroscience Institute, University of Cape Town, South Africa
| | - Mark F Cotton
- FAMCRU, Department of Paediatrics and Child Health and Tygerberg Children's Hospital, Stellenbosch University, Cape Town, South Africa
| | - Barbara Laughton
- FAMCRU, Department of Paediatrics and Child Health and Tygerberg Children's Hospital, Stellenbosch University, Cape Town, South Africa
| | - Andre van der Kouwe
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, South Africa; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA, United States
| | - Ernesta Meintjes
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, South Africa; Cape Universities Body Imaging Centre, Cape Town, South Africa; Neuroscience Institute, University of Cape Town, South Africa
| | - Francesca Little
- Department of Statistical Sciences, University of Cape Town, Private Bag X3, Rhodes Gift, 7707 Cape Town, South Africa.
| |
Collapse
|
120
|
Radebe L, Haeri Mazanderani A, Sherman GG. Indeterminate HIV PCR results within South Africa's early infant diagnosis programme, 2010-2019. Clin Microbiol Infect 2021; 28:609.e7-609.e13. [PMID: 34400341 DOI: 10.1016/j.cmi.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 07/14/2021] [Accepted: 08/01/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVES We describe the extent of, and variables associated with, indeterminate HIV-PCR results and final HIV status within South Africa's early infant diagnosis (EID) programme between 2010 and 2019. METHODS Retrospective analysis of routine paediatric HIV-PCR laboratory data from South Africa's National Health Laboratory Service Data Warehouse between 2010 and 2019. Final HIV status was determined by linking patient results (including HIV-PCR, HIV viral load, HIV serology and CD4 counts) using a probabilistic matching algorithm. Multivariate logistic regression was performed to determine variables associated with final HIV status among patients with an indeterminate HIV-PCR result. RESULTS Among 4 429 742 specimens registered for HIV-PCR testing from 3 816 166 patients, 113 209 (2.97%) tested positive and 22 899 (0.6%) tested indeterminate. As a proportion of HIV-detected results, 15.7% (23 896/151 832) of total and 31.5% (4900/15 566), 18.8% (11 400/60 794) and 10.1% (7596/75 472) among patients aged <7 days, 7 days-3 months and ≥3 months, respectively, were reported as indeterminate. Overall, 39.7% of patients with an indeterminate result had a linked HIV test to determine HIV status, of which 53.6% were positive with a median time to repeat testing of 30 days (interquartile range 15-69). Among patients who tested indeterminate, variables associated with a significantly higher odds of having a positive HIV status included testing indeterminate at birth (adjusted odds ratio (AOR) 0.63 (0.48-0.83) and 0.52 (0.39-0.69) for testing indeterminate at 7 days-3 months and ≥3 months respectively compared with birth), within a hospital (AOR 2.45 (1.99-3.03)), and in districts with an intra-uterine transmission rate ≥1.1% (AOR 3.14 (1.84-5.35)) (p < 0.001). DISCUSSION Indeterminate HIV-PCR results represent a considerable burden of missed diagnostic opportunities, diagnostic dilemmas and delays in making a definite diagnosis among HIV-infected infants within South Africa's EID programme. Alternative EID verification practices are urgently needed.
Collapse
Affiliation(s)
- Lebohang Radebe
- Paediatric HIV Diagnostics Division, Wits Health Consortium, Johannesburg, South Africa
| | - Ahmad Haeri Mazanderani
- Centre for HIV & STIs, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa; Department of Pathology, Faculty of Health Sciences, University of Limpopo, Polokwane, South Africa.
| | - Gayle G Sherman
- Paediatric HIV Diagnostics Division, Wits Health Consortium, Johannesburg, South Africa; Centre for HIV & STIs, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, South Africa; Department of Paediatrics & Child Health, Faculty of Health Sciences, University of Witwatersrand, South Africa
| |
Collapse
|
121
|
Wexler C, Kamau Y, Muchoki E, Babu S, Maosa N, Maloba M, Brown M, Goggin K, Mabachi N, Gautney B, Finocchario-Kessler S. Implementing at-birth, point-of-care HIV testing in Kenya: a qualitative study using the Consolidated Framework for Implementation Research. Implement Sci Commun 2021; 2:89. [PMID: 34380567 PMCID: PMC8359118 DOI: 10.1186/s43058-021-00188-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 07/25/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND At-birth and point-of-care (POC) testing can expedite early infant diagnosis of HIV and improve infant outcomes. Guided by the Consolidated Framework for Implementation Research (CFIR), this study describes the implementation of an at-birth POC testing pilot from the perspective of implementing providers and identifies the factors that might support and hinder the scale up of these promising interventions. METHODS We conducted 28 focus group discussions (FGDs) with 48 providers across 4 study sites throughout the course of a pilot study assessing the feasibility and impact of at-birth POC testing. FGDs were audio-recorded, transcribed, and analyzed for a priori themes related to CFIR constructs. This qualitative study was nested within a larger study to pilot and evaluate at-birth and POC HIV testing. RESULTS Out of the 39 CFIR constructs, 30 were addressed in the FGDs. While all five domains were represented, major themes revolved around constructs related to intervention characteristics, inner setting, and outer setting. Regarding intervention characteristics, the advantages of at-birth POC (rapid turnaround time resulting in improved patient management and enhanced patient motivation) were significant enough to encourage provider uptake and enthusiasm. Challenges at the intervention level (machine breakdown, processing errors), inner settings (workload, limited leadership engagement, challenges with access to information), and outer setting (patient-level challenges, limited engagement with outer setting stakeholders) hindered implementation, frustrated providers, and resulted in missed opportunities for testing. Providers discussed how throughout the course of the study adaptations to implementation (improved channels of communication, modified implementation logistics) were made to overcome some of these challenges. To improve implementation, providers cited the need for enhanced training and for greater involvement among stakeholders outside of the implementing team (i.e., other clinicians, hospital administrators and implementing partners, county and national health officials). Despite provider enthusiasm for the intervention, providers felt that the lack of engagement from leadership within the hospital and in the outer setting would preclude sustained implementation outside of a research setting. CONCLUSION Despite demonstrated feasibility and enthusiasm among implementing providers, the lack of outer setting support makes sustained implementation of at-birth POC testing unlikely at this time. The findings highlight the multi-dimensional aspect of implementation and the need to consider facilitators and barriers within each of the five CFIR domains. TRIAL REGISTRATION ClinicalTrials.gov, NCT03435887 . Retrospectively registered on 19 February 2020.
Collapse
Affiliation(s)
- Catherine Wexler
- Department of Family Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Yvonne Kamau
- Department of Family Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | | | - May Maloba
- Global Health Innovations - Kenya, Nairobi, Kenya
| | - Melinda Brown
- Department of Family Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kathy Goggin
- Health Services and Outcomes Research, Children's Mercy Kansas City, Kansas City, MO, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Natabhona Mabachi
- Department of Family Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brad Gautney
- Global Health Innovations - USA, Dallas, TX, USA
| | | |
Collapse
|
122
|
Massanella M, Puthanakit T, Leyre L, Jupimai T, Sawangsinth P, de Souza M, Suntarattiwong P, Kosalarksa P, Borkird T, Kanjanavanit S, Chokephaibulkit K, Hansudewechakul R, Petdachai W, Mitchell JL, Robb ML, Trautmann L, Ananworanich J, Chomont N. Continuous Prophylactic Antiretrovirals/Antiretroviral Therapy Since Birth Reduces Seeding and Persistence of the Viral Reservoir in Children Vertically Infected With Human Immunodeficiency Virus. Clin Infect Dis 2021; 73:427-438. [PMID: 32504081 DOI: 10.1093/cid/ciaa718] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/02/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Early antiretroviral therapy (ART) restricts the size of the human immunodeficiency virus (HIV) reservoir in infants. However, whether antiretroviral (ARV) prophylaxis given to exposed vertically infected children exerts similar effects remains unknown. METHODS We measured total and integrated HIV DNA, as well as the frequency of CD4 T cells producing multiply spliced RNA (msRNA) after stimulation (inducible reservoir) in vertically infected Thai infants. Eighty-five infants were followed longitudinally for up to 3 years. We compared the size of the reservoir in children who received continuous ARV prophylaxis since birth vs those who never received or discontinued prophylaxis before initiating ART. We used samples from a cross-sectional cohort of 37 Thai children who had initiated ART within 6 months of life to validate our findings. RESULTS Before ART, levels of HIV DNA and the frequencies of cells producing msRNA were significantly lower in infants who received continuous ARV prophylaxis since birth compared to those in whom ARV prophylaxis was discontinued or never initiated (P < .020 and P < .001, respectively). Upon ART initiation, total and integrated HIV DNA levels decayed significantly in both groups (P < .01 in all cases). Interestingly, the initial differences in the frequencies of infected cells persisted during 3 years on ART. The beneficial effect of prophylaxis on the size of the HIV reservoir was confirmed in the cross-sectional study. Importantly, no differences were observed between children who discontinued prophylactic ARVs before starting ART and those who delayed ART initiation without receiving prior prophylaxis. CONCLUSIONS Neonatal ARV prophylaxis with direct transition to ART durably limits the size of the HIV reservoir.
Collapse
Affiliation(s)
- Marta Massanella
- Centre de Recherche du Centre hospitalier de l'Université de Montréal and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Canada
| | - Thanyawee Puthanakit
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,HIV Netherlands Australia Thailand (HIV-NAT) Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Louise Leyre
- Centre de Recherche du Centre hospitalier de l'Université de Montréal and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Canada
| | - Thidarat Jupimai
- Center of Excellence for Pediatric Infectious Diseases and Vaccines, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Mark de Souza
- SEARCH, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | | | - Pope Kosalarksa
- Department of Pediatrics, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | | | - Kulkanya Chokephaibulkit
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | | | - Julie L Mitchell
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA.,United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Vaccine and Gene Therapy Institute, Oregon Health and Science University, Hillsboro, Oregon, USA
| | - Merlin L Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA.,United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Lydie Trautmann
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA.,United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Vaccine and Gene Therapy Institute, Oregon Health and Science University, Hillsboro, Oregon, USA
| | - Jintanat Ananworanich
- Queen Sirikit National Institute of Child Health, Bangkok, Thailand.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA.,United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, USA
| | - Nicolas Chomont
- Centre de Recherche du Centre hospitalier de l'Université de Montréal and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Canada
| | | |
Collapse
|
123
|
Finocchario-Kessler S, Wexler C, Brown M, Goggin K, Lwembe R, Nazir N, Gautney B, Khamadi S, Babu S, Muchoki E, Maosa N, Mabachi N, Kamau Y, Maloba M. Piloting the Feasibility and Preliminary Impact of Adding Birth HIV Polymerase Chain Reaction Testing to the Early Infant Diagnosis Guidelines in Kenya. Pediatr Infect Dis J 2021; 40:741-745. [PMID: 33990521 PMCID: PMC8274583 DOI: 10.1097/inf.0000000000003172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND In Kenya, standard early infant diagnosis (EID) with polymerase chain reaction (PCR) testing at 6-week postnatal achieves early treatment initiation (<12 weeks) in <20% of HIV+ infants. Kenya's new early infant diagnosis guidelines tentatively proposed adding PCR testing at birth, pending results from pilot studies. METHODS We piloted birth testing at 4 Kenyan hospitals between November 2017 and November 2018. Eligible HIV-exposed infants were offered both point-of-care and PCR HIV testing at birth (window 0 to <4 weeks) and 6 weeks (window 4-12 weeks). We report the: proportion of infants tested at birth, 6-week, and both birth and 6-week testing; median infant age at results; seropositivity and antiretroviral therapy initiation. RESULTS Final sample included 624 mother-infant pairs. Mean maternal age was 30.4 years, 73.2% enrolled during antenatal care and 89.9% had hospital deliveries. Among the 590 mother-infants pairs enrolled before 4 weeks postnatal, 452 (76.6%) completed birth testing before 4 weeks, with 360 (79.6%) testing within 2 weeks, and 178 (39.4%) before hospital discharge (0-2 days). Mothers were notified of birth PCR results at a median infant age of 5.4 weeks. Among all 624 enrolled infants, 575 (92.1%) were tested during the 6-week window; 417 (66.8%) received testing at both birth and 6-weeks; and 207 received incomplete testing (93.3% only 1 PCR and 6.7% no PCR). Four infants were diagnosed with HIV, and 3 infants were initiated on antiretroviral therapy early, before 12 weeks of age. CONCLUSIONS Uptake of PCR testing at birth was high and a majority of infants received repeat testing at 6 weeks of age.
Collapse
Affiliation(s)
| | - Catherine Wexler
- From the Department of Family Medicine, University of Kansas Medical Center, Kansas City, Missouri
| | - Melinda Brown
- From the Department of Family Medicine, University of Kansas Medical Center, Kansas City, Missouri
| | - Kathy Goggin
- Children’s Mercy Kansas City, Health Services and Outcomes Research, Kansas City, Missouri
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| | - Raphael Lwembe
- Center for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Niaman Nazir
- Department of Preventive Medicine, University of Kansas Medical Center, Kansas City, Missouri
| | | | - Samoel Khamadi
- Center for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | | | | | | | - Natabhona Mabachi
- From the Department of Family Medicine, University of Kansas Medical Center, Kansas City, Missouri
| | - Yvonne Kamau
- From the Department of Family Medicine, University of Kansas Medical Center, Kansas City, Missouri
| | - May Maloba
- Global Health Innovations—Kenya, Nairobi, Kenya
| |
Collapse
|
124
|
Domaoal RA, Sleeman K, Sawadogo S, Dzinamarira T, Frans N, Shatumbu SP, Kakoma LN, Shuumbwa TK, Cox MH, Stephens S, Nisbet L, Metz M, Saito S, Williams DB, Voetsch AC, Patel H, Parekh B, Duong YT. Successful Use of Near Point-of-Care Early Infant Diagnosis in NAMPHIA to Improve Turnaround Times in a National Household Survey. J Acquir Immune Defic Syndr 2021; 87:S67-S72. [PMID: 34166314 PMCID: PMC8754064 DOI: 10.1097/qai.0000000000002706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/07/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND In the population-based HIV impact assessment surveys, early infant diagnosis (EID) was provided to infants <18 months without a prior diagnosis. For the Namibia population-based HIV impact assessment (NAMPHIA), the GeneXpert platform was assessed for the feasibility of near POC EID testing compared with the standard Roche COBAS AmpliPrep/COBAS TaqMan (CAP/CTM) platform. Quality assurance measures and turnaround time were compared to improve EID results reporting. METHODS NAMPHIA participants were screened for HIV exposure using Determine HIV-1/2 rapid test; samples reactive on Determine received EID testing on the GeneXpert instrument and Xpert HIV-1 Qual assay using whole blood. Results were confirmed at the Namibia Institute of Pathology using dried blood spots on the Roche CAP/CTM platform per national guidelines. RESULTS Of the 762 screened infants, 61 (8.0%) were Determine-reactive and considered HIV-exposed. Of the 61 exposed infants, 2 were found to be HIV-infected whereas 59 were negative on both GeneXpert and Roche platforms, achieving 100% concordance. Average turnaround time was 3.4 days for the Xpert HIV-1 Qual assay, and average time from collection to testing was 1.0 days for GeneXpert compared with 10.7 days for Roche. No samples failed using GeneXpert whereas 1 sample failed using Roche and was repeated. CONCLUSION Quality POC EID testing is feasible in a national survey through extensive training and external quality assurance measures. The use of decentralized POC EID for national testing would provide rapid diagnosis and improve TATs which may prevent loss to follow-up, ensure linkage to care, and improve clinical outcomes for infants.
Collapse
Affiliation(s)
- Robert A. Domaoal
- International Laboratory Branch, Division of Global HIV & TB, CGH, CDC, Atlanta, GA, USA
| | - Katrina Sleeman
- International Laboratory Branch, Division of Global HIV & TB, CGH, CDC, Atlanta, GA, USA
| | | | | | | | | | | | | | - Mackenzie Hurlston Cox
- International Laboratory Branch, Division of Global HIV & TB, CGH, CDC, Atlanta, GA, USA
| | - Sally Stephens
- University of California San Francisco, Windhoek, Namibia
| | - Lydia Nisbet
- University of California San Francisco, Windhoek, Namibia
| | | | | | | | - Andrew C. Voetsch
- Epidemiology and Surveillance Branch, Division of Global HIV & TB, CGH, CDC, Atlanta, GA, USA
| | - Hetal Patel
- International Laboratory Branch, Division of Global HIV & TB, CGH, CDC, Atlanta, GA, USA
| | - Bharat Parekh
- International Laboratory Branch, Division of Global HIV & TB, CGH, CDC, Atlanta, GA, USA
| | | |
Collapse
|
125
|
Nuoh RD, Nyarko KM, Noora CL, Addo-Lartey A, Nortey P, Nuolabong C, Lartey M, Kenu E. Barriers to early infant diagnosis of HIV in the Wa Municipality and Lawra District of Upper West Region, Ghana. Ghana Med J 2021; 54:83-90. [PMID: 33536673 PMCID: PMC7837354 DOI: 10.4314/gmj.v54i2s.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Objective We identified socio-demographic, health system and psycho-social barriers to Early Infant Diagnosis (EID) of HIV in the Upper West Region of Ghana. Design An unmatched case control study of 96 cases and 96 controls was conducted in the ART centers in Lawra district and Wa Municipality between December 2014 and April 2015. Setting A public health facility Participants We defined a case as an HIV positive mother with an exposed infant who received EID service between January 2011 and December 2014. A control was defined as HIV Positive Mother with an exposed infant who did not receive EID services between January 2011 and December 2014. Main outcome EID by dry blood spot Deoxyribonucleic acid Polymerase chain reaction. Results A total of 192 mother-infant pairs were assessed. The mean age of infants at testing for cases was 17.3±14.9 weeks. Mother-to-child-transmission-rate was 2.3%. Factors associated with EID testing included: mother being formally employed (cOR=2.0: 95%CI:1.1–3.8), maternal formal education (cOR=2.0, 95%CI: 1.1–3.6) and maternal independent source of income (cOR 2.2, 95%CI 1.2–4.1). After adjusting for confounders, maternal independent income source was associated with EID testing (aOR 2.2, 95%CI 1.2–4.1). Median turn-around time of EID result was 11 weeks (IQR 4–27 weeks). Conclusion Women need to be empowered to gain an independent source of income. This can help maximize the benefits of e-MTCT and increase EID in the Upper West Region of Ghana. Funding This work was funded by the authors
Collapse
Affiliation(s)
- Robert D Nuoh
- Ghana Field Epidemiology and Laboratory Training Program, Department of Epidemiology, School of Public Health, College of Health Sciences, University of Ghana Legon, Accra, Ghana
| | - Kofi M Nyarko
- Namibia Field Epidemiology and Laboratory Training Program, University of Namibia, Windhoek, Namibia
| | - Charles L Noora
- Ghana Field Epidemiology and Laboratory Training Program, Department of Epidemiology, School of Public Health, College of Health Sciences, University of Ghana Legon, Accra, Ghana
| | - Adolphina Addo-Lartey
- Ghana Field Epidemiology and Laboratory Training Program, Department of Epidemiology, School of Public Health, College of Health Sciences, University of Ghana Legon, Accra, Ghana
| | - Priscillia Nortey
- Ghana Field Epidemiology and Laboratory Training Program, Department of Epidemiology, School of Public Health, College of Health Sciences, University of Ghana Legon, Accra, Ghana
| | - Culbert Nuolabong
- Ghana Field Epidemiology and Laboratory Training Program, Department of Epidemiology, School of Public Health, College of Health Sciences, University of Ghana Legon, Accra, Ghana
| | - Margaret Lartey
- Department of Medicine and Therapeutics, School of Medicine and Dentistry, College of Health Sciences, University of Ghana, Korle Bu, Accra, Ghana
| | - Ernest Kenu
- Ghana Field Epidemiology and Laboratory Training Program, Department of Epidemiology, School of Public Health, College of Health Sciences, University of Ghana Legon, Accra, Ghana.,Department of Medicine and Therapeutics, School of Medicine and Dentistry, College of Health Sciences, University of Ghana, Korle Bu, Accra, Ghana
| |
Collapse
|
126
|
Harrison MJ, Brice N, Scott C. Clinical Features of HIV Arthropathy in Children: A Case Series and Literature Review. Front Immunol 2021; 12:677984. [PMID: 34354702 PMCID: PMC8329591 DOI: 10.3389/fimmu.2021.677984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/02/2021] [Indexed: 12/02/2022] Open
Abstract
Background HIV infection has been associated with a non-erosive inflammatory arthritis in children, although few published reports exist. This study describes the clinical, laboratory and imaging features of this noncommunicable disease in a series of HIV-infected children in South Africa. Methods A database search was conducted to identify HIV-infected children enrolled in a Paediatric Rheumatology service in Cape Town, South Africa between 1 January 2010 and 31 December 2020. Retrospective data were collected from individuals classified with HIV arthropathy, based on a predefined checklist. Demographic, clinical, laboratory, sonographic, therapeutic, and outcomes data were extracted by chart review. Descriptive statistical analysis was performed using R (v4.0.3). Results Eleven cases of HIV arthropathy were included in the analysis. Cases predominantly presented in older boys with low CD4+ counts. Median age at arthritis onset was 10.3 years (IQR 6.9 – 11.6) and the male-female ratio was 3.0. The median absolute CD4+ count was 389 cells/uL (IQR 322 – 449). The clinical presentation was variable, with both oligoarthritis and polyarthritis being common. Elevated acute phase reactants were the most consistent laboratory feature, with a median ESR of 126 mL/h (IQR 67 – 136) and median CRP of 36 mg/L (IQR 25 – 68). Ultrasonography demonstrated joint effusions and synovial hypertrophy. Response to therapy was slower than has generally been described in adults, with almost all cases requiring more than one immunosuppressive agent. Five children were discharged in established remission after discontinuing immunotherapy, however outcomes data were incomplete for the remaining six cases. Conclusions In this case series, HIV arthropathy was associated with advanced immunosuppression. Therapeutic modalities included immunomodulators and antiretroviral therapy, which consistently induced disease remission although data were limited by a high rate of attrition. Prospective studies are needed to define and understand this HIV-associated noncommunicable disease.
Collapse
Affiliation(s)
- Michael J Harrison
- Fort Beaufort Provincial Hospital, Amathole District, Eastern Cape, South Africa
| | - Nicola Brice
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, Cape Town, South Africa.,University of Cape Town, Rondebosch, Cape Town, South Africa
| | - Christiaan Scott
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, Cape Town, South Africa.,University of Cape Town, Rondebosch, Cape Town, South Africa
| |
Collapse
|
127
|
Schue JL, van Dijk JH, Hamangaba F, Hamahuwa M, Moyo N, Thuma PE, Moss WJ, Sutcliffe CG. Treatment outcomes among children younger than five years living with HIV in rural Zambia, 2008-2018: a cohort study. BMC Pediatr 2021; 21:315. [PMID: 34261465 PMCID: PMC8278691 DOI: 10.1186/s12887-021-02793-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/30/2021] [Indexed: 12/01/2022] Open
Abstract
Background HIV testing and treatment guidelines for children in sub-Saharan Africa have evolved over time, such that children are now treated at younger ages. The objective of this study was to describe the treatment experience for immunologic, virologic, and growth outcomes among HIV-infected Zambian children younger than 5 years of age from 2008 to 2018. Methods Participants enrolled in a clinical cohort study in Macha, Zambia and initiating antiretroviral treatment before 5 years of age between 2008 and 2015 were included in the analysis and followed up to the end of 2018. Outcomes, including growth, CD4+ T-cell percentage, viral suppression, and mortality, were evaluated among all children using longitudinal and survival analyses. Comparisons by age at treatment initiation (< 1, 1 to < 2, and 2 to < 5 years) were also evaluated. Results Three hundred eighty-one children initiating treatment before 5 years of age between 2008 and 2015 were included in the analysis. Growth metrics and CD4+ T-cell percentage improved over time after treatment initiation. However, 20% of children remained underweight and 40% of children remained stunted after the first 36 months of treatment. 85% of children had a viral load < 400 copies/mL after 12 months of treatment. However, children < 1 year at treatment initiation were more likely to have a detectable viral load in the first 12 months of treatment and less likely to achieve viral suppression compared to older children. Mortality was highest in the first 12 months of treatment, among underweight children, and among children initiating treatment in 2008–2010 compared to 2011–2015. Conclusions Most children initiating antiretroviral treatment from 2008 to 2015 in rural Zambia responded well to treatment. However, many children remained underweight and stunted, and experienced high mortality rates during the first few months of treatment. This supports continued efforts to improve early infant diagnosis, nutritional support, and pediatric drug formulations. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-021-02793-y.
Collapse
Affiliation(s)
- Jessica L Schue
- Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, USA
| | - Janneke H van Dijk
- Erasmus University, Burgemeester Oudlaan 50, 3062, PA, Rotterdam, Netherlands
| | | | | | | | - Philip E Thuma
- Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, USA.,Macha Research Trust, Choma, Zambia
| | - William J Moss
- Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, USA
| | - Catherine G Sutcliffe
- Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, USA.
| |
Collapse
|
128
|
van Wyhe KS, Laughton B, Cotton MF, Meintjes EM, van der Kouwe AJW, Boivin MJ, Kidd M, Thomas KGF. Cognitive outcomes at ages seven and nine years in South African children from the children with HIV early antiretroviral (CHER) trial: a longitudinal investigation. J Int AIDS Soc 2021; 24:e25734. [PMID: 34259393 PMCID: PMC8278859 DOI: 10.1002/jia2.25734] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Many children living with HIV (CLWH) display impaired cognition. Although early combination antiretroviral therapy (ART) produces improved cognitive outcomes, more long-term outcome data are needed. After concluding the Children with HIV Early antiRetroviral (CHER) trial in 2011, we investigated cognitive performance, at seven and nine years of age. Participants had been randomized to deferred ART (ART-Def; n = 22); immediate time-limited ART for 40 weeks (ART-40W; n = 30) and immediate time-limited ART for 96 weeks (ART-96W; n = 18). We also recruited HIV-exposed uninfected (CHEU; n = 28) and HIV-unexposed (CHU; n = 35) children. METHODS Data were collected between May 2012 and December 2017. Mixed-model repeated-measures ANOVAs assessed differences over time between CLWH (ART-40W, ART-96W and ART-Def) and CHIV- CHEU and CHU between ART-Early (ART-40W and ART-96W), ART-Def, CHEU and CHU; and between ART-40W, ART-96W, ART-Def, CHEU and CHU. RESULTS All comparisons found significant effects of Time for most outcome variables (better scores at nine than at seven years; ps < 0.05). The first ANOVAs found that for (a) motor dexterity, CLWH performed worse than CHIV- at seven years (p < 0.001) but improved to equivalence at nine years, (b) visual-spatial processing and problem solving, only CLWH (p < 0.04) showed significant performance improvement over time and (c) working memory and executive function, CLWH performed worse than CHIV- at both seven and nine years (p = 0.03 and 0.04). The second ANOVAs found that for (a) working memory, CHU performed better than ART-Early and CHEU (p < 0.01 and <0.04), and (b) motor dexterity, ART-Def performed worse than ART-Early, CHEU and CHU at seven years (p = 0.02, <0.001 and <0.001 respectively) but improved to equivalence at nine years (ps > 0.17). Similarly, for motor dexterity, ART-Def performed worse than ART-96W, CHEU and CHU at seven years (p < 0.04, <0.001 and <0.001) but improved to equivalence at nine years (ps > 0.20). CONCLUSIONS Although neurocognitive developmental trajectories for treatment groups and controls were largely similar (i.e. performance improvements from 7 to 9), all ART-treated children, regardless of treatment arm, remain at risk for cognitive deficits over early school ages. Although the nature of these deficits may change as cognitive development proceeds, there are potential negative consequences for these children's future learning, reasoning and adaptive functioning.
Collapse
Affiliation(s)
- Kaylee S van Wyhe
- ACSENT LaboratoryDepartment of PsychologyUniversity of Cape TownCape TownSouth Africa
- Family Centre for Research with UbuntuDepartment of Paediatrics and Child HealthFaculty of Medicine and Health Sciences Stellenbosch UniversityCape TownSouth Africa
| | - Barbara Laughton
- Family Centre for Research with UbuntuDepartment of Paediatrics and Child HealthFaculty of Medicine and Health Sciences Stellenbosch UniversityCape TownSouth Africa
| | - Mark F Cotton
- Family Centre for Research with UbuntuDepartment of Paediatrics and Child HealthFaculty of Medicine and Health Sciences Stellenbosch UniversityCape TownSouth Africa
| | - Ernesta M Meintjes
- Biomedical Engineering Research CentreDivision of Biomedical EngineeringDepartment of Human BiologyUniversity of Cape TownSouth Africa
| | - Andre JW van der Kouwe
- Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalCharlestownMAUSA
| | - Michael J Boivin
- Department of Psychiatry and Neurology and OphthalmologyMichigan State UniversityEast LansingMIUSA
- Department of PsychiatryUniversity of MichiganAnn ArborMIUSA
| | - Martin Kidd
- Centre of Statistical ConsultationStellenbosch UniversityCape TownSouth Africa
| | - Kevin GF Thomas
- ACSENT LaboratoryDepartment of PsychologyUniversity of Cape TownCape TownSouth Africa
| |
Collapse
|
129
|
Evaluation of the Aptima HIV-1 Quant Dx assay for HIV diagnosis at birth in South Africa. Diagn Microbiol Infect Dis 2021; 101:115467. [PMID: 34391073 DOI: 10.1016/j.diagmicrobio.2021.115467] [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: 03/15/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 11/21/2022]
Abstract
The increased coverage of antiretroviral therapy has resulted in a decrease in the positive predictive value (PPV) and diagnostic sensitivity of early infant diagnosis assays. To evaluate the diagnostic performance of the Aptima HIV-1 Quant DX assay (Aptima) in detecting HIV infection at birth. The study was a cross-sectional laboratory based evaluation using whole blood DBS specimens. Samples were collected from HIV-exposed neonates at birth at two paediatric facilities in Gauteng between 1st March 2018 - 31st January 2020. Performance of the Aptima compared to the Cobas® AmpliPrep/Cobas® TaqMan HIV-1 Qualitative Test v2.0 was calculated using a two-by-two table and reported as proportions with 95% confidence intervals. A total of 363 infants met the inclusion criteria of which 4 (1.1%) had an Aptima result discordant with CAP/CTM HIV status: two (50%) negative and two (50%) positive. The Aptima assay had a sensitivity of 93.75% (95% CI: 79.19%-99.23%), specificity of 99.4% (95% CI: 97.83%-99.93%), PPV of 93.75% (95% CI: 78.98%-98.36%), negative predictive value of 99.4% (95% CI: 97.73%-99.84%), and overall accuracy of 98.9% (95% CI: 97.2%-99.7%). The Aptima yielded an error code on 37 (10.19%) results, of which 35 (94.59%) were resolved on repeat testing. Of the 32 HIV-detected specimens, 20 had a plasma VL result available (18 on Abbott and 2 on Cobas). The absolute median difference was 0.66 log10 (IQR: 0.36-1.71). The Aptima demonstrated good EID performance and can be considered as a qualitative EID assay.
Collapse
|
130
|
Piloya TW, Bakeera–Kitaka S, Kisitu GP, Idro R, Cusick SE. Vitamin D status and associated factors among HIV-infected children and adolescents on antiretroviral therapy in Kampala, Uganda. PLoS One 2021; 16:e0253689. [PMID: 34166428 PMCID: PMC8224887 DOI: 10.1371/journal.pone.0253689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 06/10/2021] [Indexed: 12/28/2022] Open
Abstract
Background A high prevalence of suboptimal serum vitamin D has been reported among HIV infected children even in countries with high sunshine abundance throughout the year. Vitamin D is a potent immune modulator of innate and adaptive immune responses. Vitamin D regulates immune responses through the vitamin D receptor on CD4 cells. We aimed to determine the vitamin D status of HIV infected children and factors associated with suboptimal vitamin D. Methods This was a cross sectional study. We enrolled children aged between 6 months and 12 years attending an outpatient paediatric HIV clinic. Serum 25-hydroxyvitamin D (25(OH)D) was measured using the electrochemoluminisence method. Suboptimal vitamin D was defined as 25(OH)D <30 ng/ml, vitamin D insufficiency and deficiency were 21–29 ng/ml and <20 ng/ml respectively. Anthropometry, physical exam and medical history were documented. Logistic regression was performed. Results We enrolled 376 children with mean age (sd) 8.05 years (3.03), a median (IQR) duration of ART of 5.9 years (3.2–8.4). Majority of the children (64%) had been exposed to non nucleoside reverse transcriptase inhibitors (NNRTIs). A third were severely immunosuppressed (CD4% ≤15%) at ART initiation. At the time of the study, the majority (89%) were virologically suppressed (VL <1000 copies/ml). Prevalence of 25(OH)D <30 ng/ml was 49 (13%) of 375 participants and 11 (3%) had 25(OH)D <20 ng/ml. Lopinavir/ritonavir regimen was independently associated with 25(OH)D <30 ng/ml; OR 0.27 CI (0.13–0.57), p value-0.002. Serum 25(OH)D <20 ng/ml was associated with CD4 count ≤15% at ART initiation OR 6.55(1.30–32.9), p value—0.023 and use of NNRTIs; OR 10.9(1.22–96.2), p value—0.03. Conclusion We found a low prevalence of suboptimal vitamin D compared to earlier reports. Severe immunosuppression at ART initiation and use of NNRTIs increases odds of deficiency. Vitamin D supplementation should be considered in severely immunosuppressed children initiating ART.
Collapse
Affiliation(s)
- Thereza Were Piloya
- Department of Paediatrics, College of Health Sciences, Makerere University, Kampala, Uganda
- * E-mail:
| | - Sabrina Bakeera–Kitaka
- Department of Paediatrics, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Grace Paul Kisitu
- Baylor College of Medicine, Paediatric Centre of Excellence, Kampala, Uganda
| | - Richard Idro
- Department of Paediatrics, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Sarah E. Cusick
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
131
|
Brice J, Sylla M, Desire N, Sayon S, Telly F, Bocar-Fofana D, Murphy R, Peytavin G, Diallo S, Nastouli E, Calvez V, Marcelin AG, Maiga AI, Lambert-Niclot S. Characterization of drug resistance and the defective HIV reservoir in virally suppressed vertically infected children in Mali. J Antimicrob Chemother 2021; 75:1272-1279. [PMID: 32073629 DOI: 10.1093/jac/dkaa002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/20/2019] [Accepted: 12/26/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND In the perspective of ART-free HIV remission, vertically infected children treated with suppressive ART from early infancy represent an optimal population model to better understand the genetic complexity of the reservoir. OBJECTIVES To evaluate the proportion of defective viral population and the genotypic resistance patterns in cell-associated HIV DNA. METHODS In a cohort including 93 ART-treated vertically HIV-infected (VHIV) children in Mali with plasma HIV-1 RNA ≤50 copies/mL for at least 6 months, we studied total HIV DNA, percentage of defective genomes and resistance by reverse transcriptase and protease bulk sequencing from whole blood in dried blood spots. RESULTS Children had a median age of 9.9 years at the time of inclusion (IQR = 7.6-13.4) and 3.3 years (IQR = 2-7) at ART initiation; median ART duration was 5.5 years (IQR = 3.7-7.3). The median level of total HIV DNA was 470 copies/106 cells with one patient presenting undetectable HIV DNA (<66 copies/106 cells). We observed the presence of at least one stop codon in viruses from 34 patients (37%). The presence of stop codons was not correlated with the level of HIV DNA or duration of ART. We showed a high prevalence of HIV-1 resistance in DNA with 26% of children harbouring virus resistant to at least one NRTI and 40% to at least one NNRTI. CONCLUSIONS While these VHIV children were successfully treated for a long time, they showed high prevalence of resistance in HIV DNA and a moderate defective HIV reservoir.
Collapse
Affiliation(s)
- Josephine Brice
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), AP-HP, Pitié Salpêtrière Hospital, Department of Virology, F-75013 Paris, France
| | - Mariam Sylla
- Department of Pediatrics, University Hospital Gabriel Toure, Bamako, Mali
| | - Nathalie Desire
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), AP-HP, Pitié Salpêtrière Hospital, Department of Virology, F-75013 Paris, France
| | - Sophie Sayon
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), AP-HP, Pitié Salpêtrière Hospital, Department of Virology, F-75013 Paris, France
| | - Fatoumata Telly
- Unité d'Epidémiologie Moléculaire de la Résistance du VIH aux ARV, SEREFO, FMOS, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Djeneba Bocar-Fofana
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), AP-HP, Saint Antoine Hospital, Department of Virology, F-75012 Paris, France
| | - Robert Murphy
- Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, 645 N. Michigan Avenue, Suite 900, Chicago, IL 60611, USA
| | - Gilles Peytavin
- AP-HP, Department of Pharmacology, Bichat-Claude Bernard Hospital, F-75018 Paris, France
| | - Souleymane Diallo
- Unité d'Epidémiologie Moléculaire de la Résistance du VIH aux ARV, SEREFO, FMOS, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Eleni Nastouli
- Department of Population, Policy and Practice, University College London GOS Institute of Child Health, London, UK
| | - Vincent Calvez
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), AP-HP, Pitié Salpêtrière Hospital, Department of Virology, F-75013 Paris, France
| | - Anne-Geneviève Marcelin
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), AP-HP, Pitié Salpêtrière Hospital, Department of Virology, F-75013 Paris, France
| | - Almoustapha Issiaka Maiga
- Unité d'Epidémiologie Moléculaire de la Résistance du VIH aux ARV, SEREFO, FMOS, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali.,Clinical and Microbiology Laboratory, University Hospital Gabriel Toure, Bamako, Mali
| | - Sidonie Lambert-Niclot
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), AP-HP, Saint Antoine Hospital, Department of Virology, F-75012 Paris, France
| |
Collapse
|
132
|
Bunglawala F, Rajoli RKR, Mirochnick M, Owen A, Siccardi M. Prediction of dolutegravir pharmacokinetics and dose optimization in neonates via physiologically based pharmacokinetic (PBPK) modelling. J Antimicrob Chemother 2021; 75:640-647. [PMID: 31860112 DOI: 10.1093/jac/dkz506] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/23/2019] [Accepted: 11/05/2019] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Only a few antiretroviral drugs (ARVs) are recommended for use during the neonatal period and there is a need for more to be approved to increase treatment and prophylaxis strategies. Dolutegravir, a selective integrase inhibitor, has potential for treatment of HIV infection and prophylaxis of transmission in neonates. OBJECTIVES To model the pharmacokinetics of dolutegravir in neonates and to simulate a theoretical optimal dosing regimen. METHODS The physiologically based pharmacokinetic (PBPK) model was built incorporating the age-related changes observed in neonates. Virtual neonates between 0 and 28 days were simulated. The model was validated against observed clinical data for raltegravir and midazolam in neonates, prior to the prediction of dolutegravir pharmacokinetics. RESULTS Both raltegravir and midazolam passed the criteria for model qualification, with simulated data within 1.8-fold of clinical data. The qualified model predicted the pharmacokinetics for several multidose regimens of dolutegravir. Regimen 6 involved 5 mg doses with a 48 h interval from Day 1-20, increasing to 5 mg once daily on Week 3, yielding AUC and Ctrough values of 37.2 mg·h/L and 1.3 mg/L, respectively. These exposures are consistent with those observed in paediatric patients receiving dolutegravir. CONCLUSIONS Dolutegravir pharmacokinetics were successfully simulated in the neonatal PBPK model. The predictions suggest that during the first 3 weeks of life a 5 mg dose administered every 48 h may achieve plasma exposures needed for therapy and prophylaxis.
Collapse
Affiliation(s)
- Fazila Bunglawala
- Department of Molecular and Clinical Pharmacology, University of Liverpool, 70 Pembroke Place, Liverpool L69 3GF, UK
| | - Rajith K R Rajoli
- Department of Molecular and Clinical Pharmacology, University of Liverpool, 70 Pembroke Place, Liverpool L69 3GF, UK
| | | | - Andrew Owen
- Department of Molecular and Clinical Pharmacology, University of Liverpool, 70 Pembroke Place, Liverpool L69 3GF, UK
| | - Marco Siccardi
- Department of Molecular and Clinical Pharmacology, University of Liverpool, 70 Pembroke Place, Liverpool L69 3GF, UK
| |
Collapse
|
133
|
Zijenah LS, Bandason T, Bara W, Chipiti MM, Katzenstein DA. Mother-to-child transmission of HIV-1 and infant mortality in the first six months of life, in the era of Option B Plus combination antiretroviral therapy. Int J Infect Dis 2021; 109:92-98. [PMID: 34161799 DOI: 10.1016/j.ijid.2021.06.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVES The aim of this study was to determine the contributions of intrauterine (IU), intrapartum (IP), and postpartum (PP) transmission to mother-to-child transmission of HIV-1 (MTCT) and infant mortality in the first 6 months of life, in the era of Option B Plus combination antiretroviral therapy. METHODS Plasma for virus load (VL) quantitation was obtained from 451 women enrolled into the study. VL was quantified using the Cepheid GeneXpert HIV-1 quantitative test. Dried blood spots were collected from 453 infants at birth, 4-6 weeks, 3 months, and 6 months. HIV-1 infant diagnosis was conducted using the Cepheid GeneXpert HIV-1 qualitative test. Absolute and cumulative MTCT rates, and the mortality rate by 6 months were calculated. RESULTS Seven mothers (1.55%) had transmitted HIV-1 infection to their infants by 6 months. Four infants (0.88%, 95% confidence interval (CI) 0.26-2.33%) were infected IU, one infant (0.22%, 95% CI 0-1.4%) was infected IP, and two infants (0.44%, 95% CI 0.01-1.7%) were infected PP. The infant mortality rate was 0.88% (95% CI 0.26-2.33%). CONCLUSIONS In the first 6 months of life, in the era of Option B Plus combination antiretroviral therapy, IU transmission is the major route of MTCT. The cumulative MTCT rate of 1.55% in a breastfeeding population contributes to growing evidence that complete elimination of MTCT is possible.
Collapse
Affiliation(s)
- Lynn S Zijenah
- Immunology Unit, Department of Laboratory Diagnostics and Investigative Science, University of Zimbabwe Faculty of Medicine and Health Sciences, Harare, Zimbabwe.
| | - Tsitsi Bandason
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Wilbert Bara
- Ministry of Health and Child Care, Harare, Zimbabwe
| | - Maria Mary Chipiti
- Immunology Unit, Department of Laboratory Diagnostics and Investigative Science, University of Zimbabwe Faculty of Medicine and Health Sciences, Harare, Zimbabwe
| | | |
Collapse
|
134
|
Wilson S, Bohn MK, Adeli K. POCT: An Inherently Ideal Tool in Pediatric Laboratory Medicine. EJIFCC 2021; 32:145-157. [PMID: 34421483 PMCID: PMC8343051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Point of care testing (POCT) is important in the provision of timely laboratory test results and continues to gain specific appreciation in the setting of pediatric healthcare. POCT platforms offer several advantages compared to central laboratory testing, including improved clinical outcomes, reduced time to diagnosis, length of stay, and blood volume requirements, as well as increased accessibility. These advantages are most pronounced in acute care settings such as pediatric emergency departments, intensive care units, and in remote settings, wherein rapid patient assessment and prognostication is essential to patient outcomes. The current review provides an overview and critical discussion of the evidence supporting clinical implementation of POCT systems in pediatric clinical decision-making, including but not limited to the diagnosis of viral and bacterial infection, identification of critical glucose and electrolyte dysregulation, and prognostication of post-operative inpatients. Important considerations for test result reporting and interpretation are also discussed, including analytical concordance between POCT systems and central laboratory analyzers as well as availability of pediatric reference intervals for key analytes on POCT systems. Notably, a paucity of evidence-based pediatric reference intervals for test interpretation for critical care parameters on POCT platforms is highlighted, warranting further study and unique consideration prior to clinical implementation.
Collapse
Affiliation(s)
- Siobhan Wilson
- Division of Clinical Biochemistry, Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mary Kathryn Bohn
- Division of Clinical Biochemistry, Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Khosrow Adeli
- Division of Clinical Biochemistry, Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
135
|
Abstract
Advances in perinatal HIV management have averted a significant number of infections in neonates and have made the possibility of elimination of mother-to-child transmission a reality; however, significant gaps in implementation of early testing programs as well as the expansion of therapeutic strategies to neonates are hindering prevention efforts and access to safer, more effective and easier to administer treatment. This article provides insights on the current state of perinatal HIV, recent advances, and future needs.
Collapse
Affiliation(s)
- Andres F Camacho-Gonzalez
- Division of Pediatric Infectious Diseases, Children's Healthcare of Atlanta, Emory University School of Medicine, 2015 Uppergate Drive, Suite 500, Atlanta, GA 30322, USA.
| | - Paul Palumbo
- Section of Infectious Diseases and International Health, Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 03756, USA
| |
Collapse
|
136
|
Nwosu EC, Holmes MJ, Cotton MF, Dobbels E, Little F, Laughton B, van der Kouwe A, Meintjes EM, Robertson F. Cortical structural changes related to early antiretroviral therapy (ART) interruption in perinatally HIV-infected children at 5 years of age. IBRO Neurosci Rep 2021; 10:161-170. [PMID: 34179869 PMCID: PMC8211921 DOI: 10.1016/j.ibneur.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/03/2021] [Indexed: 12/21/2022] Open
Abstract
ART interruption in children can occur especially in resource-limited settings for reasons including poor adherence, stock-outs, ART intolerance of non-pediatric formulas and pill size, as well as ultimately to test for HIV remission. Although early ART initiation is now standard of care in pediatric HIV management, very little is known on the effect of early ART initiation or subsequent interruption on brain development. This study aimed to investigate the effect of ART interruption on brain cortical thickness (CT) and folding in a subset of children from the Children with HIV Early antiRetroviral therapy (CHER) trial cohort who all started ART before 18 months of age. CHER participants in the neuroimaging follow-up study had magnetic resonance (MRI) scans on a 3T Siemens Allegra brain scanner at age 5.44 ± 0.37 years. MR images were processed using the automated cross-sectional stream in FreeSurfer v6.0 and vertex wise comparisons of CT and local gyrification indices (LGIs) were performed between HIV+ children and HIV- controls, as well as between HIV+ children on interrupted or continuous ART and controls. HIV+ children (n = 46) showed thicker cortex than HIV- children (n = 29) in bilateral frontal and left temporo-insular regions but lower LGIs in left superior and bilateral medial orbitofrontal cortex extending into rostral anterior cingulate. Children on interrupted ART (n = 21) had thicker cortex than HIV- controls in left frontal and right insular regions, but children on continuous treatment (n = 25) showed no difference from controls. Children on both interrupted and continuous ART showed region-specific alterations in LGI relative to controls. Cortical folding appears more sensitive than CT to early life events including early ART and interruption. However, immune health resilience in children can translate to long term preservation of morphometric brain development, especially for those on early and continuous treatment.
Collapse
Affiliation(s)
- Emmanuel C. Nwosu
- UCT Medical Imaging Research Unit, Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, South Africa
- Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Martha J. Holmes
- UCT Medical Imaging Research Unit, Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, South Africa
- Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Mark F. Cotton
- Family Centre for Research with Ubuntu, Department of Paediatrics & Child Health, Tygerberg Children’s Hospital and Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Els Dobbels
- Family Centre for Research with Ubuntu, Department of Paediatrics & Child Health, Tygerberg Children’s Hospital and Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Francesca Little
- Department of Statistical Sciences, Faculty of Sciences, University of Cape Town, South Africa
| | - Barbara Laughton
- Family Centre for Research with Ubuntu, Department of Paediatrics & Child Health, Tygerberg Children’s Hospital and Faculty of Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andre van der Kouwe
- A.A. Martinos Centre for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Ernesta M. Meintjes
- UCT Medical Imaging Research Unit, Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, South Africa
- Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Frances Robertson
- UCT Medical Imaging Research Unit, Division of Biomedical Engineering, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, South Africa
- Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, South Africa
| |
Collapse
|
137
|
du Plessis AM, Andronikou S, Zar HJ. Chest imaging findings of chronic respiratory disease in HIV-infected adolescents on combined anti retro viral therapy. Paediatr Respir Rev 2021; 38:16-23. [PMID: 33139219 DOI: 10.1016/j.prrv.2020.06.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 11/26/2022]
Abstract
Early treatment with combination antiretroviral therapy (cART) has improved survival of children perinatally infected with HIV into adolescence. This population is at risk of long term complications related to HIV infection, particularly chronic respiratory disease. Limited data on chest imaging findings in HIV-infected adolescents, suggest that the predominant disease is of small and large airways: predominantly bronchiolitis obliterans or bronchiectasis. Single cases of emphysema have been reported. Lung fibrosis, lymphocytic interstitial pneumonitis, post tuberculous apical fibrocystic changes and malignancies do not feature in this population. Chest radiograph (CXR) is easily accessible and widely used, especially in resource limited settings, such as sub Saharan Africa, where the greatest burden of HIV disease occurs. Lung ultrasound has been described for the diagnosis of pneumonia in children, pulmonary oedema and interstitial lung disease [1-3]. The use of this modality in chronic respiratory disease in adolescents where the predominant finding is small airway disease and bronchiectasis has however not been described. CXR is useful to evaluate structural/post infective changes, parenchymal opacification and nodules, hyperinflation or extensive bronchiectasis. CXR however, is inadequate for diagnosing small airway disease, for which high resolution computed tomography (HRCT) is the modality of choice. Where available, low dose HRCT should be used early in the course of symptomatic disease in adolescents and for follow up in children who are non responsive to treatment or clinically deteriorating. This article provides a pictorial review of the spectrum of CXR and HRCT imaging findings of chronic pulmonary disease in perinatally HIV-infected adolescents on cART and guidelines for imaging.
Collapse
Affiliation(s)
- Anne-Marie du Plessis
- Department of Paediatrics and Child Health, Red Cross Children's Hospital and SA-Medical Research Council Unit on Child & Adolescent Health, USA
| | - Savvas Andronikou
- Department of Paediatric Radiology, Children's Hospital of Philadelphia, USA
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross Children's Hospital and SA-Medical Research Council Unit on Child & Adolescent Health, USA
| |
Collapse
|
138
|
Odhiambo CO, Githuka G, Bowen N, Kingwara L, Onsase J, Ochuka B, Waweru M, Masaba R, Matu L, Mwangi E, Cohn J. Point-of-Care Early Infant Diagnosis Improves Adherence to the Testing Algorithm in Kenya. J Int Assoc Provid AIDS Care 2021; 19:2325958220906030. [PMID: 32052676 PMCID: PMC7019379 DOI: 10.1177/2325958220906030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction: We determine the level of adherence to the revised Kenya early infant diagnosis (EID)
algorithm during implementation of a point-of-care (POC) EID project. Methods: Data before (August 2016 to July 2017) and after (August 2017 to July 2018)
introduction of POC EID were collected retrospectively from the national EID database
and registers for 33 health facilities. We assessed the number of HIV-infected infants
who underwent confirmatory testing and received baseline viral load test and proportion
of infants with an initial negative result who had a subsequent test. Results and Discussion: Significantly higher number of infants accessed confirmatory testing (94.2% versus
38.6%; P < .0001) with POC EID. Baseline viral load test and
follow-up testing at 6 months, although higher with POC EID, were not significantly
different from the pre-POC EID intervention period. Conclusion: The POC EID implementation has the potential to increase proportion of infants who
receive confirmatory testing, thus reducing the risk of false-positive results.
Collapse
Affiliation(s)
| | | | - Nancy Bowen
- National Public Health Laboratories, Nairobi, Kenya
| | | | - Jared Onsase
- Elizabeth Glaser Pediatric AIDS Foundation (EGPAF), Nairobi, Kenya
| | - Bernard Ochuka
- Elizabeth Glaser Pediatric AIDS Foundation (EGPAF), Nairobi, Kenya
| | - Michael Waweru
- Elizabeth Glaser Pediatric AIDS Foundation (EGPAF), Nairobi, Kenya
| | - Rose Masaba
- Elizabeth Glaser Pediatric AIDS Foundation (EGPAF), Nairobi, Kenya
| | - Lucy Matu
- Elizabeth Glaser Pediatric AIDS Foundation (EGPAF), Nairobi, Kenya
| | - Eliud Mwangi
- Elizabeth Glaser Pediatric AIDS Foundation (EGPAF), Nairobi, Kenya
| | - Jennifer Cohn
- Elizabeth Glaser Pediatric AIDS Foundation (EGPAF), Geneva, Switzerland
| |
Collapse
|
139
|
Manglani M, Gabhale Y, Lala MM, Balakrishnan S, Bhuyan K, Rewari BB, Setia MS. Assessing the Effectiveness of a Telemedicine Initiative in Clinical Management of Children Living with HIV/AIDS in Maharashtra, India. Curr HIV Res 2021; 19:201-215. [PMID: 33397239 DOI: 10.2174/1573399817666210104102825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/26/2020] [Accepted: 11/26/2020] [Indexed: 11/22/2022]
Abstract
AIMS To evaluate the effectiveness of telemedicine in the clinical management of children living with HIV/AIDS in resource-limited settings ; Background: Telemedicine is an important mechanism for service delivery in health care settings, both in resource-rich and resource-poor settings. Such service delivery mechanisms have shown to be associated with virologic suppression and higher CD4 counts. These services are also associated with improved access, shorter visiting times, and higher patient satisfaction. ; Objective: We designed the present two-group comparison study to compare the clinical evaluation and management of children in the anti-retroviral therapy (ART) centres linked to telemedicine facility with those who are not linked to this facility in Maharashtra, India. ; Methods: We analysed clinical records from six ART centres in Maharashtra; of these, 250 children were in the linked ART centres and 301 were in the non-linked ART centres. The outcomes were classified according to investigations, management, and monitoring. For management, we evaluated: 1) Initiation of cotrimoxazole prophylaxis; 2) Children not initiated on ART when required; 3) ART regime after appropriate investigations; and 4) Change of regime (if immunologically indicated). For monitoring, we assessed the haematological monitoring of children on ART. ; Results: The mean (SD) ages of children in linked and non-linked ART centres were 10.8 (4.6) and 10.9 (4.6) years, respectively (p=0.80). After adjusting for individual and structural level variables, physical examination (OR: 2.0, 95% CI; 1.2, 3.2), screening for tuberculosis (OR: 12.9, 95% CI: 2.0, 82.9) and cotrimoxazole prophylaxis were significantly more likely in the linked centres compared with non-linked centres (OR: 1.8, 95% CI: 1.4, 2.2). A higher proportion of children eligible for ART were not initiated on treatment in the non-linked centres compared with linked centres (26% vs. 8%, p=0.06). Children were less likely to be initiated on zidovudine-based regimens without baseline haemoglobin or with baseline haemoglobin of less than 9 gm% in linked centres (OR: 0.7, 95% CI: 0.6, 0.8). Similarly, children in the linked centres were less likely to have been started on nevirapine-based regimens without baseline liver enzymes (OR: 0.8, 95% CI: 0.7, 0.9). ; Conclusion: Thus, the overall clinical management of Children Living with HIV/ AIDS (CLHA) was better in ART centres linked with the telemedicine initiative compared with those who were not linked. Children in the linked ART centres were more likely to have a complete baseline assessment (physical, hematological, radiological, and screening for TB); the presence of a pediatrician in the centres was helpful.
Collapse
Affiliation(s)
- Mamta Manglani
- Pediatric Centre of Excellence for HIV, Department of Pediatrics, LTM Medical College and General Hospital, Mumbai, India
| | - Yashwant Gabhale
- Pediatric Centre of Excellence for HIV, Department of Pediatrics, LTM Medical College and General Hospital, Mumbai, India
| | - Mamatha Murad Lala
- Pediatric Centre of Excellence for HIV, Department of Pediatrics, LTM Medical College and General Hospital, Mumbai, India
| | | | - Khanindra Bhuyan
- UNICEF, Near 73, Lodhi Gardens, Lodhi Estate, New Delhi, 110003, India
| | - Bharat Bhushan Rewari
- WHO Regional Office of South East Asea, World Health House, Indraprastha Estate, Mahatma Gandhi Marg, New Delhi 110 002, India
| | | |
Collapse
|
140
|
Palacio-Mejía LS, Hernández-Ávila JE, Molina-Vélez D, González-González L, Quezada-Sánchez AD, Hernández-Ávila M, Magis-Rodriguez C. Trends in Mortality due to HIV/AIDS in children in Mexico: Towards the elimination of mother-tochild transmission. Arch Med Res 2021; 52:746-754. [PMID: 33958214 DOI: 10.1016/j.arcmed.2021.04.008] [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: 12/06/2019] [Revised: 03/29/2021] [Accepted: 04/15/2021] [Indexed: 10/21/2022]
Abstract
AIM OF THE STUDY To examine mortality trends in children under 15 years of age due to HIV/AIDS in Mexico and describe their differences by insurance coverage. METHODS Time series analysis of deaths from 1990-2019 through a Bayesian poisson regression model with linear splines and knots in 1994, 1997, and 2003. RESULTS Overall, we observed a reduction in the mortality rate due to HIV from 2003 onwards, except in the group of 10-14 years. In the population covered with Social Security, mortality rates decreased in all age groups. However, in the group without Social Security or with Popular Security (subsidized system), mortality rates significantly decreased only for children below 5 years. of age. CONCLUSIONS Health insurance through the contributory system is associated with faster and larger reductions in HIV related infant mortality. Universal access to health insurance was not sufficient to close the gap in HIV-mortality among children under 15 years of age in Mexico.
Collapse
Affiliation(s)
| | | | - Diana Molina-Vélez
- Center for Evaluation and Surveys Research. National Institute of Public Health
| | | | | | | | | |
Collapse
|
141
|
Shalekoff S, Loubser S, Dias BDC, Strehlau R, Shiau S, Wang S, He Y, Abrams EJ, Kuhn L, Tiemessen CT. Normalization of B Cell Subsets but Not T Follicular Helper Phenotypes in Infants With Very Early Antiretroviral Treatment. Front Pediatr 2021; 9:618191. [PMID: 33996678 PMCID: PMC8118125 DOI: 10.3389/fped.2021.618191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/01/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Infant HIV-1-infection is associated with high morbidity and mortality if antiretroviral treatment (ART) is not initiated promptly. We characterized development of circulating T follicular helper cells (cTfh) and their relationship to naïve/memory B cell subsets in a cohort of neonates initiating ART within the first week of life. Methods: Infants were diagnosed within 48 hours of birth and started ART as soon as possible. The frequency and phenotype of cTfh and B cells were analyzed at enrollment (birth -19 days) and at 4, 12, and 72 weeks of age in blood of 27 HIV-1-intrauterine-infected and 25 HIV-1 exposed uninfected (HEU) infants as part of a study in Johannesburg, South Africa. cTfh cells were divided into Tfh1, Tfh2, and Tfh17 subsets. B cell phenotypes were defined as naïve, resting memory, activated memory and tissue-like memory cells. Results: HIV-1-infected infants had higher frequencies of cTfh cells than HEU infants up to 12 weeks of age and these cTfh cells were polarized toward the Tfh1 subset. Higher frequencies of Tfh1 and lower frequencies of Tfh2 and Tfh17 correlated with lower CD4+ T cell percentages. Lower frequencies of resting memory, with corresponding higher frequencies of activated memory B cells, were observed with HIV-1 infection. Importantly, dysregulations in B cell, but not cTfh cell, subsets were normalized by 72 weeks. Conclusion: Very early ART initiation in HIV-1-infected infants normalizes B cell subsets but does not fully normalize perturbations in cTfh cell subsets which remain Tfh1 polarized at 72 weeks. It remains to be determined if very early ART improves vaccine antibody responses despite the cTfh and B cell perturbations observed over the time course of this study.
Collapse
Affiliation(s)
- Sharon Shalekoff
- Centre for HIV & STIs, National Institute for Communicable Diseases and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shayne Loubser
- Centre for HIV & STIs, National Institute for Communicable Diseases and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bianca Da Costa Dias
- Centre for HIV & STIs, National Institute for Communicable Diseases and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Renate Strehlau
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephanie Shiau
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, United States
| | - Shuang Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York City, NY, United States
| | - Yun He
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York City, NY, United States
| | - Elaine J. Abrams
- ICAP at Columbia University, Mailman School of Public Health, and Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York City, NY, United States
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York City, NY, United States
| | - Caroline T. Tiemessen
- Centre for HIV & STIs, National Institute for Communicable Diseases and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
142
|
Ndongo FA, Tejiokem MC, Penda CI, Ndiang ST, Ndongo JA, Guemkam G, Sofeu CL, Tagnouokam-Ngoupo PA, Kfutwah A, Msellati P, Faye A, Warszawski J. Long-term outcomes of early initiated antiretroviral therapy in sub-Saharan children: a Cameroonian cohort study (ANRS-12140 Pediacam study, 2008-2013, Cameroon). BMC Pediatr 2021; 21:189. [PMID: 33882903 PMCID: PMC8059165 DOI: 10.1186/s12887-021-02664-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 04/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In most studies, the virological response is assessed during the first two years of antiretroviral treatment initiated in HIV-infected infants. However, early initiation of antiretroviral therapy exposes infants to very long-lasting treatment. Moreover, maintaining viral suppression in children is difficult. We aimed to assess the virologic response and mortality in HIV-infected children after five years of early initiated antiretroviral treatment (ART) and identify factors associated with virologic success in Cameroon. METHODS In the ANRS-12140 Pediacam cohort study, 2008-2013, Cameroon, we included all the 149 children who were still alive after two years of early ART. Virologic response was assessed after 5 years of treatment. The probability of maintaining virologic success between two and five years of ART was estimated using Kaplan-Meier curve. The immune status and mortality were also studied at five years after ART initiation. Factors associated with a viral load < 400 copies/mL in children still alive at five years of ART were studied using logistic regressions. RESULTS The viral load after five years of early ART was suppressed in 66.8% (60.1-73.5) of the 144 children still alive and in care. Among the children with viral suppression after two years of ART, the probability of maintaining viral suppression after five years of ART was 64.0% (54.0-74.0). The only factor associated with viral suppression after five years of ART was achievement of confirmed virological success within the first two years of ART (OR = 2.7 (1.1-6.8); p = 0.033). CONCLUSIONS The probability of maintaining viral suppression between two and five years of early initiated ART which was quite low highlights the difficulty of parents to administer drugs daily to their children in sub-Saharan Africa. It also stressed the importance of initial viral suppression for achieving and maintaining virologic success in the long-term. Further studies should focus on identifying strategies that would enhance better retention in care and improved adherence to treatment within the first two years of ART early initiated in Sub-Saharan HIV-infected children.
Collapse
Affiliation(s)
- Francis Ateba Ndongo
- Université Paris-Sud, Centre Mère et Enfant de la Fondation Chantal Biya, Francis, POB 1936, Yaounde, Cameroon.
| | | | - Calixte Ida Penda
- MPH, PH-PU, Université Douala; Hôpital Laquintinie, Douala, Cameroon
| | | | | | - Georgette Guemkam
- Centre Mère et Enfant de la Fondation Chantal Biya, Yaounde, Cameroon
| | - Casimir Ledoux Sofeu
- Université Yaoundé I; Centre Pasteur du Cameroun, Service d'Epidémiologie et de Santé Publique, Yaounde, Cameroon
| | | | - Anfumbom Kfutwah
- Centre Pasteur du Cameroun, Service de Virologie, Yaounde, Cameroon
| | | | - Albert Faye
- Université Paris Diderot, Sorbonne Paris Cité; Assistance Publique des Hôpitaux de Paris, Pédiatrie Générale, Hôpital Robert Debré, INSERM UMR 1123, ECEVE, Paris, France
| | - Josiane Warszawski
- Université Paris-Sud, Assistance Publique des Hôpitaux de Paris, CESP INSERM U1018, team 4 "HIV and STD", Hôpital Bicêtre, 94276, Le Kremlin-Bicêtre, France
| |
Collapse
|
143
|
Abstract
HIV-1 integrates its genome into the DNA of host cells. Consequently, HIV-1 genomes are copied with the host cell DNA during cellular division. Little is known about the emergence and persistence of human immunodeficiency virus (HIV)-infected T-cell clones in perinatally infected children. We analyzed peripheral blood mononuclear cells (PBMCs) for clonal expansion in 11 children who initiated antiretroviral therapy (ART) between 1.8 and 17.4 months of age and with viremia suppressed for 6 to 9 years. We obtained 8,662 HIV type 1 (HIV-1) integration sites from pre-ART samples and 1,861 sites from on-ART samples. Expanded clones of infected cells were detected pre-ART in 10/11 children. In 8 children, infected cell clones detected pre-ART persisted for 6 to 9 years on ART. A comparison of integration sites in the samples obtained on ART with healthy donor PBMCs infected ex vivo showed selection for cells with proviruses integrated in BACH2 and STAT5B. Our analyses indicate that, despite marked differences in T-cell composition and dynamics between children and adults, HIV-infected cell clones are established early in children, persist for up to 9 years on ART, and can be driven by proviral integration in proto-oncogenes.
Collapse
|
144
|
Levy B, Correia HE, Chirove F, Ronoh M, Abebe A, Kgosimore M, Chimbola O, Machingauta MH, Lenhart S, White KAJ. Modeling the Effect of HIV/AIDS Stigma on HIV Infection Dynamics in Kenya. Bull Math Biol 2021; 83:55. [PMID: 33818710 PMCID: PMC8021528 DOI: 10.1007/s11538-021-00891-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/15/2021] [Indexed: 11/25/2022]
Abstract
Stigma toward people living with HIV/AIDS (PLWHA) has impeded the response to the disease across the world. Widespread stigma leads to poor adherence of preventative measures while also causing PLWHA to avoid testing and care, delaying important treatment. Stigma is clearly a hugely complex construct. However, it can be broken down into components which include internalized stigma (how people with the trait feel about themselves) and enacted stigma (how a community reacts to an individual with the trait). Levels of HIV/AIDS-related stigma are particularly high in sub-Saharan Africa, which contributed to a surge in cases in Kenya during the late twentieth century. Since the early twenty-first century, the United Nations and governments around the world have worked to eliminate stigma from society and resulting public health education campaigns have improved the perception of PLWHA over time, but HIV/AIDS remains a significant problem, particularly in Kenya. We take a data-driven approach to create a time-dependent stigma function that captures both the level of internalized and enacted stigma in the population. We embed this within a compartmental model for HIV dynamics. Since 2000, the population in Kenya has been growing almost exponentially and so we rescale our model system to create a coupled system for HIV prevalence and fraction of individuals that are infected that seek treatment. This allows us to estimate model parameters from published data. We use the model to explore a range of scenarios in which either internalized or enacted stigma levels vary from those predicted by the data. This analysis allows us to understand the potential impact of different public health interventions on key HIV metrics such as prevalence and disease-related death and to see how close Kenya will get to achieving UN goals for these HIV and stigma metrics by 2030.
Collapse
Affiliation(s)
- Ben Levy
- Department of Mathematics, Fitchburg State University, Fitchburg, MA, USA
| | - Hannah E Correia
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.,Department of Biostatistics, Harvard University, Boston, MA, USA
| | - Faraimunashe Chirove
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Johannesburg, South Africa
| | - Marilyn Ronoh
- School of Mathematics, University of Nairobi, Nairobi, Kenya
| | - Ash Abebe
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Johannesburg, South Africa
| | - Moatlhodi Kgosimore
- Department of Biometry and Mathematics, Botswana University of Agriculture and Natural Resources, Gaborone, Botswana
| | - Obias Chimbola
- Mathematics and Statistical Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| | - M Hellen Machingauta
- Mathematics and Statistical Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| | - Suzanne Lenhart
- Mathematics Department, University of Tennessee, Knoxville, TN, USA
| | - K A Jane White
- Department of Mathematical Sciences, University of Bath, Bath, UK.
| |
Collapse
|
145
|
Matsinhe M, Bollinger T, Lee N, Loquiha O, Meggi B, Mabunda N, Mudenyanga C, Mutsaka D, Florêncio M, Mucaringua A, Macassa E, Seni A, Jani I, Buck WC. Inpatient Point-of-Care HIV Early Infant Diagnosis in Mozambique to Improve Case Identification and Linkage to Antiretroviral Therapy. GLOBAL HEALTH: SCIENCE AND PRACTICE 2021; 9:31-39. [PMID: 33684058 PMCID: PMC8087433 DOI: 10.9745/ghsp-d-20-00611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/02/2021] [Indexed: 11/23/2022]
Abstract
Introduction of point-of-care early infant diagnosis on the inpatient wards of 2 of the largest pediatric referral hospitals in Mozambique increased HIV testing volume and pediatric HIV case identification with improved linkage to antiretroviral therapy. Introduction: Novel approaches to case identification and linkage to antiretroviral therapy (ART) are needed to close gaps in early infant diagnosis (EID) of HIV. Point-of-care (POC) EID is a recent innovation that eliminates the long turnaround times of conventional EID that limit patient management in the inpatient setting. The initial deployment of POC EID in Mozambique focused primarily on outpatient clinics; however, 2 high-volume tier-4 pediatric referral hospitals were also included. Methods: To assess the impact of inpatient POC EID, a retrospective review of testing and care data from Hospital Central de Beira (HCB) and Hospital Central de Maputo (HCM) was performed for the period September 2017 to July 2018, with comparison to the 8-month pre-POC period when dried blood spots were used for conventional EID. Results: Monthly testing volume increased from 8.5 tests pre-POC to 17.6 tests with POC (P<.001). Among 511 children with POC testing, the median age was 5 months, there was ongoing breastfeeding in 326 (63.8%), and 136 (26.6%) of mothers and 146 (28.6%) of infants had not received ART or antiretroviral prophylaxis, respectively. POC tests were positive in 152 (29.7%) infants, and 52 (37.5%) had a previous negative DNA polymerase chain reaction through the conventional outpatient EID program. Linkage to ART for infants with HIV-positive tests improved 64% during the POC period (P=.002). Inpatient mortality for infected infants during the POC period was 28.2%. Excluding these deaths, 61.2% of eligible infants initiated ART as inpatients, but only 29.8% of those discharged without ART were confirmed to have initiated as outpatients. Conclusions: Inpatient wards are a high-yield site for EID and ART initiation that have historically been overlooked in programming for prevention of mother-to-child transmission. POC platforms represent a transformative opportunity to increase inpatient testing, make definitive diagnoses, and improve timely linkage to ART. Scale-up plans should prioritize pediatric wards.
Collapse
Affiliation(s)
- Mércia Matsinhe
- Hospital Central de Maputo, Maputo, Mozambique.,Instituto Nacional de Saúde, Maputo, Mozambique
| | | | - Nilza Lee
- Hospital Central de Beira, Beira, Mozambique
| | | | | | | | | | | | | | | | | | - Amir Seni
- Hospital Central de Beira, Beira, Mozambique
| | - Ilesh Jani
- Instituto Nacional de Saúde, Maputo, Mozambique
| | - W Chris Buck
- University of California Los Angeles, David Geffen School of Medicine, Los Angeles, USA.
| |
Collapse
|
146
|
Quantifying the Dynamics of HIV Decline in Perinatally Infected Neonates on Antiretroviral Therapy. J Acquir Immune Defic Syndr 2021; 85:209-218. [PMID: 32576731 DOI: 10.1097/qai.0000000000002425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Mathematical modeling has provided important insights into HIV infection dynamics in adults undergoing antiretroviral treatment (ART). However, much less is known about the corresponding dynamics in perinatally infected neonates initiating early ART. SETTING From 2014 to 2017, HIV viral load (VL) was monitored in 122 perinatally infected infants identified at birth and initiating ART within a median of 2 days. Pretreatment infant and maternal covariates, including CD4 T cell counts and percentages, were also measured. METHODS From the initial cohort, 53 infants demonstrated consistent decline and suppressed VL below the detection threshold (20 copies mL) within 1 year. For 43 of these infants with sufficient VL data, we fit a mathematical model describing the loss of short-lived and long-lived infected cells during ART. We then estimated the lifespans of infected cells and the time to viral suppression, and tested for correlations with pretreatment covariates. RESULTS Most parameters governing the kinetics of VL decline were consistent with those obtained previously from adults and other infants. However, our estimates of the lifespan of short-lived infected cells were longer than published values. This difference may reflect sparse sampling during the early stages of VL decline, when the loss of short-lived cells is most apparent. In addition, infants with higher pretreatment CD4 percentage or lower pretreatment VL trended toward more rapid viral suppression. CONCLUSIONS HIV dynamics in perinatally infected neonates initiating early ART are broadly similar to those observed in other age groups. Accelerated viral suppression is also associated with higher CD4 percentage and lower VL.
Collapse
|
147
|
Gaitho D, Kinoti F, Mwaniki L, Kemunto D, Ogoti V, Njigua C, Kubo E, Langat A, Mecha J. Factors associated with the timely uptake of initial HIV virologic test among HIV-exposed infants attending clinics within a faith-based HIV program in Kenya; a cross-sectional study. BMC Public Health 2021; 21:569. [PMID: 33757463 PMCID: PMC7986293 DOI: 10.1186/s12889-021-10587-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/08/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Early infant diagnosis (EID) of HIV, followed by effective care including antiretroviral therapy (ART), reduces infant mortality by 76% and HIV progression by 75%. In 2015, 50% of 1.2 million HIV-exposed infants (HEI) in 21 priority countries received a virologic test within the recommended 2 months of birth. We sought to identify factors associated with timely uptake of virologic EID among HEI and gain insight into missed opportunities. METHODS This was a cross-sectional study that used de-identified data from electronic medical records of 54 health facilities within the Christian Health Association of Kenya (CHAK) HIV Project database. All HEI who had their first HIV virologic test done between January 2015 and December 2017 were included in the study and categorized as either having the test within or after 8 weeks of birth. Multivariate linear mixed effects regression model was used to determine factors associated with uptake of the first HIV EID polymerase chain reaction (PCR). Predictor variables studied include sex, birth weight, the entry point into care, provision of ART prophylaxis for the infant, maternal ART at time of EID, mode of delivery, and place of delivery. RESULTS We included 2020 HEI of whom 1018 (50.4%) were female. A majority, 1596 (79.0%) had their first HIV PCR within 2 months of birth at a median age of 6.4 weeks (interquartile range 6-7.4). Overall, HIV positivity rate at initial test among this cohort was 1.2%. Delayed HIV PCR testing for EID was more likely to yield a positive result [adjusted odds ratio (aOR) = 1.29 (95% confidence interval (CI) 1.09-1.52) p = 0.003]. Infants of mothers not on ART at the time of HIV PCR test and infants who had not received prophylaxis to prevent vertical HIV transmission had significant increased odds of a delayed initial test [aOR = 1.27 (95% CI = 1.18-1.37) p = < 0.0001] and [aOR = 1.43 (95% CI 1.27-1.61) p = < 0.001] respectively. CONCLUSION An initial HIV PCR test done after 8 weeks of birth is likely to yield a positive result. Barriers to accessing ART for treatment among HIV-infected pregnant and breastfeeding women, and prophylaxis for the HEI were associated with delayed EID. In order to ensure timely EID, programs need to incorporate both facility and community strategy interventions to ensure all pregnant women seek antenatal care and deliver within health facilities.
Collapse
Affiliation(s)
- Douglas Gaitho
- Christian Health Association of Kenya (CHAK), P.O. Box 30690 – 00100, GPO, Nairobi, Kenya
| | - Freda Kinoti
- Department of Clinical Medicine & Therapeutics, University of Nairobi, P.O. Box 19676 – 00202, Nairobi, Kenya
| | - Lawrence Mwaniki
- Christian Health Association of Kenya (CHAK), P.O. Box 30690 – 00100, GPO, Nairobi, Kenya
| | - Diana Kemunto
- Christian Health Association of Kenya (CHAK), P.O. Box 30690 – 00100, GPO, Nairobi, Kenya
| | - Victor Ogoti
- Christian Health Association of Kenya (CHAK), P.O. Box 30690 – 00100, GPO, Nairobi, Kenya
| | - Catherine Njigua
- Christian Health Association of Kenya (CHAK), P.O. Box 30690 – 00100, GPO, Nairobi, Kenya
| | - Elizabeth Kubo
- Christian Health Association of Kenya (CHAK), P.O. Box 30690 – 00100, GPO, Nairobi, Kenya
| | - Agnes Langat
- Division of Global HIV and TB, Centers for Disease Control and Prevention Kenya, P.O. Box 606 – 00621, Village Market, Nairobi, Kenya
| | - Jared Mecha
- Department of Clinical medicine & Therapeutics, College of Health Sciences – University of Nairobi, P.O. Box 19676 – 00202, Nairobi, Kenya
| |
Collapse
|
148
|
High rate of loss to follow-up and virological non-suppression in HIV-infected children on antiretroviral therapy highlights the need to improve quality of care in South Africa. Epidemiol Infect 2021; 149:e88. [PMID: 33745490 PMCID: PMC8080219 DOI: 10.1017/s0950268821000637] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Provision of high-quality care and ensuring retention of children on antiretroviral therapy (ART) are essential to reduce human immunodeficiency virus (HIV)-associated morbidity and mortality. Virological non-suppression (≥1000 viral copies/ml) is an indication of suboptimal HIV care and support. This retrospective cohort study included ART-naïve children who initiated first-line ART between July 2015 and August 2017 in Johannesburg and rural Mopani district. Of 2739 children started on ART, 29.5% (807/2739) were lost to care at the point of analysis in August 2018. Among retained children, overall virological non-suppression was 30.2% (469/1554). Virological non-suppression was associated with higher loss to care 30.3% (229/755) compared with suppressed children (9.7%, 136/1399, P < 0.001). Receiving treatment in Mopani was associated with virological non-suppression in children under 5 years (adjusted odds ratio (aOR) 1.7 (95% confidence interval (CI) 1.1-2.4), 5-9 years (aOR 1.8 (1.1-3.0)) and 10-14 years (aOR 1.9 (1.2-2.8)). Virological non-suppression was associated with lower CD4 count in children 5-9 years (aOR 2.1 (1.1-4.1)) and 10-14 years (aOR 2.1 (1.2-3.8)). Additional factors included a shorter time on ART (<5 years aOR 1.8-3.7 (1.3-8.2)), and male gender (5-9 years, aOR1.5 (1.01-2.3)), and receiving cotrimoxazole prophylaxis (10-14 years aOR 2.0 (1.2-3.6)). In conclusion, virological non-suppression is a factor of subsequent programme loss in both regions, and factors affecting the quality of care need to be addressed to achieve the third UNAIDS 90 in paediatric HIV.
Collapse
|
149
|
Tagnouokam-Ngoupo PA, Penda IC, Tchatchueng Mbougua JB, Tetang Ndiang S, Yuya Septoh F, Kenne A, Ngallè JE, Jakpou S, Ateba Ndongo F, Warszawski J, Faye A, Tejiokem MC. Virological failure and antiretroviral resistance among HIV-infected children after five years follow-up in the ANRS 12225-PEDIACAM cohort in Cameroon. PLoS One 2021; 16:e0248642. [PMID: 33735301 PMCID: PMC7971859 DOI: 10.1371/journal.pone.0248642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/02/2021] [Indexed: 11/18/2022] Open
Abstract
Objective In the present study, we aimed to evaluate the virological failure (VF) and drug resistance among treated HIV-infected children after five years follow-up in the ANRS-Pediacam cohort in Cameroon. Methods From November 2007 to October 2011, HIV-infected children born to HIV-infected mothers were included in the ANRS-PEDIACAM study and followed-up for more than 5 years. Plasma viral load (VL) was measured at each visit (every three months until month 24 and every 6 months thereafter). VF was the main outcome and HIV drug resistance test was performed using the ANRS procedures and algorithm. Results Data from 155 children were analyzed. The median age at combination antiretroviral therapy (cART) initiation was 4.2 months (interquartile range (IQR): 3.2–5.8), with 103 (66.5%) children taking LPV/r-containing regimen and 51 (32.9%) children taking NVP. After five years follow-up, 63 (40.6%; CI: 32.9–48.8) children experienced VF. The median duration between cART initiation and VF was 22.1 months (IQR: 11.9–37.1) with a median VL of 4.8 log10 (IQR: 4.0–5.5). Among the 57 children with HIV drug resistance results, 40 (70.2%) had at least one drug resistance mutation. The highest resistance rates (30.4–66.1%) were obtained with Lamivudine; Efavirenz; Nevirapine and Rilpivirine. Conclusions These results show high resistance to NNRTI and emphasize the need of VL and resistance tests for optimal follow-up of HIV-infected people especially children.
Collapse
Affiliation(s)
- Paul Alain Tagnouokam-Ngoupo
- Service de Virologie, Centre Pasteur du Cameroun, Membre du Réseau International des Instituts Pasteur, Yaoundé, Cameroun
- * E-mail:
| | - Ida Calixte Penda
- Department of Clinical Sciences, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala, Cameroon
- Hôpital Laquintinie de Douala, Douala, Cameroun
| | - Jules Brice Tchatchueng Mbougua
- Service d’Epidémiologie et de Santé Publique, Centre Pasteur du Cameroun, Membre du Réseau International des Instituts Pasteur, Yaoundé, Cameroun
| | | | - Francis Yuya Septoh
- Service d’Epidémiologie et de Santé Publique, Centre Pasteur du Cameroun, Membre du Réseau International des Instituts Pasteur, Yaoundé, Cameroun
| | - Angeladine Kenne
- Service d’Epidémiologie et de Santé Publique, Centre Pasteur du Cameroun, Membre du Réseau International des Instituts Pasteur, Yaoundé, Cameroun
| | | | - Sorel Jakpou
- Service d’Epidémiologie et de Santé Publique, Centre Pasteur du Cameroun, Membre du Réseau International des Instituts Pasteur, Yaoundé, Cameroun
| | - Francis Ateba Ndongo
- Unité Pédiatrique de Jour, Centre Mère et Enfant de la Fondation Chantal Biya, Yaoundé, Cameroun
| | - Josiane Warszawski
- Center for Research in Epidemiology and Population Health U1018, Clinical Epidemiology, INSERM, Le Kremlin-Bicetre, France
- Université Paris-Sud, Public Health, Le Kremlin-Bicêtre, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Albert Faye
- Assistance Publique des Hôpitaux de Paris, Pédiatrie Générale, Hôpital Robert Debré, Paris, France
- Université Paris 7—Denis Diderot, Paris, Île-de-France, France
- INSERM UMR-S 1123 (ECEVE), Paris, France
| | - Mathurin Cyrille Tejiokem
- Service d’Epidémiologie et de Santé Publique, Centre Pasteur du Cameroun, Membre du Réseau International des Instituts Pasteur, Yaoundé, Cameroun
| | | |
Collapse
|
150
|
Goosen C, Baumgartner J, Mikulic N, Barnabas SL, Cotton MF, Zimmermann MB, Blaauw R. Examining Associations of HIV and Iron Status with Nutritional and Inflammatory Status, Anemia, and Dietary Intake in South African Schoolchildren. Nutrients 2021; 13:nu13030962. [PMID: 33809705 PMCID: PMC8002246 DOI: 10.3390/nu13030962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 12/29/2022] Open
Abstract
The etiology of multifactorial morbidities such as undernutrition and anemia in children living with the human immunodeficiency virus (HIV) (HIV+) on antiretroviral therapy (ART) is poorly understood. Our objective was to examine associations of HIV and iron status with nutritional and inflammatory status, anemia, and dietary intake in school-aged South African children. Using a two-way factorial case-control design, we compared four groups of 8 to 13-year-old South African schoolchildren: (1) HIV+ and low iron stores (inflammation-unadjusted serum ferritin ≤ 40 µg/L), n = 43; (2) HIV+ and iron sufficient non-anemic (inflammation-unadjusted serum ferritin > 40 µg/L, hemoglobin ≥ 115 g/L), n = 41; (3) children without HIV (HIV-ve) and low iron stores, n = 45; and (4) HIV-ve and iron sufficient non-anemic, n = 45. We assessed height, weight, plasma ferritin (PF), soluble transferrin receptor (sTfR), plasma retinol-binding protein, plasma zinc, C-reactive protein (CRP), α-1-acid glycoprotein (AGP), hemoglobin, mean corpuscular volume, and selected nutrient intakes. Both HIV and low iron stores were associated with lower height-for-age Z-scores (HAZ, p < 0.001 and p = 0.02, respectively), while both HIV and sufficient iron stores were associated with significantly higher CRP and AGP concentrations. HIV+ children with low iron stores had significantly lower HAZ, significantly higher sTfR concentrations, and significantly higher prevalence of subclinical inflammation (CRP 0.05 to 4.99 mg/L) (54%) than both HIV-ve groups. HIV was associated with 2.5-fold higher odds of iron deficient erythropoiesis (sTfR > 8.3 mg/L) (95% CI: 1.03–5.8, p = 0.04), 2.7-fold higher odds of subclinical inflammation (95% CI: 1.4–5.3, p = 0.004), and 12-fold higher odds of macrocytosis (95% CI: 6–27, p < 0.001). Compared to HIV-ve counterparts, HIV+ children reported significantly lower daily intake of animal protein, muscle protein, heme iron, calcium, riboflavin, and vitamin B12, and significantly higher proportions of HIV+ children did not meet vitamin A and fiber requirements. Compared to iron sufficient non-anemic counterparts, children with low iron stores reported significantly higher daily intake of plant protein, lower daily intake of vitamin A, and lower proportions of inadequate fiber intake. Along with best treatment practices for HIV, optimizing dietary intake in HIV+ children could improve nutritional status and anemia in this vulnerable population. This study was registered at clinicaltrials.gov as NCT03572010.
Collapse
Affiliation(s)
- Charlene Goosen
- Division of Human Nutrition, Department of Global Health, Stellenbosch University, Cape Town 7505, South Africa;
- Correspondence:
| | - Jeannine Baumgartner
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland; (J.B.); (N.M.); (M.B.Z.)
| | - Nadja Mikulic
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland; (J.B.); (N.M.); (M.B.Z.)
| | - Shaun L. Barnabas
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town 7505, South Africa; (S.L.B.); (M.F.C.)
| | - Mark F. Cotton
- Family Centre for Research with Ubuntu, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town 7505, South Africa; (S.L.B.); (M.F.C.)
| | - Michael B. Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland; (J.B.); (N.M.); (M.B.Z.)
| | - Renée Blaauw
- Division of Human Nutrition, Department of Global Health, Stellenbosch University, Cape Town 7505, South Africa;
| |
Collapse
|