1
|
Hiner CR, Mueller AL, Su H, Goldstein H. Interventions during Early Infection: Opening a Window for an HIV Cure? Viruses 2024; 16:1588. [PMID: 39459922 PMCID: PMC11512236 DOI: 10.3390/v16101588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Although combination antiretroviral therapy (ART) has been a landmark achievement for the treatment of human immunodeficiency virus (HIV), an HIV cure has remained elusive. Elimination of latent HIV reservoirs that persist throughout HIV infection is the most challenging barrier to an HIV cure. The progressive HIV infection is marked by the increasing size and diversity of latent HIV reservoirs until an effective immune response is mobilized, which can control but not eliminate HIV infection. The stalemate between HIV replication and the immune response is manifested by the establishment of a viral set point. ART initiation during the early stage limits HIV reservoir development, preserves immune function, improves the quality of life, and may lead to ART-free viral remission in a few people living with HIV (PLWH). However, for the overwhelming majority of PLWH, early ART initiation alone does not cure HIV, and lifelong ART is needed to sustain viral suppression. A critical area of research is focused on determining whether HIV could be functionally cured if additional treatments are provided alongside early ART. Several HIV interventions including Block and Lock, Shock and Kill, broadly neutralizing antibody (bNAb) therapy, adoptive CD8+ T cell therapy, and gene therapy have demonstrated delayed viral rebound and/or viral remission in animal models and/or some PLWH. Whether or not their application during early infection can improve the success of HIV remission is less studied. Herein, we review the current state of clinical and investigative HIV interventions and discuss their potential to improve the likelihood of post-treatment remission if initiated during early infection.
Collapse
Affiliation(s)
- Christopher R. Hiner
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - April L. Mueller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - Hang Su
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
| | - Harris Goldstein
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (C.R.H.); (A.L.M.)
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
2
|
Nazziwa J, Andrews SM, Hou MM, Bruhn CAW, Garcia-Knight MA, Slyker J, Hill S, Lohman Payne B, Moringas D, Lemey P, John-Stewart G, Rowland-Jones SL, Esbjörnsson J. Higher HIV-1 evolutionary rate is associated with cytotoxic T lymphocyte escape mutations in infants. J Virol 2024; 98:e0007224. [PMID: 38814066 PMCID: PMC11265422 DOI: 10.1128/jvi.00072-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/20/2024] [Indexed: 05/31/2024] Open
Abstract
Escape from cytotoxic T lymphocyte (CTL) responses toward HIV-1 Gag and Nef has been associated with reduced control of HIV-1 replication in adults. However, less is known about CTL-driven immune selection in infants as longitudinal studies of infants are limited. Here, 1,210 gag and 1,264 nef sequences longitudinally collected within 15 months after birth from 14 HIV-1 perinatally infected infants and their mothers were analyzed. The number of transmitted founder (T/F) viruses and associations between virus evolution, selection, CTL escape, and disease progression were determined. The analyses indicated that a paraphyletic-monophyletic relationship between the mother-infant sequences was common (80%), and that the HIV-1 infection was established by a single T/F virus in 10 of the 12 analyzed infants (83%). Furthermore, most HIV-1 CTL escape mutations among infants were transmitted from the mothers and did not revert during the first year of infection. Still, immune-driven selection was observed at approximately 3 months after HIV-1 infection in infants. Moreover, virus populations with CTL escape mutations in gag evolved faster than those without, independently of disease progression rate. These findings expand the current knowledge of HIV-1 transmission, evolution, and CTL escape in infant HIV-1 infection and are relevant for the development of immune-directed interventions in infants.IMPORTANCEDespite increased coverage in antiretroviral therapy for the prevention of perinatal transmission, paediatric HIV-1 infection remains a significant public health concern, especially in areas of high HIV-1 prevalence. Understanding HIV-1 transmission and the subsequent virus adaptation from the mother to the infant's host environment, as well as the viral factors that affect disease outcome, is important for the development of early immune-directed interventions for infants. This study advances our understanding of vertical HIV-1 transmission, and how infant immune selection pressure is shaping the intra-host evolutionary dynamics of HIV-1.
Collapse
Affiliation(s)
- Jamirah Nazziwa
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund University, Lund, Sweden
| | - Sophie M. Andrews
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Mimi M. Hou
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Miguel A. Garcia-Knight
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - Jennifer Slyker
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Sarah Hill
- Department of Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
| | - Barbara Lohman Payne
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Dorothy Moringas
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Philippe Lemey
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Global Center for Integrated Health of Women, Adolescents and Children (Global WACh), University of Washington, Seattle, Washington, USA
| | | | - Joakim Esbjörnsson
- Department of Translational Medicine, Lund University, Lund, Sweden
- Lund University Virus Centre, Lund University, Lund, Sweden
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
3
|
Ealer C, Niemczak CE, Nicol T, Magohe A, Bonacina S, Zhang Z, Rieke AuD C, Leigh S, Kobrina A, Lichtenstein J, Massawe ER, Kraus N, Buckey JC. Auditory neural processing in children living with HIV uncovers underlying central nervous system dysfunction. AIDS 2024; 38:289-298. [PMID: 37905994 PMCID: PMC10841987 DOI: 10.1097/qad.0000000000003771] [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: 11/02/2023]
Abstract
OBJECTIVE Central nervous system (CNS) damage from HIV infection or treatment can lead to developmental delays and poor educational outcomes in children living with HIV (CLWH). Early markers of central nervous system dysfunction are needed to target interventions and prevent life-long disability. The frequency following response (FFR) is an auditory electrophysiology test that can reflect the health of the central nervous system. In this study, we explore whether the FFR reveals auditory central nervous system dysfunction in CLWH. STUDY DESIGN Cross-sectional analysis of an ongoing cohort study. Data were from the child's first visit in the study. SETTING The infectious disease center in Dar es Salaam, Tanzania. METHODS We collected the FFR from 151 CLWH and 151 HIV-negative children. To evoke the FFR, three speech syllabi (/da/, /ba/, /ga/) were played monaurally to the child's right ear. Response measures included neural timing (peak latencies), strength of frequency encoding (fundamental frequency and first formant amplitude), encoding consistency (inter-response consistency), and encoding precision (stimulus-to-response correlation). RESULTS CLWH showed smaller first formant amplitudes ( P < 0.0001), weaker inter-response consistencies ( P < 0.0001) and smaller stimulus to response correlations ( P < 0.0001) than FFRs from HIV-negative children. These findings generalized across the three speech stimuli with moderately strong effect sizes (partial η2 ranged from 0.061 to 0.094). CONCLUSION The FFR shows auditory central nervous system dysfunction in CLWH. Neural encoding of auditory stimuli was less robust, more variable, and less accurate. As the FFR is a passive and objective test, it may offer an effective way to assess and detect central nervous system function in CLWH.
Collapse
Affiliation(s)
- Christin Ealer
- Space Medicine Innovations Laboratory, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Christopher E. Niemczak
- Space Medicine Innovations Laboratory, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Trent Nicol
- Auditory Neuroscience Laboratory, Department of Communication Sciences, Northwestern University, Evanston, Illinois
| | - Albert Magohe
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Silvia Bonacina
- Auditory Neuroscience Laboratory, Department of Communication Sciences, Northwestern University, Evanston, Illinois
| | - Ziyin Zhang
- Space Medicine Innovations Laboratory, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Catherine Rieke AuD
- Space Medicine Innovations Laboratory, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Samantha Leigh
- Space Medicine Innovations Laboratory, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Anastasiya Kobrina
- Space Medicine Innovations Laboratory, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Jonathan Lichtenstein
- Department of Psychiatry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Enica R. Massawe
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Nina Kraus
- Auditory Neuroscience Laboratory, Department of Communication Sciences, Northwestern University, Evanston, Illinois
- Auditory Neuroscience Laboratory, Department of Communication Sciences, Neurobiology and Otolaryngology, Northwestern University, Evanston, Illinois
| | - Jay C. Buckey
- Space Medicine Innovations Laboratory, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 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
|
5
|
Russell ML, Fish CS, Drescher S, Cassidy NAJ, Chanana P, Benki-Nugent S, Slyker J, Mbori-Ngacha D, Bosire R, Richardson B, Wamalwa D, Maleche-Obimbo E, Overbaugh J, John-Stewart G, Matsen FA, Lehman DA. Using viral sequence diversity to estimate time of HIV infection in infants. PLoS Pathog 2023; 19:e1011861. [PMID: 38117834 PMCID: PMC10732395 DOI: 10.1371/journal.ppat.1011861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/27/2023] [Indexed: 12/22/2023] Open
Abstract
Age at HIV acquisition may influence viral pathogenesis in infants, and yet infection timing (i.e. date of infection) is not always known. Adult studies have estimated infection timing using rates of HIV RNA diversification, however, it is unknown whether adult-trained models can provide accurate predictions when used for infants due to possible differences in viral dynamics. While rates of viral diversification have been well defined for adults, there are limited data characterizing these dynamics for infants. Here, we performed Illumina sequencing of gag and pol using longitudinal plasma samples from 22 Kenyan infants with well-characterized infection timing. We used these data to characterize viral diversity changes over time by designing an infant-trained Bayesian hierarchical regression model that predicts time since infection using viral diversity. We show that diversity accumulates with time for most infants (median rate within pol = 0.00079 diversity/month), and diversity accumulates much faster than in adults (compare previously-reported adult rate within pol = 0.00024 diversity/month [1]). We find that the infant rate of viral diversification varies by individual, gene region, and relative timing of infection, but not by set-point viral load or rate of CD4+ T cell decline. We compare the predictive performance of this infant-trained Bayesian hierarchical regression model with simple linear regression models trained using the same infant data, as well as existing adult-trained models [1]. Using an independent dataset from an additional 15 infants with frequent HIV testing to define infection timing, we demonstrate that infant-trained models more accurately estimate time since infection than existing adult-trained models. This work will be useful for timing HIV acquisition for infants with unknown infection timing and for refining our understanding of how viral diversity accumulates in infants, both of which may have broad implications for the future development of infant-specific therapeutic and preventive interventions.
Collapse
Affiliation(s)
- Magdalena L. Russell
- Computational Biology Program, Fred Hutch Cancer Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
| | - Carolyn S. Fish
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Sara Drescher
- University of Washington Medical Center, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
| | - Noah A. J. Cassidy
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Pritha Chanana
- Bioinformatics Shared Resource, Fred Hutch Cancer Center, Seattle, Washington, United States of America
| | - Sarah Benki-Nugent
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Jennifer Slyker
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Dorothy Mbori-Ngacha
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Rose Bosire
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Barbra Richardson
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutch Cancer Center, Seattle, Washington, United States of America
| | - Dalton Wamalwa
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | | | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Frederick A. Matsen
- Computational Biology Program, Fred Hutch Cancer Center, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
- Department of Statistics, University of Washington, Seattle, Washington, United States of America
| | - Dara A. Lehman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
6
|
Schröter J, de Boer RJ. What explains the poor contraction of the viral load during paediatric HIV infection? J Theor Biol 2023; 570:111521. [PMID: 37164225 DOI: 10.1016/j.jtbi.2023.111521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/03/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023]
Abstract
An acute HIV infection in young children differs markedly from that in adults: Children have higher viral loads (VL), and a poor contraction to a setpoint VL that is not much lower than the peak VL. As a result, children progress faster towards AIDS in the absence of treatment. We used a classical ordinary differential equation model for viral infection dynamics to study why children have a lower viral contraction ratio than adults. We performed parameter sweeps to identify factors explaining the observed difference between children and adults. We grouped parameters associated with the host, the infection, or the immune response. Based on paediatric data available from datasets within the EPIICAL project (https://www.epiical.org/), we refuted that viral replication rates differ between young children and adults, and therefore these cannot be responsible for the low VL contraction ratios seen in children. The major differences in lowering VL contraction ratio resulted from sweeping the parameters linked to the immune response. Thus, we postulate that an "ineffective" (late and/or weak) immune response is the most parsimonious explanation for the higher setpoint VL in young children, and hence the reason for their fast disease progression.
Collapse
Affiliation(s)
- Juliane Schröter
- Theoretical Biology & Bioinformatics, Utrecht University, Utrecht, The Netherlands.
| | - Rob J de Boer
- Theoretical Biology & Bioinformatics, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
7
|
Faris JG, Orbidan D, Wells C, Petersen BK, Sprenger KG. Moving the needle: Employing deep reinforcement learning to push the boundaries of coarse-grained vaccine models. Front Immunol 2022; 13:1029167. [PMID: 36405722 PMCID: PMC9670804 DOI: 10.3389/fimmu.2022.1029167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Highly mutable infectious disease pathogens (hm-IDPs) such as HIV and influenza evolve faster than the human immune system can contain them, allowing them to circumvent traditional vaccination approaches and causing over one million deaths annually. Agent-based models can be used to simulate the complex interactions that occur between immune cells and hm-IDP-like proteins (antigens) during affinity maturation-the process by which antibodies evolve. Compared to existing experimental approaches, agent-based models offer a safe, low-cost, and rapid route to study the immune response to vaccines spanning a wide range of design variables. However, the highly stochastic nature of affinity maturation and vast sequence space of hm-IDPs render brute force searches intractable for exploring all pertinent vaccine design variables and the subset of immunization protocols encompassed therein. To address this challenge, we employed deep reinforcement learning to drive a recently developed agent-based model of affinity maturation to focus sampling on immunization protocols with greater potential to improve the chosen metrics of protection, namely the broadly neutralizing antibody (bnAb) titers or fraction of bnAbs produced. Using this approach, we were able to coarse-grain a wide range of vaccine design variables and explore the relevant design space. Our work offers new testable insights into how vaccines should be formulated to maximize protective immune responses to hm-IDPs and how they can be minimally tailored to account for major sources of heterogeneity in human immune responses and various socioeconomic factors. Our results indicate that the first 3 to 5 immunizations, depending on the metric of protection, should be specially tailored to achieve a robust protective immune response, but that beyond this point further immunizations require only subtle changes in formulation to sustain a durable bnAb response.
Collapse
Affiliation(s)
- Jonathan G. Faris
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Daniel Orbidan
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, United States
| | - Charles Wells
- Department of Computer Science, Rice University, TX, Houston, United States
| | - Brenden K. Petersen
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Kayla G. Sprenger
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
8
|
Kjaldgaard L, Claude KM, Mukadi-Bamuleka D, Kitenge-Omasumbu R, Dixit D, Edidi-Atani F, Kuamfumu MM, Bulabula-Penge J, Mambu-Mbika F, Tshiani-Mbaya O, Diaz J, Mulangu S, Legand A, Mbala-Kingebeni P, Formenty P, Ahuka-Mundeke S, Muyembe-Tamfum JJ, Hawkes MT. Virus kinetics and biochemical derangements among children with Ebolavirus disease. EClinicalMedicine 2022; 53:101638. [PMID: 36105872 PMCID: PMC9465268 DOI: 10.1016/j.eclinm.2022.101638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND A paucity of data is available on virologic and biochemical characteristics of paediatric Ebolavirus disease (EVD), compared to adults. METHODS We conducted a retrospective chart review of children (<16 years old) and a comparator group of young adults (16-44 years) from two treatment centres during the 2018-2020 EVD epidemic in Eastern Democratic Republic of the Congo. Statistical methods included chi-squared and Fisher's exact tests (dichotomous and categorical variables), Mann-Whitney U-test (continuous variables), multivariable linear regression (for determinants of admission viral load), linear mixed-effects models (for analysis of longitudinal viral load), and Cox proportional hazard models (to examine risk factors for mortality). FINDINGS We included 73 children and 234 adults admitted from April to October 2019. Paediatric patients commonly had electrolytes imbalances: hypokalaemia in 26/73 (36%), hyperkalaemia in 38/73 (52%), and hyponatraemia in 54/73 (74%). Hypoglycaemia occurred in 20/73 (27%), acute kidney injury in 43/73 (59%), and rhabdomyolysis in 35/73 (48%). Biochemical abnormalities were detected in a similar proportion of children and adults. The viral load (VL, log10 copies/mL) at admission (7.2 versus 6.5, p=0.0001), the peak viral load (7.5 versus 6.7, p=<0.0001), and the time for viraemia clearance (16 days versus 12 days, p=<0.0001) were significantly different in children. The duration of hospital stay was prolonged in children (20 versus 16 days, p=<0.0001). Risk factors for mortality in children were: VL >7.6 log10copies/mL, alanine transaminase >525 U/L, C-reactive protein >100 mg/L, blood urea nitrogen >7.5 mmol/L, rhabdomyolysis, and.acute kidney injury. INTERPRETATION Paediatric EVD patients, like adults, experience multiorgan dysfunction with life-threatening electrolyte imbalances, hypoglycaemia, kidney injury, liver injury, and rhabdomyolysis. Paediatric patients have significantly higher VLs throughout the course of EVD than adults. FUNDING This study was not funded.
Collapse
Affiliation(s)
- Lindsey Kjaldgaard
- Department of Paediatrics, University of Alberta, Edmonton, AB, Canada
- Member, Women and Children's Research Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Daniel Mukadi-Bamuleka
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Richard Kitenge-Omasumbu
- Programme National d'Urgences et Actions Humanitaires (PNUAH), Ministry of Health of the Democratic Republic of the Congo (DRC), Democratic Republic of the Congo
| | - Devika Dixit
- Department of Medicine, Division of Infectious Diseases, University of Saskatchewan, Saskatoon, SK, Canada
| | - François Edidi-Atani
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Meris Matondo Kuamfumu
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Junior Bulabula-Penge
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Fabrice Mambu-Mbika
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Olivier Tshiani-Mbaya
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Janet Diaz
- World Health Organization (WHO), Geneva, Switzerland
| | - Sabue Mulangu
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | - Anais Legand
- World Health Organization (WHO), Geneva, Switzerland
| | - Placide Mbala-Kingebeni
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | | | - Steve Ahuka-Mundeke
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of the Congo
| | | | - Michael T. Hawkes
- Member, Women and Children's Research Institute, University of Alberta, Edmonton, AB, Canada
- Department of Paediatrics, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- School of Public Health, University of Alberta, Edmonton, AB, Canada
- Distinguished Researcher, Stollery Science Lab, University of Alberta, Edmonton, AB, Canada
- Corresponding author at: Department of Paediatrics, University of Alberta, 3-588D Edmonton Clinic Health Academy, 11405 87 Ave NW, Edmonton, AB T6G 1C9, Canada.
| |
Collapse
|
9
|
Jordan MR, Bikinesi L, Ashipala L, Mutenda N, Brantuo M, Hunt G, Shiningavamwe A, Mutandi G, Beukes A, Beard S, Battey K, Dziuban EJ, Raizes E, Adjei P, Tang A, Giron A, Hong SY. Pretreatment HIV drug resistance among treatment naïve infants newly diagnosed with HIV in 2016 in Namibia: results of a nationally representative study. Open Forum Infect Dis 2022; 9:ofac102. [PMID: 35434174 PMCID: PMC9007920 DOI: 10.1093/ofid/ofac102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/14/2022] [Indexed: 12/02/2022] Open
Abstract
Background The World Health Organization (WHO) recommends routine surveillance of pretreatment human immunodeficiency virus (HIV) drug resistance (HIVDR) in children <18 months of age diagnosed with HIV through early infant diagnosis (EID). In 2016, 262 children <18 months of age were diagnosed with HIV in Namibia through EID. Levels of HIVDR in this population are unknown. Methods In 2016, Namibia surveyed pretreatment HIVDR among children aged <18 months following WHO guidance. Reverse transcriptase, protease, and integrase regions of HIV-1 were genotyped from remnant dried blood spot specimens from all infants diagnosed with HIV in Namibia in 2016. HIVDR was predicted using the Stanford HIVdb algorithm. Results Of 262 specimens genotyped, 198 HIV-1 protease and reverse transcriptase sequences and 118 HIV-1 integrase sequences were successfully amplified and analyzed. The prevalence of efavirenz/nevirapine (EFV/NVP), abacavir (ABC), zidovudine, lamivudine/emtricitabine (3TC/FTC), and tenofovir (TDF) resistance was 62.6%, 17.7%, 5.6%, 15.7%, and 10.1%, respectively. No integrase inhibitor resistance was detected. Conclusions The high level of EFV/NVP resistance is unsurprising; however, levels of ABC and TDF resistance are among the highest observed to date in infants in sub-Saharan Africa. The absence of resistance to dolutegravir (DTG) is reassuring but underscores the need to further study the impact of ABC and 3TC/FTC resistance on pediatric protease inhibitor– and DTG-based regimens and accelerate access to other antiretroviral drugs. Results underscore the need for antiretroviral therapy optimization and prompt management of high viral loads in infants and pregnant and breastfeeding women.
Collapse
Affiliation(s)
- Michael R Jordan
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA, USA and Levy Center for Integrated Management of Antimicrobial Resistance, Tufts University School of Medicine, Boston, MA, USA
| | - Leonard Bikinesi
- Directorate of Special Programmes, Republic of Namibia Ministry of Health and Social Services, Windhoek, Namibia
| | - Laimi Ashipala
- Directorate of Special Programmes, Republic of Namibia Ministry of Health and Social Services, Windhoek, Namibia
| | - Nicholus Mutenda
- Directorate of Special Programmes, Republic of Namibia Ministry of Health and Social Services, Windhoek, Namibia
| | | | - Gillian Hunt
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa
| | | | - Gram Mutandi
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Windhoek, Namibia
| | - Anita Beukes
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Windhoek, Namibia
| | - Suzanne Beard
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Katherine Battey
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA. USA
| | - Eric J Dziuban
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Windhoek, Namibia
| | - Elliot Raizes
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Paul Adjei
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA, USA
| | - Alice Tang
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, USA
| | - Amalia Giron
- Department of Global HIV, Hepatitis and STI Programmes, World Health Organization, Geneva, Switzerland
| | - Steven Y Hong
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Windhoek, Namibia
| |
Collapse
|
10
|
Derivation of an HIV Risk Score for African Women Who Engage in Sex Work. AIDS Behav 2021; 25:3292-3302. [PMID: 33861378 DOI: 10.1007/s10461-021-03235-7] [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: 03/13/2021] [Indexed: 10/21/2022]
Abstract
No tool exists to stratify HIV risk in contemporary African female sex worker (FSW) populations. Data from a cohort of HIV-negative FSWs in Mombasa, Kenya from 2010 to 2017 were used to conduct a survival analysis assessing predictors of HIV infection. Stepwise regression was used to construct a multivariable model that formed the basis for the score. Seventeen HIV infections occurred over 1247 person-years of follow-up contributed by 670 women. Using depot medroxyprogesterone acetate (DMPA), having a curable sexually transmitted infection (STI), and being married contributed points to the score. HIV incidence was 0.85/100 person-years in a lower-risk group and 3.10/100 person-years in a higher-risk group. In a cohort with overall HIV incidence < 1.50/100 person-years, this risk score identified a subgroup of FSWs with HIV incidence > 3.00/100 person-years, which is the threshold used by the World Health Organization for initiating pre-exposure prophylaxis (PrEP). If validated in an external population, this tool could be useful for targeted PrEP promotion among higher-risk FSWs.
Collapse
|
11
|
Genetic Control of Neonatal Immune Tolerance to an Exogenous Retrovirus. J Virol 2020; 94:JVI.01608-20. [PMID: 32999021 DOI: 10.1128/jvi.01608-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Abstract
Viruses, including retroviruses, can be passed from mothers to their progeny during birth and breastfeeding. It is assumed that newborns may develop immune tolerance to milk-transmitted pathogens similarly to food antigens. I/LnJ mice are uniquely resistant to retroviruses acquired as newborns or as adults as they produce virus-neutralizing antibodies (Abs). A loss-of-function allele of H2-Ob (Ob), originally mapped within the virus infectivity controller 1 (vic1) locus, is responsible for production of antiretrovirus Abs in I/LnJ mice. Importantly, Ob-deficient and vic1 I/LnJ congenic mice on other genetic backgrounds produce antivirus Abs when infected as adults, but not as newborns. We report here that I/LnJ mice carry an additional genetic locus, virus infectivity controller 2 (vic2), that abrogates neonatal immune tolerance to retroviruses. Further genetic analysis mapped the vic2 locus to the telomeric end of chromosome 15. Identification of the vic2 gene and understanding of the related signaling pathways would make blocking of neonatal immune tolerance to retroviruses an achievable goal.IMPORTANCE This work describes a previously unknown genetic mechanism that allows neonates to respond to infections as efficiently as adults.
Collapse
|
12
|
Kageyama S, Amolong Hinay A, Telan EFO, Samonte GMJ, Leano PSA, Tsuneki-Tokunaga A, Kanai K. Intrinsic Replication Competences of HIV Strains After Zidovudine/Lamivudine/Nevirapine Treatment in the Philippines. J Int Assoc Provid AIDS Care 2020; 18:2325958219856579. [PMID: 31216920 PMCID: PMC6748504 DOI: 10.1177/2325958219856579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although drug-resistant HIV variants are considered to be less fit than drug-susceptible viruses, replication competence of these variants harbored by patients has not yet been elucidated in detail. We herein assessed the replication competence of strains obtained from individuals receiving antiretroviral therapy. Among 11 306 participants in a drug resistance surveillance in the Philippines, 2629 plasma samples were obtained from individuals after a 12-month treatment with zidovudine (ZDV)/lamivudine (3TC)/nevirapine (NVP). The replication competence of HIV isolates was then assessed by reinoculation into seronegative peripheral blood mononuclear cells in the absence of drugs in vitro. The drug resistance rate was estimated to be 9.2%. Drug-resistant strains were still a minority of closely related strains in a phylogenetic cluster. Among the available 295 samples, 37 HIV strains were successfully isolated. Progeny viruses were produced at a wide range (5.1 × 106 to 3.4 × 109 copies/mL) in primary culture of peripheral blood mononuclear cells. The viral yields were higher than the corresponding plasma viral load (1300 to 3.4 × 106 copies/mL) but correlated with those (r = 0.4). These results suggest that strains with higher intrinsic replication competence are one of the primary targets of newly selected drugs at the increasing phase of the plasma viral load during antiretroviral therapy.
Collapse
Affiliation(s)
- Seiji Kageyama
- 1 Division of Virology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Alfredo Amolong Hinay
- 1 Division of Virology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | | | | | - Prisca Susan Agustin Leano
- 2 National Reference Laboratory, STD AIDS Cooperative Central Laboratory, San Lazaro Hospital, Manila, Philippines
| | - Akeno Tsuneki-Tokunaga
- 1 Division of Virology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kyosuke Kanai
- 1 Division of Virology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
13
|
Pollara J, Edwards RW, Jha S, Lam CYK, Liu L, Diedrich G, Nordstrom JL, Huffman T, Pickeral JA, Denny TN, Permar SR, Ferrari G. Redirection of Cord Blood T Cells and Natural Killer Cells for Elimination of Autologous HIV-1-Infected Target Cells Using Bispecific DART® Molecules. Front Immunol 2020; 11:713. [PMID: 32373131 PMCID: PMC7186435 DOI: 10.3389/fimmu.2020.00713] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022] Open
Abstract
Mother-to-child transmission of HIV-1 remains a major global health challenge. Currently, HIV-1-infected infants require strict lifelong adherence to antiretroviral therapy to prevent replication of virus from reservoirs of infected cells, and to halt progression of disease. There is a critical need for immune interventions that can be deployed shortly after infection to eliminate HIV-1-infected cells in order to promote long-term remission of viremia, or to potentially cure pediatric HIV-1-infection. Bispecific HIV × CD3 DART® molecules able to co-engage the HIV-1 envelope protein on the surface of infected cells and CD3 on cytolytic T cells have been previously shown to eliminate HIV-1 infected cells in vitro and are candidates for passive immunotherapy to reduce the virus reservoir. However, their potential utility as therapy for infant HIV-1 infection is unclear as the ability of these novel antibody-based molecules to work in concert with cells of the infant immune system had not been assessed. Here, we use human umbilical cord blood as a model of the naïve neonatal immune system to evaluate the ability of HIV x CD3 DART molecules to recruit and redirect neonatal effector cells for elimination of autologous CD4+ T cells infected with HIV-1 encoding an envelope gene sequenced from a mother-to-child transmission event. We found that HIV × CD3 DART molecules can redirect T cells present in cord blood for elimination of HIV-infected CD4+ T cells. However, we observed reduced killing by T cells isolated from cord blood when compared to cells isolated from adult peripheral blood-likely due to the absence of the memory and effector CD8+ T cells that are most cytolytic when redirected by bispecific DART molecules. We also found that newly developed HIV × CD16 DART molecules were able to recruit CD16-expressing natural killer cells from cord blood to eliminate HIV-infected cells, and the activity of cord blood natural killer cells could be substantially increased by priming with IL-15. Our results support continued development of HIV-specific DART molecules using relevant preclinical animal models to optimize strategies for effective use of this immune therapy to reduce HIV-1 infection in pediatric populations.
Collapse
Affiliation(s)
- Justin Pollara
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - R Whitney Edwards
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Shalini Jha
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | | | - Liqin Liu
- Macrogenics, Inc., Rockville, MD, United States
| | | | | | - Tori Huffman
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Joy A Pickeral
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Thomas N Denny
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Sallie R Permar
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States.,Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Guido Ferrari
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States.,Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
14
|
Fokam J, Sosso SM, Yagai B, Billong SC, Djubgang Mbadie RE, Kamgaing Simo R, Edimo SV, Nka AD, Tiga Ayissi A, Yimga JF, Takou D, Moudourou S, Ngo Nemb M, Nfetam Elat JB, Santoro MM, Perno CF, Colizzi V, Ndjolo A. Viral suppression in adults, adolescents and children receiving antiretroviral therapy in Cameroon: adolescents at high risk of virological failure in the era of "test and treat". AIDS Res Ther 2019; 16:36. [PMID: 31744517 PMCID: PMC6864925 DOI: 10.1186/s12981-019-0252-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/09/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND After the launching of the « Test & Treat » strategy and the wider accessibility to viral load (VL), evaluating virological success (VS) would help in meeting the UNAIDS targets by 2020 in Cameroon. SETTING AND METHODS Cross-sectional study conducted in the Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management (CIRCB), Yaoundé, Cameroon; data generated between October 2016 and August 2017 amongst adults, adolescents and children at 12, 24, 36 and ≥ 48 months on ART. VS was defined as < 1000 copies/mL of blood plasma and controlled viremia as VL < 50 copies/mL. Data were analysed by SPSS; p < 0.05 considered as significant. RESULTS 1946 patients (70% female) were enrolled (1800 adults, 105 adolescents, 41 children); 1841 were on NNRTI-based and 105 on PI-based therapy; with 346 patients at M12, 270 at M24, 205 at M36 and 1125 at ≥ M48. The median (IQR) duration on was 48 months (24-48). Overall, VS was 79.4% (95% CI 77.6-81.2) and 67.1% (95% CI 64.9-69.1) had controlled viral replication. On NNRTI-based, VS was 79.9% vs. 71.4% on PIs-based, p = 0.003. By ART duration, VS was 84.1% (M12), 85.9% (M24), 75.1% (M36) and 77.2% (≥ M48), p = 0.001. By age, VS was 75.6% (children), 53.3% (adolescents) and 81.1% (adults), p < 0.001. CONCLUSIONS In this sub-population of patients receiving ART in Cameroon, about 80% might be experiencing VS, with declining performance at adolescence, with NNRTI-based regimens, and as from 36 months on ART. Thus, improving VS may require an adapted adherence support mechanism, especially for adolescents with long-term treatment in resource-limited settings.
Collapse
Affiliation(s)
- Joseph Fokam
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon.
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon.
- National HIV Drug Resistance Working Group, Ministry of Public Health, Yaounde, Republic of Cameroon.
| | - Samuel Martin Sosso
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon.
| | - Bouba Yagai
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Serge Clotaire Billong
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- National HIV Drug Resistance Working Group, Ministry of Public Health, Yaounde, Republic of Cameroon
- Central Technical Group, National AIDS Control Committee, Yaoundé, Cameroon
| | | | - Rachel Kamgaing Simo
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
| | - Serge Valery Edimo
- Central Technical Group, National AIDS Control Committee, Yaoundé, Cameroon
| | - Alex Durand Nka
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
| | - Aline Tiga Ayissi
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
| | - Junie Flore Yimga
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
| | - Désiré Takou
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
| | - Sylvie Moudourou
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
| | - Marinette Ngo Nemb
- Central Technical Group, National AIDS Control Committee, Yaoundé, Cameroon
| | - Jean-Bosco Nfetam Elat
- National HIV Drug Resistance Working Group, Ministry of Public Health, Yaounde, Republic of Cameroon
- Central Technical Group, National AIDS Control Committee, Yaoundé, Cameroon
| | - Maria-Mercedes Santoro
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Carlo-Federico Perno
- Department of Experimental Medicine, Faculty of Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
- Department of Microbiology, University of Milan, Milan, Italy
| | - Vittorio Colizzi
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
- UNESCO Multidisciplinary Board of Biotechnology, University of Rome Tor Vergata, Rome, Italy
- Faculty of Biomedical Sciences, Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Alexis Ndjolo
- Chantal BIYA International Reference Centre for Research On HIV/AIDS Prevention and Management (CIRCB), Melen Road, PO BOX 3077, Yaounde, Cameroon
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| |
Collapse
|
15
|
Nelson AN, Goswami R, Dennis M, Tu J, Mangan RJ, Saha PT, Cain DW, Curtis AD, Shen X, Shaw GM, Bar K, Hudgens M, Pollara J, De Paris K, Van Rompay KKA, Permar SR. Simian-Human Immunodeficiency Virus SHIV.CH505-Infected Infant and Adult Rhesus Macaques Exhibit Similar Env-Specific Antibody Kinetics, despite Distinct T-Follicular Helper and Germinal Center B Cell Landscapes. J Virol 2019; 93:e00168-19. [PMID: 31092583 PMCID: PMC6639294 DOI: 10.1128/jvi.00168-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/02/2019] [Indexed: 12/29/2022] Open
Abstract
Global elimination of pediatric human immunodeficiency virus (HIV) infections will require the development of novel immune-based approaches, and understanding infant immunity to HIV is critical to guide the rational design of these intervention strategies. Despite their immunological immaturity, chronically HIV-infected children develop broadly neutralizing antibodies (bnAbs) more frequently and earlier than adults do. However, the ontogeny of humoral responses during acute HIV infection is poorly defined in infants and challenging to study in human cohorts due to the presence of maternal antibodies. To further our understanding of age-related differences in the development of HIV-specific immunity during acute infection, we evaluated the generation of virus-specific humoral immune responses in infant (n = 6) and adult (n = 12) rhesus macaques (RMs) infected with a transmitted/founder (T/F) simian-human immunodeficiency virus (SHIV) (SHIV.C.CH505 [CH505]). The plasma HIV envelope-specific IgG antibody kinetics were similar in SHIV-infected infant and adult RMs, with no significant differences in the magnitude or breadth of these responses. Interestingly, autologous tier 2 virus neutralization responses also developed with similar frequencies and kinetics in infant and adult RMs, despite infants exhibiting significantly higher follicular T helper cell (Tfh) and germinal center B cell frequencies than adults. Finally, we show that plasma viral load was the strongest predictor of the development of autologous virus neutralization in both age groups. Our results indicate that the humoral immune response to SHIV infection develops with similar kinetics among infant and adult RMs, suggesting that the early-life immune system is equipped to respond to HIV-1 and promote the production of neutralizing HIV antibodies.IMPORTANCE There is a lack of understanding of how the maturation of the infant immune system influences immunity to HIV infection or how these responses differ from those of adults. Improving our knowledge of infant HIV immunity will help guide antiviral intervention strategies that take advantage of the unique infant immune environment to successfully elicit protective immune responses. We utilized a rhesus macaque model of SHIV infection as a tool to distinguish the differences in HIV humoral immunity in infants versus adults. Here, we demonstrate that the kinetics and quality of the infant humoral immune response to HIV are highly comparable to those of adults during the early phase of infection, despite distinct differences in their Tfh responses, indicating that slightly different mechanisms may drive infant and adult humoral immunity.
Collapse
Affiliation(s)
- Ashley N Nelson
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Ria Goswami
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Maria Dennis
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Joshua Tu
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Riley J Mangan
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Pooja T Saha
- Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Derek W Cain
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Alan D Curtis
- Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Xiaoying Shen
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - George M Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katharine Bar
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Hudgens
- Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Justin Pollara
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Kristina De Paris
- Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, California, USA
| | - Sallie R Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
16
|
Mavigner M, Habib J, Deleage C, Rosen E, Mattingly C, Bricker K, Kashuba A, Amblard F, Schinazi RF, Lawson B, Vanderford TH, Jean S, Cohen J, McGary C, Paiardini M, Wood MP, Sodora DL, Silvestri G, Estes J, Chahroudi A. Simian Immunodeficiency Virus Persistence in Cellular and Anatomic Reservoirs in Antiretroviral Therapy-Suppressed Infant Rhesus Macaques. J Virol 2018; 92:e00562-18. [PMID: 29997216 PMCID: PMC6146711 DOI: 10.1128/jvi.00562-18] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/29/2018] [Indexed: 11/20/2022] Open
Abstract
Worldwide, nearly two million children are infected with human immunodeficiency virus (HIV), with breastfeeding accounting for the majority of contemporary HIV transmissions. Antiretroviral therapy (ART) has reduced HIV-related morbidity and mortality but is not curative. The main barrier to a cure is persistence of latent HIV in long-lived reservoirs. However, our understanding of the cellular and anatomic sources of the HIV reservoir during infancy and childhood is limited. Here, we developed a pediatric model of ART suppression in orally simian immunodeficiency virus (SIV)-infected rhesus macaque (RM) infants, with measurement of virus persistence in blood and tissues after 6 to 9 months of ART. Cross-sectional analyses were conducted to compare SIV RNA and DNA levels in adult and infant RMs naive to treatment and on ART. We demonstrate efficient viral suppression following ART initiation in SIV-infected RM infants with sustained undetectable plasma viral loads in the setting of heterogeneous penetration of ART into lymphoid and gastrointestinal tissues and low drug levels in the brain. We further show reduction in SIV RNA and DNA on ART in lymphoid tissues of both infant and adult RMs but stable (albeit low) levels of SIV RNA and DNA in the brains of viremic and ART-suppressed infants. Finally, we report a large contribution of naive CD4+ T cells to the total CD4 reservoir of SIV in blood and lymph nodes of ART-suppressed RM infants that differs from what we show in adults. These results reveal important aspects of HIV/SIV persistence in infants and provide insight into strategic targets for cure interventions in a pediatric population.IMPORTANCE While antiretroviral therapy (ART) can reduce HIV replication, the virus cannot be eradicated from an infected individual, and our incomplete understanding of HIV persistence in reservoirs greatly complicates the generation of a cure for HIV infection. Given the immaturity of the infant immune system, it is critically important to study HIV reservoirs specifically in this population. Here, we established a pediatric animal model to simulate breastfeeding transmission and study SIV reservoirs in rhesus macaque (RM) infants. Our study demonstrates that ART can be safely administered to infant RMs for prolonged periods and that it efficiently controls viral replication in this model. SIV persistence was shown in blood and tissues, with similar anatomic distributions of SIV reservoirs in infant and adult RMs. However, in the peripheral blood and lymph nodes, a greater contribution of the naive CD4+ T cells to the SIV reservoir was observed in infants than in adults.
Collapse
Affiliation(s)
- Maud Mavigner
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jakob Habib
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Elias Rosen
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Cameron Mattingly
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Katherine Bricker
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Angela Kashuba
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Franck Amblard
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Raymond F Schinazi
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Benton Lawson
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Thomas H Vanderford
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sherrie Jean
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Joyce Cohen
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Colleen McGary
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Mirko Paiardini
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Matthew P Wood
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Donald L Sodora
- Center for Infectious Disease Research, Seattle, Washington, USA
| | - Guido Silvestri
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Jacob Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Ann Chahroudi
- Department of Pediatrics and Center for AIDS Research, Emory University School of Medicine, Atlanta, Georgia, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Emory+Children's Center for Childhood Infections and Vaccines, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Jordan MR, Penazzato M, Cournil A, Vubil A, Jani I, Hunt G, Carmona S, Maphalala G, Mthethwa N, Watera C, Kaleebu P, Musanhu CC, Mtapuri-Zinyowera S, Dzangare J, Peeters M, Yang C, Parkin N, Bertagnolio S. Human Immunodeficiency Virus (HIV) Drug Resistance in African Infants and Young Children Newly Diagnosed With HIV: A Multicountry Analysis. Clin Infect Dis 2018; 65:2018-2025. [PMID: 29020335 DOI: 10.1093/cid/cix698] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/03/2017] [Indexed: 11/12/2022] Open
Abstract
Background Programs for the prevention of mother-to-child transmission (PMTCT) of human immunodeficiency virus (HIV) have been scaled up in many low- and middle-income countries. However, HIV drug resistance (HIVDR) data among HIV-1-infected young children remain limited. Methods Surveys of pretreatment HIVDR among children aged <18 months who were diagnosed with HIV through early infant diagnosis were conducted in 5 sub-Saharan African countries (Mozambique, Swaziland, South Africa, Uganda, and Zimbabwe) between 2011 and 2014 following World Health Organization (WHO) guidance. Deidentified demographic and clinical data were used to explore risk factors associated with nonnucleoside reverse transcriptase inhibitor (NNRTI) resistance. Results Among the 1450 genotypes analyzed, 1048 had accompanying demographic and clinical data. The median age of children was 4 months; 50.4% were female. HIV from 54.1% showed resistance to 1 or more antiretroviral (ARV) drugs, with 53.0% and 8.8% having resistance to 1 or more NNRTI or nucleoside reverse transcriptase inhibitors, respectively. NNRTI resistance was particularly high in children exposed to ARV drugs through PMTCT; adjusted odds ratios were 1.8 (95% confidence interval [CI], 1.3-2.6) for maternal exposure only and 2.4 (CI, 1.6-3.6) for neonatal exposure only. Conclusions Protease inhibitor-based regimens in children aged <3 years are currently recommended by WHO, but the implementation of this recommendation is suboptimal. These results reinforce the urgent need to overcome barriers to scaling up pediatric protease inhibitor-based regimens in sub-Saharan Africa and underscore the need to accelerate the study and approval of integrase inhibitors for use in young children.
Collapse
Affiliation(s)
- Michael R Jordan
- Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts
| | - Martina Penazzato
- Department of HIV/AIDS, World Health Organization, Geneva, Switzerland
| | - Amandine Cournil
- IRD UMI 233, INSERM U1175, Université de Montpellier, Unité TransVIHMI, France
| | | | - Ilesh Jani
- Instituto Nacional de Saúde, Maputo, Mozambique
| | | | | | | | | | | | | | | | | | | | - Martine Peeters
- IRD UMI 233, INSERM U1175, Université de Montpellier, Unité TransVIHMI, France
| | - Chunfu Yang
- Division of Global HIV and TB, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | |
Collapse
|
18
|
Subbaraman H, Schanz M, Trkola A. Broadly neutralizing antibodies: What is needed to move from a rare event in HIV-1 infection to vaccine efficacy? Retrovirology 2018; 15:52. [PMID: 30055627 PMCID: PMC6064177 DOI: 10.1186/s12977-018-0433-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/21/2018] [Indexed: 02/06/2023] Open
Abstract
The elicitation of broadly neutralizing antibodies (bnAbs) is considered crucial for an effective, preventive HIV-1 vaccine. Led by the discovery of a new generation of potent bnAbs, the field has significantly advanced over the past decade. There is a wealth of knowledge about the development of bnAbs in natural infection, their specificity, potency, breadth and function. Yet, devising immunogens and vaccination regimens that evoke bnAb responses has not been successful. Where are the roadblocks in their development? What can we learn from natural infection, where bnAb induction is possible but rare? Herein, we will reflect on key discoveries and discuss open questions that may bear crucial insights needed to move towards creating effective bnAb vaccines.
Collapse
Affiliation(s)
- Harini Subbaraman
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Merle Schanz
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Aggarwal H, Khan L, Chaudhary O, Kumar S, Makhdoomi MA, Singh R, Sharma K, Mishra N, Lodha R, Srinivas M, Das BK, Kabra SK, Luthra K. Alterations in B Cell Compartment Correlate with Poor Neutralization Response and Disease Progression in HIV-1 Infected Children. Front Immunol 2017; 8:1697. [PMID: 29250072 PMCID: PMC5717014 DOI: 10.3389/fimmu.2017.01697] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
Several B cell defects are reported in HIV-1 infected individuals including variation in B cell subsets, polyclonal B cell activation and exhaustion, with broadly neutralizing antibodies elicited in less than 10–20% of the infected population. HIV-1 disease progression is faster in children than adults. B Lymphocyte Stimulator (BLyS), expressed on dendritic cells (DCs), is a key regulator of B cell homeostasis. Understanding how DCs influence B cell phenotype and functionality (viral neutralization), thereby HIV-1 disease outcome in infected children, is important to develop interventional strategies for restoration of B cell function. In this study, a total of 38 vertically transmitted HIV-1 infected antiretroviral therapy (ART) naïve children and 25 seronegative controls were recruited. Based on the CD4 counts and years post-infection, infected children were categorized as long-term non-progressors (LTNPs) (n = 20) and progressors (n = 18). Eight of these progressors were followed up at 6–12 months post-ART. Percentages (%) of DCs, B cell subsets, and expression of BLyS on DCs were analyzed by flow-cytometry. Plasma levels of B cell growth factors were measured by ELISA and viral neutralization activity was determined using TZM-bl assay. Lower (%) of myeloid DCs (mDCs), plasmacytoid DCs, and high expression of BLyS on mDCs were observed in HIV-1 infected progressors than seronegative controls. Progressors showed lower % of naive B cells, resting memory B cells and higher % of mature activated, tissue-like memory B cells as compared to seronegative controls. Higher plasma levels of IL-4, IL-6, IL-10, and IgA were observed in progressors vs. seronegative controls. Plasma levels of IgG were high in progressors and in LTNPs than seronegative controls, suggesting persistence of hypergammaglobulinemia at all stages of disease. High plasma levels of BLyS in progressors positively correlated with poor viral neutralizing activity. Interestingly on follow up, treatment naïve progressors, post-ART showed increase in resting memory B cells along with reduction in plasma BLyS levels that correlated with improvement in viral neutralization. This is the first study to demonstrate that reduction in plasma BLyS levels correlates with restoration of B cell function, in terms of viral neutralization in HIV-1-infected children.
Collapse
Affiliation(s)
- Heena Aggarwal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Lubina Khan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Omkar Chaudhary
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjeev Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Ravinder Singh
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Kanika Sharma
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Nitesh Mishra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Lodha
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Maddur Srinivas
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Bimal Kumar Das
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - Sushil Kumar Kabra
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
20
|
Kumar S, Kumar R, Khan L, Makhdoomi MA, Thiruvengadam R, Mohata M, Agarwal M, Lodha R, Kabra SK, Sinha S, Luthra K. CD4-Binding Site Directed Cross-Neutralizing scFv Monoclonals from HIV-1 Subtype C Infected Indian Children. Front Immunol 2017; 8:1568. [PMID: 29187855 PMCID: PMC5694743 DOI: 10.3389/fimmu.2017.01568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/01/2017] [Indexed: 12/12/2022] Open
Abstract
Progression of human immunodeficiency virus type-1 (HIV-1) infection in children is faster than adults. HIV-1 subtype C is responsible for more than 50% of the infections globally and more than 90% infections in India. To date, there is no effective vaccine against HIV-1. Recent animal studies and human Phase I trials showed promising results of the protective effect of anti-HIV-1 broadly neutralizing antibodies (bnAbs). Interaction between CD4 binding site (CD4bs) on the HIV-1 envelope glycoprotein and CD4 receptor on the host immune cells is the primary event leading to HIV-1 infection. The CD4bs is a highly conserved region, comprised of a conformational epitope, and is a potential target of bnAbs such as VRC01 that is presently under human clinical trials. Recombinant scFvs can access masked epitopes due to their small size and have shown the potential to inhibit viral replication and neutralize a broad range of viruses. Pediatric viruses are resistant to many of the existing bnAbs isolated from adults. Therefore, in this study, pooled peripheral blood mononuclear cells from 9 chronically HIV-1 subtype C infected pediatric cross-neutralizers whose plasma antibodies exhibited potent and cross-neutralizing activity were used to construct a human anti-HIV-1 scFv phage library of 9 × 108 individual clones. Plasma mapping using CD4bs-specific probes identified the presence of CD4bs directed antibodies in 4 of these children. By extensive biopanning of the library with CD4bs-specific antigen RSC3 core protein, we identified two cross-neutralizing scFv monoclonals 2B10 and 2E4 demonstrating a neutralizing breadth and GMT of 77%, 17.9 µg/ml and 32%, 51.2 µg/ml, respectively, against a panel of 49 tier 1, 2 and 3 viruses. Both scFvs competed with anti-CD4bs bnAb VRC01 confirming their CD4bs epitope specificity. The 2B10 scFv was effective in neutralizing the 7 subtype C and subtype A pediatric viruses tested. Somatic hypermutations in the VH gene of scFvs (10.1–11.1%) is comparable with that of the adult antibodies. These cross-neutralizing CD4bs-directed scFvs can serve as potential reagents for passive immunotherapy. A combination of cross-neutralizing scFvs of diverse specificities with antiretroviral drugs may be effective in suppressing viremia at an early stage of HIV-1 infection and prevent disease progression.
Collapse
Affiliation(s)
- Sanjeev Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rajesh Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Lubina Khan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | | | - Madhav Mohata
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Mudit Agarwal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Lodha
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Sushil Kumar Kabra
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
21
|
Makhdoomi MA, Khan L, Kumar S, Aggarwal H, Singh R, Lodha R, Singla M, Das BK, Kabra SK, Luthra K. Evolution of cross-neutralizing antibodies and mapping epitope specificity in plasma of chronic HIV-1-infected antiretroviral therapy-naïve children from India. J Gen Virol 2017; 98:1879-1891. [PMID: 28696188 DOI: 10.1099/jgv.0.000824] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Delineating the factors leading to the development of broadly neutralizing antibodies (bnAbs) during natural HIV-1 infection and dissecting their epitope specificities generates useful information for vaccine design. This is the first longitudinal study to assess the plasma-neutralizing antibody response and neutralizing determinants in HIV-1-infected children from India. We enrolled 26 and followed up 20 antiretroviral therapy (ART)-naïve, asymptomatic, chronic HIV-1-infected children. Five (19.2 %) baseline and 10 (50 %) follow-up plasma samples neutralized ≥50 % of subtypes A, B and C tier 2 viruses at an ID50 titre ≥150. A modest improvement in neutralization breadth and potency was observed with time. At baseline, subtype C-specific neutralization predominated (P=0.026); interestingly, follow-up samples exhibited cross-neutralizing activity. Epitope mapping revealed V3C reactive antibodies with significantly increased Max50 binding titres in follow-up samples from five infected children; patient #4's plasma antibodies exhibited V3-directed neutralization. A salient observation was the presence of CD4 binding site (CD4bs)-specific NAbs in patient #18 that improved with time (1.76-fold). The RSC3 wild-type (RSC3WT) protein-depleted plasma eluate of patient #18 demonstrated a more than 50% ID50 decrease in neutralization capacity against five HIV-1 pseudoviruses. Further, the presence of CD4bs-neutralizing determinants in patient #18's plasma was confirmed by the neutralizing activity demonstrated by the CD4bs-directed IgG fraction purified from this plasma, and competition with sCD4 against JRFLgp120, identifying this paediatric donor as a potential candidate for the isolation of CD4bs-directed bnAbs. Overall, we observed a relative increase in plasma-neutralizing activity with time in HIV-1-infected children, which suggests that the bnAbs evolve.
Collapse
Affiliation(s)
- Muzamil A Makhdoomi
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Lubina Khan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sanjeev Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Heena Aggarwal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ravinder Singh
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Rakesh Lodha
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mohit Singla
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Bimal K Das
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sushil K Kabra
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
22
|
Sex and gender differences in HIV-1 infection. Clin Sci (Lond) 2017; 130:1435-51. [PMID: 27389589 DOI: 10.1042/cs20160112] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/06/2016] [Indexed: 01/03/2023]
Abstract
The major burden of the human immunodeficiency (HIV) type 1 pandemic is nowadays carried by women from sub-Saharan Africa. Differences in the manifestations of HIV-1 infection between women and men have been long reported, and might be due to both socio-economic (gender) and biological (sex) factors. Several studies have shown that women are more susceptible to HIV-1 acquisition than men. Following HIV-1 infection, women have lower viral loads during acute infection and exhibit stronger antiviral responses than men, which may contribute to differences in the size of viral reservoirs. Oestrogen receptor signalling could represent an important mediator of sex differences in HIV-1 reservoir size and may represent a potential therapeutic target. Furthermore, immune activation, a hallmark of HIV-1 infection, is generally higher in women than in men and could be a central mechanism in the sex difference observed in the speed of HIV-1 disease progression. Here, we review the literature regarding sex-based differences in HIV-1 infection and discuss how a better understanding of the underlying mechanisms could improve preventive and therapeutic strategies.
Collapse
|
23
|
Benki-Nugent S, Wamalwa D, Langat A, Tapia K, Adhiambo J, Chebet D, Okinyi HM, John-Stewart G. Comparison of developmental milestone attainment in early treated HIV-infected infants versus HIV-unexposed infants: a prospective cohort study. BMC Pediatr 2017; 17:24. [PMID: 28095807 PMCID: PMC5240280 DOI: 10.1186/s12887-017-0776-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/02/2017] [Indexed: 11/18/2022] Open
Abstract
Background Infant HIV infection is associated with delayed milestone attainment. The extent to which effective antiretroviral therapy (ART) prevents these delays is not well defined. Methods Ages at attainment of milestones were compared between HIV-infected (initiated ART by age <5 months), and HIV-unexposed uninfected (HUU) infants. Kaplan Meier analyses were used to estimate and compare (log-rank tests) ages at milestones between groups. Adjusted analyses were performed using Cox proportional hazards models. Results Seventy-three HIV-infected on ART (median enrollment age 3.7 months) and 92 HUU infants (median enrollment age 1.6 months) were followed prospectively. HIV-infected infants on ART had delays in developmental milestone attainment compared to HUU: median age at attainment of sitting with support, sitting unsupported, walking with support, walking unsupported, monosyllabic speech and throwing toys were each delayed (all p-values <0.0005). Compared with HUU, the subset of HIV-infected infants with both virologic suppression and immune recovery at 6 months had delays for speech (delay: 2.0 months; P = 0.0002) and trend to later walking unsupported. Among HIV-infected infants with poor 6-month post-ART responses (lacking viral suppression and immune recovery) there were greater delays versus HUU for: walking unsupported (delay: 4.0 months; P = 0.0001) and speech (delay: 5.0 months; P < 0.0001). Conclusions HIV infected infants with viral suppression on ART had better recovery of developmental milestones than those without suppression, however, deficits persisted compared to uninfected infants. Earlier ART may be required for optimized cognitive outcomes in perinatally HIV-infected infants. Trial registration NCT00428116; January 22, 2007. Electronic supplementary material The online version of this article (doi:10.1186/s12887-017-0776-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sarah Benki-Nugent
- Department of Global Health, University of Washington, Box 359909, 325 9th Ave., Seattle, WA, 98104, USA.
| | - Dalton Wamalwa
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Agnes Langat
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Kenneth Tapia
- Department of Global Health, University of Washington, Box 359909, 325 9th Ave., Seattle, WA, 98104, USA
| | - Judith Adhiambo
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Daisy Chebet
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Helen Moraa Okinyi
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Box 359909, 325 9th Ave., Seattle, WA, 98104, USA
| |
Collapse
|
24
|
Differences in virologic and immunologic response to antiretroviral therapy among HIV-1-infected infants and children. AIDS 2016; 30:2835-2843. [PMID: 27603293 DOI: 10.1097/qad.0000000000001244] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Virologic and immunologic responses to antiretroviral treatment (ART) in infants may differ from older children due to immunologic, clinical, or epidemiologic characteristics. METHODS Longitudinal ART responses were modeled and compared in HIV-infected infants and children enrolled in cohorts in Nairobi, Kenya. Participants were enrolled soon after HIV diagnosis, started on ART, and followed for 2 years. Viral load decline was compared between infant and child cohorts using a nonlinear mixed effects model and CD4% reconstitution using a linear mixed effects model. RESULTS Among 121 infants, median age at ART was 3.9 months; among 124 children, median age was 4.8 years. At baseline, viral load was higher among infants than children (6.47 vs. 5.91 log10 copies/ml, P < 0.001). Infants were less likely than children to suppress viral load to less than 250 copies/ml following 6 months of ART (32% infants vs. 73% children, P < 0.0001). CD4% was higher at baseline in infants than children (19 vs. 7.3%, P < 0.001). Older children had more rapid CD4% reconstitution than infants, but failed to catch up to infant CD4%. CONCLUSION Despite substantially higher CD4% at ART initiation, viral suppression was significantly slower among infants than older children. New strategies are needed to optimize infant outcomes on ART.
Collapse
|
25
|
Abstract
Globally, 240,000 infants are newly infected with HIV-1 each year and 3.2 million children are living with the infection. Combination antiretroviral therapy (cART) has reduced HIV-1-related disease and mortality in children but is not curative owing to the early generation of a latent reservoir of long-lived memory CD4(+) T cells bearing replication-competent HIV-1 provirus integrated into cellular DNA. This review focuses on recent advances in our understanding of the establishment of HIV-1 persistence in children and how early initiation of cART in the setting of the developing infant immune system limits the formation of the long-lived latent CD4(+) cell reservoir that remains a barrier to remission or cure.
Collapse
Affiliation(s)
- Katherine Luzuriaga
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605;
| |
Collapse
|
26
|
Garcia-Knight MA, Slyker J, Payne BL, Pond SLK, de Silva TI, Chohan B, Khasimwa B, Mbori-Ngacha D, John-Stewart G, Rowland-Jones SL, Esbjörnsson J. Viral Evolution and Cytotoxic T Cell Restricted Selection in Acute Infant HIV-1 Infection. Sci Rep 2016; 6:29536. [PMID: 27403940 PMCID: PMC4941567 DOI: 10.1038/srep29536] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/17/2016] [Indexed: 01/21/2023] Open
Abstract
Antiretroviral therapy-naive HIV-1 infected infants experience poor viral containment and rapid disease progression compared to adults. Viral factors (e.g. transmitted cytotoxic T- lymphocyte (CTL) escape mutations) or infant factors (e.g. reduced CTL functional capacity) may explain this observation. We assessed CTL functionality by analysing selection in CTL-targeted HIV-1 epitopes following perinatal infection. HIV-1 gag, pol and nef sequences were generated from a historical repository of longitudinal specimens from 19 vertically infected infants. Evolutionary rate and selection were estimated for each gene and in CTL-restricted and non-restricted epitopes. Evolutionary rate was higher in nef and gag vs. pol, and lower in infants with non-severe immunosuppression vs. severe immunosuppression across gag and nef. Selection pressure was stronger in infants with non-severe immunosuppression vs. severe immunosuppression across gag. The analysis also showed that infants with non-severe immunosuppression had stronger selection in CTL-restricted vs. non-restricted epitopes in gag and nef. Evidence of stronger CTL selection was absent in infants with severe immunosuppression. These data indicate that infant CTLs can exert selection pressure on gag and nef epitopes in early infection and that stronger selection across CTL epitopes is associated with favourable clinical outcomes. These results have implications for the development of paediatric HIV-1 vaccines.
Collapse
Affiliation(s)
- Miguel A Garcia-Knight
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom.,KEMRI-Wellcome Trust Research Program, Centre for Geographical Medicine Research, Kilifi, Kenya
| | - Jennifer Slyker
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Barbara Lohman Payne
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya.,Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Sergei L Kosakovsky Pond
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
| | - Thushan I de Silva
- Section of Paediatrics, Department of Medicine, Wright Fleming Institute, Imperial College London, London, United Kingdom
| | - Bhavna Chohan
- Department of Global Health, University of Washington, Seattle, Washington, United States of America.,Kenyan Medical Research Institute, Nairobi, Kenya
| | | | - Dorothy Mbori-Ngacha
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, Washington, United States of America.,Department of Medicine, University of Washington, Seattle, Washington, United States of America.,Department of Epidemiology, University of Washington, Seattle, Washington, United States of America.,Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Sarah L Rowland-Jones
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Joakim Esbjörnsson
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
27
|
Robb ML, Eller LA, Kibuuka H, Rono K, Maganga L, Nitayaphan S, Kroon E, Sawe FK, Sinei S, Sriplienchan S, Jagodzinski LL, Malia J, Manak M, de Souza MS, Tovanabutra S, Sanders-Buell E, Rolland M, Dorsey-Spitz J, Eller MA, Milazzo M, Li Q, Lewandowski A, Wu H, Swann E, O'Connell RJ, Peel S, Dawson P, Kim JH, Michael NL. Prospective Study of Acute HIV-1 Infection in Adults in East Africa and Thailand. N Engl J Med 2016; 374:2120-30. [PMID: 27192360 PMCID: PMC5111628 DOI: 10.1056/nejmoa1508952] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Acute human immunodeficiency virus type 1 (HIV-1) infection is a major contributor to transmission of HIV-1. An understanding of acute HIV-1 infection may be important in the development of treatment strategies to eradicate HIV-1 or achieve a functional cure. METHODS We performed twice-weekly qualitative plasma HIV-1 RNA nucleic acid testing in 2276 volunteers who were at high risk for HIV-1 infection. For participants in whom acute HIV-1 infection was detected, clinical observations, quantitative measurements of plasma HIV-1 RNA levels (to assess viremia) and HIV antibodies, and results of immunophenotyping of lymphocytes were obtained twice weekly. RESULTS Fifty of 112 volunteers with acute HIV-1 infection had two or more blood samples collected before HIV-1 antibodies were detected. The median peak viremia (6.7 log10 copies per milliliter) occurred 13 days after the first sample showed reactivity on nucleic acid testing. Reactivity on an enzyme immunoassay occurred at a median of 14 days. The nadir of viremia (4.3 log10 copies per milliliter) occurred at a median of 31 days and was nearly equivalent to the viral-load set point, the steady-state viremia that persists durably after resolution of acute viremia (median plasma HIV-1 RNA level, 4.4 log10 copies per milliliter). The peak viremia and downslope were correlated with the viral-load set point. Clinical manifestations of acute HIV-1 infection were most common just before and at the time of peak viremia. A median of one symptom of acute HIV-1 infection was recorded at a median of two study visits, and a median of one sign of acute HIV-1 infection was recorded at a median of three visits. CONCLUSIONS The viral-load set point occurred at a median of 31 days after the first detection of plasma viremia and correlated with peak viremia. Few symptoms and signs were observed during acute HIV-1 infection, and they were most common before peak viremia. (Funded by the Department of Defense and the National Institute of Allergy and Infectious Diseases.).
Collapse
Affiliation(s)
- Merlin L Robb
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Leigh A Eller
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Hannah Kibuuka
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Kathleen Rono
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Lucas Maganga
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Sorachai Nitayaphan
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Eugene Kroon
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Fred K Sawe
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Samuel Sinei
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Somchai Sriplienchan
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Linda L Jagodzinski
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Jennifer Malia
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Mark Manak
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Mark S de Souza
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Sodsai Tovanabutra
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Eric Sanders-Buell
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Morgane Rolland
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Julie Dorsey-Spitz
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Michael A Eller
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Mark Milazzo
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Qun Li
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Andrew Lewandowski
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Hao Wu
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Edith Swann
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Robert J O'Connell
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Sheila Peel
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Peter Dawson
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Jerome H Kim
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| | - Nelson L Michael
- From the U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring (M.L.R., L.A.E., L.L.J., J.M., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L., S.P., J.H.K., N.L.M.), Henry M. Jackson Foundation for the Advancement of Military Medicine (M.L.R., L.A.E., M. Manak, S.T., E.S.-B., M.R., J.D.-S., M.A.E., M. Milazzo, Q.L.), the Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health (E.S.), Bethesda, and Emmes, Rockville (A.L., H.W., P.D.) - all in Maryland; Makerere University Walter Reed Project, Kampala, Uganda (H.K.); Walter Reed Project, Kericho, Kenya (K.R., F.K.S., S. Sinei); Mbeya Medical Research Centre, Mbeya, Tanzania (L.M.); the Armed Forces Research Institute of Medical Sciences (S.N., E.K., S. Sriplienchan, M.S.S., R.J.O.) and SEARCH, Thai Red Cross AIDS Research Center (E.K., M.S.S.) - both in Bangkok, Thailand; and the International Vaccine Institute, Seoul, South Korea (J.H.K.)
| |
Collapse
|
28
|
Affiliation(s)
- Katherine Luzuriaga
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester (K.L.); and the Elizabeth Glaser Pediatric AIDS Foundation, Washington, DC (L.M.M.)
| | - Lynne M Mofenson
- From the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester (K.L.); and the Elizabeth Glaser Pediatric AIDS Foundation, Washington, DC (L.M.M.)
| |
Collapse
|
29
|
Lillis L, Lehman DA, Siverson JB, Weis J, Cantera J, Parker M, Piepenburg O, Overbaugh J, Boyle DS. Cross-subtype detection of HIV-1 using reverse transcription and recombinase polymerase amplification. J Virol Methods 2016; 230:28-35. [PMID: 26821087 DOI: 10.1016/j.jviromet.2016.01.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 01/21/2016] [Accepted: 01/21/2016] [Indexed: 11/29/2022]
Abstract
A low complexity diagnostic test that rapidly and reliably detects HIV infection in infants at the point of care could facilitate early treatment, improving outcomes. However, many infant HIV diagnostics can only be performed in laboratory settings. Recombinase polymerase amplification (RPA) is an isothermal amplification technology that can rapidly amplify proviral DNA from multiple subtypes of HIV-1 in under twenty minutes without complex equipment. In this study we added reverse transcription (RT) to RPA to allow detection of both HIV-1 RNA and DNA. We show that this RT-RPA HIV-1 assay has a limit of detection of 10-30 copies of an exact sequence matched DNA or RNA, respectively. In addition, at 100 copies of RNA or DNA, the assay detected 171 of 175 (97.7%) sequence variants that represent all the major subtypes and recombinant forms of HIV-1 Groups M and O. This data suggests that the application of RT-RPA for the combined detection of HIV-1 viral RNA and proviral DNA may prove a highly sensitive tool for rapid and accurate diagnosis of infant HIV.
Collapse
Affiliation(s)
| | - Dara A Lehman
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | | | - Julie Weis
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - Jason Cantera
- PATH, 2201 Westlake Ave Suite 200, Seattle, WA 98121, USA
| | - Mathew Parker
- TwistDx Limited, Minerva Building, Babraham Research Campus, Babraham, Cambridge CB22, UK
| | - Olaf Piepenburg
- TwistDx Limited, Minerva Building, Babraham Research Campus, Babraham, Cambridge CB22, UK
| | - Julie Overbaugh
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | - David S Boyle
- PATH, 2201 Westlake Ave Suite 200, Seattle, WA 98121, USA.
| |
Collapse
|
30
|
High rate of lymphoma among a UK cohort of adolescents with vertically acquired HIV-1 infection transitioning to adult care in the era of antiretroviral therapy. AIDS 2016; 30:153-6. [PMID: 26558727 DOI: 10.1097/qad.0000000000000942] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Among an inner London UK cohort of 147 adolescents transitioning from paediatric into adult care between 2007 and 2015, a new diagnosis of lymphoma was made in five patients; incidence rate = 0.425/100 person-years (95% confidence interval = 0.424-0.426). Previously described risk factors, including low nadir CD4 cell count and ongoing HIV-1 viraemia, appeared to be important. These data suggest that careful surveillance and a low threshold for investigating relevant symptoms continue to be essential for such patients.
Collapse
|
31
|
Milligan C, Richardson BA, John-Stewart G, Nduati R, Overbaugh J. Passively acquired antibody-dependent cellular cytotoxicity (ADCC) activity in HIV-infected infants is associated with reduced mortality. Cell Host Microbe 2016; 17:500-6. [PMID: 25856755 DOI: 10.1016/j.chom.2015.03.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/11/2014] [Accepted: 02/18/2015] [Indexed: 12/11/2022]
Abstract
In addition to direct effects on virus infectivity, antibodies mediate antibody-dependent cellular cytotoxicity (ADCC), the killing of an antibody-coated virus-infected cell by cytotoxic effector cells. Although ADCC has been suggested to protect against HIV, the relationship between HIV-specific ADCC antibodies at the time of HIV exposure and infection outcome in humans remains to be assessed. We evaluated the ADCC activity of passively acquired antibodies in infants born to HIV-infected mothers. ADCC levels were higher in uninfected than infected infants, although not significantly. Increase in ADCC antibody activity in infected infants was associated with reduced mortality risk. Infant ADCC positively correlated with the magnitude of IgG1 binding, and IgG1 levels were associated with survival in infected infants. Infant IgG3-binding antibodies were not associated with infected infant survival. These data suggest a therapeutic benefit of pre-existing HIV-specific ADCC antibodies and support a role for eliciting ADCC-mediating IgG1 in HIV vaccines.
Collapse
Affiliation(s)
- Caitlin Milligan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Barbra A Richardson
- Department of Global Health, University of Washington, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Ruth Nduati
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
32
|
Milligan C, Richardson BA, John-Stewart G, Nduati R, Overbaugh J. FCGR2A and FCGR3A Genotypes in Human Immunodeficiency Virus Mother-to-Child Transmission. Open Forum Infect Dis 2015; 2:ofv149. [PMID: 26613093 PMCID: PMC4653957 DOI: 10.1093/ofid/ofv149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/02/2015] [Indexed: 12/31/2022] Open
Abstract
Background. Fc-mediated effector functions have been suggested to influence human immunodeficiency virus (HIV) acquisition and disease progression. Analyzing the role of host Fc gamma receptor (FcγR) polymorphisms on HIV outcome in mother-to-child transmission (MTCT) will increase our understanding of how host genetics may alter immune responses in prevention, therapy, and disease. This study analyzed the impact of FCGR2A and FCGR3A genotypes on MTCT in a cohort in which Fc-mediated antibody functions are predictive of infant HIV outcome. Methods. Human immunodeficiency virus-positive mothers and their infants from a historical MTCT cohort were genotyped for FCGR2A and FCGR3A. We assessed the impact of these genotypes on transmission and acquisition of HIV and disease progression using χ(2) tests, survival analyses, and logistic regression. Results. Among 379 mother-infant pairs, infant FCGR2A and FCGR3A genotypes were not associated with infant HIV infection or disease progression. Maternal FCGR2A was not associated with transmission, but there was a trend between maternal FCGR3A genotype and transmission (P = .07). When dichotomizing mothers into FCGR3A homozygotes and heterozygotes, heterozygotes had a 64.5% higher risk of transmission compared with homozygotes (P = .02). This risk was most evident in the early breastfeeding window, but a trend was only observed when restricting analyses to breastfeeding mothers (hazards ratio, 1.64; P = .064). Conclusions. Infant FCGR2A and FCGR3A genotypes were not associated with HIV infection or disease progression, and, thus, host FcγR genotype may not significantly impact vaccination or therapeutic regimens that depend on Fc-mediated antibody functions. Maternal FCGR3A genotype may influence early breastfeeding transmission risk, but more studies should be conducted to clarify this association and its mechanism.
Collapse
Affiliation(s)
- Caitlin Milligan
- Division of Human Biology ; Medical Scientist Training Program , University of Washington School of Medicine ; Departments of Global Health
| | - Barbra A Richardson
- Vaccine and Infectious Disease Division , Fred Hutchinson Cancer Research Center ; Departments of Global Health ; Biostatistics
| | - Grace John-Stewart
- Departments of Global Health ; Medicine ; Epidemiology ; Pediatrics , University of Washington , Seattle
| | - Ruth Nduati
- Department of Pediatrics and Child Health , University of Nairobi , Kenya
| | - Julie Overbaugh
- Division of Human Biology ; Medical Scientist Training Program , University of Washington School of Medicine
| |
Collapse
|
33
|
Abstract
More than 75 million people worldwide have been infected with human immunodeficiency virus (HIV), and there are now approximately 37 million individuals living with the infection. Untreated HIV replication causes progressive CD4(+) T cell loss and a wide range of immunological abnormalities, leading to an increased risk of infectious and oncological complications. HIV infection also contributes to cardiovascular disease, bone disease, renal and hepatic dysfunction and several other common morbidities. Antiretroviral drugs are highly effective at inhibiting HIV replication, and for individuals who can access and adhere to these drugs, combination antiretroviral therapy leads to durable (and probably lifelong) suppression of viral replication. Viral suppression enables immune recovery and the near elimination of the risk for developing acquired immune deficiency syndrome (AIDS). Despite effective treatment, HIV-infected individuals have a higher than expected risk of heart, bone, liver, kidney and neurological disease. When used optimally by an infected (or by an uninfected) person, antiretroviral drugs can virtually eliminate the risk of HIV transmission. Despite major advances in prevention sciences, HIV transmission remains common in many vulnerable populations, including men who have sex with men, injection drug users and sex workers. Owing to a lack of widespread HIV testing and the costs and toxicities associated with antiretroviral drugs, the majority of the infected population is not on effective antiretroviral therapy. To reverse the pandemic, improved prevention, treatment and implementation approaches are necessary.
Collapse
Affiliation(s)
- Steven G Deeks
- University of California, San Francisco, Department of Medicine, 995 Potrero Avenue, San Francisco, California 94110, USA
| | - Julie Overbaugh
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andrew Phillips
- Department of Infection and Population Health, University College London, London, UK
| | - Susan Buchbinder
- University of California, San Francisco, Department of Medicine, 995 Potrero Avenue, San Francisco, California 94110, USA.,San Francisco Department of Health, San Francisco, California, USA
| |
Collapse
|
34
|
Chohan BH, Tapia K, Benki-Nugent S, Khasimwa B, Ngayo M, Maleche-Obimbo E, Wamalwa D, Overbaugh J, John-Stewart G. Nevirapine Resistance in Previously Nevirapine-Unexposed HIV-1-Infected Kenyan Infants Initiating Early Antiretroviral Therapy. AIDS Res Hum Retroviruses 2015; 31:783-91. [PMID: 25819584 DOI: 10.1089/aid.2014.0370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nevirapine (NVP) resistance occurs frequently in infants following NVP use in prevention of mother-to-child transmission (PMTCT) regimens. However, among previously NVP-unexposed infants treated with NVP-antiretroviral therapy (ART), the development and impact of NVP resistance have not been well characterized. In a prospective clinical trial providing early ART to HIV-infected infants <5 months of age in Kenya (OPH03 study), we followed NVP-unexposed infants who initiated NVP-ART for 12 months. Viral loads were assessed and resistance determined using a population-based genotypic resistance assay. Of 99 infants screened, 33 had no prior NVP exposure, 22 of whom were initiated on NVP-ART. Among 19 infants with follow-up, seven (37%) infants developed resistance: one at 3 months and six at 6 months after ART initiation. The cumulative probability of NVP resistance was 5.9% at 3 months and 43.5% at 6 months. Baseline HIV RNA levels (p=0.7) and other characteristics were not associated with developing resistance. Post-ART, higher virus levels at visits preceding the detection of resistance were significantly associated with increased detection of resistance (p=0.004). Virus levels after 6 and 12 months of ART were significantly higher in infants with resistance than those without (p=0.007, p=0.030, respectively). Among infants without previous NVP exposure, development of NVP resistance was frequent and was associated with virologic failure during the first year of ART. Earlier development of NVP resistance in infants than in adults initiating NVP-ART may be due to longer viremia following ART or inadequate NVP levels resulting from NVP lead-in dosing. The development of NVP resistance may, in part, explain the superiority of protease inhibitor-based ART in infants.
Collapse
Affiliation(s)
- Bhavna H Chohan
- 1 Department of Medical Microbiology, University of Nairobi , Nairobi, Kenya
- 2 Department of Global Health, University of Washington , Seattle, Washington
- 3 Kenya Medical Research Institute , Nairobi, Kenya
| | - Kenneth Tapia
- 2 Department of Global Health, University of Washington , Seattle, Washington
| | - Sarah Benki-Nugent
- 2 Department of Global Health, University of Washington , Seattle, Washington
| | - Brian Khasimwa
- 4 Department of Pediatrics, University of Nairobi , Nairobi, Kenya
| | - Musa Ngayo
- 3 Kenya Medical Research Institute , Nairobi, Kenya
| | | | - Dalton Wamalwa
- 4 Department of Pediatrics, University of Nairobi , Nairobi, Kenya
| | - Julie Overbaugh
- 5 Fred Hutchinson Cancer Research Center , Seattle, Washington
| | - Grace John-Stewart
- 2 Department of Global Health, University of Washington , Seattle, Washington
- 6 Department of Epidemiology, University of Washington , Seattle, Washington
- 7 Department of Medicine, University of Washington , Seattle, Washington
- 8 Department of Pediatrics, University of Washington , Seattle, Washington
| |
Collapse
|
35
|
Lohman-Payne B, Sandifer T, OhAinle M, Crudder C, Lynch J, Omenda MM, Maroa J, Fowke K, John-Stewart GC, Farquhar C. In-utero infection with HIV-1 associated with suppressed lymphoproliferative responses at birth. Clin Exp Immunol 2014; 178:86-93. [PMID: 24853045 DOI: 10.1111/cei.12386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2014] [Indexed: 11/28/2022] Open
Abstract
In-utero exposure to HIV-1 may affect the immune system of the developing child and may induce HIV-1-specific immune responses, even in the absence of HIV-1 infection. We evaluated lymphoproliferative capacity at birth among 40 HIV-1-uninfected infants born to HIV-1-infected mothers and 10 infants who had acquired HIV-1 in utero. Cord blood mononuclear cells were assayed using [(3) H]-thymidine incorporation for proliferation in response to HIV-1 p55-gag and the control stimuli phytohaemagglutinin (PHA), Staphylococcus enterotoxin B (SEB) and allogeneic cells. In response to HIV-1 p55-gag, eight (20%) HIV-1-exposed, uninfected (EU) infants had a stimulation index (SI) ≥ 2 and three (30%) in-utero HIV-1 infected infants had SI ≥2. The frequency and magnitude of responses to HIV-1 p55-gag were low overall, and did not differ statistically between groups. However, proliferative responses to control stimuli were significantly higher in EU infants than in infants infected in utero, with a median SI in response to PHA of 123 [interquartile range (IQR) 77-231] versus 18 (IQR 4-86) between EU and infected infants, respectively (P < 0·001). Among infected infants, gestational maturity was associated with the strength of HIV-1 p55-gag response (P < 0·001); neither maternal nor infant HIV-1 viral load was associated. In summary, EU and HIV-1-infected infants mounted HIV-1-specific lymphoproliferative responses at similar rates (20-30%), and although global immune function was preserved among EU infants, neonatal immune responses were significantly compromised by HIV-1 infection. Such early lymphoproliferative compromise may, in part, explain rapid progression to AIDS and death among HIV-1-infected infants.
Collapse
Affiliation(s)
- B Lohman-Payne
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya; Department of Medicine, University of Washington, Seattle, Washington, USA; Department of Global Health, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Luzuriaga K, Tabak B, Garber M, Chen YH, Ziemniak C, McManus MM, Murray D, Strain MC, Richman DD, Chun TW, Cunningham CK, Persaud D. HIV type 1 (HIV-1) proviral reservoirs decay continuously under sustained virologic control in HIV-1-infected children who received early treatment. J Infect Dis 2014; 210:1529-38. [PMID: 24850788 PMCID: PMC4215073 DOI: 10.1093/infdis/jiu297] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 04/14/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Early initiation of combination antiretroviral therapy (cART) to human immunodeficiency virus type 1 (HIV-1)-infected infants controls HIV-1 replication and reduces mortality. METHODS Plasma viremia (lower limit of detection, <2 copies/mL), T-cell activation, HIV-1-specific immune responses, and the persistence of cells carrying replication-competent virus were quantified during long-term effective combination antiretroviral therapy (cART) in 4 perinatally HIV-1-infected youth who received treatment early (the ET group) and 4 who received treatment late (the LT group). Decay in peripheral blood mononuclear cell (PBMC) proviral DNA levels was also measured over time in the ET youth. RESULTS Plasma viremia was not detected in any ET youth but was detected in all LT youth (median, 8 copies/mL; P = .03). PBMC proviral load was significantly lower in ET youth (median, 7 copies per million PBMCs) than in LT youth (median, 181 copies; P = .03). Replication-competent virus was recovered from all LT youth but only 1 ET youth. Decay in proviral DNA was noted in all 4 ET youth in association with limited T-cell activation and with absent to minimal HIV-1-specific immune responses. CONCLUSIONS Initiation of early effective cART during infancy significantly limits circulating levels of proviral and replication-competent HIV-1 and promotes continuous decay of viral reservoirs. Continued cART with reduction in HIV-1 reservoirs over time may facilitate HIV-1 eradication strategies.
Collapse
Affiliation(s)
- Katherine Luzuriaga
- Program in Molecular Medicine
- Department of Pediatrics
- Center for Clinical and Translational Science, University of Massachusetts Medical School, Worcester
| | - Barbara Tabak
- Program in Molecular Medicine
- Bioinformatics and Integrative Biology
| | - Manuel Garber
- Program in Molecular Medicine
- Bioinformatics and Integrative Biology
| | - Ya Hui Chen
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore
| | - Carrie Ziemniak
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore
| | | | - Danielle Murray
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Matthew C. Strain
- University of California San Diego, La Jolla
- Veterans Affairs San Diego Healthcare System, California
| | - Douglas D. Richman
- University of California San Diego, La Jolla
- Veterans Affairs San Diego Healthcare System, California
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Coleen K. Cunningham
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Deborah Persaud
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore
| |
Collapse
|
37
|
Njuguna IN, Ambler G, Reilly M, Ondondo B, Kanyugo M, Lohman-Payne B, Gichuhi C, Borthwick N, Black A, Mehedi SR, Sun J, Maleche-Obimbo E, Chohan B, John-Stewart GC, Jaoko W, Hanke T. PedVacc 002: a phase I/II randomized clinical trial of MVA.HIVA vaccine administered to infants born to human immunodeficiency virus type 1-positive mothers in Nairobi. Vaccine 2014; 32:5801-8. [PMID: 25173484 PMCID: PMC4414927 DOI: 10.1016/j.vaccine.2014.08.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/07/2014] [Accepted: 08/15/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND A safe, effective vaccine for breastfeeding infants born to HIV-1-positive mothers could complement antiretroviral therapy (ART) for prevention of mother-to-child transmission of HIV-1. To date, only a few HIV-1 vaccine candidates have been tested in infants. TRIAL DESIGN A phase I/II randomized controlled trial PedVacc 002 was conducted to determine the safety and immunogenicity of a single, low dose of MVA.HIVA vaccine delivered intramuscularly to healthy 20-week-old infants born to HIV-1-positive mothers in Nairobi, Kenya. METHODS Pregnant HIV-1-positive women in the 2nd/3rd trimester of gestation were enrolled, provided with ART and self-selected their infant-feeding modality. Infants received nevirapine and cotrimoxazole prophylaxis. At 20 weeks of age, eligible HIV-1-negative infants were randomized to vaccine versus no-treatment arms and followed to 48 weeks of age for assessments of vaccine safety, HIV-1-specific T-cell responses and antibodies to routine childhood vaccines. RESULTS Between February and November 2010, 182 mothers were screened, 104 were eligible and followed on ART during pregnancy/postpartum, of whom 73 had eligible infants at 20 weeks postpartum. Thirty-six infants were randomized to vaccine and 37 to no treatment. Eighty-four percent of infants breastfed, and retention at 48 weeks was 99%. Adverse events were rare and similar between the two arms. HIV-1-specific T-cell frequencies in interferon-γ ELISPOT assay were transiently higher in the MVA.HIVA arm (p=0.002), but not above the threshold for a positive assay. Protective antibody levels were adequate and similar between arms for all routine childhood vaccines except HBV, where 71% of MVA.HIVA subjects compared to 92% of control subjects were protected (p=0.05). CONCLUSIONS This trial tested for the first time an MVA-vectored candidate HIV-1 vaccine in HIV-1-exposed infants in Africa, demonstrating trial feasibility and vaccine safety, low immunogenicity, and compatibility with routine childhood vaccinations. These results are reassuring for use of the MVA vector in more potent prime-boost regimens.
Collapse
Affiliation(s)
- Irene N Njuguna
- Department of Pediatrics and Child Health, University of Nairobi, PO Box 19676, 00202 Nairobi, Kenya
| | - Gwen Ambler
- Department of Global Health, University of Washington, Seattle, WA 98104, USA
| | - Marie Reilly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, SE-17177 Stockholm, Sweden
| | | | - Mercy Kanyugo
- Department of Pediatrics and Child Health, University of Nairobi, PO Box 19676, 00202 Nairobi, Kenya
| | - Barbara Lohman-Payne
- Department of Pediatrics and Child Health, University of Nairobi, PO Box 19676, 00202 Nairobi, Kenya
| | - Christine Gichuhi
- Department of Clinical Medicine and Therapeutics, University of Nairobi, PO Box 19676, 00202 Nairobi, Kenya
| | | | - Antony Black
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Shams-Rony Mehedi
- Statistics and Data Management Department, Medical Research Council Unit, Fajara, The Gambia
| | - Jiyu Sun
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Elizabeth Maleche-Obimbo
- Department of Pediatrics and Child Health, University of Nairobi, PO Box 19676, 00202 Nairobi, Kenya
| | - Bhavna Chohan
- Department of Pediatrics and Child Health, University of Nairobi, PO Box 19676, 00202 Nairobi, Kenya
| | - Grace C John-Stewart
- Departments of Pediatrics, Medicine, Epidemiology, and Global Health, University of Washington, Seattle, WA 98104, USA
| | - Walter Jaoko
- KAVI-Institute of Clinical Research, University of Nairobi, PO Box 19676, 00202 Nairobi, Kenya
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; The Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
38
|
Qin M, Brummel S, Singh KK, Fenton T, Spector SA. Associations of host genetic variants on CD4⁺ lymphocyte count and plasma HIV-1 RNA in antiretroviral naïve children. Pediatr Infect Dis J 2014; 33:946-52. [PMID: 24797997 PMCID: PMC4216611 DOI: 10.1097/inf.0000000000000330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [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
BACKGROUND CD4 T-lymphocyte (CD4) counts and HIV plasma RNA concentration (RNA) are 2 key HIV disease markers. The complex interplay between virus and host genetics may contribute to differences in viral set point and CD4 status. Determining the effects of host genetic variation on HIV disease markers is often complicated by the use of antiretroviral therapy. In this study, the association between genetic variants and baseline HIV RNA and CD4 counts was examined in a large cohort of antiretroviral naïve children. METHODS Specimens from 1053 HIV-infected children were screened for single nucleotide polymorphisms in 78 regions from 17 genes. Linear regression with a robust variance estimator was used to test the association between genetic markers with HIV RNA and CD4 count, controlling for age, race/ethnicity and study. False discovery rate (FDR) controlling was used to adjust for multiple testing. RESULTS The study population was 60% black, 26% Hispanic and 13% white; median age 2.35 years; 55% female. Baseline median CD4 count was 780/mm; median log10 HIV RNA was 5.17 copies/mL. For analyses of the associations of genetic makers with baseline CD4 count, 6 HLA and 4 additional markers exhibited P < 0.05, but none met the criteria for statistical significance with FDR controlled at 0.05. For baseline HIV RNA, HLA DRB1*15, DRB1*10, B-27/57, B-14, Cw-8, B-57 were statistically significant with FDR controlled at 0.05. CONCLUSIONS These results provide strong evidence that HLA DRB1*15, DRB1*10, B-27/57, B-14, Cw-8, B-57 are associated with HIV RNA and play a role in HIV pathogenesis in infected children.
Collapse
Affiliation(s)
- Min Qin
- Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, MA
| | - Sean Brummel
- Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, MA
| | - Kumud K. Singh
- University of California, San Diego, La Jolla, CA and Rady Children’s Hospital, San Diego, CA
| | - Terry Fenton
- Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, MA
| | - Stephen A. Spector
- University of California, San Diego, La Jolla, CA and Rady Children’s Hospital, San Diego, CA
| |
Collapse
|
39
|
Early development of broadly neutralizing antibodies in HIV-1-infected infants. Nat Med 2014; 20:655-8. [PMID: 24859529 PMCID: PMC4060046 DOI: 10.1038/nm.3565] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/14/2014] [Indexed: 02/07/2023]
Abstract
Eliciting protective neutralizing antibodies (NAbs) against HIV-1 is daunting because of the extensive genetic and antigenic diversity of HIV-1. Moreover, broad and potent responses are uncommon even during persistent infection, with only a subset of adults developing broadly neutralizing antibodies (bNAbs) that recognize variants from different HIV-1 clades1–8. It is not known whether bNAbs can also arise in HIV-1-infected infants, who typically progress to disease faster than adults9, presumably in part due to an immature immune system10. Here, we show that bNAbs develop at least as commonly in infants as in adults. Cross-clade NAb responses were detected in 20/28 infected infants, in some cases, within 1 year of infection. Among infants with the top quartile of responses, neutralization of Tier 2–3 variants from multiple clades was detected at 20 months post-infection. These findings suggest that, even in early life, there is sufficient B-cell functionality to mount bNAbs against HIV-1. Additionally, the relatively early appearance of bNAbs in infants may provide a unique setting for understanding the pathways of B-cell maturation leading to bNAbs.
Collapse
|
40
|
Meriki HD, Tufon KA, Afegenwi MH, Nyindem BA, Atanga PN, Anong DN, Cho-Ngwa F, Nkuo-Akenji T. Immuno-haematologic and virologic responses and predictors of virologic failure in HIV-1 infected adults on first-line antiretroviral therapy in Cameroon. Infect Dis Poverty 2014; 3:5. [PMID: 24479873 PMCID: PMC3922096 DOI: 10.1186/2049-9957-3-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 01/27/2014] [Indexed: 11/10/2022] Open
Abstract
Background Contemporary data on the immunologic, haematologic and virologic responses and predictors of virologic failure after initiation of free antiretroviral treatment in Cameroon are needed to evaluate the current treatment-monitoring algorithm and to complement efforts to scale-up and improve on the management of HIV infections. Methods This was a cross-sectional study conducted between October 2010 and June 2012. A total of 951 participants aged 18–74 years were recruited from selected approved HIV treatment centres of the Northwest and Southwest regions. This comprised 247 males and 704 females. Demographic, self-reported risk behaviours and socioeconomic data were obtained using a structured questionnaire. Full blood and CD4 + T-cell counts were done using standard automated techniques. Determination of viral load (VL) was done using Abbott RealTime HIV-1 m2000™ system. Data was analysed using SPSS version 17. The statistical significance level was P < 0.05. Results The median duration of antiretroviral therapy (ART) was 24 months. The population mean CD4 + T-cell count was 255.3 cells/μL [95% CI, 236.8 – 273.9]. Overall, 45.9%, 43.8% and 10.2% of the participants had CD4 + T-cell counts of < 200 cells/μL, 200–499 cells/μL and > 500 cells/μL respectively. Anaemia was present in 26.2% of the participants with 62.3%, 25.7% and 12% described as mild, moderate and severe anaemia respectively. Virologic failure occurred in 23.2% of the participants with 12.3% having VL > 10,000 RNA copies/mL. Meanwhile 76.8% of patients attained adequate viral suppression with 40.8% having undetectable viral load. The age group 18–29 years (p = 0.024), co-infection with tuberculosis (p = 0.014), anaemia (p = 0.028) and distance from the treatment centre (p = 0.011) independently predicted virologic failure. Conclusion The majority of the participants achieved adequate viral suppression after ≥ 6 months of ART. Despite these favourable immuno-haematologic and virologic outcomes, the National AIDS Control Program should step-up efforts to improve on antiretroviral drug distribution, as well as proper assessment and management of anaemia, foster early diagnosis and treatment of tuberculosis and enhance treatment adherence counselling especially in younger patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Theresa Nkuo-Akenji
- Department of Microbiology and Parasitology, University of Buea, P,O, Box 63, Buea, Cameroon.
| |
Collapse
|
41
|
Afolabi MO, Ndure J, Drammeh A, Darboe F, Mehedi SR, Rowland-Jones SL, Borthwick N, Black A, Ambler G, John-Stewart GC, Reilly M, Hanke T, Flanagan KL. A phase I randomized clinical trial of candidate human immunodeficiency virus type 1 vaccine MVA.HIVA administered to Gambian infants. PLoS One 2013; 8:e78289. [PMID: 24205185 PMCID: PMC3813444 DOI: 10.1371/journal.pone.0078289] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 09/07/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND A vaccine to decrease transmission of human immunodeficiency virus type 1 (HIV-1) during breast-feeding would complement efforts to eliminate infant HIV-1 infection by antiretroviral therapy. Relative to adults, infants have distinct immune development, potentially high-risk of transmission when exposed to HIV-1 and rapid progression to AIDS when infected. To date, there have been only three published HIV-1 vaccine trials in infants. TRIAL DESIGN We conducted a randomized phase I clinical trial PedVacc 001 assessing the feasibility, safety and immunogenicity of a single dose of candidate vaccine MVA.HIVA administered intramuscularly to 20-week-old infants born to HIV-1-negative mothers in The Gambia. METHODS Infants were followed to 9 months of age with assessment of safety, immunogenicity and interference with Expanded Program on Immunization (EPI) vaccines. The trial is the first stage of developing more complex prime-boost vaccination strategies against breast milk transmission of HIV-1. RESULTS From March to October 2010, 48 infants (24 vaccine and 24 no-treatment) were enrolled with 100% retention. The MVA.HIVA vaccine was safe with no difference in adverse events between vaccinees and untreated infants. Two vaccine recipients (9%) and no controls had positive ex vivo interferon-γ ELISPOT assay responses. Antibody levels elicited to the EPI vaccines, which included diphtheria, tetanus, whole-cell pertussis, hepatitis B virus, Haemophilus influenzae type b and oral poliovirus, reached protective levels for the vast majority and were similar between the two arms. CONCLUSIONS A single low-dose of MVA.HIVA administered to 20-week-old infants in The Gambia was found to be safe and without interference with the induction of protective antibody levels by EPI vaccines, but did not alone induce sufficient HIV-1-specific responses. These data support the use of MVA carrying other transgenes as a boosting vector within more complex prime-boost vaccine strategies against transmission of HIV-1 and/or other infections in this age group. TRIAL REGISTRATION ClinicalTrials.gov NCT00982579. The Pan African Clinical Trials Registry PACTR2008120000904116.
Collapse
Affiliation(s)
| | - Jorjoh Ndure
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Abdoulie Drammeh
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Fatoumatta Darboe
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| | - Shams-Rony Mehedi
- Statistics and Data Management Department, Medical Research Council Unit, Fajara, The Gambia
| | | | - Nicola Borthwick
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Antony Black
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Gwen Ambler
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Grace C. John-Stewart
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
| | - Marie Reilly
- Department of Medical Epidemiology and Biostatistics, Stockholm, Sweden
| | - Tomáš Hanke
- Departments of Biostatistics, Medicine, and Epidemiology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Katie L. Flanagan
- Vaccinology Theme, Medical Research Council Unit, Fajara, The Gambia
| |
Collapse
|
42
|
Shirreff G, Alizon S, Cori A, Günthard HF, Laeyendecker O, van Sighem A, Bezemer D, Fraser C. How effectively can HIV phylogenies be used to measure heritability? EVOLUTION MEDICINE AND PUBLIC HEALTH 2013; 2013:209-24. [PMID: 24481201 PMCID: PMC3850537 DOI: 10.1093/emph/eot019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background and objectives: The severity of HIV-1 infection, measured by set-point viral load (SPVL), is highly variable between individuals. Its heritability between infections quantifies the control the pathogen genotype has over disease severity. Heritability estimates vary widely between studies, but differences in methods make comparison difficult. Phylogenetic comparative analysis offers measures of phylogenetic signal, but it is unclear how to interpret them in terms of the fraction of variance in SPVL controlled by the virus genotype. Methodology: We present computational methods which link statistics summarizing phylogenetic signal to heritability, h2 in order to test for and quantify it. We re-analyse data from Switzerland and Uganda, and apply it to new data from the Netherlands. We systematically compare established and new (e.g. phylogenetic pairs, PP) phylogenetic signal statistics. Results: Heritability estimates varied by method and dataset. Several methods were consistently able to detect simulated heritability above , but none below. Pagel’s λ was the most robust and sensitive. The PP method found no heritability in the Netherlands data, whereas Pagel’s λ found significant heritability only in a narrow subdivision (P =0.038). Heritability was estimated at h2=0.52 (95% confidence interval 0.00–0.63). Conclusions and implications: This standardized measure, h2, allows comparability of heritability between cohorts. We confirm high heritability in Swiss data, but neither in Ugandan data nor in the Netherlands, where it is barely significant or undetectable. Existing phylogenetic methods are ill-suited for detecting heritability below , which may nonetheless be biologically important.
Collapse
Affiliation(s)
- George Shirreff
- Medical Research Council Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College, London, UK; Institute for Integrative Biology, ETH Zürich, Zürich, Switzerland; Lab MIVEGEC UMR CNRS 5290, IRD 224, UM1, UM2, Montpellier, France; Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, University of Zürich, Zürich, Switzerland; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, MD, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; and Stichting HIV Monitoring, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Patel RR, Patel S, Clarke E, Khan AW, Doshi B, Radcliffe KW. Guidance and practice on frequency of HIV and sexually transmitted infection testing in men who have sex with men - what is the European situation? Int J STD AIDS 2013; 25:213-8. [PMID: 24216033 DOI: 10.1177/0956462413497700] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Men who have sex with men (MSM) are at particular risk for HIV/sexually transmitted infections (STI). To investigate the European guidance used for MSM STI and HIV screening, risk level profiling and how this translated to practice, we conducted a questionnaire survey of leading physicians in the European branch of the International Union against Sexually Transmitted Infections (IUSTI). We identified that most European countries have limited guidance on screening intervals for MSM. Where risk profiling is advised, it is often left to clinicians to weight different behaviours and decide on screening frequency. Our results suggest that European MSM STI and HIV testing guidelines be developed with clear and specific recommendations around screening intervals and risk profiling. These guidelines will be particularly helpful due to rapidly evolving models of sexual healthcare, and the emergence of new providers who may benefit from guidelines that require less interpretation.
Collapse
|
44
|
Precise determination of time to reach viral load set point after acute HIV-1 infection. J Acquir Immune Defic Syndr 2013; 61:448-54. [PMID: 23143525 DOI: 10.1097/qai.0b013e31827146e0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The HIV viral load set point has long been used as a prognostic marker of disease progression and more recently as an end-point parameter in HIV vaccine clinical trials. The definition of set point, however, is variable. Moreover, the earliest time at which the set point is reached after the onset of infection has never been clearly defined. METHODS In this study, we obtained sequential plasma viral load data from 60 acutely HIV-infected Chinese patients among a cohort of men who have sex with men, mathematically determined viral load set point levels, and estimated time to attain set point after infection. We also compared the results derived from our models and that obtained from an empirical method. RESULTS With novel uncomplicated mathematic model, we discovered that set points may vary from 21 to 119 days dependent on the patients' initial viral load trajectory. The viral load set points were 4.28 ± 0.86 and 4.25 ± 0.87 log10 copies per milliliter (P = 0.08), respectively, as determined by our model and an empirical method, suggesting an excellent agreement between the old and new methods. CONCLUSIONS We provide a novel method to estimate viral load set point at the very early stage of HIV infection. Application of this model can accurately and reliably determine the set point, thus providing a new tool for physicians to better monitor early intervention strategies in acutely infected patients and scientists to rationally design preventative vaccine studies.
Collapse
|
45
|
King CC, Ellington SR, Kourtis AP. The role of co-infections in mother-to-child transmission of HIV. Curr HIV Res 2013; 11:10-23. [PMID: 23305198 PMCID: PMC4411038 DOI: 10.2174/1570162x11311010003] [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] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/11/2012] [Accepted: 12/14/2012] [Indexed: 01/27/2023]
Abstract
In HIV-infected women, co-infections that target the placenta, fetal membranes, genital tract, and breast tissue, as well as systemic maternal and infant infections, have been shown to increase the risk for mother-to-child transmission of HIV (MTCT). Active co-infection stimulates the release of cytokines and inflammatory agents that enhance HIV replication locally or systemically and increase tissue permeability, which weakens natural defenses to MTCT. Many maternal or infant co-infections can affect MTCT of HIV, and particular ones, such as genital tract infection with herpes simplex virus, or systemic infections such as hepatitis B, can have substantial epidemiologic impact on MTCT. Screening and treatment for co-infections that can make infants susceptible to MTCT in utero, peripartum, or postpartum can help reduce the incidence of HIV infection among infants and improve the health of mothers and infants worldwide.
Collapse
Affiliation(s)
- Caroline C King
- Division of Reproductive Health, NCCDPHP, Centers for Disease Control and Prevention, 4770 Buford Highway, NE, MS-K34, Atlanta, GA 30341, USA.
| | | | | |
Collapse
|
46
|
Slyker JA, Lohman-Payne B, John-Stewart GC, Dong T, Mbori-Ngacha D, Tapia K, Atzberger A, Taylor S, Rowland-Jones SL, Blish CA. The impact of HIV-1 infection and exposure on natural killer (NK) cell phenotype in Kenyan infants during the first year of life. Front Immunol 2012; 3:399. [PMID: 23293640 PMCID: PMC3533178 DOI: 10.3389/fimmu.2012.00399] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 12/10/2012] [Indexed: 01/30/2023] Open
Abstract
Natural killer (NK) cells play an important role in the containment of HIV replication during primary infection, though their functions are impaired during chronic HIV infection. Infants experience more rapid HIV disease progression than adults, but contributions of infant NK cells to containing HIV infection are unknown. The aim of this study was to determine the impact of HIV infection on infant NK cell phenotype by evaluating samples and data from a cohort study of women and their infants, conducted in Nairobi, Kenya between 1999 and 2003. The percentage and phenotype of NK cells was evaluated longitudinally by multi-parameter flow cytometry over the first year of life in HIV-infected (HIV+, = 16), HIV-exposed uninfected (HIV-EU, n = 6), and healthy unexposed controls (HIV–, n = 4). At birth, NK subset distributions based on expression of CD56 and CD16 did not differ between HIV+, HIV-EU, or HIV– infants. However, HIV infection was associated with a subsequent decline in NK cells as a percentage of total lymphocytes (p < 0.001), and an expanding proportion of CD56-CD16+ NK cells (p < 0.001). Activated CD38brightCD69+ NK cells were more frequent in the HIV+ infants, followed by HIV-EU and HIV- infants, in both CD56dim (p = 0.005) and CD56bright compartments (p = 0.03). HIV infection and exposure was also associated with a significant decline in the percentage of perforin-expressing NK cells in the CD56dim compartment over the first year of life, with HIV+ infants losing approximately 2.5% (p < 0.001) and HIV-EU infants losing 3.0% (p = 0.01) of perforin+ cells per month. Thus, infant HIV infection is associated with alterations in NK cell subsets, activation, and cytolytic potential that could contribute to their poor control over HIV infection. Furthermore, exposure to HIV infection in infants who escaped infection is also associated with alterations in NK cells that may contribute to the reduced ability to fight infections that is observed in HIV-EU infants.
Collapse
Affiliation(s)
- Jennifer A Slyker
- Department of Global Health, University of Washington Seattle, WA, USA ; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University Oxford, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Laser cutting eliminates nucleic acid cross-contamination in dried-blood-spot processing. J Clin Microbiol 2012; 50:4128-30. [PMID: 23052309 DOI: 10.1128/jcm.02549-12] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dried blood spots (DBS) are useful for molecular assays but are prone to false positives from cross-contamination. In our malaria DBS assay, cross-contamination was encountered despite cleaning techniques suitable for HIV-1. We therefore developed a contact-free laser cutting system that effectively eliminated cross-contamination during DBS processing.
Collapse
|
48
|
Low-frequency nevirapine resistance at multiple sites may predict treatment failure in infants on nevirapine-based treatment. J Acquir Immune Defic Syndr 2012; 60:225-33. [PMID: 22395670 DOI: 10.1097/qai.0b013e3182515730] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Resistance commonly arises in infants exposed to single-dose nevirapine (sdNVP) for prevention of mother to child transmission. Although K103N and Y181C are common following sdNVP, multiple other mutations also confer NVP resistance. It remains unclear whether specific NVP resistance mutations or combinations of mutations predict virologic failure in infants when present at low frequencies before NVP-based treatment. METHODS Twenty sdNVP-exposed infants who were subsequently treated with NVP-based highly active antiretroviral therapy were examined. Pretreatment plasma samples were tested for the presence of NVP resistance mutations by allele-specific polymerase chain reaction for K103N and Y181C and ultradeep pyrosequencing (UDPS) for all primary NVP mutations. Viral levels were determined every 3 months for up to 24 months on NVP-highly active antiretroviral therapy. Cox proportional hazard models were used to determine correlates of viral failure. RESULTS The NVP resistance mutations K103N or Y181C were detected in pretreatment plasma samples in 6 infants by allele-specific polymerase chain reaction. NVP resistance at these or other sites was detectable by UDPS in 10 of 20 infants tested. Virologic failure occurred in 50% of infants with any NVP resistance mutations detected, whereas only 20% of infants without resistance experienced viral failure, but the difference was not significant (P = 0.19). An increase in the number of NVP resistance mutations detectable by UDPS in an infant was significantly associated with an increased risk of virologic failure [HR = 1.79 (95% confidence interval: 1.07 to 2.99), P = 0.027]. CONCLUSIONS Low frequencies of multiple NVP resistance mutations, in addition to K103N and Y181C, present in infants before NVP-based treatment may predict treatment outcome.
Collapse
|
49
|
Abstract
BACKGROUND Reducing rates of partner change and increasing condom usage among gay men are obvious targets for potentially reducing syphilis transmission among gay men. METHODS We developed an agent-based stochastic model to examine syphilis transmission among a population of gay men, representative of gay men in Australia. This model was used to explore the potential impact of changes in sexual behavior over 1 month, 3 month, and indefinite time frames on syphilis epidemics. RESULTS Simulations of interventions showed that short-term reductions in rates of partner change and increased condom use would have negligible impact on the long-term trends of syphilis epidemics. If no interventions are introduced, then the model forecasts that the syphilis prevalence in the population could continue to rise, with an increase of 80% in the number of men infected with syphilis during the next decade. However, if changes in sexual behavior are maintained in the long-term, then syphilis epidemics can be mitigated. If condom use is sustained at 80% in partnerships that are HIV discordant or of unknown status, then the prevalence of syphilis is estimated to decrease by 9% over 10 years. Similarly, if partner acquisition rates decrease by 25%, then there will be a 22% reduction in syphilis prevalence. CONCLUSIONS Interventions promoting partner reduction or increased condom use would be ineffective in the short-term, and would have limited prospects for success in the long-term unless very large changes in behavior are sustained. Complementary social research indicates that such long-term changes in behavior are unlikely to be adopted, and therefore other intervention strategies need to be developed to reduce syphilis among gay men.
Collapse
|
50
|
Lohman-Payne B, Slyker J, Rowland-Jones SL. Immune approaches for the prevention of breast milk transmission of HIV-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 743:185-95. [PMID: 22454350 DOI: 10.1007/978-1-4614-2251-8_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Barbara Lohman-Payne
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya, 00202.
| | | | | |
Collapse
|