1
|
Tiemessen CT. Human models that inform antiretroviral therapy-free remission with perinatally acquired HIV infection. Curr Opin HIV AIDS 2025; 20:249-256. [PMID: 39946194 PMCID: PMC11970615 DOI: 10.1097/coh.0000000000000918] [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] [Indexed: 02/19/2025]
Abstract
PURPOSE OF REVIEW Rare persons who achieve disease-control despite high viral loads (viraemic nonprogressors) or maintain virologic control in the absence of antiretroviral therapy (ART) (elite controllers) or following ART interruption (posttreatment controllers) possess protective factors that can be harnessed for interventions to achieve ART-free remission. This review broadly summarizes these phenotypes in adults and children, and updates on findings important in informing strategies for ART-free remission in children with HIV. RECENT FINDINGS To date, only a few individual cases of posttreatment control have been described in children. Smaller HIV reservoir size with very early ART initiation in neonates with in-utero acquired HIV associates with improved virological and immunological outcomes. Nine new cases of ART-free remission in children were recently described - 4 from the P1115 trial, and 5 males from the Ucwaningo Lwabantwana study in South Africa. A striking reduction in the decay of intact proviruses was observed over three decades on suppressive ART in two early-treated twins with HIV. SUMMARY The unique environment of perinatal HIV infection favours effective restriction and decay of the HIV-1 reservoir with suppressive ART initiated very early. Sex and population differences require consideration in ongoing studies to inform ART-free remission.
Collapse
Affiliation(s)
- Caroline T Tiemessen
- Centre for HIV and STIs, National Institute for Communicable Diseases, a division of the National Health Laboratory Service, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
2
|
Bayón-Gil Á, Martinez-Picado J, Puertas MC. Viremic non-progression in HIV/SIV infection: A tied game between virus and host. Cell Rep Med 2025; 6:101921. [PMID: 39842407 PMCID: PMC11866547 DOI: 10.1016/j.xcrm.2024.101921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/16/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
High-efficacy antiretroviral treatment (ART) has been a game-changer for HIV/AIDS pandemic, but incomplete CD4+ T cell recovery and persistent chronic immune activation still affect HIV-suppressed people. Exceptional cases of HIV infection that naturally exhibit delayed disease progression provide invaluable insights into protective biological mechanisms with potential clinical application. Viremic non-progressors (VNPs) represent an extremely rare population of individuals with HIV, characterized by preservation of the CD4+ T cell compartment despite persistent high levels of viral load (>10,000 copies/mL). While only a few studies have investigated the immunovirological characteristics of adult and pediatric VNPs, most of our knowledge about this phenotype stems from its non-human-primate counterpart, the natural simian immunodeficiency virus (SIV) hosts. In this review, we synthesize the insights gained from recent studies of natural SIV hosts and VNPs and evaluate the potential similarities and differences in the mechanisms that underlie the absence of pathogenesis, with special focus on the control of immune activation.
Collapse
Affiliation(s)
- Ángel Bayón-Gil
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain; Germans Trias i Pujol Research Institute, Badalona, Spain; CIBERINFEC, Institute of Health Carlos III, Madrid, Spain; University of Vic-Central University of Catalonia, Vic, Spain; Catalan Institution for Research and Advanced Studies, Barcelona, Spain.
| | - Maria C Puertas
- IrsiCaixa Immunopathology Research Institute, Badalona, Spain; Germans Trias i Pujol Research Institute, Badalona, Spain; CIBERINFEC, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Fonseca JA, King AC, Chahroudi A. More than the Infinite Monkey Theorem: NHP Models in the Development of a Pediatric HIV Cure. Curr HIV/AIDS Rep 2024; 21:11-29. [PMID: 38227162 PMCID: PMC10859349 DOI: 10.1007/s11904-023-00686-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2023] [Indexed: 01/17/2024]
Abstract
PURPOSE OF REVIEW An HIV cure that eliminates the viral reservoir or provides viral control without antiretroviral therapy (ART) is an urgent need in children as they face unique challenges, including lifelong ART adherence and the deleterious effects of chronic immune activation. This review highlights the importance of nonhuman primate (NHP) models in developing an HIV cure for children as these models recapitulate the viral pathogenesis and persistence. RECENT FINDINGS Several cure approaches have been explored in infant NHPs, although knowledge gaps remain. Broadly neutralizing antibodies (bNAbs) show promise for controlling viremia and delaying viral rebound after ART interruption but face administration challenges. Adeno-associated virus (AAV) vectors hold the potential for sustained bNAb expression. Therapeutic vaccination induces immune responses against simian retroviruses but has yet to impact the viral reservoir. Combining immunotherapies with latency reversal agents (LRAs) that enhance viral antigen expression should be explored. Current and future cure approaches will require adaptation for the pediatric immune system and unique features of virus persistence, for which NHP models are fundamental to assess their efficacy.
Collapse
Affiliation(s)
- Jairo A Fonseca
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexis C King
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
- Emory+Children's Center for Childhood Infections and Vaccines, Atlanta, GA, USA.
| |
Collapse
|
4
|
Vieira V, Lim N, Singh A, Leitman E, Dsouza R, Adland E, Muenchhoff M, Roider J, Marin Lopez M, Carabelli J, Giandhari J, Groll A, Jooste P, Prado JG, Thobakgale C, Dong K, Kiepiela P, Prendergast AJ, Tudor-Williams G, Frater J, Walker BD, Ndung’u T, Ramsuran V, Leslie A, Kløverpris HN, Goulder P. Slow progression of pediatric HIV associates with early CD8+ T cell PD-1 expression and a stem-like phenotype. JCI Insight 2023; 8:e156049. [PMID: 36602861 PMCID: PMC9977437 DOI: 10.1172/jci.insight.156049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
HIV nonprogression despite persistent viremia is rare among adults who are naive to antiretroviral therapy (ART) but relatively common among ART-naive children. Previous studies indicate that ART-naive pediatric slow progressors (PSPs) adopt immune evasion strategies similar to those described in natural hosts of SIV. However, the mechanisms underlying this immunophenotype are not well understood. In a cohort of early-treated infants who underwent analytical treatment interruption (ATI) after 12 months of ART, expression of PD-1 on CD8+ T cells immediately before ATI was the main predictor of slow progression during ATI. PD-1+CD8+ T cell frequency was also negatively correlated with CCR5 and HLA-DR expression on CD4+ T cells and predicted stronger HIV-specific T lymphocyte responses. In the CD8+ T cell compartment of PSPs, we identified an enrichment of stem-like TCF-1+PD-1+ memory cells, whereas pediatric progressors and viremic adults had a terminally exhausted PD-1+CD39+ population. TCF-1+PD-1+ expression on CD8+ T cells was associated with higher proliferative activity and stronger Gag-specific effector functionality. These data prompted the hypothesis that the proliferative burst potential of stem-like HIV-specific cytotoxic cells could be exploited in therapeutic strategies to boost the antiviral response and facilitate remission in infants who received early ART with a preserved and nonexhausted T cell compartment.
Collapse
Affiliation(s)
- Vinicius Vieira
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Nicholas Lim
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Alveera Singh
- Africa Health Research Institute, Durban, South Africa
| | - Ellen Leitman
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Reena Dsouza
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Maximilian Muenchhoff
- Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University, Munich, Germany
- German Center for Infection Research, Munich, Germany
| | - Julia Roider
- German Center for Infection Research, Munich, Germany
- Department of Infectious Diseases, Ludwig-Maximilians-University, Munich, Germany
| | | | | | - Jennifer Giandhari
- KwaZulu-Natal Research Innovation and Sequencing Platform, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Andreas Groll
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - Pieter Jooste
- Department of Paediatrics, Kimberley Hospital, Kimberley, South Africa
| | - Julia G. Prado
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute, Badalona, Spain; Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Christina Thobakgale
- Faculty of Health Sciences, Centre for HIV and STIs, National Institute for Communicable Diseases, University of the Witwatersrand, Johannesburg, South Africa
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Krista Dong
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Photini Kiepiela
- South African Medical Research Council, Durban, South Africa
- Wits Health Consortium, Johannesburg, South Africa
| | - Andrew J. Prendergast
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Gareth Tudor-Williams
- Centre for Paediatrics and Child Health, Imperial College London, London, United Kingdom
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom
| | - Bruce D. Walker
- Africa Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Thumbi Ndung’u
- Africa Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Veron Ramsuran
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Henrik N. Kløverpris
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
5
|
Arije O, Titus R, Olaniran A, Dadi A, Garba D, Okeke E, Godpower O, Anyanti J, Idogho O, Roebersen C, Vrolings E, Onayade A. Effectiveness of community mobilisation models in improving HIV testing services uptake among women and children in Nigeria: A quasi-experimental study. Glob Public Health 2023; 18:2284880. [PMID: 38015746 DOI: 10.1080/17441692.2023.2284880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023]
Abstract
Nigeria carries a high burden of HIV infections, with Taraba State having a prevalence of 2.49%. This quasi-experimental study evaluated the impact of the Lafiyan Yara project, which utilised various community-based mobilisation models, on the enhancement of HTS uptake among women during pregnancy, and children. The intervention involved the implementation of mobilisation by Traditional Birth Attendants (TBA), Village Health Workers (VHW), Patent and Proprietary Medicine Vendors (PPMVs), and a combination of the three in four study local government areas (LGA) in Taraba State. Baseline and end-line surveys were conducted focused on women aged 15-49 years who delivered a child in the past 1 year, and children in their households, in the study and a control LGA. A difference-in-difference (DID) approach was applied by using a probit regression model with interaction terms for treatment status (intervention vs. control) and survey timing to compute the DID estimates of uptake of HTS. The TBA model showed the highest impact in the referral of women to HTS, while the combined model demonstrated the greatest impact in referrals for children. Scaling up and strengthening these community mobilisation efforts can improve access to HIV testing and contribute to HIV/AIDS prevention and control in the region.
Collapse
Affiliation(s)
- Olujide Arije
- Institute of Public Health, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Rachel Titus
- Institute of Public Health, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Akintayo Olaniran
- Institute of Public Health, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Aisha Dadi
- Society for Family Health, Abuja, Nigeria
| | | | | | | | | | | | | | | | - Adedeji Onayade
- Institute of Public Health, Obafemi Awolowo University, Ile-Ife, Nigeria
| |
Collapse
|
6
|
Vieira VA, Herbert N, Cromhout G, Adland E, Goulder P. Role of Early Life Cytotoxic T Lymphocyte and Natural Killer Cell Immunity in Paediatric HIV Cure/Remission in the Anti-Retroviral Therapy Era. Front Immunol 2022; 13:886562. [PMID: 35634290 PMCID: PMC9130627 DOI: 10.3389/fimmu.2022.886562] [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: 02/28/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Only three well-characterised cases of functional cure have been described in paediatric HIV infection over the past decade. This underlines the fact that early initiation of combination antiretroviral therapy (cART), whilst minimising the size of the viral reservoir, is insufficient to achieve cure, unless other factors contribute. In this review, we consider these additional factors that may facilitate functional cure in paediatric infection. Among the early life immune activity, these include HIV-specific cytotoxic T-lymphocyte (CTL) and natural killer (NK) cell responses. The former have less potent antiviral efficacy in paediatric compared with adult infection, and indeed, in early life, NK responses have greater impact in suppressing viral replication than CTL. This fact may contribute to a greater potential for functional cure to be achieved in paediatric versus adult infection, since post-treatment control in adults is associated less with highly potent CTL activity, and more with effective antiviral NK cell responses. Nonetheless, antiviral CTL responses can play an increasingly effective role through childhood, especially in individuals expressing then 'protective' HLA-I molecules HLA-B*27/57/58:01/8101. The role of the innate system on preventing infection, in shaping the particular viruses transmitted, and influencing outcome is discussed. The susceptibility of female fetuses to in utero mother-to-child transmission, especially in the setting of recent maternal infection, is a curiosity that also provides clues to mechanisms by which cure may be achieved, since initial findings are that viral rebound is less frequent among males who interrupt cART. The potential of broadly neutralising antibody therapy to facilitate cure in children who have received early cART is discussed. Finally, we draw attention to the impact of the changing face of the paediatric HIV epidemic on cure potential. The effect of cART is not limited to preventing AIDS and reducing the risk of transmission. cART also affects which mothers transmit. No longer are mothers who transmit those who carry genes associated with poor immune control of HIV. In the cART era, a high proportion (>70% in our South African study) of transmitting mothers are those who seroconvert in pregnancy or who for social reasons are diagnosed late in pregnancy. As a result, now, genes associated with poor immune control of HIV are not enriched in mothers who transmit HIV to their child. These changes will likely influence the effectiveness of HLA-associated immune responses and therefore cure potential among children.
Collapse
Affiliation(s)
- Vinicius A. Vieira
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Nicholas Herbert
- Africa Health Research Institute (AHRI), Nelson R Mandela School of Medicine, Durban, South Africa
| | - Gabriela Cromhout
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Emily Adland
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom,Africa Health Research Institute (AHRI), Nelson R Mandela School of Medicine, Durban, South Africa,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa,*Correspondence: Philip Goulder,
| |
Collapse
|
7
|
OUP accepted manuscript. J Appl Lab Med 2022; 7:1120-1130. [DOI: 10.1093/jalm/jfac026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/01/2022] [Indexed: 11/12/2022]
|
8
|
Vieira VA, Adland E, Malone DFG, Martin MP, Groll A, Ansari MA, Garcia-Guerrero MC, Puertas MC, Muenchhoff M, Guash CF, Brander C, Martinez-Picado J, Bamford A, Tudor-Williams G, Ndung’u T, Walker BD, Ramsuran V, Frater J, Jooste P, Peppa D, Carrington M, Goulder PJR. An HLA-I signature favouring KIR-educated Natural Killer cells mediates immune control of HIV in children and contrasts with the HLA-B-restricted CD8+ T-cell-mediated immune control in adults. PLoS Pathog 2021; 17:e1010090. [PMID: 34793581 PMCID: PMC8639058 DOI: 10.1371/journal.ppat.1010090] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/02/2021] [Accepted: 11/04/2021] [Indexed: 12/30/2022] Open
Abstract
Natural Killer (NK) cells contribute to HIV control in adults, but HLA-B-mediated T-cell activity has a more substantial impact on disease outcome. However, the HLA-B molecules influencing immune control in adults have less impact on paediatric infection. To investigate the contribution NK cells make to immune control, we studied >300 children living with HIV followed over two decades in South Africa. In children, HLA-B alleles associated with adult protection or disease-susceptibility did not have significant effects, whereas Bw4 (p = 0.003) and low HLA-A expression (p = 0.002) alleles were strongly associated with immunological and viral control. In a comparator adult cohort, Bw4 and HLA-A expression contributions to HIV disease outcome were dwarfed by those of protective and disease-susceptible HLA-B molecules. We next investigated the immunophenotype and effector functions of NK cells in a subset of these children using flow cytometry. Slow progression and better plasma viraemic control were also associated with high frequencies of less terminally differentiated NKG2A+NKp46+CD56dim NK cells strongly responsive to cytokine stimulation and linked with the immunogenetic signature identified. Future studies are indicated to determine whether this signature associated with immune control in early life directly facilitates functional cure in children.
Collapse
Affiliation(s)
- Vinicius A. Vieira
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Maureen P. Martin
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD and Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Andreas Groll
- Department of Statistics, TU Dortmund University, Dortmund, Germany
| | - M. Azim Ansari
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Mari C. Puertas
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- CIBER en Enfermedades Infecciosas, Madrid, Spain
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, LMU München, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Claudia Fortuny Guash
- Infectious Diseases and Systemic Inflammatory Response in Pediatrics, Infectious Diseases Unit, Department of Pediatrics, Sant Joan de Déu Hospital Research Foundation, Barcelona, Spain
- Center for Biomedical Network Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Pediatrics, University of Barcelona, Barcelona, Spain
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- CIBER en Enfermedades Infecciosas, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- CIBER en Enfermedades Infecciosas, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Alasdair Bamford
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | - Thumbi Ndung’u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute (AHRI), Durban, South Africa
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
- Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Bruce D. Walker
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute (AHRI), Durban, South Africa
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Veron Ramsuran
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - John Frater
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford, United Kingdom
| | - Pieter Jooste
- Department of Paediatrics, Kimberley Hospital, Kimberley, South Africa
| | - Dimitra Peppa
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Mary Carrington
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD and Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Philip J. R. Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
9
|
Attaf M, Roider J, Malik A, Rius Rafael C, Dolton G, Prendergast AJ, Leslie A, Ndung'u T, Kløverpris HN, Sewell AK, Goulder PJ. Cytomegalovirus-Mediated T Cell Receptor Repertoire Perturbation Is Present in Early Life. Front Immunol 2020; 11:1587. [PMID: 33101265 PMCID: PMC7554308 DOI: 10.3389/fimmu.2020.01587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Human cytomegalovirus (CMV) is a highly prevalent herpesvirus, particularly in sub-Saharan Africa, where it is endemic from infancy. The T cell response against CMV is important in keeping the virus in check, with CD8 T cells playing a major role in the control of CMV viraemia. Human leukocyte antigen (HLA) B*44:03-positive individuals raise a robust response against the NEGVKAAW (NW8) epitope, derived from the immediate-early-2 (IE-2) protein. We previously showed that the T cell receptor (TCR) repertoire raised against the NW8-HLA-B*44:03 complex was oligoclonal and characterised by superdominant clones, which were shared amongst unrelated individuals (i.e., "public"). Here, we address the question of how stable the CMV-specific TCR repertoire is over the course of infection, and whether substantial differences are evident in TCR repertoires in children, compared with adults. We present a longitudinal study of four HIV/CMV co-infected mother-child pairs, who in each case express HLA-B*44:03 and make responses to the NW8 epitope, and analyse their TCR repertoire over a period spanning more than 10 years. Using high-throughput sequencing, the paediatric CMV-specific repertoire was found to be highly diverse. In addition, paediatric repertoires were remarkably similar to adults, with public TCR responses being shared amongst children and adults alike. The CMV-specific repertoire in both adults and children displayed strong fluctuations in TCR clonality and repertoire architecture over time. Previously characterised superdominant clonotypes were readily identifiable in the children at high frequency, suggesting that the distortion of the CMV-specific repertoire is incurred as a direct result of CMV infection rather than a product of age-related "memory inflation." Early distortion of the TCR repertoire was particularly apparent in the case of the TCR-β chain, where oligoclonality was low in children and positively correlated with age, a feature we did not observe for TCR-α. This discrepancy between TCR-α and -β chain repertoire may reflect differential contribution to NW8 recognition. Altogether, the results of the present study provide insight into the formation of the TCR repertoire in early life and pave the way to better understanding of CD8 T cell responses to CMV at the molecular level.
Collapse
MESH Headings
- Adolescent
- Adult
- Age Factors
- Antigens, Viral/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Child
- Child, Preschool
- Coinfection
- Cytomegalovirus/immunology
- Cytomegalovirus Infections/immunology
- Cytomegalovirus Infections/metabolism
- Cytomegalovirus Infections/virology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Female
- HIV Infections/immunology
- HIV Infections/virology
- HLA Antigens/immunology
- High-Throughput Nucleotide Sequencing
- Humans
- Infant
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Peptides/chemistry
- Peptides/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Cell Antigen Receptor Specificity
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Viral Load
- Young Adult
Collapse
Affiliation(s)
- Meriem Attaf
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Julia Roider
- Human Immunodeficiency Virus Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- German Centre for Infection Research, Munich, Germany
- Department of Infectious Diseases, Ludwig-Maximilians-University, Munich, Germany
| | - Amna Malik
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Cristina Rius Rafael
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Andrew J. Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, United Kingdom
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Alasdair Leslie
- Africa Health Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Infection and Immunity, University College London, London, United Kingdom
| | - Thumbi Ndung'u
- Human Immunodeficiency Virus Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Infection and Immunity, University College London, London, United Kingdom
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Boston, MA, United States
- Virology and Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Henrik N. Kløverpris
- Africa Health Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Infection and Immunity, University College London, London, United Kingdom
| | - Andrew K. Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Philip J. Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Boston, MA, United States
| |
Collapse
|
10
|
Millar JR, Bengu N, Fillis R, Sprenger K, Ntlantsana V, Vieira VA, Khambati N, Archary M, Muenchhoff M, Groll A, Grayson N, Adamson J, Govender K, Dong K, Kiepiela P, Walker BD, Bonsall D, Connor T, Bull MJ, Nxele N, Roider J, Ismail N, Adland E, Puertas MC, Martinez-Picado J, Matthews PC, Ndung'u T, Goulder P. HIGH-FREQUENCY failure of combination antiretroviral therapy in paediatric HIV infection is associated with unmet maternal needs causing maternal NON-ADHERENCE. EClinicalMedicine 2020; 22:100344. [PMID: 32510047 PMCID: PMC7264978 DOI: 10.1016/j.eclinm.2020.100344] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Early combination antiretroviral therapy (cART) reduces the size of the viral reservoir in paediatric and adult HIV infection. Very early-treated children may have higher cure/remission potential. METHODS In an observational study of 151 in utero (IU)-infected infants in KwaZulu-Natal, South Africa, whose treatment adhered strictly to national guidelines, 76 infants diagnosed via point-of-care (PoC) testing initiated cART at a median of 26 h (IQR 18-38) and 75 infants diagnosed via standard-of-care (SoC) laboratory-based testing initiated cART at 10 days (IQR 8-13). We analysed mortality, time to suppression of viraemia, and maintenance of aviraemia over the first 2 years of life. FINDINGS Baseline plasma viral loads were low (median 8000 copies per mL), with 12% of infants having undetectable viraemia pre-cART initiation. However, barely one-third (37%) of children achieved suppression of viraemia by 6 months that was maintained to >12 months. 24% had died or were lost to follow up by 6 months. Infant mortality was 9.3%. The high-frequency virological failure in IU-infected infants was associated not with transmitted or acquired drug-resistant mutations but with cART non-adherence (plasma cART undetectable/subtherapeutic, p<0.0001) and with concurrent maternal cART failure (OR 15.0, 95%CI 5.6-39.6; p<0.0001). High-frequency virological failure was observed in PoC- and SoC-tested groups of children. INTERPRETATION The success of early infant testing and cART initiation strategies is severely limited by subsequent cART non-adherence in HIV-infected children. Although there are practical challenges to administering paediatric cART formulations, these are overcome by mothers who themselves are cART-adherent. These findings point to the ongoing obligation to address the unmet needs of the mothers. Eliminating the particular barriers preventing adequate treatment for these vulnerable women and infants need to be prioritised in order to achieve durable suppression of viraemia on cART, let alone HIV cure/remission, in HIV-infected children. FUNDING Wellcome Trust, National Institutes of Health.
Collapse
Affiliation(s)
- Jane R Millar
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Nomonde Bengu
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
| | - Rowena Fillis
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
| | - Ken Sprenger
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
| | | | - Vinicius A Vieira
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Nisreen Khambati
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Moherndran Archary
- Department of Paediatrics, University of KwaZulu-Natal, Durban, South Africa
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich, Germany
- German Center for Infection Research (DZIF), Partner site Munich, Germany
| | - Andreas Groll
- TU Dortmund University, Department of Statistics, Vogelpothsweg 87, 44227 Dortmund
| | - Nicholas Grayson
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - John Adamson
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Katya Govender
- Africa Health Research Institute (AHRI), Durban, South Africa
| | - Krista Dong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Photini Kiepiela
- South African Medical Research Council, Durban 4001, SC Africa
- Wits Health Consortium, Johannesburg 2193, SC Africa
| | - Bruce D Walker
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Institute for Medical Engineering and Sciences and Department of Biology, Massachusetts Institute of Technology, Cambridge MA 02139, United States
- Howard Hughes Medical Institute, Chevy Chase MD 20815, United States
| | - David Bonsall
- Oxford Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas Connor
- Cardiff University School of Biosciences, The Sir Martin Evans Building, Cardiff University, Cardiff, United Kingdom
| | - Matthew J Bull
- Pathogen Genomics Unit, Public Health Wales Microbiology Cardiff, University Hospital of Wales, Cardiff, United Kingdom
| | - Nelisiwe Nxele
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Julia Roider
- German Center for Infection Research (DZIF), Partner site Munich, Germany
- Department of Infectious Diseases, Ludwig-Maximilians-University, Munich
| | - Nasreen Ismail
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Philippa C Matthews
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Microbiology and Infectious Diseases, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Oxford BRC, John Radcliffe Hospital, Oxford, United Kingdom
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Africa Health Research Institute (AHRI), Durban, South Africa
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Philip Goulder
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| |
Collapse
|
11
|
Lou H, Ge X, Xu B, Liu W, Zhou YH. Prevention of Mother-to-Child Transmission of HIV: Data Analysis Based on Pregnant Women Population from 2012 to 2018, in Nantong City, China. Curr HIV Res 2020; 18:458-465. [PMID: 32778030 DOI: 10.2174/1570162x18666200810134025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND China has implemented a nation-wide policy to control mother-to-child transmission (MTCT) of the human immunodeficiency virus (HIV) since 2011, yet the efficacy of the control policy is less studied. The aim of the present study was to report the data in the prevention of MTCT of HIV in Nantong city, China. METHODS The screening and prevalence of HIV in pregnant women and the efficacy of prophylaxis in Nantong city, China, January 2012 through December 2018, were analyzed. RESULTS Among a total population of 410,044 pregnant women, anti-HIV was tested prenatally in 393,658 (96.0%) women and in 16,287 (3.97%) women at delivery. In total, 51 women were confirmed with HIV infection. After the exclusion of repeat pregnancies, the overall prevalence of HIV infection was 1.20/10 000 (48/400,377). The prevalence (6.75/10,000) in women tested at delivery was >5-fold higher than that (1.02/10,000) in prenatally screened women. Of 48 HIV-infected women, 12 terminated their pregnancies and 36 others delivered 36 neonates, of whom 35 were followed up. No HIV infection occurred in 24 children born to mothers with antiretroviral therapy (ART) during pregnancy along with other preventive measures. Among 11 children born to mothers who did not receive ART during pregnancy because of the absence of a prenatal anti-HIV test, none of the 6 children who were delivered by cesarean section and timely administered neonatal antiretroviral prophylaxis was infected, but 2 (40%) of 5 children who were spontaneously delivered and administered delayed antiretroviral prophylaxis were infected. CONCLUSION Prenatal identification of HIV infection and timely administration of all preventive measures can completely block MTCT of HIV. The data indicate that more efforts must be taken to ensure that all pregnant women are tested for anti-HIV during pregnancy. For pregnant women who missed the prenatal screening, a positive result in rapid anti-HIV test at delivery should be sufficient to take preventive measures to prevent MTCT of HIV.
Collapse
Affiliation(s)
- Haiqin Lou
- Department of Mass Health Care, Nantong Municipal Maternal and Child Health Hospital, Nantong, Jiangsu, China
| | - Xiaoyun Ge
- Department of Mass Health Care, Nantong Municipal Maternal and Child Health Hospital, Nantong, Jiangsu, China
| | - Biyun Xu
- Department of Biostatistics, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Weiwei Liu
- Department of Mass Health Care, Nantong Municipal Maternal and Child Health Hospital, Nantong, Jiangsu, China
| | - Yi-Hua Zhou
- Departments of Laboratory Medicine and Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
12
|
CD71 + Erythroid Cells Exacerbate HIV-1 Susceptibility, Mediate trans-Infection, and Harbor Infective Viral Particles. mBio 2019; 10:mBio.02767-19. [PMID: 31772057 PMCID: PMC6879723 DOI: 10.1128/mbio.02767-19] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Immature red blood cells (erythroid precursors or CD71+ erythroid cells) have a wide range of immunomodulatory properties. In this study, we found that these erythroid precursors are abundant in the human cord blood/placental tissues, in the blood of HIV-infected and anemic individuals. We observed that these cells exacerbate HIV-1 replication/infection in target cells and even make HIV target cells more permissible to HIV infection. In addition, we found that HIV gets a free ride by binding on the surface of these cells and thus can travel to different parts of the body. In agreement, we noticed a positive correlation between the plasma viral load and the frequency of these cells in HIV patients. More importantly, we observed that infective HIV particles reside inside these erythroid precursors but not mature red blood cells. Therefore, these cells by harboring HIV can play an important role in HIV pathogenesis. CD71+ erythroid cells (CECs) have a wide range of immunomodulatory properties. Here, we show that CECs are expanded in the peripheral blood of HIV patients, with a positive correlation between their frequency and the plasma viral load. CECs from HIV patients and human cord blood/placenta exacerbate HIV-1 infection/replication when cocultured with CD4+ T cells, and that preexposure of CD4+ T cells to CECs enhances their permissibility to HIV infection. However, mature red blood cells (RBCs) do not enhance HIV replication when cocultured with CD4+ T cells. We also found CECs express substantial levels of the NOX2 gene and via a mitochondrial reactive oxygen species (ROS)-dependent mechanism possibly upregulate NF-κB in CD4+ T cells once cocultured, which affects the cell cycle machinery to facilitate HIV-1 replication. The complement receptor-1 (CD35) and the Duffy antigen receptor for chemokines (DARC) as potential HIV target molecules are expressed significantly higher on CECs compared to mature red blood cells. Blocking CD35 or DARC substantially abolishes HIV-1 transmission by RBCs to uninfected CD4+ T cells but not by CECs. In contrast, we observed CECs bind to HIV-1 via CD235a and subsequently transfer the virus to uninfected CD4+ T cells, which can be partially blocked by the anti-CD235a antibody. More importantly, we found that CECs from HIV-infected individuals in the presence of antiretroviral therapy harbor infective viral particles, which mediate HIV-1 trans-infection of CD4+ T cells. Therefore, our findings provide a novel insight into the role of CECs in HIV pathogenesis as potential contributing cells in viral persistence and transmission.
Collapse
|
13
|
Dakum P, Tola M, Iboro N, Okolo CA, Anuforom O, Chime C, Peters S, Jumare J, Ogbanufe O, Ahmad A, Ndembi N. Correlates and determinants of Early Infant Diagnosis outcomes in North-Central Nigeria. AIDS Res Ther 2019; 16:27. [PMID: 31521170 PMCID: PMC6744629 DOI: 10.1186/s12981-019-0245-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/30/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A negative status following confirmatory Early Infant Diagnosis (EID) is the desired pediatric outcome of prevention of Mother to Child Transmission (PMTCT) programs. EID impacts epidemic control by confirming non-infected HIV-exposed infants (HEIs) and prompting timely initiation of ART in HIV-infected babies which improves treatment outcomes. OBJECTIVES We explored factors associated with EID outcomes among HEI in North-Central Nigeria. METHOD This is a cross-sectional study using EID data of PMTCT-enrollees matched with results of HEI's dried blood samples (DBS), processed for DNA-PCR from January 2015 through July 2017. Statistical analyses were done using SPSS version 20.0 to generate frequencies and examine associations, including binomial logistic regression with p < 0.05 being statistically significant. RESULTS Of 14,448 HEI in this analysis, 51.8% were female and 95% (n = 12,801) were breastfed. The median age of the infants at sample collection was 8 weeks (IQR 6-20), compared to HEI tested after 20 weeks of age, those tested earlier had significantly greater odds of a negative HIV result (≤ 6 weeks: OR = 3.8; 6-8 weeks: OR = 2.1; 8-20 weeks: OR = 1.5) with evidence of a significant linear trend (p < 0.001). Similarly, HEI whose mothers received combination antiretroviral therapy (cART) before (OR = 11.8) or during the index pregnancy (OR = 8.4) had significantly higher odds as compared to those whose mothers did not receive cART. In addition, HEI not breastfed had greater odds of negative HIV result as compared to those breastfed (OR = 1.9). CONCLUSIONS cART prior to and during pregnancy, earlier age of HEI at EID testing and alternative feeding other than breastfeeding were associated with an increased likelihood of being HIV-negative on EID. Therefore, strategies to scale-up PMTCT services are needed to mitigate the burden of HIV among children.
Collapse
|
14
|
Millar JR, Mvo Z, Bengu N, Fillis R, Sprenger K, Matthews PC, Archary M, Ndung'u T, Adland E, Puertas MC, Martinez-Picado J, Goulder P. Increasing Diagnostic Uncertainties in Children With In Utero HIV Infection. Pediatr Infect Dis J 2019; 38:e166-e168. [PMID: 31033913 PMCID: PMC7612888 DOI: 10.1097/inf.0000000000002315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We present a case of an in utero HIV-infected child, who on day 1 of life had a positive whole blood total nucleic acid test but viral load <20 RNA copies/mL. Dried blood spot total nucleic acid testing was negative on day 1, 10 and at 3 months, while on ART prophylaxis then positive at 5 months after prophylaxis ended. Retrospective peripheral blood mononuclear cells HIV DNA testing from day 1 of life was positive, confirming in utero infection.
Collapse
Affiliation(s)
- Jane R Millar
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Zodumo Mvo
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
| | - Nomonde Bengu
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
| | - Rowena Fillis
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
| | - Ken Sprenger
- Umkhuseli Innovation and Research Management, Pietermaritzburg, South Africa
| | | | - Moherndran Archary
- Department of Paediatrics, University of KwaZulu-Natal, Durban, South Africa
| | - Thumbi Ndung'u
- Ragon Institute of MGH, MIT and Havard, Cambridge, MA, USA
- Africa Health Research Institute, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Maria C Puertas
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Javier Martinez-Picado
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- University of Vic-Central University of Catalonia, Vic, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
D'Souza RR, Gopalan BP, Rajnala N, Phetsouphanh C, Shet A. Increased monocyte activation with age among HIV-infected long term non-progressor children: implications for early treatment initiation. HIV Med 2019; 20:513-522. [PMID: 31131542 DOI: 10.1111/hiv.12751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The key to newer therapeutic and eradication approaches often lies in understanding slow disease progression in HIV infection. The paediatric population has been poorly studied in this regard. We aimed to describe a cohort of perinatally infected long-term nonprogressor (LTNP) children living with HIV in India and to evaluate the immune biomarkers of disease progression. METHODS LTNPs (ART-naïve, with a CD4 count ≥ 500 cells/μL at age ≥ 7 years) among the cohort of HIV-infected children were identified and monitored longitudinally, and their CD4 T-cell counts and plasma viral loads were measured every 6 months. The plasma monocyte/macrophage activation markers, namely soluble CD14 (sCD14), soluble CD163 (sCD163) and interferon-inducible protein-10 (IP-10) were measured by enzyme-linked immunosorbent assay (ELISA) in LTNPs and progressors. The Mann-Whitney U-test was used to compare the two groups and P values < 0.05 were considered statistically significant. Spearman's rank or Pearson's correlation coefficient (r) was calculated to determine the associations between variables. RESULTS Among 378 children living with HIV-1 surveyed in our cohort, 40 (10.6%) were LTNPs. Longitudinal analysis of the LTNP data showed that both CD4 count and viral load declined significantly with age (P < 0.0001 for both). Plasma sCD14 levels were significantly (P < 0.005) higher in progressors and sCD163 levels were significantly (P < 0.0001) higher in LTNPs. CONCLUSIONS The prevalence of LTNPs in our cohort of perinatally infected children living with HIV was 10.6%. We observed a trend for associations between the increasing sCD163 monocyte/macrophage activation marker levels, declining CD4 counts and the gradual loss of nonprogressor status with age in the LTNPs. These findings underscore the need for early antiretroviral therapy in those children with proven slow disease progression.
Collapse
Affiliation(s)
- R R D'Souza
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK.,Division of Infectious Diseases, St John's Research Institute, Bangalore, India
| | - B P Gopalan
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India.,The University of Trans-disciplinary Health Sciences and Technology, Bangalore, India
| | - N Rajnala
- Division of Infectious Diseases, St John's Research Institute, Bangalore, India
| | - C Phetsouphanh
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - A Shet
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
16
|
Abstract
Background: Reports of posttreatment control following antiretroviral therapy (ART) have prompted the question of how common immune control of HIV infection is in the absence of ART. In contrast to adult infection, where elite controllers have been very well characterized and constitute approximately 0.5% of infections, very few data exist to address this question in paediatric infection. Methods: We describe 11 ART-naive elite controllers from 10 cohorts of HIV-infected children being followed in South Africa, Brazil, Thailand, and Europe. Results: All but one of the elite controllers (91%) are females. The median age at which control of viraemia was achieved was 6.5 years. Five of these 11 (46%) children lost control of viraemia at a median age of 12.9 years. Children who maintained control of viraemia had significantly higher absolute CD4+ cell counts in the period of elite control than those who lost viraemic control. On the basis of data available from these cohorts, the prevalence of elite controllers in paediatric infection is estimated to be 5–10-fold lower than in adults. Conclusion: Although conclusions are limited by the study design, these data suggest that, whilst paediatric elite control can be achieved, compared with adult elite controllers, this occurs rarely, and takes some years after infection to achieve. Also, loss of immune control arises in a high proportion of children and often relatively rapidly. These findings are consistent with the more potent antiviral immune responses observed in adults and in females.
Collapse
|
17
|
Abstract
In this brief review and perspective, we address the question of whether the immune responses that bring about immune control of acute HIV infection are the same as, or distinct from, those that maintain long-term viral suppression once control of viremia has been achieved. To this end, we describe the natural history of elite and post-treatment control, noting the lack of data regarding what happens acutely. We review the evidence suggesting that the two clinical phenotypes may differ in terms of the mechanisms required to achieve and maintain control, as well as the level of inflammation that persists once a steady state is achieved. We then describe the evidence from longitudinal studies of controllers who fail and studies of biologic sex (male versus female), age (children versus adults), and simian immunodeficiency virus (SIV) (pathogenic/experimental versus nonpathogenic/natural infection). Collectively, these studies demonstrate that the battle between the inflammatory and anti-inflammatory pathways during acute infection has long-term consequences, both for the degree to which control is maintained and the health of the individual. Potent and stringent control of HIV may be required acutely, but once control is established, the chronic inflammatory response can be detrimental. Interventional approaches designed to bring about HIV cure and/or remission should be nuanced accordingly.
Collapse
Affiliation(s)
- Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco, California, United States of America
| |
Collapse
|
18
|
Recovery of effective HIV-specific CD4+ T-cell activity following antiretroviral therapy in paediatric infection requires sustained suppression of viraemia. AIDS 2018; 32:1413-1422. [PMID: 29734220 PMCID: PMC6039399 DOI: 10.1097/qad.0000000000001844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Background: The success of increasing access to antiretroviral therapy (ART) in paediatric HIV infection prompts the question of the potential for eradication of HIV infection in this age group. ‘Shock-and-kill’ HIV cure approaches, currently in development, may depend upon an effective antiviral T-cell response to eradicate virus-infected cells. Method: We here investigate the ability of HIV-infected children receiving ART from early childhood (median 24 months’ age) to generate effective HIV-specific CD4+ and CD8+ T-cell immune responses that would facilitate future immune-based cure therapies. Results: Initial analysis of ART-naive HIV-infected children demonstrated that maintenance of normal-for-age absolute CD4+ T-cell counts was strongly linked to high IL-2 production and polyfunctional HIV-specific CD4+ T-cell responses (P < 0.0001 in each case). Low viral load was, similarly, strongly associated with markedly low IFN-γ and high IL-2 HIV-specific CD4+ T-cell responses (P < 0.0001). In children receiving ART, establishment of this immune profile (high IL-2 and low IFN-γ HIV-specific T-cell production) was strongly related to the duration of viraemic suppression. Failure to suppress viraemia on ART, and even the successful suppression of viraemia interrupted by the occurrence of transient viraemia of more than 1000 HIV copies/ml, was associated with an immune profile of high IFN-γ and low IL-2 HIV-specific T-cell responses and low polyfunctionality. Conclusion: These data are consistent with recovery of functional CD4+ T-cell responses in ART-treated children, in contrast to relative lack of CD4+ T-cell function recovery described in ART-treated adults. However, the challenges of achieving long-term suppression of viraemia in ART-treated children through adolescence remain daunting.
Collapse
|
19
|
Muenchhoff M, Adland E, Karimanzira O, Crowther C, Pace M, Csala A, Leitman E, Moonsamy A, McGregor C, Hurst J, Groll A, Mori M, Sinmyee S, Thobakgale C, Tudor-Williams G, Prendergast AJ, Kloverpris H, Roider J, Leslie A, Shingadia D, Brits T, Daniels S, Frater J, Willberg CB, Walker BD, Ndung'u T, Jooste P, Moore PL, Morris L, Goulder P. Nonprogressing HIV-infected children share fundamental immunological features of nonpathogenic SIV infection. Sci Transl Med 2017; 8:358ra125. [PMID: 27683550 DOI: 10.1126/scitranslmed.aag1048] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022]
Abstract
Disease-free infection in HIV-infected adults is associated with human leukocyte antigen-mediated suppression of viremia, whereas in the sooty mangabey and other healthy natural hosts of simian immunodeficiency virus (SIV), viral replication continues unabated. To better understand factors preventing HIV disease, we investigated pediatric infection, where AIDS typically develops more rapidly than in adults. Among 170 nonprogressing antiretroviral therapy-naïve children aged >5 years maintaining normal-for-age CD4 T cell counts, immune activation levels were low despite high viremia (median, 26,000 copies/ml). Potent, broadly neutralizing antibody responses in most of the subjects and strong virus-specific T cell activity were present but did not drive pediatric nonprogression. However, reduced CCR5 expression and low HIV infection in long-lived central memory CD4 T cells were observed in pediatric nonprogressors. These children therefore express two cardinal immunological features of nonpathogenic SIV infection in sooty mangabeys-low immune activation despite high viremia and low CCR5 expression on long-lived central memory CD4 T cells-suggesting closer similarities with nonpathogenetic mechanisms evolved over thousands of years in natural SIV hosts than those operating in HIV-infected adults.
Collapse
Affiliation(s)
- Maximilian Muenchhoff
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Max von Pettenkofer-Institute, Department of Virology, Ludwig-Maximilians-University Munich, Munich, Germany. German Center for Infection Research (DZIF), Partner Site Munich, Germany
| | - Emily Adland
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Owen Karimanzira
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Carol Crowther
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Matthew Pace
- Institute for Emerging Infections, Oxford Martin School, University of Oxford, Oxford, U.K. Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Anna Csala
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Ellen Leitman
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Angeline Moonsamy
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Callum McGregor
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Jacob Hurst
- Institute of Cancer Research, Old Brompton Road, London SW7 3RP, U.K
| | - Andreas Groll
- Department of Mathematics, Ludwig-Maximilians-University Munich, Theresienstrasse 39, 80333 Munich, Germany
| | - Masahiko Mori
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Smruti Sinmyee
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K
| | - Christina Thobakgale
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | | | | | - Henrik Kloverpris
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa. Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Julia Roider
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa
| | - Alasdair Leslie
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa
| | - Delane Shingadia
- Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children, London, U.K
| | - Thea Brits
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Samantha Daniels
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - John Frater
- Institute for Emerging Infections, Oxford Martin School, University of Oxford, Oxford, U.K. Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Christian B Willberg
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, South Parks Road, Oxford OX1 3SY, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, U.K
| | - Bruce D Walker
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139-4307, USA
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal (UKZN), 4001 Durban, South Africa. Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139-4307, USA. Max Planck Institute for Infection Biology, Berlin, Germany
| | - Pieter Jooste
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Penny L Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa. Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. Center for the AIDS Programme of Research in South Africa (CAPRISA), 4001 Durban, South Africa
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa. Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. Center for the AIDS Programme of Research in South Africa (CAPRISA), 4001 Durban, South Africa
| | - Philip Goulder
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Road, University of Oxford, Oxford OX1 3SY, U.K. HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal (UKZN), Durban, South Africa. Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children, London, U.K.
| |
Collapse
|
20
|
Abstract
INTRODUCTION Recent goals of antiretroviral treatment of neonates have expanded from reducing morbidity and mortality to also aiming to facilitate HIV remission. Areas covered: In this review, we present and discuss the rationale and evidence-bases for each of these distinct goals. Next we discuss the challenges of how to identify HIV-infected neonates. Finally, we discuss the specific antiretroviral drugs that are preferred for this group, making distinctions between the use of these agents in prevention and treatment. Expert commentary: The clinical and scientific challenges of pharmacological treatment of acute HIV infection in neonates are complicated by externalities beyond biology. At the same time, these challenges are energized by the unique biological opportunities afforded by investigating this population, including a unique immune profile, ability to study both mother and neonate as well as transmitted and acquired virus, and time period spanning both the period soon after infection as well as the period of viral reservoir establishment and related damage. Given the unique scientific opportunities afforded by study of pharmacologic treatment of acute HIV infection in neonates, we hypothesize that over the next five years breakthroughs may occur that may lead to new interventions effective at achieving HIV remission.
Collapse
Affiliation(s)
- Louise Kuhn
- Professor of Epidemiology, Gertrude H. Sergievsky Center, College of Physicians and Surgeons 622, West 168 Street, PH 19-113 New York, NY, 10032
| | - Stephanie Shiau
- Postdoctoral Research Scientist Gertrude H. Sergievsky Center College of Physicians and Surgeons 622 West 168 Street, PH 19-118 New York, NY, 10032,
| |
Collapse
|
21
|
Leitman EM, Thobakgale CF, Adland E, Ansari MA, Raghwani J, Prendergast AJ, Tudor-Williams G, Kiepiela P, Hemelaar J, Brener J, Tsai MH, Mori M, Riddell L, Luzzi G, Jooste P, Ndung'u T, Walker BD, Pybus OG, Kellam P, Naranbhai V, Matthews PC, Gall A, Goulder PJR. Role of HIV-specific CD8 + T cells in pediatric HIV cure strategies after widespread early viral escape. J Exp Med 2017; 214:3239-3261. [PMID: 28983013 PMCID: PMC5679167 DOI: 10.1084/jem.20162123] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 06/22/2017] [Accepted: 08/30/2017] [Indexed: 11/04/2022] Open
Abstract
Recent studies have suggested greater HIV cure potential among infected children than adults. A major obstacle to HIV eradication in adults is that the viral reservoir is largely comprised of HIV-specific cytotoxic T lymphocyte (CTL) escape variants. We here evaluate the potential for CTL in HIV-infected slow-progressor children to play an effective role in "shock-and-kill" cure strategies. Two distinct subgroups of children were identified on the basis of viral load. Unexpectedly, in both groups, as in adults, HIV-specific CTL drove the selection of escape variants across a range of epitopes within the first weeks of infection. However, in HIV-infected children, but not adults, de novo autologous variant-specific CTL responses were generated, enabling the pediatric immune system to "corner" the virus. Thus, even when escape variants are selected in early infection, the capacity in children to generate variant-specific anti-HIV CTL responses maintains the potential for CTL to contribute to effective shock-and-kill cure strategies in pediatric HIV infection.
Collapse
Affiliation(s)
- Ellen M Leitman
- Department of Paediatrics, University of Oxford, Oxford, England, UK
| | - Christina F Thobakgale
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, England, UK
| | - M Azim Ansari
- Oxford Martin School, University of Oxford, Oxford, England, UK
| | - Jayna Raghwani
- Department of Zoology, University of Oxford, Oxford, England, UK
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, England, UK.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Gareth Tudor-Williams
- Division of Medicine, Department of Paediatrics, Imperial College London, London, England, UK
| | - Photini Kiepiela
- Medical Research Council, Durban, South Africa.,Witwatersrand Health Consortium, Johannesburg, South Africa
| | - Joris Hemelaar
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford, England, UK.,Linacre Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, University of Witwatersrand, Johannesburg, South Africa
| | - Jacqui Brener
- Department of Paediatrics, University of Oxford, Oxford, England, UK
| | - Ming-Han Tsai
- Department of Paediatrics, University of Oxford, Oxford, England, UK
| | - Masahiko Mori
- Department of Paediatrics, University of Oxford, Oxford, England, UK.,Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Lynn Riddell
- Northampton Healthcare NHS Foundation Trust, Cliftonville, England, UK
| | - Graz Luzzi
- Buckinghampshire Healthcare NHS Foundation Trust, High Wycombe, England, UK
| | - Pieter Jooste
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Thumbi Ndung'u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA
| | - Bruce D Walker
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.,Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford, England, UK
| | - Paul Kellam
- Kymab Ltd., Babraham Research Campus, Babraham, England, UK.,Department of Medicine, Division of Infectious Diseases, Imperial College Faculty of Medicine, London, England, UK
| | - Vivek Naranbhai
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA.,Centre for the AIDS Programme of Research in South Africa, University of KwaZulu Natal, Durban, South Africa
| | - Philippa C Matthews
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, England, UK
| | - Astrid Gall
- Wellcome Trust Sanger Institute, Hinxton, England, UK
| | - Philip J R Goulder
- Department of Paediatrics, University of Oxford, Oxford, England, UK .,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
22
|
CD71 + erythroid suppressor cells impair adaptive immunity against Bordetella pertussis. Sci Rep 2017; 7:7728. [PMID: 28798335 PMCID: PMC5552872 DOI: 10.1038/s41598-017-07938-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/05/2017] [Indexed: 01/03/2023] Open
Abstract
Infant’s immune system cannot control infection or respond to vaccination as efficiently as older individuals, a phenomenon that has been attributed to immunological immaturity. Recently, we challenged this notion and proposed the presence of actively immunosuppressive and physiologically enriched CD71+ erythroid cells in neonates. Here we utilized Bordetella pertussis, a common neonatal respiratory tract pathogen, as a proof of concept to investigate the role of these cells in adaptive immunity. We observed that CD71+ cells have distinctive immunosuppressive properties and prevent recruitment of immune cells to the mucosal site of infection. CD71+ cells ablation unleashed induction of B. pertussis-specific protective cytokines (IL-17 and IFN-γ) in the lungs and spleen upon re-infection or vaccination. We also found that CD71+ cells suppress systemic and mucosal B. pertussis-specific antibody responses. Enhanced antigen-specific adaptive immunity following CD71+ cells depletion increased resistance of mice to B. pertussis infection. Furthermore, we found that human cord blood CD71+ cells also suppress T and B cell functions in vitro. Collectively, these data provide important insight into the role of CD71+ erythroid cells in adaptive immunity. We anticipate our results will spark renewed investigation in modulating the function of these cells to enhance host defense to infections in newborns.
Collapse
|
23
|
Mother-to-Child HIV Transmission Bottleneck Selects for Consensus Virus with Lower Gag-Protease-Driven Replication Capacity. J Virol 2017. [PMID: 28637761 DOI: 10.1128/jvi.00518-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In the large majority of cases, HIV infection is established by a single variant, and understanding the characteristics of successfully transmitted variants is relevant to prevention strategies. Few studies have investigated the viral determinants of mother-to-child transmission. To determine the impact of Gag-protease-driven viral replication capacity on mother-to-child transmission, the replication capacities of 148 recombinant viruses encoding plasma-derived Gag-protease from 53 nontransmitter mothers, 48 transmitter mothers, and 47 infected infants were assayed in an HIV-1-inducible green fluorescent protein reporter cell line. All study participants were infected with HIV-1 subtype C. There was no significant difference in replication capacities between the nontransmitter (n = 53) and transmitter (n = 44) mothers (P = 0.48). Infant-derived Gag-protease NL4-3 recombinant viruses (n = 41) were found to have a significantly lower Gag-protease-driven replication capacity than that of viruses derived from the mothers (P < 0.0001 by a paired t test). High percent similarities to consensus subtype C Gag, p17, p24, and protease sequences were also found in the infants (n = 28) in comparison to their mothers (P = 0.07, P = 0.002, P = 0.03, and P = 0.02, respectively, as determined by a paired t test). These data suggest that of the viral quasispecies found in mothers, the HIV mother-to-child transmission bottleneck favors the transmission of consensus-like viruses with lower viral replication capacities.IMPORTANCE Understanding the characteristics of successfully transmitted HIV variants has important implications for preventative interventions. Little is known about the viral determinants of HIV mother-to-child transmission (MTCT). We addressed the role of viral replication capacity driven by Gag, a major structural protein that is a significant determinant of overall viral replicative ability and an important target of the host immune response, in the MTCT bottleneck. This study advances our understanding of the genetic bottleneck in MTCT by revealing that viruses transmitted to infants have a lower replicative ability as well as a higher similarity to the population consensus (in this case HIV subtype C) than those of their mothers. Furthermore, the observation that "consensus-like" virus sequences correspond to lower in vitro replication abilities yet appear to be preferentially transmitted suggests that viral characteristics favoring transmission are decoupled from those that enhance replicative capacity.
Collapse
|
24
|
Girma M, Wendaferash R, Shibru H, Berhane Y, Hoelscher M, Kroidl A. Uptake and performance of prevention of mother-to-child transmission and early infant diagnosis in pregnant HIV-infected women and their exposed infants at seven health centres in Addis Ababa, Ethiopia. Trop Med Int Health 2017; 22:765-775. [PMID: 28407452 DOI: 10.1111/tmi.12881] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To assess the uptake of WHO-recommended PMTCT procedures in Ethiopia's health services. METHODS Prospective observational study of HIV-positive pregnant mothers and their newborns attending PMTCT services at seven health centres in Addis Ababa. Women were recruited during antenatal care and followed up with their newborns at delivery, Day 6 and Week 6 post-partum. Retention to PMCTC procedures, self-reported antiretroviral treatment (ART) adherence and HIV infant outcome were assessed. Turnaround times of HIV early infant diagnosis (EID) procedures were extracted from health registers. RESULTS Of 494 women enrolled, 4.9% did not complete PMTCT procedures due to active denial or loss to follow-up. HIV was first diagnosed in 223 (45.1%) and ART initiated in 321 (65.0%) women during pregnancy. ART was initiated in a median of 1.3 weeks (IQR 0-4.3) after HIV diagnosis. Poor self-reported treatment adherence was higher post-partum than during pregnancy (12.5% vs. 7.0%, P = 0.002) and significantly associated with divorced/separated marital status (RR 2.2, 95% CI 1.3-3.8), low family income (RR 2.1, 95% CI 1.1-4.1), low CD4 count (RR 1.7, 95% CI 1.0-3.0) and ART initiation during delivery (RR 2.5, 95% CI 1.1-5.6). Of 435 infants born alive, 98.6% received nevirapine prophylaxis. The mother-to-child HIV transmission rate was 0.7% after a median of 6.7 weeks (IQR 6.4-10.4), but EID results were received for only 46.6% within 3 months of birth. CONCLUSION High retention in PMTCT services, triple maternal ART and high infant nevirapine prophylaxis coverage were associated with low mother-to-child HIV transmission. Declining post-partum ART adherence and challenges of EID linkage require attention.
Collapse
Affiliation(s)
- Marshet Girma
- Addis Continental Institute of Public Health, Addis Ababa, Ethiopia.,Center for International Health, University of Munich, Munich, Germany
| | | | | | - Yemane Berhane
- Addis Continental Institute of Public Health, Addis Ababa, Ethiopia
| | - Michael Hoelscher
- Center for International Health, University of Munich, Munich, Germany.,Division of Infectious Diseases and Tropical Medicine, University of Munich, Munich, Germany.,German Centre for Infection Research, Munich, Germany
| | - Arne Kroidl
- Division of Infectious Diseases and Tropical Medicine, University of Munich, Munich, Germany.,German Centre for Infection Research, Munich, Germany
| |
Collapse
|
25
|
Crux NB, Elahi S. Human Leukocyte Antigen (HLA) and Immune Regulation: How Do Classical and Non-Classical HLA Alleles Modulate Immune Response to Human Immunodeficiency Virus and Hepatitis C Virus Infections? Front Immunol 2017; 8:832. [PMID: 28769934 PMCID: PMC5513977 DOI: 10.3389/fimmu.2017.00832] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/30/2017] [Indexed: 12/13/2022] Open
Abstract
The genetic factors associated with susceptibility or resistance to viral infections are likely to involve a sophisticated array of immune response. These genetic elements may modulate other biological factors that account for significant influence on the gene expression and/or protein function in the host. Among them, the role of the major histocompatibility complex in viral pathogenesis in particular human immunodeficiency virus (HIV) and hepatitis C virus (HCV), is very well documented. We, recently, added a novel insight into the field by identifying the molecular mechanism associated with the protective role of human leukocyte antigen (HLA)-B27/B57 CD8+ T cells in the context of HIV-1 infection and why these alleles act as a double-edged sword protecting against viral infections but predisposing the host to autoimmune diseases. The focus of this review will be reexamining the role of classical and non-classical HLA alleles, including class Ia (HLA-A, -B, -C), class Ib (HLA-E, -F, -G, -H), and class II (HLA-DR, -DQ, -DM, and -DP) in immune regulation and viral pathogenesis (e.g., HIV and HCV). To our knowledge, this is the very first review of its kind to comprehensively analyze the role of these molecules in immune regulation associated with chronic viral infections.
Collapse
Affiliation(s)
- Nicole B. Crux
- Faculty of Medicine and Dentistry, Department of Dentistry, University of Alberta, Edmonton, AB, Canada
- Faculty of Medicine and Dentistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- Faculty of Medicine and Dentistry, Department of Dentistry, University of Alberta, Edmonton, AB, Canada
- Faculty of Medicine and Dentistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
26
|
Wei HH, Tsai LP, Wu PS. Adolescent onset of vertically transmitted untreated AIDS: A report of one case. Tzu Chi Med J 2016; 28:173-175. [PMID: 28757752 PMCID: PMC5442908 DOI: 10.1016/j.tcmj.2015.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 07/28/2015] [Accepted: 12/03/2015] [Indexed: 11/03/2022] Open
Abstract
A 12-year-old adolescent girl with intractable pneumonia and desaturation was sent to our hospital. An immunocompromised state was highly suspected because of an oral thrush persisting for a year and pneumonia of unusual severity. Laboratory tests confirmed she had human immunodeficiency virus (HIV) infection and full-blown AIDS. She lived with her adopted parents and reported no history of sexual abuse, drug abuse, or blood transfusion. We contacted the Center of Disease Control and discovered that her mother had HIV and had passed away a few years ago, thus confirming that she was a case of vertically transmitted HIV patient who had only developed AIDS recently. Even though her mother had HIV, our public health department failed to follow her as a potential HIV victim, probably because routine HIV examinations for pregnant women only started in 2005, 4 years after she was born.
Collapse
Affiliation(s)
- Hsi-Hsien Wei
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Li-Ping Tsai
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Ping-Sheng Wu
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| |
Collapse
|
27
|
Tsai MH, Muenchhoff M, Adland E, Carlqvist A, Roider J, Cole DK, Sewell AK, Carlson J, Ndung'u T, Goulder PJR. Paediatric non-progression following grandmother-to-child HIV transmission. Retrovirology 2016; 13:65. [PMID: 27608713 PMCID: PMC5016918 DOI: 10.1186/s12977-016-0300-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/23/2016] [Indexed: 11/15/2022] Open
Abstract
Background In contrast to adult HIV infection, where slow disease progression is strongly linked to immune control of HIV mediated by protective HLA class I molecules such as HLA-B*81:01, the mechanisms by which a minority of HIV-infected children maintain normal-for-age CD4 counts and remain clinically healthy appear to be HLA class I-independent and are largely unknown. To better understand these mechanisms, we here studied a HIV-infected South African female, who remained a non-progressor throughout childhood.
Results
Phylogenetic analysis of viral sequences in the HIV-infected family members, together with the history of grand-maternal breast-feeding, indicated that, unusually, the non-progressor child had been infected via grandmother-to-child transmission. Although HLA-B*81:01 was expressed by both grandmother and grand-daughter, autologous virus in each subject encoded an escape mutation L188F within the immunodominant HLA-B*81:01-restricted Gag-specific epitope TL9 (TPQDLNTML, Gag 180–188). Since the transmitted virus can influence paediatric and adult HIV disease progression, we investigated the impact of the L188F mutant on replicative capacity. When this variant was introduced into three distinct HIV clones in vitro, viral replicative capacity was abrogated altogether. However, a virus constructed using the gag sequence of the non-progressor child replicated as efficiently as wildtype virus. Conclusion These findings suggest alternative sequences of events: the transmission of the uncompensated low fitness L188F to both children, potentially contributing to slow progression in both, consistent with previous studies indicating that disease progression in children can be influenced by the replicative capacity of the transmitted virus; or the transmission of fully compensated virus, and slow progression here principally the result of HLA-independent host-specific factors, yet to be defined.
Electronic supplementary material The online version of this article (doi:10.1186/s12977-016-0300-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M-H Tsai
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford, OX1 3SY, UK
| | - M Muenchhoff
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford, OX1 3SY, UK
| | - E Adland
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford, OX1 3SY, UK
| | - A Carlqvist
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford, OX1 3SY, UK
| | - J Roider
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford, OX1 3SY, UK
| | - D K Cole
- Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - A K Sewell
- Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - J Carlson
- Microsoft Research, eScience Group, Los Angeles, CA, USA
| | - T Ndung'u
- HIV Pathogenesis Program, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.,Max Planck Institute for Infection Biology, Berlin, Germany
| | - P J R Goulder
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, South Parks Road, Oxford, OX1 3SY, UK. .,HIV Pathogenesis Program, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
28
|
Kebede Y, Fonjungo PN, Tibesso G, Shrivastava R, Nkengasong JN, Kenyon T, Kebede A, Gadde R, Ayana G. Improved Specimen-Referral System and Increased Access to Quality Laboratory Services in Ethiopia: The Role of the Public-Private Partnership. J Infect Dis 2016; 213 Suppl 2:S59-64. [PMID: 27025700 DOI: 10.1093/infdis/jiv576] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Nonstandardized specimen-transport logistics, lack of laboratory personnel to transport specimens, lack of standard specimen containers, and long turnaround time (TAT) hindered access to quality laboratory services. The objective of the Becton, Dickinson, and Company (BD)-US President's Emergency Plan for AIDS Relief (PEPFAR) Public-Private Partnership (PPP) was to support country-specific programs to develop integrated laboratory systems, services, and quality improvement strategies, with an emphasis on strengthening the specimen-referral system (SRS). METHODS In 2007, through the Centers for Disease Control and Prevention (CDC), the Ethiopian Public Health Institute (EPHI) joined with the BD-PEPFAR PPP to strengthen laboratory systems. A joint planning and assessment committee identified gaps in the SRS for prioritization and intervention and piloted the system in Addis Ababa and Amhara Region. RESULTS The PPP established standardized, streamlined specimen logistics, using the Ethiopian Postal Service Enterprise to support a laboratory network in which 554 facilities referred specimens to 160 laboratories. The PPP supported procuring 400 standard specimen containers and the training of 586 laboratory personnel and 81 postal workers. The average TAT was reduced from 7 days (range, 2-14 days) to 2 days (range, 1-3 days) in Addis Ababa and from 10 days (range, 6-21 days) to 5 days (range, 2-6 days) in Amhara Region. CONCLUSIONS This study highlights the feasibility and untapped potential of PPPs to strengthen laboratory systems. This planned and structured approach to improving specimen referral enhanced access to quality laboratory services.
Collapse
Affiliation(s)
- Yenew Kebede
- Centers for Disease Control and Prevention (CDC)
| | - Peter N Fonjungo
- Centers for Disease Control and Prevention (CDC) CDC, Atlanta, Georgia
| | - Gudeta Tibesso
- International Center for AIDS Care and Treatment Programs, Columbia University
| | | | | | - Thomas Kenyon
- Centers for Disease Control and Prevention (CDC) CDC, Atlanta, Georgia
| | - Amha Kebede
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Renuka Gadde
- Becton, Dickinson, and Company, Franklin Lakes, New Jersey
| | - Gonfa Ayana
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| |
Collapse
|
29
|
Nguefack F, Dongmo R, Touffic Othman CL, Tatah S, Njiki Kinkela MN, Koki Ndombo PO. Obstetrical, maternal characteristics and outcome of HIV-infected rapid progressor infants at Yaounde: a retrospective study. Transl Pediatr 2016; 5:46-54. [PMID: 27186521 PMCID: PMC4855195 DOI: 10.21037/tp.2016.04.04] [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] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Rapid progressors are exposed to HIV infection at an early stage of life, and the prognosis is poor without treatment. Reducing the proportion of infants who are rapid progressors, require strengthening strategies to achieve the highest level of performance for the PMTCT program. METHODS This was a retrospective study carried out on HIV infected infants aged less than 12 months, clinically classified stage 4 (WHO) or having CD4 count <25%. We described maternal and obstetrical characteristics of HIV-infected rapid progressors using univariate and bivariate analysis. Patients' survival was monitored from the inclusion time to the end of the study. We then estimated their probability of survival with or without anti-retroviral (ARV) treatment from birth using the Kaplan-Meier method. RESULTS The characteristics of the mothers of the 150 rapid progressors infants we included were: low level of education (OR=3.87; P=0.016), CD4 count less than 200/mm(3) (OR=43.3; P=0.000), absence of ARV prophylaxis (OR=6.02; P=0.043), or treatment with HAART (OR=5.74; P=0.000) during pregnancy. In the children, the most important findings were lack of co-trimoxazole prophylaxis (OR=11.61; P=0.000) and antiretroviral prophylaxis (OR=2.70; P=0.0344). The survival rate was 84.3% in infants who were receiving HAART as opposed to 43.3% in those who were not (P<0.05). CONCLUSIONS HIV infected women who are eligible should start antiretroviral treatment prior to a pregnancy, in order to improve their immunological status. This measure associated to cotrimoxazole prophylaxis and ART could improve their survival.
Collapse
Affiliation(s)
- Félicitée Nguefack
- 1 Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, P.O. Box 1364, Cameroon ; 2 Gynaeco-Obstetric and Paediatric Hospital, P.o. Box 4362, Yaoundé, Cameroon ; 3 District Hospital Efoulan PO. Box 1113-Yaoundé, Cameroon ; 4 Mother and Child Centre of the Chantal Biya Foundation, P.O. Box 1444, Yaoundé, Cameroon
| | - Roger Dongmo
- 1 Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, P.O. Box 1364, Cameroon ; 2 Gynaeco-Obstetric and Paediatric Hospital, P.o. Box 4362, Yaoundé, Cameroon ; 3 District Hospital Efoulan PO. Box 1113-Yaoundé, Cameroon ; 4 Mother and Child Centre of the Chantal Biya Foundation, P.O. Box 1444, Yaoundé, Cameroon
| | - Carole Leïla Touffic Othman
- 1 Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, P.O. Box 1364, Cameroon ; 2 Gynaeco-Obstetric and Paediatric Hospital, P.o. Box 4362, Yaoundé, Cameroon ; 3 District Hospital Efoulan PO. Box 1113-Yaoundé, Cameroon ; 4 Mother and Child Centre of the Chantal Biya Foundation, P.O. Box 1444, Yaoundé, Cameroon
| | - Sandra Tatah
- 1 Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, P.O. Box 1364, Cameroon ; 2 Gynaeco-Obstetric and Paediatric Hospital, P.o. Box 4362, Yaoundé, Cameroon ; 3 District Hospital Efoulan PO. Box 1113-Yaoundé, Cameroon ; 4 Mother and Child Centre of the Chantal Biya Foundation, P.O. Box 1444, Yaoundé, Cameroon
| | - Mina Ntoto Njiki Kinkela
- 1 Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, P.O. Box 1364, Cameroon ; 2 Gynaeco-Obstetric and Paediatric Hospital, P.o. Box 4362, Yaoundé, Cameroon ; 3 District Hospital Efoulan PO. Box 1113-Yaoundé, Cameroon ; 4 Mother and Child Centre of the Chantal Biya Foundation, P.O. Box 1444, Yaoundé, Cameroon
| | - Paul Olivier Koki Ndombo
- 1 Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, P.O. Box 1364, Cameroon ; 2 Gynaeco-Obstetric and Paediatric Hospital, P.o. Box 4362, Yaoundé, Cameroon ; 3 District Hospital Efoulan PO. Box 1113-Yaoundé, Cameroon ; 4 Mother and Child Centre of the Chantal Biya Foundation, P.O. Box 1444, Yaoundé, Cameroon
| |
Collapse
|
30
|
Abstract
Recent anecdotal reports of HIV-infected children who received early antiretroviral therapy (ART) and showed sustained control of viral replication even after ART discontinuation have raised the question of whether there is greater intrinsic potential for HIV remission, or even eradication ('cure'), in paediatric infection than in adult infection. This Review describes the influence of early initiation of ART, of immune ontogeny and of maternal factors on the potential for HIV cure in children and discusses the unique immunotherapeutic opportunities and obstacles that paediatric infection may present.
Collapse
Affiliation(s)
- Philip J Goulder
- Department of Paediatrics, University of Oxford, Oxford OX1 3SY, UK
| | - Sharon R Lewin
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne 3004, Australia
| | - Ellen M Leitman
- Department of Paediatrics, University of Oxford, Oxford OX1 3SY, UK
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW The paediatric HIV epidemic is changing. Over the past decade, new infections have substantially reduced, whereas access to antiretroviral therapy (ART) has increased. Overall this success means that numbers of children living with HIV are climbing. In addition, the problems observed in adult infection resulting from chronic inflammation triggered by persistent immune activation even following ART mediated suppression of viral replication are magnified in children infected from birth. RECENT FINDINGS Features of immune ontogeny favour low immune activation in early life, whereas specific aspects of paediatric HIV infection tend to increase it. A subset of ART-naïve nonprogressing children exists in whom normal CD4 cell counts are maintained in the setting of persistent high viremia and yet in the context of low immune activation. This sooty mangabey-like phenotype contrasts with nonprogressing adult infection which is characterized by the expression of protective HLA class I molecules and low viral load. The particular factors contributing to raised or lowered immune activation in paediatric infection, which ultimately influence disease outcome, are discussed. SUMMARY Novel strategies to circumvent the unwanted long-term consequences of HIV infection may be possible in children in whom natural immune ontogeny in early life militates against immune activation. Defining the mechanisms underlying low immune activation in natural HIV infection would have applications beyond paediatric HIV.
Collapse
Affiliation(s)
- Julia M Roider
- aDepartment of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, UK bHIV Pathogenesis Programme, The Doris Duke Medical Research Institute cKwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | | |
Collapse
|
32
|
Danaviah S, de Oliveira T, Gordon M, Govender S, Chelule P, Pillay S, Naicker T, Cassol S, Ndung'u T. Analysis of Dominant HIV Quasispecies Suggests Independent Viral Evolution Within Spinal Granulomas Coinfected with Mycobacterium tuberculosis and HIV-1 Subtype C. AIDS Res Hum Retroviruses 2016; 32:262-70. [PMID: 26564424 DOI: 10.1089/aid.2015.0189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Extrapulmonary tuberculosis (TB) is a significant public health challenge in South Africa and worldwide, largely fuelled by the HIV epidemic. In spinal TB, Mycobacteria infect the spinal column without dissemination to the spinal cord. The immune microenvironment, target cell characteristics, and other evolutionary forces within granulomas during HIV/TB coinfection are poorly characterized. We investigated whether spinal TB granulomas represent a sequestered anatomical site where independent HIV evolution occurs, and assessed the role of macrophages as a target cell for both HIV and mycobacteria. RNA was extracted from plasma and granulomatous tissue from six antiretroviral-naive HIV-1/spinal TB-coinfected patients, RT-PCR amplified, and the C2-V5 env segment was cloned and sequenced. Analysis of genetic diversity, phylogeny and coalescence patterns was performed on clonal sequences. To investigate their role in HIV sequestration, macrophages and the HIV-1 p24 protein were immune localized and ultrastructural features were studied. Intercompartment diversity measurements and phylogenetic reconstruction revealed anatomically distinct monophyletic HIV-1 clusters in four of six patients. Genotypic CCR5-tropic variants were predominant (98.9%) with conservation of putative N-linked glycosylation sites in both compartments. CD68(+) reactivity was associated with higher tissue viral load (r = 1.0; p < 0.01) but not greater intrapatient diversity (r = 0.60; p > 0.05). Ultrastructural imaging revealed the presence of bacterial and virus-like particles within membrane-bound intracellular compartments of macrophages. Spinal tuberculosis granulomas may form anatomically discreet sites of divergent viral evolution. Macrophages in these granulomas harbored both pathogens, suggesting that they may facilitate the process of viral sequestration within this compartment.
Collapse
Affiliation(s)
- Sivapragashini Danaviah
- Africa Centre for Health and Population Studies, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Tulio de Oliveira
- Africa Centre for Health and Population Studies, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Michelle Gordon
- Department of Virology, Nelson R. Mandela School of Medicine, UKZN, Durban, South Africa
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), UKZN, Durban, South Africa
| | - Shunmugam Govender
- Department of Orthopedics, Nelson R. Mandela School of Medicine, UKZN, Durban, South Africa
| | - Paul Chelule
- School of Public Health, Sefako Makgatho Health Sciences University, Medunsa, South Africa
| | - Sureshnee Pillay
- Africa Centre for Health and Population Studies, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal (UKZN), Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Nelson R. Mandela School of Medicine, UKZN, Durban, South Africa
| | - Sharon Cassol
- MRC Inflammation and Immunity Unit, Department of Immunology, University of Pretoria, Pretoria, South Africa
| | - Thumbi Ndung'u
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), UKZN, Durban, South Africa
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, UKZN, Durban, South Africa
- Max Planck Institute for Infection Biology, Chariteplatz, Berlin, Germany
- The Ragon Institute of MGH, MIT and Harvard University, Cambridge, Massachusetts
| |
Collapse
|
33
|
Kløverpris HN, Leslie A, Goulder P. Role of HLA Adaptation in HIV Evolution. Front Immunol 2016; 6:665. [PMID: 26834742 PMCID: PMC4716577 DOI: 10.3389/fimmu.2015.00665] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/27/2015] [Indexed: 01/22/2023] Open
Abstract
Killing of HIV-infected cells by CD8+ T-cells imposes strong selection pressure on the virus toward escape. The HLA class I molecules that are successful in mediating some degree of control over the virus are those that tend to present epitopes in conserved regions of the proteome, such as in p24 Gag, in which escape also comes at a significant cost to viral replicative capacity (VRC). In some instances, compensatory mutations can fully correct for the fitness cost of such an escape variant; in others, correction is only partial. The consequences of these events within the HIV-infected host, and at the population level following transmission of escape variants, are discussed. The accumulation of escape mutants in populations over the course of the epidemic already shows instances of protective HLA molecules losing their impact, and in certain cases, a modest decline in HIV virulence in association with population-level increase in mutants that reduce VRC.
Collapse
Affiliation(s)
- Henrik N Kløverpris
- KwaZulu-Natal Research Institute for Tuberculosis and HIV, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Alasdair Leslie
- KwaZulu-Natal Research Institute for Tuberculosis and HIV, Nelson R Mandela School of Medicine, University of KwaZulu-Natal , Durban , South Africa
| | - Philip Goulder
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa; Department of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Mori M, Adland E, Paioni P, Swordy A, Mori L, Laker L, Muenchhoff M, Matthews PC, Tudor-Williams G, Lavandier N, van Zyl A, Hurst J, Walker BD, Ndung’u T, Prendergast A, Goulder P, Jooste P. Sex Differences in Antiretroviral Therapy Initiation in Pediatric HIV Infection. PLoS One 2015; 10:e0131591. [PMID: 26151555 PMCID: PMC4494714 DOI: 10.1371/journal.pone.0131591] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 06/02/2015] [Indexed: 01/17/2023] Open
Abstract
The incidence and severity of infections in childhood is typically greater in males. The basis for these observed sex differences is not well understood, and potentially may facilitate novel approaches to reducing disease from a range of conditions. We here investigated sex differences in HIV-infected children in relation to antiretroviral therapy (ART) initiation and post-treatment outcome. In a South African cohort of 2,101 HIV-infected children, we observed that absolute CD4+ count and CD4% were significantly higher in ART-naïve female, compared to age-matched male, HIV-infected children. Absolute CD4 count and CD4% were also significantly higher in HIV-uninfected female versus male neonates. We next showed that significantly more male than female children were initiated on ART (47% female); and children not meeting criteria to start ART by >5 yrs were more frequently female (59%; p<0.001). Among ART-treated children, immune reconstitution of CD4 T-cells was more rapid and more complete in female children, even after adjustment for pre-ART absolute CD4 count or CD4% (p=0.011, p=0.030, respectively). However, while ART was initiated as a result of meeting CD4 criteria less often in females (45%), ART initiation as a result of clinical disease in children whose CD4 counts were above treatment thresholds occurred more often in females (57%, p<0.001). The main sex difference in morbidity observed in children initiating ART above CD4 thresholds, above that of TB disease, was as a result of wasting and stunting observed in females with above-threshold CD4 counts (p=0.002). These findings suggest the possibility that optimal treatment of HIV-infected children might incorporate differential CD4 treatment thresholds for ART initiation according to sex.
Collapse
Affiliation(s)
- Masahiko Mori
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Emily Adland
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Paolo Paioni
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Alice Swordy
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Luisa Mori
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Leana Laker
- Kimberley Hospital, Kimberley, Durban, South Africa
| | | | | | | | - Nora Lavandier
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Jacob Hurst
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, United States of America
- Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Thumbi Ndung’u
- Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), University of KwaZulu-Natal, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Andrew Prendergast
- Centre for Paediatrics, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Philip Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | | |
Collapse
|
35
|
Adland E, Paioni P, Thobakgale C, Laker L, Mori L, Muenchhoff M, Csala A, Clapson M, Flynn J, Novelli V, Hurst J, Naidoo V, Shapiro R, Huang KHG, Frater J, Prendergast A, Prado JG, Ndung’u T, Walker BD, Carrington M, Jooste P, Goulder PJR. Discordant Impact of HLA on Viral Replicative Capacity and Disease Progression in Pediatric and Adult HIV Infection. PLoS Pathog 2015; 11:e1004954. [PMID: 26076345 PMCID: PMC4468173 DOI: 10.1371/journal.ppat.1004954] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/13/2015] [Indexed: 11/18/2022] Open
Abstract
HLA class I polymorphism has a major influence on adult HIV disease progression. An important mechanism mediating this effect is the impact on viral replicative capacity (VRC) of the escape mutations selected in response to HLA-restricted CD8+ T-cell responses. Factors that contribute to slow progression in pediatric HIV infection are less well understood. We here investigate the relationship between VRC and disease progression in pediatric infection, and the effect of HLA on VRC and on disease outcome in adult and pediatric infection. Studying a South African cohort of >350 ART-naïve, HIV-infected children and their mothers, we first observed that pediatric disease progression is significantly correlated with VRC. As expected, VRCs in mother-child pairs were strongly correlated (p = 0.004). The impact of the protective HLA alleles, HLA-B*57, HLA-B*58:01 and HLA-B*81:01, resulted in significantly lower VRCs in adults (p<0.0001), but not in children. Similarly, in adults, but not in children, VRCs were significantly higher in subjects expressing the disease-susceptible alleles HLA-B*18:01/45:01/58:02 (p = 0.007). Irrespective of the subject, VRCs were strongly correlated with the number of Gag CD8+ T-cell escape mutants driven by HLA-B*57/58:01/81:01 present in each virus (p = 0.0002). In contrast to the impact of VRC common to progression in adults and children, the HLA effects on disease outcome, that are substantial in adults, are small and statistically insignificant in infected children. These data further highlight the important role that VRC plays both in adult and pediatric progression, and demonstrate that HLA-independent factors, yet to be fully defined, are predominantly responsible for pediatric non-progression.
Collapse
Affiliation(s)
- Emily Adland
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
| | - Paolo Paioni
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
| | - Christina Thobakgale
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Leana Laker
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Luisa Mori
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Maximilian Muenchhoff
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Anna Csala
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
| | - Margaret Clapson
- Department of Paediatric Infectious Diseases, Great Ormond St Hospital for Children, London, United Kingdom
| | - Jacquie Flynn
- Department of Paediatric Infectious Diseases, Great Ormond St Hospital for Children, London, United Kingdom
| | - Vas Novelli
- Department of Paediatric Infectious Diseases, Great Ormond St Hospital for Children, London, United Kingdom
| | - Jacob Hurst
- The Institute for Emerging Infections, The Oxford Martin School, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
- Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, United Kingdom
| | - Vanessa Naidoo
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Roger Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Kuan-Hsiang Gary Huang
- The Institute for Emerging Infections, The Oxford Martin School, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
| | - John Frater
- The Institute for Emerging Infections, The Oxford Martin School, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
- Oxford National Institute of Health Research, Biomedical Research Centre, Oxford, United Kingdom
| | - Andrew Prendergast
- Centre for Paediatrics, Blizard Institute, Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Julia G. Prado
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Thumbi Ndung’u
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Boston, Massachusetts, United States of America
- KwaZulu-Natal Research Institute for Tuberculosis and HIV, University of KwaZulu-Natal, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Bruce D. Walker
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Boston, Massachusetts, United States of America
| | - Mary Carrington
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Boston, Massachusetts, United States of America
- Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Pieter Jooste
- Paediatric Department, Kimberley Hospital, Northern Cape, South Africa
| | - Philip J. R. Goulder
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, United Kingdom
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Paediatric Infectious Diseases, Great Ormond St Hospital for Children, London, United Kingdom
| |
Collapse
|
36
|
Davies MA. Research gaps in neonatal HIV-related care. South Afr J HIV Med 2015; 16:375. [PMID: 29568592 PMCID: PMC5843028 DOI: 10.4102/sajhivmed.v16i1.375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/16/2015] [Indexed: 12/19/2022] Open
Abstract
The South African prevention of mother to child transmission programme has made excellent progress in reducing vertical HIV transmission, and paediatric antiretroviral therapy programmes have demonstrated good outcomes with increasing treatment initiation in younger children and infants. However, both in South Africa and across sub-Saharan African, lack of boosted peri-partum prophylaxis for high-risk vertical transmission, loss to follow-up, and failure to initiate HIV-infected infants on antiretroviral therapy (ART) before disease progression are key remaining gaps in neonatal HIV-related care. In this issue of the Southern African Journal of HIV Medicine, experts provide valuable recommendations for addressing these gaps. The present article highlights a number of areas where evidence is lacking to inform guidelines and programme development for optimal neonatal HIV-related care.
Collapse
Affiliation(s)
- Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, South Africa
| |
Collapse
|
37
|
Sherman GG. HIV testing during the neonatal period. South Afr J HIV Med 2015; 16:362. [PMID: 29568587 PMCID: PMC5843063 DOI: 10.4102/sajhivmed.v16i1.362] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/10/2015] [Indexed: 02/06/2023] Open
Affiliation(s)
- Gayle G Sherman
- Department of Paediatrics and Child Health, University of the Witwatersrand, South Africa.,Centre for HIV and STI, National Institute for Communicable Diseases, South Africa
| |
Collapse
|
38
|
A mathematical model evaluating the timing of early diagnostic testing in HIV-exposed infants in South Africa. J Acquir Immune Defic Syndr 2014; 67:341-8. [PMID: 25118910 DOI: 10.1097/qai.0000000000000307] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Antiretroviral therapy is often initiated too late to impact early HIV-related infant mortality. Earlier treatment requires an earlier diagnosis, and the currently recommended 6-week HIV polymerase chain reaction (PCR) test needs reconsideration. This study aims to identify (1) optimal testing intervals to maximize the number of perinatal HIV infections diagnosed and (2) programmatic issues that impact diagnosis. METHODS A mathematical model was developed to simulate antiretroviral prophylaxis uptake and health outcomes in 240,000 HIV-exposed South African infants. The model considered routine early testing with 1 PCR (at birth, 6, 10, or 14 weeks of age) and with 2 PCR tests (at birth and at 6, 10, or 14 weeks of age). RESULTS A single 6-week test would diagnose the same number of perinatal HIV infections as birth testing (P = 0.92) but fewer infections than a 10-week test (P < 0.01). Ten-week testing identifies the highest number of perinatally infected infants (P < 0.01 compared with a single test at all other ages) but does not save additional life years compared with birth testing (P = 0.27). Performing 2 PCR tests (at birth and 10 weeks) would identify the highest number of perinatal infections (P < 0.01 versus a second 6- or 14-week test). However, 25% of perinatal HIV infections would remain undiagnosed, largely because of failure to return PCR test results to caregivers. CONCLUSIONS Six weeks may no longer be the optimal age to diagnose perinatal HIV infections. Two early PCR tests (at birth and 10 weeks) would likely be the ideal diagnostic algorithm, but must be coupled with improved program coverage.
Collapse
|
39
|
Mutwa PR, Boer KR, Asiimwe-Kateera B, Tuyishimire D, Muganga N, Lange JMA, van de Wijgert J, Asiimwe A, Reiss P, Geelen SPM. Safety and effectiveness of combination antiretroviral therapy during the first year of treatment in HIV-1 infected Rwandan children: a prospective study. PLoS One 2014; 9:e111948. [PMID: 25365302 PMCID: PMC4218827 DOI: 10.1371/journal.pone.0111948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 10/08/2014] [Indexed: 11/19/2022] Open
Abstract
Background With increased availability of paediatric combination antiretroviral therapy (cART) in resource limited settings, cART outcomes and factors associated with outcomes should be assessed. Methods HIV-infected children <15 years of age, initiating cART in Kigali, Rwanda, were followed for 18 months. Prospective clinical and laboratory assessments included weight-for-age (WAZ) and height-for-age (HAZ) z-scores, complete blood cell count, liver transaminases, creatinine and lipid profiles, CD4 T-cell count/percent, and plasma HIV-1 RNA concentration. Clinical success was defined as WAZ and WAZ >−2, immunological success as CD4 cells ≥500/mm3 and ≥25% for respectively children over 5 years and under 5 years, and virological success as a plasma HIV-1 RNA concentration <40 copies/mL. Results Between March 2008 and December 2009, 123 HIV-infected children were included. The median (interquartile (IQR) age at cART initiation was 7.4 (3.2, 11.5) years; 40% were <5 years and 54% were female. Mean (95% confidence interval (95%CI)) HAZ and WAZ at baseline were −2.01 (−2.23, −1.80) and −1.73 (−1.95, −1.50) respectively and rose to −1.75 (−1.98, −1.51) and −1.17 (−1.38, −0.96) after 12 months of cART. The median (IQR) CD4 T-cell values for children <5 and ≥5 years of age were 20% (13, 28) and 337 (236, 484) cells/mm3respectively, and increased to 36% (28, 41) and 620 (375, 880) cells/mm3. After 12 months of cART, 24% of children had a detectable viral load, including 16% with virological failure (HIV-RNA>1000 c/mL). Older age at cART initiation, poor adherence, and exposure to antiretrovirals around birth were associated with virological failure. A third (33%) of children had side effects (by self-report or clinical assessment), but only 9% experienced a severe side effect requiring a cART regimen change. Conclusions cART in Rwandan HIV-infected children was successful but success might be improved further by initiating cART as early as possible, optimizing adherence and optimizing management of side effects.
Collapse
Affiliation(s)
- Philippe R. Mutwa
- Kigali University Teaching Hospital, Department of Pediatrics, Kigali, Rwanda
- Department of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| | - Kimberly R. Boer
- Department of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, Amsterdam, The Netherlands
- Biomedical Research, Epidemiology Unit, Royal Tropical Institute, Amsterdam, The Netherlands
| | - Brenda Asiimwe-Kateera
- Department of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, Amsterdam, The Netherlands
| | - Diane Tuyishimire
- Outpatients Clinic, Treatment and Research on HIV/AIDS Centre, Kigali, Rwanda
| | - Narcisse Muganga
- Kigali University Teaching Hospital, Department of Pediatrics, Kigali, Rwanda
| | - Joep M. A. Lange
- Department of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, Amsterdam, The Netherlands
| | - Janneke van de Wijgert
- Department of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, Amsterdam, The Netherlands
- Institute of Infection and Global Health, University of Liverpool, Liverpool, United of Kingdom
- Rinda Ubuzima, Kigali, Rwanda
| | | | - Peter Reiss
- Department of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, Amsterdam, The Netherlands
| | - Sibyl P. M. Geelen
- Department of Global Health and Amsterdam Institute for Global Health and Development, Academic Medical Center, Amsterdam, The Netherlands
- Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
40
|
Tobin NH, Aldrovandi GM. Are infants unique in their ability to be "functionally cured" of HIV-1? Curr HIV/AIDS Rep 2014; 11:1-10. [PMID: 24390641 DOI: 10.1007/s11904-013-0189-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The recent report of an infant that appears to have achieved a "functional cure" of HIV-1 following receipt of antiretroviral therapy (ART) within 30 hours of birth raises many questions: was the child infected? Was this result due to unique features of this particular infant's immune system, the immune system of infants or the very early initiation of effective ART? In this manuscript, we discuss the pathogenesis of HIV-1 in infants, highlighting the unique features of infant immune development and how these may inform efforts to cure HIV infection. We will also compare the path to infant "cure" to cures in adults.
Collapse
Affiliation(s)
- Nicole H Tobin
- Department of Pediatrics, Children's Hospital Los Angeles, 4650 Sunset Blvd., MS#51, Los Angeles, CA, 90027, USA
| | | |
Collapse
|
41
|
Muenchhoff M, Prendergast AJ, Goulder PJR. Immunity to HIV in Early Life. Front Immunol 2014; 5:391. [PMID: 25161656 PMCID: PMC4130105 DOI: 10.3389/fimmu.2014.00391] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/30/2014] [Indexed: 01/14/2023] Open
Abstract
The developing immune system is adapted to the exposure to a plethora of pathogenic and non-pathogenic antigens encountered in utero and after birth, requiring a fine balance between protective immunity and immune tolerance. In early stages of life, this tolerogenic state of the innate and adaptive immune system and the lack of immunological memory render the host more susceptible to infectious pathogens like HIV. HIV pathogenesis is different in children, compared to adults, with more rapid disease progression and a substantial lack of control of viremia compared to adults. Plasma viral load remains high during infancy and only declines gradually over several years in line with immune maturation, even in rare cases where children maintain normal CD4 T-lymphocyte counts for several years without antiretroviral therapy (ART). These pediatric slow progressors also typically show low levels of immune activation despite persistently high viremia, resembling the phenotype of natural hosts of SIV infection. The lack of immunological memory places the fetus and the newborn at higher risk of infections; however, it may also provide an opportunity for unique interventions. Frequencies of central memory CD4+ T-lymphocytes, one of the main cellular reservoirs of HIV, are very low in the newborn child, so immediate ART could prevent the establishment of persistent viral reservoirs and result in "functional cure." However, as recently demonstrated in the case report of the "Mississippi child" who experienced viral rebound after more than 2 years off ART, additional immunomodulatory strategies might be required for sustained viral suppression after ART cessation. In this review, we discuss the interactions between HIV and the developing immune system in children and the potential implications for therapeutic and prophylactic interventions.
Collapse
Affiliation(s)
- Maximilian Muenchhoff
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research , Oxford , UK
| | - Andrew J Prendergast
- Centre for Paediatrics, Blizard Institute, Queen Mary University of London , London , UK ; Zvitambo Institute for Maternal and Child Health Research , Harare , Zimbabwe
| | - Philip Jeremy Renshaw Goulder
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research , Oxford , UK ; HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal , Durban , South Africa
| |
Collapse
|
42
|
If you text them, they will come: using the HIV infant tracking system to improve early infant diagnosis quality and retention in Kenya. AIDS 2014; 28 Suppl 3:S313-21. [PMID: 24991904 DOI: 10.1097/qad.0000000000000332] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The objective of this study is to evaluate the impact of the HIV Infant Tracking System (HITSystem) for quality improvement of early infant diagnosis (EID) of HIV services. DESIGN AND SETTING This observational pilot study compared 12 months of historical preintervention EID outcomes at one urban and one peri-urban government hospital in Kenya to 12 months of intervention data to assess retention and time throughout the EID cascade of care. PARTICIPANTS Mother-infant pairs enrolled in EID at participating hospitals before (n = 320) and during (n = 523) the HITSystem pilot were eligible to participate. INTERVENTION The HITSystem utilizes Internet-based coordination of the multistep PCR cycle, automated alerts to trigger prompt action from providers and laboratory technicians, and text messaging to notify mothers when results are ready or additional action is needed. MAIN OUTCOME MEASURES The main outcome measures were retention throughout EID services, meeting time-sensitive targets and improving results turn-around time, and increasing early antiretroviral therapy (ART) initiation among HIV-infected infants. RESULTS The HITSystem was associated with an increase in the proportion of HIV-exposed infants retained in EID care at 9 months postnatal (45.1-93.0% urban; 43.2-94.1% peri-urban), a decrease in turn-around times between sample collection, PCR results and notification of mothers in both settings, and a significant increase in the proportion of HIV-infected infants started on antiretroviral therapy at each hospital(14 vs. 100% urban; 64 vs. 100% peri-urban). CONCLUSION The HITSystem maximizes the use of easily accessible technology to improve the quality and efficiency of EID services in resource-limited settings.
Collapse
|
43
|
Promising antibody testing strategies for early infant HIV infection diagnosis in China. PLoS One 2014; 9:e99935. [PMID: 24971594 PMCID: PMC4074049 DOI: 10.1371/journal.pone.0099935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 05/20/2014] [Indexed: 01/12/2023] Open
Abstract
Background In China, 1.1% of people living with HIV were transmitted vertically, causing a heavy burden on families and society. Early infant diagnosis (EID) is critical for improving neonatal survival. The purpose of this study is to suggest improvement in antibody testing strategies with dried blood spots (DBSs) for EID in China through analysis of anti-HIV seroreversion of infants. Methods A total of 280 infants born to HIV infected mothers in four diverse provinces of China where multiple subtypes coexist were enrolled. The status of the infants' infection was determined by HIV antibody enzyme immunoassay and Western blot analysis at ≥18 months of age or by convincing clinical and epidemiologic data for deceased infants. A total of 1028 DBSs were collected during follow-up, which were tested to obtain anti-HIV signal to cut-off ratio (S/CO) data. Results For uninfected infants, anti-HIV S/CO decreased with age. Seropositivity percentage declined most rapidly at 6 months to 9 months of age and 98.7% children seroreverted by 12 months of age. For most infected infants, minimum S/CO values were obtained at ≤6 months of age. Antibody negative predictive value was 100% at ≥6 months of age. An S/CO increase ≥1.86 after three months follow-up can determine HIV infection. S/CO threshold of 3.17 can differentiate infected from uninfected infants for exposed kids at 9 months or older with sensitivity as 100% and specificity ≥94.2%. Significance Suggestions obtained through studying seroreversion data of Chinese HIV-exposed infants help improve antibody strategies for HIV EID in China. The infection can be determined as early as 3 months of age and excluded as early as 6 months of age.
Collapse
|
44
|
Virologic response in children treated with abacavir-compared with stavudine-based antiretroviral treatment: a South African multi-cohort analysis. Pediatr Infect Dis J 2014; 33:617-22. [PMID: 24378944 PMCID: PMC4024348 DOI: 10.1097/inf.0000000000000222] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Initiation criteria and pediatric antiretroviral treatment regimens have changed over the past few years in South Africa. We reported worse early virological outcomes associated with the use of abacavir (ABC)-based regimens at 1 large site: here, we expand this analysis to multiple sites in the IeDEA-Southern Africa collaboration. METHODS Data for 9543 antiretroviral treatment-naïve children <16 years at treatment initiation started on either stavudine/lamivudine (d4T/3TC) or ABC/3TC with efavirenz (EFV) or ritonavir-boosted lopinavir (LPV/r) treated at 6 clinics in Johannesburg and Cape Town, South Africa, were analyzed with χ tests and logistic regression to evaluate viral suppression at 6 and 12 months. RESULTS Prevalence of viral suppression at 6 months in 2174 children started on a d4T-based LPV/r regimen was greater (70%) than among 438 children started on an ABC-based LPV/r regimen (54%, P < 0.0001). Among 3189 children started on a d4T-based EFV regimen, a higher proportion (86%) achieved suppression at 6 months compared with 391 children started on ABC-containing EFV regimens (78%, P < 0.0001). Relative benefit of d4T versus ABC on 6-month suppression remained in multivariate analysis after adjustment for pretreatment characteristics, cohort and year of program [LPV/r: odds ratio = 0.57 (confidence interval: 0.46-0.72); EFV: odds ratio = 0.46 (confidence interval: 0.32-0.65)]. CONCLUSIONS This expanded analysis is consistent with our previous report of worse virological outcomes after ABC was introduced as part of first-line antiretroviral treatment in South Africa. Whether due to the drug itself or coincident with other changes over time, continued monitoring and analyses must clarify causes and prevent suboptimal long-term outcomes.
Collapse
|
45
|
Penazzato M, Prendergast AJ, Muhe LM, Tindyebwa D, Abrams E, Cochrane HIV/AIDS Group. Optimisation of antiretroviral therapy in HIV-infected children under 3 years of age. Cochrane Database Syst Rev 2014; 2014:CD004772. [PMID: 24852077 PMCID: PMC11022182 DOI: 10.1002/14651858.cd004772.pub4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND In the absence of antiretroviral therapy (ART), over 50% of HIV-infected infants progress to AIDS and death by 2 years of age. However, there are challenges to initiation of ART in early life, including the possibility of drug resistance in the context of prevention of mother-to-child transmission (PMTCT) programs, a paucity of drug choices , uncertain dosing for some medications and long-term toxicities. Key management decisions include when to start ART, what regimen to start, and whether and when to substitute drugs or interrupt therapy. This review, an update of a previous review, aims to summarize the currently available evidence on this topic and inform the ART management in HIV-infected children less than 3 years of age. OBJECTIVES To evaluate 1) when to start ART in young children (less than 3 years); 2) what ART to start with, comparing first-line non-nucleoside reverse transcriptase inhibitor (NNRTI) and protease inhibitor (PI)-based regimens; and 3) whether alternative strategies should be used to optimize antiretroviral treatment in this population: induction (initiation with 4 drugs rather than 3 drugs) followed by maintenance ART, interruption of ART and substitution of PI with NNRTI drugs once virological suppression is achieved on a PI-based regimen. SEARCH METHODS Search methodsWe searched for published studies in the Cochrane HIV/AIDS Review Group Trials Register, The Cochrane Library, Pubmed, EMBASE and CENTRAL. We screened abstracts from relevant conference proceedings and searched for unpublished and ongoing trials in clinical trial registries (ClinicalTrials.gov and the WHO International Clinical Trials Registry Platform). SELECTION CRITERIA We identified RCTs that recruited perinatally HIV-infected children under 3 years of age without restriction of setting. We rejected trials that did not include children less than 3 years of age, did not provide stratified outcomes for those less than 3 years or did not evaluate either timing of ART initiation, choice of drug regimen or treatment switch/interruption strategy. DATA COLLECTION AND ANALYSIS Two reviewers independently applied study selection criteria, assessed study quality and extracted data. Effects were assessed using the hazard ratio (HR) for time-to-event outcomes, relative risk for dichotomous outcomes and weighted mean difference for continuous outcomes. MAIN RESULTS A search of the databases identified a total of 735 unique, previously unreviewed studies, of which 731 were excluded to leave 4 new studies to incorporate into the review. Four additional studies were identified in conference proceedings, for a total of 8 studies addressing when to start treatment (n=2), what to start (n=3), whether to substitute lopinavir/ritonavir (LPV/r) with nevirapine (NVP) (n=1), whether to use an induction-maintenance ART strategy (n=1) and whether to interrupt treatment (n=1).Treatment initiation in asymptomatic infants with good immunological status was associated with a 75% reduction (HR=0.25; 95%CI 0.12-0.51; p=0.0002) in mortality or disease progression in the one trial with sufficient power to address this question. In a smaller pilot trial, median CD4 cell count was not significantly different between early and deferred treatment groups 12 months after ART.Regardless of previous exposure to nevirapine for PMTCT, the hazard for treatment failure at 24 weeks was 1.79 (95%CI 1.33, 2.41) times higher in children starting ART with a NVP-based regimen compared to those starting with a LPV/r-based regimen (p=0.0001) with no clear difference in the effect observed for children younger or older than 1 year. The hazard for virological failure at 24 weeks was overall 1.84 (95%CI 1.29, 2.63) times higher for children starting ART with a NVP-based regimen compared to those starting with a LPV/r-based regimen (p=0.0008) with a larger difference in time to virological failure (or death) between the NVP and LPV/r-based regimens when ART was initiated in the first year of life.Infants starting a LPV/r regimen and achieving sustained virological suppression who then substituted LPV/r with NVP after median 9 months on LPV/r were less likely to develop virological failure (defined as at least one VL greater than 50 copies/mL) compared with infants who started and stayed on LPV/r (HR=0.62, 95%CI 0.41, 0.92, p=0.02). However the hazard for confirmed failure at a higher viral load (>1000 copies/mL) was greater among children who switched to NVP compared to those who remained on LPV/r (HR=10.19, 95% CI 2.36, 43.94, p=0.002).Children undergoing an induction-maintenance ART approach with a 4-drug NNRTI-based regimen for 36 weeks, followed by 3-drug ART, had significantly greater CD4 rise than children receiving a standard 3-drug NNRTI-based ART at 36 weeks (mean difference 1.70 [95%CI 0.61, 2.79] p=0.002) and significantly better viral load response at 24 weeks (OR 1.99 [95%CI 1.09, 3.62] p=0.02). However, the immunological and virological benefits were short-term.The one trial of treatment interruption that compared children initiating continuous ART from infancy with children interrupting ART was terminated early because the duration of treatment interruption was less than 3 months in most infants. Children interrupting treatment had similar growth and occurrence of serious adverse events as those in the continuous arm. AUTHORS' CONCLUSIONS ART initiation in asymptomatic children under 1 year of age reduces morbidity and mortality, but it remains unclear whether there are clinical benefits to starting ART in asymptomatic children diagnosed with HIV infection between 1-3 years.The available evidence shows that a LPV/r-based first-line regimen is more efficacious than a NVP-based regimen, regardless of PMTCT exposure status. New formulations of LPV/r are urgently required to enable new WHO recommendations to be implemented. An alternative approach to long-term LPV/r is substituting LPV/r with NVP once virological suppression is achieved. This strategy looked promising in the one trial undertaken, but may be difficult to implement in the absence of routine viral load testing.A 4-drug induction-maintenance approach showed short-term virological and immunological benefits during the induction phase but, in the absence of sustained benefits, is not recommended as a routine treatment strategy. Treatment interruption following early ART initiation in infancy was challenging for children who were severely immunocompromised in the context of poor clinical immunological condition at ART initiation due to the short duration of interruption, and is therefore not practical in ART treatment programmes where close monitoring is not feasible.
Collapse
Affiliation(s)
| | | | - Lulu M Muhe
- World Health Organization20 Avenue AppiaGenevaSwitzerland1211
| | - Denis Tindyebwa
- African Network for the Care of Children Affected by HIV/AIDS (ANECCA)KampalaUganda
| | - Elaine Abrams
- Columbia University. Mailman School of Public HealthEpidemiology and Pediatrics722 W 168th StNew YorkNew YorkUSA10032
| | | |
Collapse
|
46
|
Callens SFJ, McKellar MS, Colebunders R. HIV care and treatment for children in resource-limited settings. Expert Rev Anti Infect Ther 2014; 6:181-90. [DOI: 10.1586/14787210.6.2.181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
47
|
Validation and calibration of a computer simulation model of pediatric HIV infection. PLoS One 2013; 8:e83389. [PMID: 24349503 PMCID: PMC3862684 DOI: 10.1371/journal.pone.0083389] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 11/04/2013] [Indexed: 11/30/2022] Open
Abstract
Background Computer simulation models can project long-term patient outcomes and inform health policy. We internally validated and then calibrated a model of HIV disease in children before initiation of antiretroviral therapy to provide a framework against which to compare the impact of pediatric HIV treatment strategies. Methods We developed a patient-level (Monte Carlo) model of HIV progression among untreated children <5 years of age, using the Cost-Effectiveness of Preventing AIDS Complications model framework: the CEPAC-Pediatric model. We populated the model with data on opportunistic infection and mortality risks from the International Epidemiologic Database to Evaluate AIDS (IeDEA), with mean CD4% at birth (42%) and mean CD4% decline (1.4%/month) from the Women and Infants’ Transmission Study (WITS). We internally validated the model by varying WITS-derived CD4% data, comparing the corresponding model-generated survival curves to empirical survival curves from IeDEA, and identifying best-fitting parameter sets as those with a root-mean square error (RMSE) <0.01. We then calibrated the model to other African settings by systematically varying immunologic and HIV mortality-related input parameters. Model-generated survival curves for children aged 0-60 months were compared, again using RMSE, to UNAIDS data from >1,300 untreated, HIV-infected African children. Results In internal validation analyses, model-generated survival curves fit IeDEA data well; modeled and observed survival at 16 months of age were 91.2% and 91.1%, respectively. RMSE varied widely with variations in CD4% parameters; the best fitting parameter set (RMSE = 0.00423) resulted when CD4% was 45% at birth and declined by 6%/month (ages 0-3 months) and 0.3%/month (ages >3 months). In calibration analyses, increases in IeDEA-derived mortality risks were necessary to fit UNAIDS survival data. Conclusions The CEPAC-Pediatric model performed well in internal validation analyses. Increases in modeled mortality risks required to match UNAIDS data highlight the importance of pre-enrollment mortality in many pediatric cohort studies.
Collapse
|
48
|
Fonjungo PN, Girma M, Melaku Z, Mekonen T, Tanuri A, Hailegiorgis B, Tegbaru B, Mengistu Y, Ashenafi A, Mamo W, Abreha T, Tibesso G, Ramos A, Ayana G, Freeman R, Nkengasong JN, Zewdu S, Kebede Y, Abebe A, Kenyon TA, Messele T. Field expansion of DNA polymerase chain reaction for early infant diagnosis of HIV-1: The Ethiopian experience. Afr J Lab Med 2013; 2:31. [PMID: 26855901 PMCID: PMC4740918 DOI: 10.4102/ajlm.v2i1.31] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Early diagnosis of infants infected with HIV (EID) and early initiation of treatment significantly reduces the rate of disease progression and mortality. One of the challenges to identification of HIV-1-infected infants is availability and/or access to quality molecular laboratory facilities which perform molecular virologic assays suitable for accurate identification of the HIV status of infants. Method We conducted a joint site assessment and designed laboratories for the expansion of DNA polymerase chain reaction (PCR) testing based on dried blood spot (DBS) for EID in six regions of Ethiopia. Training of appropriate laboratory technologists and development of required documentation including standard operating procedures (SOPs) was carried out. The impact of the expansion of EID laboratories was assessed by the number of tests performed as well as the turn-around time. Results DNA PCR for EID was introduced in 2008 in six regions. From April 2006 to April 2008, a total of 2848 infants had been tested centrally at the Ethiopian Health and Nutrition Research Institute (EHNRI) in Addis Ababa, and which was then the only laboratory with the capability to perform EID; 546 (19.2%) of the samples were positive. By November 2010, EHNRI and the six laboratories had tested an additional 16 985 HIV-exposed infants, of which 1915 (11.3%) were positive. The median turn-around time for test results was 14 days (range 14–21 days). Conclusion Expansion of HIV DNA PCR testing facilities that can provide quality and reliable results is feasible in resource-limited settings. Regular supervision and monitoring for quality assurance of these laboratories is essential to maintain accuracy of testing.
Collapse
Affiliation(s)
- Peter N Fonjungo
- Center for Disease Control and Prevention (CDC), Addis Ababa, Ethiopia
| | - Mulu Girma
- Ethiopian Health and Nutrition Research Institute (EHNRI), Addis Ababa, Ethiopia
| | | | - Teferi Mekonen
- Center for Disease Control and Prevention (CDC), Addis Ababa, Ethiopia
| | | | | | - Belete Tegbaru
- Ethiopian Health and Nutrition Research Institute (EHNRI), Addis Ababa, Ethiopia
| | - Yohannes Mengistu
- Center for Disease Control and Prevention (CDC), Addis Ababa, Ethiopia
| | | | - Wubshet Mamo
- University of Washington, ITECH Program, Addis Ababa, Ethiopia
| | | | - Gudetta Tibesso
- Ethiopian Health and Nutrition Research Institute (EHNRI), Addis Ababa, Ethiopia
| | - Artur Ramos
- Center for Global Health, Centers for Disease Control and Prevention (CDC), Atlanta, USA
| | - Gonfa Ayana
- Ethiopian Health and Nutrition Research Institute (EHNRI), Addis Ababa, Ethiopia
| | - Richard Freeman
- Clinton HIV/AIDS Access Initiative (CHAI), Addis Ababa, Ethiopia
| | - John N Nkengasong
- Center for Global Health, Centers for Disease Control and Prevention (CDC), Atlanta, USA
| | - Solomon Zewdu
- John Hopkins University, TSEHAI program, Addis Ababa, Ethiopia
| | - Yenew Kebede
- Center for Disease Control and Prevention (CDC), Addis Ababa, Ethiopia
| | - Almaz Abebe
- Ethiopian Health and Nutrition Research Institute (EHNRI), Addis Ababa, Ethiopia
| | - Thomas A Kenyon
- Center for Disease Control and Prevention (CDC), Addis Ababa, Ethiopia
| | - Tsehaynesh Messele
- Ethiopian Health and Nutrition Research Institute (EHNRI), Addis Ababa, Ethiopia
| |
Collapse
|
49
|
Prendergast AJ, Klenerman P, Goulder PJR. The impact of differential antiviral immunity in children and adults. Nat Rev Immunol 2012; 12:636-48. [PMID: 22918466 DOI: 10.1038/nri3277] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The course of immune maturation has evolved to favour survival at each stage of development in early life. Fetal and neonatal immune adaptations facilitate intrauterine survival and provide early postnatal protection against extracellular pathogens, but they leave infants susceptible to intracellular pathogens such as viruses that are acquired perinatally. This Review focuses on three such pathogens--HIV, hepatitis B virus and cytomegalovirus--and relates the differential impact of these infections in infants and adults to the antiviral immunity that is generated at different ages. A better understanding of age-specific antiviral immunity may inform the development of integrated prevention, treatment and vaccine strategies to minimize the global disease burden resulting from these infections.
Collapse
Affiliation(s)
- Andrew J Prendergast
- Centre for Paediatrics, Blizard Institute, Queen Mary University of London, Newark Street, London E1 2AT, UK
| | | | | |
Collapse
|
50
|
Madhi SA, Izu A, Violari A, Cotton MF, Panchia R, Dobbels E, Sewraj P, van Niekerk N, Jean-Philippe P, Adrian PV. Immunogenicity following the first and second doses of 7-valent pneumococcal conjugate vaccine in HIV-infected and -uninfected infants. Vaccine 2012; 31:777-83. [PMID: 23228814 DOI: 10.1016/j.vaccine.2012.11.076] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/14/2012] [Accepted: 11/26/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND The immunogenicity of pneumococcal conjugate vaccine (PCV) has not been evaluated in HIV-infected infants following the first and second PCV-doses. We studied antibody kinetics of serotypes included in 7-valent PCV in HIV-infected and HIV-uninfected infants prior to and following each of three PCV-doses. METHODS HIV-uninfected infants born to HIV-uninfected (HUU) and HIV-infected mothers (HEU); and perinatal HIV-infected children with CD(4+)<25% randomized to initiate antiretroviral treatment (ART) when clinically and/or immunologically indicated (ART-) or immediately (ART+) were enrolled. Vaccination occurred at approximately 7.4, 11.5 and 15.5 weeks of age. Serotype-specific antibody was measured by ELISA following each PCV-dose and opsonophagocytic activity (OPA) to three serotypes following the second and third doses. RESULTS Pre-vaccination, antibody geometric mean concentrations (GMCs) were higher in HUU compared to HIV-exposed groups for most serotypes. GMCs and proportion of infants with antibody ≥0.35 μg/ml were similar in HUU compared to other groups following the second PCV-dose. In all groups, GMCs were greater following the third compared to post-second dose; and a higher proportion within each group had antibody ≥0.35 μg/ml to 6B and 23F. OPA GMTs increased after the third compared to post-second dose for studied-serotypes; as did the proportion with OPA ≥8 to 23F. CONCLUSION A two-dose primary-series of PCV probably confers similar protection against invasive pneumococcal disease in HIV-infected compared to HUU children. The inferior response to serotypes 6B and 23F, and lower GMCs and OPA GMTs, following two compared to after three PCV-doses may have implications in the prevention of pneumococcal disease in high-burden countries.
Collapse
Affiliation(s)
- Shabir A Madhi
- National Institute for Communicable Diseases - Division of National Health Laboratory Service, Centre for Vaccines and Immunology, Sandringham, South Africa.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|