1
|
Wedderburn CJ, Musiime GM, McHenry MS. Care of the Child Perinatally Exposed to Human Immunodeficiency Virus. Clin Perinatol 2024; 51:881-894. [PMID: 39487026 DOI: 10.1016/j.clp.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
Substantial progress in preventing vertical human immunodeficiency virus (HIV) transmission has led to a dramatic decline in new pediatric HIV infections. Alongside this success, a growing population of children who are HIV-exposed but uninfected face unique health challenges due to a variety of risk factors. Recommendations for healthcare providers caring for this population include optimizing and integrating general and HIV-related care for both mother and child through comprehensive care packages. Further research and multidisciplinary approaches are needed to address the long-term health implications for this vulnerable population.
Collapse
Affiliation(s)
- Catherine J Wedderburn
- Department of Paediatrics and Child Health and Neuroscience Institute, University of Cape Town, Cape Town, South Africa.
| | - Grace M Musiime
- Department of Paediatrics, AIC Kijabe Hospital, Kiambu, Kenya
| | - Megan S McHenry
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, 705 Riley Hospital Drive, Room 5900, Indianapolis, IN 46208, USA. https://twitter.com/MeganS_McHenry
| |
Collapse
|
2
|
Tshiambara P, Hoffman M, Legodi H, Balakrishna Y, Feucht U. Dietary intake and growth of HIV exposed and unexposed 6-12 months old infants in South Africa. MATERNAL & CHILD NUTRITION 2024:e13740. [PMID: 39400932 DOI: 10.1111/mcn.13740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/23/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
Factors affecting the growth of HIV-exposed-uninfected (HEU) children are multi-factorial, with limited information available on the dietary intake from 6 months. This study compared the dietary intake, micronutrient composition of breastmilk, and growth of HEU and HIV-unexposed-uninfected (HUU) infants aged 6 and 12 months in an urban setting. A repeated cross-sectional study used structured questionnaires to collect socio-demographic, dietary intake, food group data, and anthropometric measurements in the Siyakhula study. The HEU (48%) and HUU (52%) infants were included (total n = 181). At 6 months, HEU infants had lower weight-for-age z-scores (WAZ) (-0.6 ± 1.1 vs. 0.1 ± 1.2; p < 0.001), length-for-age z-scores (-0.8 ± 1.4 vs. -0.1 ± 1.2; p < 0.001), and mid-upper-arm circumference-for-age z-scores (MUACAZ) (0.5 ± 1.1 vs. 1.0 ± 0.9; p < 0.001) than HUU infants. At 12 months, HEU infants had lower WAZ, MUACAZ, and weight-for-length z-scores compared to HUU infants (p < 0.05). Stunting was found at 6 (15%) and 12 (12%) months in HEU infants. The micronutrient composition of breastmilk fed to both groups was similar. Breastfeeding rates were lower in HEU than in HUU infants at 6 (49% vs. 64%; p = 0.005) and 12 (24% vs. 46%; p = 0.002) months. Less than 3% of HEU and HUU infants achieved minimal dietary diversity scores at 12 months. Dietary intake of fat was similar in all breastfed infants, but iron and vitamin B12 were higher in non-breastfed HEU infants at 12 months. HEU infants had lower breastfeeding rates than HUU infants. A lack of dietary diversity was found in all infants. Nutrition education and counselling in the complementary feeding phase are essential for optimal growth.
Collapse
Affiliation(s)
- Phumudzo Tshiambara
- Department of Human Nutrition, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Consumer and Food Sciences, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
- Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Cape Town, South Africa
| | - Marinel Hoffman
- Department of Consumer and Food Sciences, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
- Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Cape Town, South Africa
| | - Heather Legodi
- Department of Human Nutrition and Dietetics, Sefako Makgatho Health Sciences University, Ga-Rankuwa, South Africa
| | - Yusentha Balakrishna
- Biostatistics Research Unit, South African Medical Research Council, Durban, South Africa
| | - Ute Feucht
- Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Pretoria, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Cape Town, South Africa
- Department of Paediatrics, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
3
|
Abrams EJ, Jao J, Madlala HP, Zerbe A, Catalano P, Gerschenson M, Goedecke JH, Gomba Y, Josefson J, Kurland IJ, Legbedze J, McComsey GA, Matyesini S, Mukonda E, Robinson D, Myer L. An observational cohort study to investigate the impact of dolutegravir in pregnancy and its obesogenic effects on the metabolic health of women living with HIV and their children: Study protocol. PLoS One 2024; 19:e0307296. [PMID: 39159183 PMCID: PMC11332920 DOI: 10.1371/journal.pone.0307296] [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: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Dolutegravir (DTG)-based antiretroviral therapy is the World Health Organization's preferred first-line regimen for all persons with HIV, including pregnant women. While DTG has been implicated as an obesogen associated with greater weight gain compared to other antiretrovirals, there is a paucity of data in pregnant women and their children. The Obesogenic oRigins of maternal and Child metabolic health Involving Dolutegravir (ORCHID) study is investigating associations between DTG, weight gain, and metabolic outcomes in the context of HIV. MATERIALS & METHODS ORCHID is a prospective observational study taking place in Cape Town, South Africa (NCT04991402). A total of 1920 pregnant women with and without HIV infection are being followed from ≤18 weeks gestational age to 24 months postpartum with their children. Participants attend eleven study visits: 3 antenatal, delivery, and 7 postnatal visits. Several embedded sub-studies address specific scientific aims. Primary outcome measurements in mothers include anthropometry, blood pressure, body composition, dysglycemia, insulin resistance (IR), and dyslipidemia. Other maternal measures include demographics, resting energy expenditure, viral load, physical activity, dietary intake, hepatic steatosis, and repository specimens. Sub-study measurements include markers of adipose inflammation, gut integrity, and satiety/hunger, subcutaneous adipose tissue morphology and mitochondrial function, and metabolomics. Primary outcome measurements in children include anthropometry, adipose tissue mass, dysglycemia, IR, and dyslipidemia. Other variables include fetal growth, birth outcomes, medical/breastfeeding history, caloric intake, neurodevelopment, and repository specimens. Sub-study measurements include metabolites/lipid subspecies in umbilical cord blood, as well as breast milk composition and DTG exposure. DISCUSSION ORCHID will play a pivotal role in defining obesogenic mechanisms and clinical consequences of DTG use in pregnancy in women with HIV and their children. It will provide insights into metabolic disease risk reduction in the context of HIV/DTG, identify intervention targets, and inform public health approaches to diminish chronic metabolic co-morbidities for women and children.
Collapse
Affiliation(s)
- Elaine J. Abrams
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States of America
- Department of Pediatrics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, United States of America
| | - Jennifer Jao
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Hlengiwe P. Madlala
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Allison Zerbe
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY, United States of America
| | - Patrick Catalano
- Maternal Infant Research Institute, Obstetrics and Gynecology Research, Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University School of Medicine, Tufts University, Boston, MA, United States of America
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Julia H. Goedecke
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
- Health through Physical Activity, Lifestyle and Sport Research Centre (HPALS), Division of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Yolanda Gomba
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Jami Josefson
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Irwin J. Kurland
- Department of Medicine, Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Justine Legbedze
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Grace A. McComsey
- University Hospitals Health System, Cleveland, OH, United States of America
- Department of Medicine and Pediatrics of Case Western Reserve University, Cleveland, OH, United States of America
| | - Sandisiwe Matyesini
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Elton Mukonda
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| | - Daniel Robinson
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Western Cape, Cape Town, South Africa
| |
Collapse
|
4
|
Wand H, Moodley J, Reddy T, Naidoo S. Geospatial correlations and variations in child mortality and stunting in South Africa: Evaluating distal vs structural determinants. Spat Spatiotemporal Epidemiol 2024; 50:100653. [PMID: 39181600 DOI: 10.1016/j.sste.2024.100653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 03/10/2024] [Accepted: 04/22/2024] [Indexed: 08/27/2024]
Abstract
South Africa has one of the highest child mortality and stunting rates in the world. Flexible geoadditive models were used to investigate the geospatial variations in child mortality and stunting in South Africa. We used consecutive rounds of national surveys (2008-2017). The child mortality declined from 31 % to 24 % over time. Lack of medical insurance, black ethnicity, low-socioeconomic conditions, and poor housing conditions were identified as the most significant correlates of child mortality. The model predicted degrees of freedom which was estimated as 19.55 (p < 0.001), provided compelling evidence for sub-geographical level variations in child mortality which ranged from 6 % to 35 % across the country. Population level impact of the distal characteristics on child mortality and stunting exceeded that of other risk factors. Geospatial analysis can help in monitoring trends in child mortality over time and in evaluating the impact of health interventions.
Collapse
Affiliation(s)
- Handan Wand
- Kirby Institute, University of New South Wales, Kensington, 2052, New South Wales, Australia.
| | | | - Tarylee Reddy
- Biostatistics Unit, South African Medical Research Council, Durban, Kwazulu-Natal, South Africa
| | | |
Collapse
|
5
|
Barrios-Tascon A, Strehlau R, Patel F, Burke M, Shiau S, Shen Y, Arpadi SM, Abrams EJ, Tiemessen CT, Kuhn L. Growth Trajectories Over the First Year of Life Among Early-Treated Infants with Human Immunodeficiency Virus and Infants Who are Human Immunodeficiency Virus-Exposed Uninfected. J Pediatr 2024; 270:114018. [PMID: 38508485 PMCID: PMC11176027 DOI: 10.1016/j.jpeds.2024.114018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/10/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024]
Abstract
OBJECTIVE To investigate the role of early antiretroviral therapy (ART) on growth trajectories of infants with human immunodeficiency virus (IHIV) in the first year of life. STUDY DESIGN As part of a clinical trial of early ART in Johannesburg, South Africa (2015-2018), 116 IHIV diagnosed within 48 hours of birth were started on ART as soon as possible, and 80 uninfected infants born to mothers living with HIV (IHEU) were enrolled. Both groups were followed prospectively from birth through 48 weeks and growth parameters collected. The groups were compared and risk factors for poor growth investigated, in the full cohort and among IHIV separately. RESULTS IHIV had lower mean weight-for-age Z-scores (WAZ) than IHEU at 4 and 8 weeks (-1.17 [SE:0.14] vs -0.72 [0.14], P = .035 and -1.23 [0.15] vs -0.67 [0.14], P = .012). Although there was some closing of the gap over time, means remained lower in IHIV through 48 weeks. In length-for-age Z-scores (LAZ), differences widened over time and IHIV had lower Z-scores by 48 weeks (-1.41 [0.15] vs -0.80 [0.18], P = .011). Deficits in WAZ and LAZ in IHIV vs IHEU were most marked among girls. IHIV with pre-ART viral load ≥1000 copies/ml had significantly lower weight-for-length and mid-upper arm circumference Z-scores across all time points through 48 weeks. CONCLUSIONS IHIV on early ART had deficits in WAZ over the first 8 weeks of life and lower LAZ at 48 weeks than IHEU. Among IHIV, higher pre-ART viral load was associated with worse anthropometric indicators through 48 weeks.
Collapse
Affiliation(s)
- Ana Barrios-Tascon
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY.
| | - Renate Strehlau
- VIDA Nkanyezi Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Faeezah Patel
- Wits RHI, Shandukani Research Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Megan Burke
- VIDA Nkanyezi Research Unit, Rahima Moosa Mother and Child Hospital, Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephanie Shiau
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ
| | - Yanhan Shen
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Stephen M Arpadi
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY; Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| | - Elaine J Abrams
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY; ICAP, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| | - Caroline T Tiemessen
- Centre for HIV and STIs, National Institutes for Communicable Diseases, A Division of the National Health Laboratory Service, and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY; Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
6
|
Njuguna IN, King'e M, Moraa H, Kumar M, Benki-Nugent S, Wagner AD, McGrath CJ, Dorsey S, Ndegwa S, Onyango A, Wamalwa D, John-Stewart G. Cohort profile: longitudinal and population comparison of children who are HIV-exposed uninfected and children who are HIV unexposed in Kenya (HOPE study). BMJ Open 2024; 14:e081975. [PMID: 38844397 PMCID: PMC11163661 DOI: 10.1136/bmjopen-2023-081975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/22/2024] [Indexed: 06/12/2024] Open
Abstract
PURPOSE Globally, the number of children/adolescents exposed to HIV but uninfected (HIV-exposed uninfected, HEU) is growing. The HEU outcomes: population-evaluation and screening strategies study was designed to provide population-level evidence of the impact of HIV and recent antiretroviral therapy regimen exposure on neurodevelopmental, hearing and mental health outcomes from infancy to adolescence. PARTICIPANTS The study includes a prospective mother-infant cohort and cross-sectional child/youth-caregiver cohorts conducted in Kenya.Between 2021 and 2022, the study enrolled 2000 mother-infant pairs (1000 HEU and 1000 HIV-unexposed uninfected (HUU)) for longitudinal follow-up. Infants were eligible if they were aged 4-10 weeks and healthy. Mothers were eligible if their HIV status was known and were ≥18 years. Study visits are 6 monthly until the child reaches age 3 years.Cross-sectional cohorts spanning ages 3-18 years started enrolment in 2022. Target enrolment is 4400 children/youth (4000 HEU and 400 HUU). Children and youth are eligible if they are HIV negative, maternal HIV status and timing of diagnosis is known, and caregivers are ≥18 years.Data on infant/child/youth growth, neurodevelopment, mental health, morbidity and hearing are collected at enrolment using standardised tools. Dry blood spots samples are collected for telomere length assessment at baseline and yearly for the longitudinal cohort. Growth z-scores, neurodevelopmental scores, telomere length and prevalence of developmental and hearing problems will be compared between HEU/HUU populations. FINDINGS TO DATE Full cohort enrolment for the longitudinal cohort is complete and participants are in follow-up. At 1 year of age, comparing HEU to HUU neurodevelopment using the Malawi developmental assessment tool, we found that HEU infants had higher language scores and comparable scores in fine motor, gross motor and social scores. The cross-sectional cohort has enrolled over 2000 participants and recruitment is ongoing. FUTURE PLANS Longitudinal cohort follow-up and enrolment to the cross-sectional study will be completed in June 2024.
Collapse
Affiliation(s)
- Irene N Njuguna
- Department of Medical Research, Kenyatta National Hospital, Nairobi, Kenya
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Maureen King'e
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Helen Moraa
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Manasi Kumar
- Grossman School of Medicine, New York University, New York City, New York, USA
- Department of Psychiatry, University of Nairobi, Nairobi, Kenya
| | - Sarah Benki-Nugent
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Anjuli Dawn Wagner
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Christine J McGrath
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Shannon Dorsey
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Psychiatry, University of Washington, Seattle, Washington, USA
| | - Serah Ndegwa
- Departnment of Surgery, University of Nairobi, Nairobi, Kenya
| | - Alvin Onyango
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Dalton Wamalwa
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Deaprtment of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Johnson M, Lazarus SK, Bennett AE, Tovar-Salazar A, Robertson CE, Kofonow JM, Li S, McCollister B, Nunes MC, Madhi SA, Frank DN, Weinberg A. Gut Microbiota and Other Factors Associated With Increased Regulatory T Cells in Hiv-exposed Uninfected Infants. RESEARCH SQUARE 2024:rs.3.rs-3909424. [PMID: 38352510 PMCID: PMC10862973 DOI: 10.21203/rs.3.rs-3909424/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
HIV-exposed uninfected infants (HEU) have higher infectious morbidity than HIV-unexposed infants (HUU). HEU have multiple immune defects of unknown origin. We hypothesized that HEU have higher regulatory T cells (Treg) than HUU, which may dampen their immune defenses against pathogens. We compared 25 Treg subsets between HEU and HUU and sought the factors that may affect Treg frequencies. At birth, 3 Treg subsets, including CD4 + FOXP3 + and CD4 + FOXP3 + CD25+, had higher frequencies in 123 HEU than 117 HUU and 3 subsets were higher in HUU. At 28 and 62 weeks of life, 5 Treg subsets were higher in HEU, and none were higher in HUU. The frequencies of the discrepant Treg subsets correlated at birth with differential abundances of bacterial taxas in maternal gut microbiome and at subsequent visits in infant gut microbiomes. In vitro, bacterial taxa most abundant in HEU expanded Treg subsets with higher frequencies in HEU, recapitulating the in vivo observations. Other factors that correlated with increased Treg were low maternal CD4 + T cells in HEU at birth and male sex in HUU at 28 weeks. We conclude that maternal and infant gut dysbiosis are central to the Treg increase in HEU and may be targeted by mitigating interventions.
Collapse
|
8
|
Matjuda EN, Engwa GA, Mungamba MM, Sewani-Rusike CR, Goswami N, Nkeh-Chungag BN. Cardio-Metabolic Health of Offspring Exposed in Utero to Human Immuno-Deficiency Virus and Anti-Retroviral Treatment: A Systematic Review. BIOLOGY 2024; 13:32. [PMID: 38248463 PMCID: PMC10813696 DOI: 10.3390/biology13010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
BACKGROUND Antiretroviral treatment (ART) use during pregnancy continues to rise as it is known to decrease the likelihood of HIV transmission from mother to child. However, it is still unknown whether foetal exposure to (ART) may affect the foetal environment, predisposing the offspring to cardiometabolic risk. Therefore, the aim of this study was to systematically review the cardio-metabolic effects of in utero exposure to HIV/ART on offspring. METHODS We carried out a systematic review and obtained literature from the Google scholar, PubMed, ProQuest, Web of Science, and Scopus databases. Two independent reviewers evaluated the titles, abstracts, and full-length English contents. Data from the eligible studies were included. RESULTS The search yielded 7596 records. After assessing all of these records, 35 of the full-length articles were included in this systematic review. Several studies showed that low birth weight, small head circumference, and altered mitochondrial content were more common among HIV-exposed uninfected (HEU) children compared to HIV-unexposed uninfected children (HUU). A few studies demonstrated elevated triglyceride levels, lower levels of insulin, and increased blood pressure, oxidative stress, vascular dysfunction, cardiac damage, and myocardial dysfunction among HEU children compared with HUU children. CONCLUSION Most findings showed that there were cardio-metabolic health risk factors among HEU children, indicating that maternal exposure to HIV and ART may negatively affect foetal health, which may lead to cardio-metabolic morbidity later in life.
Collapse
Affiliation(s)
- Edna Ngoakoana Matjuda
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (E.N.M.); (M.M.M.); (C.R.S.-R.)
| | - Godwill Azeh Engwa
- Department of Biological and Environmental Sciences, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (G.A.E.); (N.G.)
| | - Muhulo Muhau Mungamba
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (E.N.M.); (M.M.M.); (C.R.S.-R.)
| | - Constance Rufaro Sewani-Rusike
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (E.N.M.); (M.M.M.); (C.R.S.-R.)
| | - Nandu Goswami
- Department of Biological and Environmental Sciences, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (G.A.E.); (N.G.)
- Physiology Division, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Neue Stiftingtalstrasse 6, D-5 A, 8036 Graz, Austria
- Department of Health Sciences, Alma Mater Europaea, 2000 Maribor, Slovenia
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Benedicta Ngwenchi Nkeh-Chungag
- Department of Biological and Environmental Sciences, Faculty of Health Sciences, Walter Sisulu University PBX1, Mthatha 5117, South Africa; (G.A.E.); (N.G.)
| |
Collapse
|
9
|
Bulterys MA, Njuguna I, King'e M, Chebet D, Moraa H, Gomez L, Onyango A, Malavi K, Nzia G, Chege M, Neary J, Wagner AD, Lawley KA, Wamalwa D, Benki‐Nugent S, John‐Stewart G. Neurodevelopment of children who are HIV-exposed and uninfected in Kenya. J Int AIDS Soc 2023; 26 Suppl 4:e26149. [PMID: 37909174 PMCID: PMC10618871 DOI: 10.1002/jia2.26149] [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: 01/25/2023] [Accepted: 08/14/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION Predictors of neurodevelopment among children who are HIV-exposed uninfected (CHEU) are poorly understood. METHODS Mothers with and without HIV and their children were enrolled during 6-week postnatal care visits across seven sites in Kenya between March 2021 and June 2022. Infant neurodevelopment was assessed using the Malawi Developmental Assessment Tool, including social, language, fine motor and gross motor domains. We used multivariate linear mixed effects models to identify associations between 1-year neurodevelopment scores, HIV and antiretroviral therapy (ART) exposures, and household factors, adjusted for potential confounders and clustered by the site. RESULTS At 1-year evaluation, CHEU (n = 709) and children who are HIV-unexposed uninfected (CHUU) (n = 715) had comparable median age (52 weeks) and sex distribution (49% vs. 52% female). Mothers living with HIV were older (31 vs. 27 years), had lower education (50% vs. 26% primary) and were more likely to be report moderate-to-severe food insecurity (26% vs. 9%) (p < 0.01 for all). Compared to CHUU, CHEU had higher language scores (adjusted coeff: 0.23, 95% CI: 0.06, 0.39) and comparable social, fine and gross motor scores. Among all children, preterm birth was associated with lower gross motor scores (adjusted coeff: -1.38, 95% CI: -2.05, -0.71), food insecurity was associated with lower social scores (adjusted coeff: -0.37, 95% CI: -0.73, -0.01) and maternal report of intimate partner violence (IPV) was associated with lower fine motor (adjusted coeff: -0.76, 95% CI: -1.40, -0.13) and gross motor scores (adjusted coeff: -1.07, 95% CI: -1.81, -0.33). Among CHEU, in utero efavirenz (EFV) exposure during pregnancy was associated with lower gross motor scores compared to dolutegravir (DTG) exposure (adjusted coeff: -0.51, 95% CI: -1.01, -0.03). Lower fine and gross motor scores were also associated with having a single or widowed mother (adjusted coeff: -0.45, 95% CI: -0.87, -0.03) or a deceased or absent father (adjusted coeff: -0.81, 95% CI: -1.58, -0.05), respectively. CONCLUSIONS Biologic and social factors were associated with child neurodevelopment. Despite socio-demographic differences between CHEU and CHUU, 1-year neurodevelopment was similar. Addressing IPV and food insecurity may provide benefits regardless of maternal HIV status. DTG use was associated with higher neurodevelopmental scores in CHEU, compared to EFV regimens, potentially contributing to a lack of neurodevelopmental difference between CHEU and CHUU.
Collapse
Affiliation(s)
- Michelle A. Bulterys
- Department of Global HealthUniversity of WashingtonSeattleWashingtonUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Irene Njuguna
- Department of Global HealthUniversity of WashingtonSeattleWashingtonUSA
- Kenyatta National HospitalNairobiKenya
| | - Maureen King'e
- Department of Pediatrics and Child HealthUniversity of NairobiNairobiKenya
| | - Daisy Chebet
- Department of Pediatrics and Child HealthUniversity of NairobiNairobiKenya
| | | | - Laurén Gomez
- Department of Global HealthUniversity of WashingtonSeattleWashingtonUSA
| | | | | | | | | | - Jillian Neary
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Anjuli D. Wagner
- Department of Global HealthUniversity of WashingtonSeattleWashingtonUSA
| | - Kendall A. Lawley
- Department of Global HealthUniversity of WashingtonSeattleWashingtonUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Dalton Wamalwa
- Department of Pediatrics and Child HealthUniversity of NairobiNairobiKenya
| | | | - Grace John‐Stewart
- Department of Global HealthUniversity of WashingtonSeattleWashingtonUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
- Departments of Pediatrics, MedicineUniversity of WashingtonSeattleWashingtonUSA
- Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
10
|
Bulterys MA, Njuguna I, Mahy M, Gulaid LA, Powis KM, Wedderburn CJ, John-Stewart G. Neurodevelopment among children exposed to HIV and uninfected in sub-Saharan Africa. J Int AIDS Soc 2023; 26 Suppl 4:e26159. [PMID: 37909232 PMCID: PMC10618877 DOI: 10.1002/jia2.26159] [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: 01/27/2023] [Accepted: 08/21/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION The population of 16 million children exposed to HIV and uninfected (CHEU) under 15 years of age continues to expand rapidly, and the estimated prevalence of CHEU exceeds 20% in several countries in sub-Saharan Africa with high HIV prevalence. Some evidence suggests that CHEU experience suboptimal neurodevelopmental outcomes compared to children born to women without HIV. In this commentary, we discuss the latest research on biologic and socio-behavioural factors associated with neurodevelopmental outcomes among CHEU. DISCUSSION Some but not all studies have noted that CHEU are at risk of poorer neurodevelopment across multiple cognitive domains, most notably in language and motor skills, in diverse settings, ages and using varied assessment tools. Foetal HIV exposure can adversely influence infant immune function, structural brain integrity and growth trajectories. Foetal exposure to antiretrovirals may also influence outcomes. Moreover, general, non-CHEU-specific risk factors for poor neurodevelopment, such as preterm birth, food insecurity, growth faltering and household violence, are amplified among CHEU; addressing these factors will require multi-factorial solutions. There is a need for rigorous harmonised approaches to identify children at the highest risk of delay. In high-burden HIV settings, existing maternal child health programmes serving the general population could adopt structured early child development programmes that educate healthcare workers on CHEU-specific risk factors and train them to conduct rapid neurodevelopmental screening tests. Community-based interventions targeting parent knowledge of optimal caregiving practices have shown to be successful in improving neurodevelopmental outcomes in children and should be adapted for CHEU. CONCLUSIONS CHEU in sub-Saharan Africa have biologic and socio-behavioural factors that may influence their neurodevelopment, brain maturation, immune system and overall health and wellbeing. Multidisciplinary research is needed to disentangle complex interactions between contributing factors. Common environmental and social risk factors for suboptimal neurodevelopment in the general population are disproportionately magnified within the CHEU population, and it is, therefore, important to draw on existing knowledge when considering the socio-behavioural pathways through which HIV exposure could impact CHEU neurodevelopment. Approaches to identify children at greatest risk for poor outcomes and multisectoral interventions are needed to ensure optimal outcomes for CHEU in sub-Saharan Africa.
Collapse
Affiliation(s)
- Michelle A Bulterys
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Irene Njuguna
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Kenyatta National Hospital, Nairobi, Kenya
| | | | - Laurie A Gulaid
- UNICEF, eastern and southern Africa Regional Office, Nairobi, Kenya
| | - Katheen M Powis
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Catherine J Wedderburn
- Department of Pediatrics and Child Health and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Grace John-Stewart
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- School of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
le Roux SM, Abrams EJ, Zerbe A, Phillips TK, Myer L. Children of a syndemic: co-occurring and mutually reinforcing adverse child health exposures in a prospective cohort of HIV-affected mother-infant dyads in Cape Town, South Africa. J Int AIDS Soc 2023; 26 Suppl 4:e26152. [PMID: 37909170 PMCID: PMC10618899 DOI: 10.1002/jia2.26152] [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: 02/08/2023] [Accepted: 08/14/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION Several HIV-related syndemics have been described among adults. We investigated syndemic vulnerability to hazardous drinking (HD), intimate partner violence (IPV) and household food insecurity (HFIS) in breastfed children born without HIV in urban South Africa. We compared those who were perinatally HIV exposed (CHEU) to those who were not (CHU), under conditions of universal maternal antiretroviral therapy (ART) and breastfeeding. METHODS A prospective cohort of pregnant women living with HIV (WLHIV), and without HIV, were enrolled and followed with their infants for 12 months postpartum (2013-2017). All WLHIV initiated antenatal efavirenz-based ART. Measurements of growth (∼3 monthly), infectious cause hospitalisation, ambulatory childhood illness (2-week recall) and neurodevelopment (BSID-III, measured at ∼12 months' age) were compared across bio-social strata using generalised linear regression models, with interaction terms; maternal data included interview-based measures for HD (AUDIT-C), IPV (WHO VAW) and HFIS. RESULTS Among 872 breastfeeding mother-infant pairs (n = 461 CHEU, n = 411 CHU), WLHIV (vs. HIV negative) reported more unemployment (279/461, 60% vs. 217/411, 53%; p = 0.02), incomplete secondary education (347/461, 75% vs. 227/411, 55%; p < 0.0001), HD (25%, 117/459 vs. 7%, 30/411; p < 0.0001) and IPV (22%, 101/457 vs. 8%, 32/411; p < 0.0001) at enrolment; and HFIS at 12 months (45%, 172/386 vs. 30%, 105/352; p > 0.0001). There were positive interactions between maternal HIV and other characteristics. Compared to food secure CHU, the mean difference (95% CI) in weight-for-age Z-score (WAZ) was 0.06 (-0.14; 0.25) for food insecure CHU; -0.26 (-0.42; -0.10) for food secure CHEU; and -0.43 (-0.61; -0.25), for food insecure CHEU. Results were similar for underweight (WAZ < -2), infectious-cause hospitalisation, cognitive and motor delay. HIV-IPV interactions were evident for ambulatory diarrhoea and motor delay. There were HIV-HD interactions for odds of underweight, stunting, cognitive and motor delay. Compared to HD-unexposed CHU, the odds ratios (95% CI) of underweight were 2.31 (1.11; 4.82) for HD-exposed CHU; 3.57 (0.84; 15.13) for HD-unexposed CHEU and 6.01 (2.22; 16.22) for HD-exposed CHEU. CONCLUSIONS These data suggest that maternal HIV-related syndemics may partly drive excess CHEU health risks, highlighting an urgent need for holistic maternal and family care and support alongside ART to optimise the health of CHEU.
Collapse
Affiliation(s)
- Stanzi M le Roux
- Division of Epidemiology & Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Elaine J Abrams
- Mailman School of Public Health, ICAP at Columbia University, Columbia University, New York, New York, USA
- Department of Pediatrics, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA
| | - Allison Zerbe
- Mailman School of Public Health, ICAP at Columbia University, Columbia University, New York, New York, USA
| | - Tamsin K Phillips
- Division of Epidemiology & Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Landon Myer
- Division of Epidemiology & Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
12
|
Zhang Z, Duri K, Duisters KLW, Schoeman JC, Chandiwana P, Lindenburg P, Jaeger J, Ziegler S, Altfeld M, Kohler I, Harms A, Gumbo FZ, Hankemeier T, Bunders MJ. Altered methionine-sulfone levels are associated with impaired growth in HIV-exposed-uninfected children. AIDS 2023; 37:1367-1376. [PMID: 37070556 DOI: 10.1097/qad.0000000000003574] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
OBJECTIVE To determine immune-metabolic dysregulation in children born to women living with HIV. METHODS Longitudinal immune-metabolomic analyses of plasma of 32 pregnant women with HIV (WHIV) and 12 uninfected women and their children up to 1.5 years of age were performed. RESULTS Using liquid chromatography-mass spectrometry and a multiplex bead assay, 280 metabolites (57 amino acids, 116 positive lipids, 107 signalling lipids) and 24 immune mediators (e.g. cytokines) were quantified. combinational antiretroviral therapy (cART) exposure was categorized as cART initiation preconception (long), cART initiation postconception up to 4 weeks before birth (medium) and cART initiation within 3 weeks of birth (short). Plasma metabolite profiles differed between HIV-exposed-uninfected (HEU)-children with long cART exposure compared to HIV-unexposed-children (HUU). Specifically, higher levels of methionine-sulfone, which is associated with oxidative stress, were detected in HEU-children with long cART exposure compared to HUU-children. High infant methionine-sulfone levels were reflected by high prenatal plasma levels in the mother. Increased methionine-sulfone levels in the children were associated with decreased growth, including both weight and length. CONCLUSION These findings based on longitudinal data demonstrate that dysregulation of metabolite networks associated with oxidative stress in children born to WHIV is associated with restricted infant growth.
Collapse
Affiliation(s)
- Zhengzheng Zhang
- Metabolomics and Analytics Centre, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Kerina Duri
- Immunology Unit, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | | | - Johannes C Schoeman
- Metabolomics and Analytics Centre, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Panashe Chandiwana
- Immunology Unit, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Peter Lindenburg
- Metabolomics and Analytics Centre, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
- Research Group Metabolomics, Faculty Science & Technology, University of Applied Sciences Leiden, Hogeschool Leiden, Leiden, The Netherlands
| | | | | | | | - Isabelle Kohler
- Metabolomics and Analytics Centre, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
- Division of BioAnalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Amy Harms
- Metabolomics and Analytics Centre, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Felicity Z Gumbo
- Department of Primary Health Sciences, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Madeleine J Bunders
- Leibniz Institute of Virology, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
13
|
Wedderburn CJ, Evans C, Slogrove AL, Rehman AM, Gibb DM, Prendergast AJ, Penazzato M. Co-trimoxazole prophylaxis for children who are HIV-exposed and uninfected: a systematic review. J Int AIDS Soc 2023; 26:e26079. [PMID: 37292018 PMCID: PMC10251133 DOI: 10.1002/jia2.26079] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 03/08/2023] [Indexed: 06/10/2023] Open
Abstract
INTRODUCTION Co-trimoxazole prophylaxis is recommended for children born to women with HIV to protect those who acquire HIV from opportunistic infections, severe bacterial infections and malaria. With scale-up of maternal antiretroviral therapy, most children remain HIV-exposed uninfected (HEU) and the benefits of universal co-trimoxazole are uncertain. We assessed the effect of co-trimoxazole on mortality and morbidity of children who are HEU. METHODS We performed a systematic review (PROSPERO number: CRD42021215059). We systematically searched MEDLINE, Embase, Cochrane CENTRAL, Global Health, CINAHL Plus, Africa-Wide Information, SciELO and WHO Global Index Medicus for peer-reviewed articles from inception to 4th January 2022 without limits. Ongoing randomized controlled trials (RCTs) were identified through registries. We included RCTs reporting mortality or morbidity in children who are HEU receiving co-trimoxazole versus no prophylaxis/placebo. The risk of bias was assessed using the Cochrane 2.0 tool. Data were summarized using narrative synthesis and findings were stratified by malaria endemicity. RESULTS We screened 1257 records and included seven reports from four RCTs. Two trials from Botswana and South Africa of 4067 children who are HEU found no difference in mortality or infectious morbidity in children randomized to co-trimoxazole prophylaxis started at 2-6 weeks of age compared to those randomized to placebo or no treatment, although event rates were low. Sub-studies found that antimicrobial resistance was higher in infants receiving co-trimoxazole. Two trials in Uganda investigating prolonged co-trimoxazole after breastfeeding cessation showed protection against malaria but no other morbidity or mortality differences. All trials had some concerns or a high risk of bias, which limited the certainty of evidence. DISCUSSION Studies show no clinical benefit of co-trimoxazole prophylaxis in children who are HEU, except to prevent malaria. Potential harms were identified for co-trimoxazole prophylaxis leading to antimicrobial resistance. The trials in non-malarial regions were conducted in populations with low mortality potentially reducing generalizability to other settings. CONCLUSIONS In low-mortality settings with few HIV transmissions and well-performing early infant diagnosis and treatment programmes, universal co-trimoxazole may not be required.
Collapse
Affiliation(s)
- Catherine J. Wedderburn
- Department of Paediatrics and Child Health and Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
- Medical Research Council Clinical Trials Unit at University College LondonLondonUK
- Department of Clinical ResearchLondon School of Hygiene & Tropical MedicineLondonUK
| | - Ceri Evans
- Blizard InstituteQueen Mary University of LondonLondonUK
- Zvitambo Institute for Maternal and Child Health ResearchHarareZimbabwe
- Department of Clinical InfectionMicrobiology and ImmunologyUniversity of LiverpoolLiverpoolUK
| | - Amy L. Slogrove
- Department of Paediatrics and Child HealthFaculty of Medicine & Health SciencesStellenbosch UniversityWorcesterSouth Africa
| | - Andrea M. Rehman
- MRC International Statistics & Epidemiology GroupDepartment of Infectious Disease EpidemiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Diana M. Gibb
- Medical Research Council Clinical Trials Unit at University College LondonLondonUK
| | - Andrew J. Prendergast
- Blizard InstituteQueen Mary University of LondonLondonUK
- Zvitambo Institute for Maternal and Child Health ResearchHarareZimbabwe
| | - Martina Penazzato
- Department of Global HIVHepatitis and Sexually Transmitted Infections ProgrammesWorld Health OrganizationGenevaSwitzerland
| |
Collapse
|
14
|
Cherkos AS, LaCourse SM, Enquobahrie DA, Richardson BA, Bradford S, Montepiedra G, Mmbaga BT, Mbengeranwa T, Masheto G, Jean–Phillippe P, Chakhtoura N, Theron G, Weinberg A, Cassim H, Raesi MS, Jean E, Wabwire D, Nematadzira T, Stranix-Chibanda L, Hesseling AC, Aurpibul L, Gupta A, John-Stewart G. Effect of pregnancy versus postpartum maternal isoniazid preventive therapy on infant growth in HIV-exposed uninfected infants: a post-hoc analysis of the TB APPRISE trial. EClinicalMedicine 2023; 58:101912. [PMID: 36969345 PMCID: PMC10031034 DOI: 10.1016/j.eclinm.2023.101912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
Background Isoniazid preventive therapy (IPT) initiation during pregnancy was associated with increased incidence of adverse pregnancy outcomes in the TB APPRISE trial. Effects of in utero IPT exposure on infant growth are unknown. Methods This post-hoc analysis used data from the TB APPRISE trial, a multicentre, double-blind, placebo-controlled trial, which randomised women to 28-week IPT starting in pregnancy (pregnancy-IPT) or postpartum week 12 (postpartum-IPT) in eight countries with high tuberculosis prevalence. Participants were enrolled between August 2014 and April 2016. Based on modified intent-to-treat analyses, we analysed only live-born babies who had at least one follow-up after birth and compared time to infant growth faltering between arms to 12 weeks and 48 weeks postpartum in overall and sex-stratified multivariable Cox proportional hazards regression. Factors adjusted in the final models include sex of infant, mother's baseline BMI, age in years, ART regimen, viral load, CD4 count, education, and household food insecurity. Results Among 898 HIV-exposed uninfected (HEU) infants, 447 (49.8%) were females. Infants in pregnancy-IPT had a 1.47-fold higher risk of becoming underweight by 12 weeks (aHR 1.47 [95% CI: 1.06, 2.03]) than infants in the postpartum-IPT; increased risk persisted to 48 weeks postpartum (aHR 1.34 [95% CI: 1.01, 1.78]). Maternal IPT timing was not associated with stunting or wasting. In sex-stratified analyses, male infants in the pregnancy-IPT arm experienced an increased risk of low birth weight (LBW) (aRR 2.04 [95% CI: 1.16, 3.68), preterm birth (aRR 1.81 [95% CI: 1.04, 3.21]) and becoming underweight by 12 weeks (aHR 2.02 [95% CI: 1.29, 3.18]) and 48 weeks (aHR 1.82 [95% CI: 1.23, 2.69]). Maternal IPT timing did not influence growth in female infants. Interpretation Maternal IPT during pregnancy was associated with an increased risk of LBW, preterm birth, and becoming underweight among HEU infants, particularly male infants. These data add to prior TB APPRISE data, suggesting that IPT during pregnancy impacts infant growth, which could inform management, and warrants further examination of mechanisms. Funding The TB APPRISE study Supported by the National Institutes of Health (NIH) (award numbers, UM1AI068632 [IMPAACT LOC], UM1AI068616 [IMPAACT SDMC], and UM1AI106716 [IMPAACT LC]) through the National Institute of Allergy and Infectious Diseases, with cofunding from the Eunice Kennedy Shriver National Institute of Child Health and Human Development (contract number, HHSN275201800001I) and the National Institute of Mental Health.
Collapse
Affiliation(s)
- Ashenafi S. Cherkos
- Biostatistics and Epidemiology Department, School of Public Health, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sylvia M. LaCourse
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Global Health, School of Public Health, University of Washington, Seattle, WA, USA
| | - Daniel A. Enquobahrie
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Barbra A. Richardson
- Department of Global Health, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Grace Montepiedra
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Blandina T. Mmbaga
- Kilimanjaro Clinical Research Institute -Kilimanjaro Christian Medical Centre and Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Tapiwa Mbengeranwa
- University of Zimbabwe College of Health Sciences-Clinical Trials Research Centre, Harare, Zimbabwe
| | | | | | - Nahida Chakhtoura
- NIH, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD, Bethesda, MD, USA
| | - Gerhard Theron
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Adriana Weinberg
- Departments of Pediatrics, Medicine and Pathology, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, CO, USA
| | - Haseena Cassim
- Perinatal HIV Research Unit, University of the Witwatersrand, South Africa
| | - Mpho S. Raesi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Elsie Jean
- Department of Pediatrics, GHESKIO Centers, Port-au-Prince, Haiti
| | - Deo Wabwire
- Makerere University – Johns Hopkins University Research Collaboration, Kampala, Uganda
| | | | - Lynda Stranix-Chibanda
- Department of Paediatrics and Child Health, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Anneke C. Hesseling
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Stellenbosch University, South Africa
| | - Linda Aurpibul
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Amita Gupta
- Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Grace John-Stewart
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
- Department of Global Health, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
15
|
Tshiambara P, Hoffman M, Legodi H, Botha T, Mulol H, Pisa P, Feucht U. Comparison of Feeding Practices and Growth of Urbanized African Infants Aged 6-12 Months Old by Maternal HIV Status in Gauteng Province, South Africa. Nutrients 2023; 15:nu15061500. [PMID: 36986230 PMCID: PMC10053312 DOI: 10.3390/nu15061500] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/15/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Appropriate feeding practices are protective against malnutrition and poor growth. We compared feeding practices and growth in HIV-exposed-uninfected (HEU) and HIV-unexposed-uninfected (HUU) between 6-12 months of age in urbanized African infants in South Africa. A repeated cross-sectional analysis was used to determine differences in infant feeding practices and anthropometric measures by HIV exposure status at 6, 9, and 12 months in the Siyakhula study. The study included 181 infants (86 HEU; 95 HUU). Breastfeeding rates were lower in HEU vs. HUU infants at 9 (35.6% vs. 57.3%; p = 0.013) and 12 months (24.7% vs. 48.0%; p = 0.005). Introduction to early complementary foods was common (HEU = 16.2 ± 11.0 vs. HUU = 12.8 ± 9.3 weeks; p = 0.118). Lower weight-for-age Z-scores (WAZ) and head circumference-for-age Z-scores (HCZ) were found in HEU infants at birth. At 6 months, WAZ, length-for-age Z-scores (LAZ), HCZ, and mid-upper-arm circumference-for-age Z-scores (MUACAZ) were lower in HEU vs. HUU infants. At 9 months, lower WAZ, LAZ, and MUACAZ were found in HEU vs. HUU infants. At 12 months, lower WAZ, MUACAZ, and weight-for-length Z-scores (-0.2 ± 1.2 vs. 0.2 ± 1.2; p = 0.020) were observed. HEU infants had lower rates of breastfeeding and poorer growth compared to HUU infants. Maternal HIV exposure affects the feeding practices and growth of infants.
Collapse
Affiliation(s)
- Phumudzo Tshiambara
- Department of Human Nutrition, Faculty of Health Sciences, University of Pretoria, Prinshof Campus, Pretoria 0084, South Africa
- Department of Consumer and Food Sciences, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield Campus, Pretoria 0028, South Africa
- Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
| | - Marinel Hoffman
- Department of Consumer and Food Sciences, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield Campus, Pretoria 0028, South Africa
- Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
| | - Heather Legodi
- Department of Human Nutrition, Faculty of Health Sciences, University of Pretoria, Prinshof Campus, Pretoria 0084, South Africa
| | - Tanita Botha
- Department of Statistics, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield Campus, Pretoria 0028, South Africa
| | - Helen Mulol
- Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Paediatrics, Faculty of Health Sciences, University of Pretoria, Prinshof Campus, Pretoria 0084, South Africa
| | - Pedro Pisa
- Department of Human Nutrition, Faculty of Health Sciences, University of Pretoria, Prinshof Campus, Pretoria 0084, South Africa
| | - Ute Feucht
- Research Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Paediatrics, Faculty of Health Sciences, University of Pretoria, Prinshof Campus, Pretoria 0084, South Africa
| |
Collapse
|
16
|
Floridia M, Orlando S, Andreotti M, Mphwere R, Kavalo T, Ciccacci F, Scarcella P, Marazzi MC, Giuliano M. A 12-month Prospective Study of HIV-infected and HIV-uninfected Women and Their Infants in Malawi: Comparative Analysis of Clinical Events and Infant Growth. Am J Trop Med Hyg 2023; 108:394-402. [PMID: 36535249 PMCID: PMC9896343 DOI: 10.4269/ajtmh.22-0482] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/16/2022] [Indexed: 12/23/2022] Open
Abstract
Few studies have compared clinical outcomes in HIV-exposed uninfected (HEU) and HIV-unexposed uninfected (HUU) infants in the current scenario of universal and lifelong maternal antiretroviral therapy (ART). HIV-uninfected and HIV-infected Malawian women receiving ART and their breastfed infants were followed for 12 months postpartum, analyzing the rates of infectious and noninfectious events and assessing infant growth at 6 weeks, 6 months, and 12 months. The cohorts included 227 mothers (70 HIV-negative, 157 HIV-positive) and 235 infants (72 HUU, 163 HEU). No maternal or infant deaths occurred during follow-up. HIV-negative women were less likely to complete follow-up (48.6% versus 91.1%). Mothers with and without HIV had similar rates of both infectious and noninfectious events per person-month. Infants who were HEU, compared with HUU, had higher rates of events of any type, lower respiratory tract infections (LRTI), and noninfectious events. HEU had lower body mass index (BMI) at 6 weeks but did not differ from HUU in all anthropometric measures at 6 and 12 months; in growth between 6 weeks and 12 months; and in occurrence of stunting, underweight, and wasting at 6 weeks, 6 months, and 12 months. During the first year of life, infants who were HEU, compared with HUU, showed a transiently lower BMI and an increased risk of LRTI.
Collapse
Affiliation(s)
- Marco Floridia
- Istituto Superiore di Sanità, National Center for Global Health, Rome, Italy
| | - Stefano Orlando
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Mauro Andreotti
- Istituto Superiore di Sanità, National Center for Global Health, Rome, Italy
| | | | - Thom Kavalo
- DREAM Program, Community of S. Egidio, Blantyre, Malawi
| | - Fausto Ciccacci
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Paola Scarcella
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Marina Giuliano
- Istituto Superiore di Sanità, National Center for Global Health, Rome, Italy
| |
Collapse
|
17
|
Abstract
Prior to widespread availability of antiretroviral therapy (ART) in sub-Saharan Africa, children who were HIV-exposed but uninfected (HEU) had increased mortality, morbidity and undernutrition compared with children who were HIV-unexposed. Scale-up of ART has led to impressive declines in vertical HIV transmission, but over 15 million children are now HEU, 90% of whom live in sub-Saharan Africa. There are ongoing health disparities among children who are HEU, with higher mortality, morbidity and stunting and modest impairments in early child development, which collectively hamper health and human capital in high prevalence countries. The underlying causes are multifactorial and include exposure to HIV, co-infections and a skewed antenatal inflammatory milieu, particularly if mothers start ART once they have advanced disease, as well as socioeconomic risk factors, which may cluster in HIV-affected households. Improving maternal health through early and sustained ART, ensuring optimal breastfeeding, and implementing evidence-based priority interventions for all children in areas of high HIV prevalence, will likely improve outcomes. A more comprehensive intervention package based on the Nurturing Care Framework may have particular benefits for children who are HEU, to close health gaps and ensure that the next generation of HIV-free children survive and thrive, and lead healthy and productive lives.
Collapse
|
18
|
Slogrove AL, de Beer ST, Kalk E, Boulle A, Cotton M, Cupido H, Laughton B, Marlow M, Mehta U, Msolo N, Myer L, Powis KM, Schoeman E, Tomlinson M, Zunza M, Williams P, Davies MA. Survival and health of children who are HIV-exposed uninfected: study protocol for the CHERISH (Children HIV-Exposed Uninfected - Research to Inform Survival and Health) dynamic, prospective, maternal-child cohort study. BMJ Open 2023; 13:e070465. [PMID: 36593001 PMCID: PMC9809249 DOI: 10.1136/bmjopen-2022-070465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/15/2022] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION CHERISH is designed to establish a long-term sustainable system for measurement of in utero and postnatal exposures and outcomes in children who are HIV-exposed uninfected (HEU) and HIV-unexposed to compare survival, hospitalisation, growth and neurodevelopment in the Western Cape, South Africa. METHODS AND ANALYSIS During 2022-2025, the CHERISH dynamic cohort is prospectively enrolling pregnant people with and without HIV at 24-36 weeks gestation from one urban and one rural community, following mother-child pairs, including children who are HEU (target N=1200) and HIV-unexposed (target N=600) for 3 years from the child's birth. In-person visits occur at enrolment, delivery, 12 months, 24 months and 36 months with intervening 3-monthly telephone data collection. Children and mothers without HIV are tested for HIV at all in-person visits. Data on exposures and outcomes are collected from routine standardised healthcare documentation, maternal interview, measurement (growth and neurodevelopment) at in-person visits and linkage to the Western Cape Provincial Health Data Centre (survival and hospitalisation). A priori adverse birth outcomes, advanced maternal HIV and maternal mental health are considered potential mediators of outcome disparities in children who are HEU and will be evaluated as such in multivariable models appropriate for each outcome. ETHICS AND DISSEMINATION Mothers interested in joining the study are taken through a visual informed consent document for their and their child's participation, with the option to consent to anonymised de-identified data being contributed to a public data repository. All data is captured directly into an electronic database using alphanumeric identifiers devoid of identifying information. The cohort study is approved by Human Research Ethics Committees of Stellenbosch University (N20/08/084), University of Cape Town (723/2021) and Western Cape Government (WC_2021_09_007). Findings will be shared with participants, participating communities, local and provincial stakeholders, child health clinicians, researchers and policymakers at local, national and international forums and submitted for publication in peer-reviewed journals.
Collapse
Affiliation(s)
- Amy L Slogrove
- Department of Paediatrics and Child Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
- Department of Global Health, Ukwanda Centre for Rural Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
| | - Shani Tamlyn de Beer
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
- Division of Population Health Sciences, University of Bristol, Bristol, UK
| | - Emma Kalk
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Andrew Boulle
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
- Health Intelligence Directorate, Western Cape Provincial Government, Cape Town, South Africa
| | - Mark Cotton
- Department of Paediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Heinrich Cupido
- Department of Paediatrics and Child Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
| | - Barbara Laughton
- Department of Paediatrics and Child Health, Family Centre for Research with Ubuntu, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Marguerite Marlow
- Department of Global Health, Institute for Life Course Health Research, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Ushma Mehta
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Ncumisa Msolo
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Landon Myer
- Division of Epidemiology and Biostatistics, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
| | - Kathleen M Powis
- Departments of Internal Medicine and Paediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Elisma Schoeman
- Department of Paediatrics and Child Health, Stellenbosch University Faculty of Medicine and Health Sciences, Worcester, South Africa
| | - Mark Tomlinson
- Department of Global Health, Institute for Life Course Health Research, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Moleen Zunza
- Department of Global Health, Division of Epidemiology and Biostatistics, Stellenbosch University Faculty of Medicine and Health Sciences, Cape Town, South Africa
| | - Paige Williams
- Department of Biostatistics, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town Faculty of Health Sciences, Observatory, South Africa
- Health Intelligence Directorate, Western Cape Provincial Government, Cape Town, South Africa
| |
Collapse
|
19
|
Rickman RR, Lane CE, Collins SM, Miller JD, Onono M, Wekesa P, Nichols AR, Foster SF, Shiau S, Young SL, Widen EM. Body Composition Trajectories During the First 23 Months of Life Differ by HIV Exposure Among Infants in Western Kenya: A Prospective Study. J Nutr 2023; 153:331-339. [PMID: 36913469 PMCID: PMC10196592 DOI: 10.1016/j.tjnut.2022.11.010] [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: 08/11/2022] [Revised: 10/29/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Infants who are HIV-exposed and uninfected have suboptimal growth patterns compared to those who are HIV-unexposed and uninfected. However, little is known about how these patterns persist beyond 1 year of life. OBJECTIVES This study aimed to examine whether infant body composition and growth trajectories differed by HIV exposure during the first 2 years of life among Kenyan infants using advanced growth modeling. METHODS Repeated infant body composition and growth measurements (mean: 6; range: 2-7) were obtained from 6 weeks to 23 months in the Pith Moromo cohort in Western Kenya (n = 295, 50% HIV-exposed and uninfected, 50% male). Body composition trajectory groups were fitted using latent class mixed modeling (LCMM) and associations between HIV exposure and growth trajectories were examined using logistic regression analysis. RESULTS All infants exhibited poor growth. However, HIV-exposed infants generally grew suboptimally than unexposed infants. Across all body composition models except for the sum of skinfolds, HIV-exposed infants had a higher likelihood of belonging to the suboptimal growth groups identified by LCMM than the HIV-unexposed infants. Notably, HIV-exposed infants were 3.3 times more likely (95% CI: 1.5-7.4) to belong to the length-for-age z-score growth class that remained at a z-score of < -2, indicating stunted growth. HIV-exposed infants were also 2.6 times more likely (95% CI: 1.2-5.4) to belong to the weight-for-length-for-age z-score growth class that remained between 0 and -1, and were 4.2 times more likely (95% CI: 1.9-9.3) to belong to the weight-for-age z-score growth class that indicated poor weight gain besides stunted linear growth. CONCLUSIONS In a cohort of Kenyan infants, HIV-exposed infants grew suboptimally compared to HIV-unexposed infants beyond 1 year of age. These growth patterns and longer-term effects should be further investigated to support the ongoing efforts to reduce early-life HIV exposure-related health disparities.
Collapse
Affiliation(s)
- Rachel R Rickman
- Department of Nutritional Sciences, University of Texas, Austin, TX, USA
| | - Charlotte E Lane
- International Initiative for Impact Evaluation Inc (3ie), Washington, DC, USA
| | - Shalean M Collins
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Joshua D Miller
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | | | - Pauline Wekesa
- Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Amy R Nichols
- Department of Nutritional Sciences, University of Texas, Austin, TX, USA
| | - Saralyn F Foster
- Department of Nutritional Sciences, University of Texas, Austin, TX, USA
| | - Stephanie Shiau
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA
| | - Sera L Young
- Department of Anthropology, Northwestern University, Evanston, IL, USA; Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | - Elizabeth M Widen
- Department of Nutritional Sciences, University of Texas, Austin, TX, USA; Department of Women's Health and Pediatrics, Dell Pediatric Research Institute, University of Texas, Austin, TX, USA.
| |
Collapse
|
20
|
Toledo G, Landes M, van Lettow M, Tippett Barr BA, Bailey H, Crichton S, Msungama W, Thorne C. Risk factors for stunting in children who are HIV-exposed and uninfected after Option B+ implementation in Malawi. MATERNAL & CHILD NUTRITION 2023; 19:e13451. [PMID: 36349962 PMCID: PMC9749602 DOI: 10.1111/mcn.13451] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/10/2022]
Abstract
Evidence suggests children HIV-exposed and uninfected (CHEU) experience poor growth. We analysed child anthropometrics and explored factors associated with stunting among Malawian CHEU. Mothers with HIV and their infants HIV-exposed were enroled in a nationally representative prospective cohort within the National Evaluation of Malawi's Prevention of Mother-to-Child HIV Transmission Programme after Option B+ implementation (2014-2018). Anthropometry was measured at enrolment (age 1-6 months), visit 1 (approximately 12 months), and visit 2 (approximately 24 months). Weight-for-age (WAZ) and length-for-age (LAZ) z-scores were calculated using World Health Organization Growth Standards; underweight and stunting were defined as WAZ and LAZ more than 2 standard deviations below the reference median. Multivariable logistic regression restricted to CHEU aged 24 months (±3 months) was used to identify factors associated with stunting. Among 1211 CHEU, 562/1211 attended visit 2, of which 529 were aged 24 months (±3 months) and were included. At age 24 months, 40.4% of CHEU were stunted and/or underweight, respectively. In multi-variable analysis, adjusting for child age and sex, the odds of stunting were higher among CHEU with infectious disease diagnosis compared to those with no diagnosis (adjusted odds ratio = 3.35 [95% confidence interval: 1.82-6.17]), which was modified by co-trimoxazole prophylaxis (p = 0.028). Infant low birthweight was associated with an increased odds of stunting; optimal feeding and maternal employment were correlated with reduced odds. This is one of the first studies examining CHEU growth since Option B+. Interventions to improve linear growth among CHEU should address their multi-faceted health risks, alongside maternal ART prescription, and follow-up of mother-child pairs.
Collapse
Affiliation(s)
- Gabriela Toledo
- Population, Policy, and Practice Research & Teaching Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Megan Landes
- Department of Family and Community MedicineUniversity of TorontoTorontoOntarioCanada
- Dalla Lana School of Public HealthUniversity of TorontoTorontoOntarioCanada
| | - Monique van Lettow
- Dalla Lana School of Public HealthUniversity of TorontoTorontoOntarioCanada
| | | | - Heather Bailey
- Institute for Global Health, University College LondonLondonUK
| | - Siobhan Crichton
- Medical Research Council Clinical Trials UnitUniversity College LondonLondonUK
| | - Wezi Msungama
- Division of Global HIV and TuberculosisCenters for Disease Control and PreventionLilongweMalawi
| | - Claire Thorne
- Population, Policy, and Practice Research & Teaching Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| |
Collapse
|
21
|
In-utero HIV exposure and cardiometabolic health among children 5-8 years: findings from a prospective birth cohort in South Africa. AIDS 2023; 37:173-182. [PMID: 36476456 PMCID: PMC9751971 DOI: 10.1097/qad.0000000000003412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To evaluate if in-utero HIV exposure is associated with adverse cardiometabolic health outcomes at 5-8 years of age. DESIGN Prospective cohort study. METHODS We enrolled a random sample of HIV-exposed but uninfected (HEU) and HIV-unexposed children from the Drakenstein Child Health study, a longitudinal birth cohort study in Cape Town, South Africa, in a cardiometabolic health pilot study. Outcomes were assessed by trained study staff and included: anthropometry, body composition and size, blood pressure, fasting plasma glucose, HbA1c, lipids, and insulin resistance using HOMA-IR. We used multivariable linear and log-binomial regression to estimate associations between HIV-exposure and cardiometabolic outcomes, adjusted for child age, sex, height, body size, and maternal factors as appropriate. RESULTS We included 260 children (HEU n = 100, HIV-unexposed n = 160). HEU children had older mothers (median age 30 vs. 26 years), with minimal differences in gestational age and size at birth by HIV-exposure status. In multivariable analyses, HEU children had lower weight-for-age (mean difference -0.35, 95% confidence interval -0.66, -0.05), and height-for-age (mean difference -0.29, 95% confidence interval -0.56, -0.03; z-scores). There were no differences in adiposity, impaired glucose metabolism, or lipid levels by HIV-exposure status. Overall, 12% of children had blood pressure more than 90th percentile, with no differences by HIV-exposure status. CONCLUSION Overall, there were few differences in cardiometabolic outcomes between HEU and HIV-unexposed children in this South African cohort. Although these findings are reassuring, monitoring of cardiometabolic health is important as HEU and HIV-unexposed children enter adolescence and cardiometabolic risk trajectories become established.
Collapse
|
22
|
Nyofane M, Hoffman M, Mulol H, Botha T, Vannevel V, Pattinson R, Feucht U. Early Childhood Growth Parameters in South African Children with Exposure to Maternal HIV Infection and Placental Insufficiency. Viruses 2022; 14:v14122745. [PMID: 36560749 PMCID: PMC9782627 DOI: 10.3390/v14122745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/24/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Maternal HIV exposure and intrauterine growth restriction (IUGR) due to placental insufficiency both carry major risks to early child growth. We compared the growth outcomes of children aged 18 months who had abnormal umbilical artery resistance indices (UmA-RI), as a marker of placental insufficiency, with a comparator group of children with normal UmA-RI during pregnancy, as mediated by maternal HIV infection. The cross-sectional study included 271 children, grouped into four subgroups based on HIV exposure and history of normal/abnormal UmA-RI, using available pregnancy and birth information. Standard procedures were followed to collect anthropometric data, and z-scores computed as per World Health Organization growth standards. Lower length-for-age z-scores (LAZ) were observed in children who were HIV-exposed-uninfected (CHEU) (-0.71 ± 1.23; p = 0.004) and who had abnormal UmA-RI findings (-0.68 ± 1.53; p < 0.001). CHEU with abnormal UmA-RI had lower LAZ (-1.3 ± 1.3; p < 0.001) and weight-for-age z-scores (WAZ) (-0.64 ± 0.92; p = 0.014) compared to the control group. The prevalence of stunting was 40.0% in CHEU with abnormal UmA-RI and 16.0% in CHEU with normal UmA-RI (p < 0.001; p = 0.016, respectively). In conclusion, maternal HIV exposure and placental insufficiency are independent risk factors for childhood stunting, with this risk potentiated when these two risk factors overlap.
Collapse
Affiliation(s)
- Mothusi Nyofane
- Department of Consumer and Food Sciences, University of Pretoria, Pretoria 0002, South Africa
- Department of Nutrition, National University of Lesotho, Maseru 100, Lesotho
- Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
- Correspondence: ; Tel.: +266-5775-1718
| | - Marinel Hoffman
- Department of Consumer and Food Sciences, University of Pretoria, Pretoria 0002, South Africa
- Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
| | - Helen Mulol
- Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Paediatrics, University of Pretoria, Pretoria 0002, South Africa
| | - Tanita Botha
- Department of Statistics, University of Pretoria, Pretoria 0002, South Africa
| | - Valerie Vannevel
- Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Obstetrics and Gynaecology, University of Pretoria, Pretoria 0002, South Africa
| | - Robert Pattinson
- Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Obstetrics and Gynaecology, University of Pretoria, Pretoria 0002, South Africa
| | - Ute Feucht
- Centre for Maternal, Fetal, Newborn and Child Health Care Strategies, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa
- Research Unit for Maternal and Infant Health Care Strategies, South African Medical Research Council, Pretoria 0001, South Africa
- Department of Paediatrics, University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
23
|
Ramokolo V, Kuhn L, Lombard C, Jackson D, Goga AE. Impact of antenatal antiretroviral drug exposure on the growth of children who are HIV-exposed uninfected: the national South African Prevention of Mother to Child Evaluation cohort study. BMC Infect Dis 2022; 22:908. [PMID: 36474212 PMCID: PMC9724324 DOI: 10.1186/s12879-022-07847-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 11/06/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The relationship between in-utero antiretroviral (ARV) drug exposure and child growth needs further study as current data provide mixed messages. We compared postnatal growth in the first 18-months of life between children who are HIV-exposed uninfected (CHEU) with fetal exposure to ARV drugs (prophylaxis or triple-drug therapy (ART)) and CHEU not exposed to ARVs. We also examined other independent predictors of postnatal growth. METHODS We analysed data from a national prospective cohort study of 2526 CHEU enrolled at 6-weeks and followed up 3-monthly till 18-months postpartum, between October 2012 and September 2014. Infant anthropometry was measured, and weight-for-age (WAZ) and length-for-age (LAZ) Z-scores calculated. Generalized estimation equation models were used to compare Z-scores between groups. RESULTS Among 2526 CHEU, 617 (24.4%) were exposed to ART since -pregnancy (pre-conception ART), 782 (31.0%) to ART commencing post-conception, 879 (34.8%) to maternal ARV prophylaxis (Azidothymidine (AZT)), and 248 (9.8%) had no ARV exposure. In unadjusted analyses, preterm birth rates were higher among CHEU with no ARV exposure than in other groups. Adjusting for infant age, the mean WAZ profile was lower among CHEU exposed to pre-conception ART [-0.13 (95% confidence interval - 0.26; - 0.01)] than the referent AZT prophylaxis group; no differences in mean WAZ profiles were observed for the post-conception ART (- 0.05 (- 0.16; 0.07)), None (- 0.05 (- 0.26; 0.16)) and newly-infected (- 0.18 (- 0.48; 0.13)) groups. Mean LAZ profiles were similar across all groups. In multivariable analyses, mean WAZ and LAZ profiles for the ARV exposure groups were completely aligned. Several non-ARV factors including child, maternal, and socio-demographic factors independently predicted mean WAZ. These include child male (0.45 (0.35; 0.56)) versus female, higher maternal education grade 7-12 (0.28 (0.14; 0.42) and 12 + (0.36 (0.06; 0.66)) versus ≤ grade7, employment (0.16 (0.04; 0.28) versus unemployment, and household food security (0.17 (0.03; 0.31). Similar predictors were observed for mean LAZ. CONCLUSION Findings provide evidence for initiating all pregnant women living with HIV on ART as fetal exposure had no demonstrable adverse effects on postnatal growth. Several non-HIV-related maternal, child and socio-demographic factors were independently associated with growth, highlighting the need for multi-sectoral interventions. Longer-term monitoring of CHEU children is recommended.
Collapse
Affiliation(s)
- Vundli Ramokolo
- grid.415021.30000 0000 9155 0024HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, 1 Francie Van Zyl Drive, Tygerberg, South Africa ,grid.239585.00000 0001 2285 2675Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY USA ,grid.239585.00000 0001 2285 2675Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY USA
| | - Louise Kuhn
- grid.239585.00000 0001 2285 2675Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY USA ,grid.239585.00000 0001 2285 2675Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY USA
| | - Carl Lombard
- grid.415021.30000 0000 9155 0024Biostatistics Unit, South African Medical Research Council, Tygerberg, South Africa ,grid.11956.3a0000 0001 2214 904XDivision of Epidemiology and Biostatistics Unit, Department of Global Health, University of Stellenbosch, Tygerberg, South Africa
| | - Debra Jackson
- grid.8991.90000 0004 0425 469XCentre for Maternal Adolescent Reproductive & Child Health (MARCH), London School of Hygiene and Tropical Medicine, London, UK ,grid.8974.20000 0001 2156 8226School of Public Health, University of the Western Cape, Cape Town, South Africa
| | - Ameena E. Goga
- grid.415021.30000 0000 9155 0024HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, 1 Francie Van Zyl Drive, Tygerberg, South Africa ,grid.49697.350000 0001 2107 2298Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
24
|
Pavlinac PB, Singa B, Huang ML, Shrestha L, Li V, Atlas HE, Diakhate MM, Brander R, Meshak L, Bogonko G, Tickell KD, McGrath CJ, Machuara IM, Ounga DO, Berkley JA, Richardson BA, John-Stewart G, Walson JL, Slyker J. Cytomegalovirus Viremia Predicts Postdischarge Mortality in Kenyan HIV-Exposed Uninfected Children. J Infect Dis 2022; 226:1519-1527. [PMID: 35152295 PMCID: PMC9624454 DOI: 10.1093/infdis/jiac047] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/11/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) viremia is associated with mortality in severely ill immunocompetent adults and hospitalized children with HIV (CWH). We measured CMV viremia in HIV-exposed and -unexposed Kenyan children aged 1-59 months discharged from hospital and determined its relationship with postdischarge mortality. METHODS CMV DNA levels were measured in plasma from 1024 children (97 of which were HIV exposed uninfected [HEU], and 15 CWH). Poisson and Cox proportional hazards regression models were used to identify correlates of CMV viremia ≥ 1000 IU/mL
and estimate associations with 6-month mortality, respectively. RESULTS CMV viremia was detected in 31% of children, with levels ≥ 1000 IU/mL in 5.8%. HIV infection, age < 2 years, breastfeeding, and midupper arm circumference < 12.5 cm were associated with CMV viremia ≥ 1000 IU/mL. Among HEU children, CMV ≥ 1000 IU/mL (hazard ratio [HR] = 32.0; 95% confidence interval [CI], 2.9-354.0; P = .005) and each 1-log increase in CMV viral load (HR = 5.04; 95% CI, 1.7-14.6; P = .003) were associated with increased risk of mortality. CMV viremia was not significantly associated with mortality in HIV-unexposed children. CONCLUSIONS CMV levels at hospital postdischarge predict increased risk of 6-month mortality in Kenyan HEU children. CMV suppression may be a novel target to reduce mortality in HEU children. CLINICAL TRIAL REGISTRATION NCT02414399.
Collapse
Affiliation(s)
- Patricia B Pavlinac
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Benson Singa
- Kenya Medical Research Institute, Nairobi, Kenya
- Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
| | - Meei-Li Huang
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Lasata Shrestha
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Vanessa Li
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Hannah E Atlas
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | | | - Rebecca Brander
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Liru Meshak
- Homa Bay Teaching and Referral Hospital, Homa Bay, Kenya
| | | | - Kirkby D Tickell
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
| | - Christine J McGrath
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
| | | | | | - James A Berkley
- Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Barbra A Richardson
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - 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, Allergy, and Infectious Disease, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Judd L Walson
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- Department of Medicine, Allergy, and Infectious Disease, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jennifer Slyker
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
25
|
Szanyi J, Walles JK, Tesfaye F, Gudeta AN, Björkman P. Intrauterine
HIV
exposure is associated with linear growth restriction among Ethiopian children in the first 18 months of life. Trop Med Int Health 2022; 27:823-830. [DOI: 10.1111/tmi.13805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Joshua Szanyi
- Department of Translational Medicine Lund University Sweden
| | - John König Walles
- Department of Translational Medicine Lund University Sweden
- Department of Infectious Diseases Central Hospital Kristianstad Sweden
| | - Fregenet Tesfaye
- Department of Translational Medicine Lund University Sweden
- Armauer Hansen Research Institute Addis Ababa Ethiopia
| | | | - Per Björkman
- Department of Translational Medicine Lund University Sweden
- Department of Infectious Diseases Skåne University Hospital Malmö Sweden
| |
Collapse
|
26
|
Bengtson AM, le Roux SM, Phillips TK, Brittain K, Zerbe A, Madlala HP, Malaba TR, Petro G, Abrams EJ, Myer L. Relationship between pre-pregnancy maternal body mass index and infant weight trajectories in HIV-exposed and HIV-unexposed infants. Paediatr Perinat Epidemiol 2022; 36:536-547. [PMID: 34859468 PMCID: PMC9163208 DOI: 10.1111/ppe.12825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/08/2021] [Accepted: 09/19/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Maternal HIV and antiretroviral therapy (ART) exposure in utero may influence infant weight, but the contribution of maternal y body mass index (BMI) to early life overweight and obesity is not clear. OBJECTIVE To estimate associations between maternal BMI at entry to antenatal care (ANC) and infant weight through approximately 1 year of age and to evaluate whether associations were modified by maternal HIV status, maternal HIV and viral load, breastfeeding intensity through 6 months or timing of entry into ANC. METHODS We followed HIV-uninfected and -infected pregnant women initiating efavirenz-based ART from first antenatal visit through 12 months postpartum. Infant weight was assessed via World Health Organization BMI and weight-for-length z-scores (WLZ) at 6 weeks, 3, 6, 9 and 12 months. We used multivariable linear mixed-effects models to estimate associations between maternal BMI and infant z-scores over time. RESULTS In 861 HIV-uninfected infants (454 HIV-exposed; 407 HIV-unexposed), nearly 20% of infants were overweight or obese by 12 months of age, regardless of HIV exposure status. In multivariable analyses, increasing maternal BMI category was positively associated with higher infant BMIZ and WLZ scores between 6 weeks and 12 months of age and did not differ by HIV exposure status. However, HIV-exposed infants had slightly lower BMIZ and WLZ trajectories through 12 months of age, compared with HIV-unexposed infants across all maternal BMI categories. Differences in BMIZ and WLZ scores by HIV exposure were not explained by timing of entry into ANC or maternal viral load pre-ART initiation, but z-scores were slightly higher for HIV-exposed infants who were predominantly or exclusively versus partially breastfed. CONCLUSIONS These findings suggest maternal BMI influences early infant weight gain, regardless of infant HIV exposure status. Intervention to reduce maternal BMI may help to address growing concerns about obesity among HIV-uninfected children.
Collapse
Affiliation(s)
- Angela M. Bengtson
- Department of Epidemiology, Brown University School of Public Health, Providence, RI
| | - Stanzi M le Roux
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Tamsin K Phillips
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa, Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Kirsty Brittain
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa, Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Allison Zerbe
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY
| | - Hlengiwe P Madlala
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Thokozile R. Malaba
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa, Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Gregory Petro
- Department of Obstetrics & Gynaecology, University of Cape Town and New Somerset Hospital, Cape Town, South Africa
| | - Elaine J. Abrams
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY, Department of Pediatrics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa, Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
27
|
Jones S, Coelho Nunes M. Beyond COVID-19: Equitable epidemiology for studying the impact of maternal infections on neonatal mortality and morbidity. Paediatr Perinat Epidemiol 2022; 36:553-555. [PMID: 35768341 PMCID: PMC9328205 DOI: 10.1111/ppe.12892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 01/31/2023]
Affiliation(s)
- Stephanie Jones
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Marta Coelho Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
28
|
Wedderburn CJ, Weldon E, Bertran-Cobo C, Rehman AM, Stein DJ, Gibb DM, Yeung S, Prendergast AJ, Donald KA. Early neurodevelopment of HIV-exposed uninfected children in the era of antiretroviral therapy: a systematic review and meta-analysis. THE LANCET. CHILD & ADOLESCENT HEALTH 2022; 6:393-408. [PMID: 35483380 PMCID: PMC9090907 DOI: 10.1016/s2352-4642(22)00071-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND There are 15·4 million children who are HIV-exposed and uninfected worldwide. Early child development crucially influences later academic and socioeconomic factors. However, the neurodevelopmental outcomes of HIV-exposed uninfected (HEU) children in the era of maternal antiretroviral therapy (ART) remain unclear. We aimed to examine the effects of in-utero exposure to HIV and ART on child neurodevelopment. METHODS For this systematic review and meta-analysis, we searched MEDLINE, Embase, PubMed, Africa-Wide Information, PsycInfo, and Global Health databases from inception to May 27, 2020, for studies from the past two decades reporting neurodevelopment of HEU children aged 0-5 years compared with HIV-unexposed (HU) children (aim 1), and effects of different maternal ART regimens on neurodevelopment of HEU children (aim 2). We did narrative syntheses for both aims, and a random-effects meta-analysis of high-quality studies comparing HEU children and HU children, to obtain weighted pooled estimates of effect sizes. This study was registered with PROSPERO, CRD42018075910. FINDINGS We screened 35 527 records and included 45 articles from 31 studies. Overall, 12 (57%) of 21 studies comparing HEU children and HU children found worse neurodevelopment in HEU children in at least one domain. Study design and methodological quality were variable, with heterogeneity across populations. Meta-analysis included eight high-quality studies comparing 1856 HEU children with 3067 HU children at ages 12-24 months; among HEU children with available data, 1709 (99%) of 1732 were exposed to ART. HEU children had poorer expressive language (effect size -0·17 [95% CI -0·27 to -0·07], p=0·0013) and gross motor function (-0·13 [-0·20 to -0·07], p<0·0001) than HU children, but similar cognitive development (-0·06 [-0·19 to 0·06], p=0·34), receptive language development (-0·10 [-0·23 to 0·03], p=0·14), and fine motor skills (-0·05 [-0·15 to 0·06], p=0·36). Results suggested little or no evidence of an effect of specific maternal ART regimens on neurodevelopment; study heterogeneity prevented meta-analysis. INTERPRETATION HEU children are at risk of subtle impairments in expressive language and gross motor development by age 2 years. We found no consistent effect of maternal ART regimens analysed, although evidence was scarce. We highlight the need for large high-quality longitudinal studies to assess the neurodevelopmental trajectories of HEU children and to investigate underlying mechanisms to inform intervention strategies. FUNDING Wellcome Trust and Medical Research Council.
Collapse
Affiliation(s)
- Catherine J Wedderburn
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK; MRC Clinical Trials Unit, University College London, London, UK.
| | - Ella Weldon
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Cesc Bertran-Cobo
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Andrea M Rehman
- MRC International Statistics & Epidemiology Group, London School of Hygiene & Tropical Medicine, London, UK
| | - Dan J Stein
- The Neuroscience Institute, University of Cape Town, Cape Town, South Africa; Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa; MRC Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| | - Diana M Gibb
- MRC Clinical Trials Unit, University College London, London, UK
| | - Shunmay Yeung
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kirsten A Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; The Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
29
|
Grant-Beurmann S, Jumare J, Ndembi N, Matthew O, Shutt A, Omoigberale A, Martin OA, Fraser CM, Charurat M. Dynamics of the infant gut microbiota in the first 18 months of life: the impact of maternal HIV infection and breastfeeding. MICROBIOME 2022; 10:61. [PMID: 35414043 PMCID: PMC9004197 DOI: 10.1186/s40168-022-01230-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Access to antiretroviral therapy (ART) during pregnancy and breastfeeding for mothers with HIV has resulted in fewer children acquiring HIV peri- and postnatally, resulting in an increase in the number of children who are exposed to the virus but are not infected (HEU). HEU infants have an increased likelihood of childhood infections and adverse growth outcomes, as well as increased mortality compared to their HIV-unexposed (HUU) peers. We explored potential differences in the gut microbiota in a cohort of 272 Nigerian infants born to HIV-positive and negative mothers in this study during the first 18 months of life. RESULTS The taxonomic composition of the maternal vaginal and gut microbiota showed no significant differences based on HIV status, and the composition of the infant gut microbiota at birth was similar between HUU and HEU. Longitudinal taxonomic composition of the infant gut microbiota and weight-for-age z-scores (WAZ) differed depending on access to breast milk. HEU infants displayed overall lower WAZ than HUU infants at all time points. We observed a significantly lower relative abundance of Bifidobacterium in HEU infants at 6 months postpartum. Breast milk composition also differed by time point and HIV infection status. The antiretroviral therapy drugs, lamivudine and nevirapine, as well as kynurenine, were significantly more abundant in the breast milk of mothers with HIV. Levels of tiglyl carnitine (C5) were significantly lower in the breast milk of mothers without HIV. ART drugs in the breast milk of mothers with HIV were associated with a lower relative abundance of Bifidobacterium longum. CONCLUSIONS Maternal HIV infection was associated with adverse growth outcomes of HEU infants in this study, and these differences persist from birth through at least 18 months, which is a critical window for the development of the immune and central nervous systems. We observed that the relative abundance of Bifidobacterium spp. was significantly lower in the gut microbiota of all HEU infants over the first 6 months postpartum, even if HEU infants were receiving breast milk. Breastfeeding was of benefit in our HEU infant cohort in the first weeks postpartum; however, ART drug metabolites in breast milk were associated with a lower abundance of Bifidobacterium. Video abstract.
Collapse
Affiliation(s)
- Silvia Grant-Beurmann
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jibreel Jumare
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Ashley Shutt
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Olivia A Martin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Claire M Fraser
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Man Charurat
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
Benki-Nugent SF, Yunusa R, Mueni A, Laboso T, Tamasha N, Njuguna I, Gómez L, Wamalwa DC, Tapia K, Maleche-Obimbo E, Bangirana P, Boivin MJ, John-Stewart GC. Lower Neurocognitive Functioning in HIV-Exposed Uninfected Children Compared With That in HIV-Unexposed Children. J Acquir Immune Defic Syndr 2022; 89:441-447. [PMID: 35202050 PMCID: PMC8873990 DOI: 10.1097/qai.0000000000002881] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/29/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Perinatal HIV and antiretroviral therapy exposure may influence neurocognitive outcomes, although evidence is mixed and most studies are limited to outcomes in the first 24 months. We compared neurocognitive outcomes in school-aged children who were HIV exposed uninfected (CHEU) with those in children who were HIV unexposed uninfected (CHUU). SETTING Children were recruited from a health center in Nairobi, Kenya. METHODS Key inclusion criteria were children aged 5-12 years and confirmed child and maternal HIV status; for CHEU, mothers reported knowing HIV-positive status before or at delivery of the index child. Children underwent a detailed battery of neuropsychological tests and behavioral assessment, and comparisons of scores between CHEU and CHUU were conducted using linear regression. RESULTS Among 56 CHEU and 65 CHUU, the median age and sex distributions were 6.8 and 7.0 years (P = 0.8) and 48% and 60% girls (P = 0.2), respectively. In analyses adjusted for child's age and sex and caregiver's age, education, and household rent, CHEU had significantly lower mean z scores for global cognitive ability than CHUU [-0.35, 95% confidence interval (CI): -0.64 to -0.05; P = 0.02], short-term memory (-0.44, 95% CI: -0.76 to -0.12; P = 0.008), delayed memory (-0.43, 95% CI: -0.79 to -0.08; P = 0.02), attention (-0.41, 95% CI: -0.78 to -0.05; P = 0.03), and processing speed (-0.76, 95% CI: -1.37 to -0.16; P = 0.01). Models adjusted for child nutritional status, household food security, and orphanhood yielded similar results. CONCLUSIONS Children exposed to HIV had poorer long-term neurocognitive outcomes than CHUU. These data suggest that long-term studies of neurocognitive and educational attainment in CHEU are warranted.
Collapse
Affiliation(s)
| | - Rabi Yunusa
- Department of Global Health, University of Washington, Seattle, WA
| | - Alice Mueni
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Tony Laboso
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Nancy Tamasha
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Irene Njuguna
- Department of Global Health, University of Washington, Seattle, WA
| | - Laurén Gómez
- Department of Global Health, University of Washington, Seattle, WA
| | - Dalton C Wamalwa
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Kenneth Tapia
- Department of Global Health, University of Washington, Seattle, WA
| | | | - Paul Bangirana
- Department of Psychiatry, Makerere University, Kampala, Uganda
| | - Michael J Boivin
- Departments of Psychiatry and of Neurology & Ophthalmology, Michigan State University, East Lansing
- Department of Psychiatry, University of Michigan, Ann Arbor, MI
| | - Grace C John-Stewart
- Department of Global Health, University of Washington, Seattle, WA
- Departments of Pediatrics
- Epidemiology; and
- Medicine, University of Washington, Seattle, WA
| |
Collapse
|
31
|
Glenn Fowler M, Aizire J, Sikorskii A, Atuhaire P, Wambuzi Ogwang L, Mutebe A, Katumbi C, Maliwichi L, Familiar I, Taha T, Boivin MJ. Growth deficits in antiretroviral and HIV-exposed uninfected versus unexposed children in Malawi and Uganda persist through 60 months of age. AIDS 2022; 36:573-582. [PMID: 34750297 PMCID: PMC9097628 DOI: 10.1097/qad.0000000000003122] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To compare childhood physical growth among antiretroviral drug and maternal HIV-exposed uninfected (AHEU) compared with HIV-unexposed uninfected (HUU) children. DESIGN Longitudinal follow-up of PROMISE trial (NCT01061151) AHEU and age-matched and sex-matched HUU children, enrolled (September 2013 to October 2014) in Malawi and Uganda. METHOD We compared WHO population standardized z-scores [height-for-age (HAZ), weight-for-age (WAZ), weight-for-height (WHZ), head-circumference-for-age (HCAZ) at 12, 24, 36, 48, and 60 months of age]. We evaluated HUU versus AHEU [in-utero combination antiretroviral treatment (cART) versus Zidovudine (ZDV) alone]; stratified by country, using longitudinal linear and generalized linear mixed models. RESULTS Of 466 Malawian and 477 Ugandan children, median maternal age at enrollment was 24.5 years (Malawi) and 27.8 years (Uganda); more than 90% were breastfed through 12 months except Uganda AHEU (64.0%). HAZ scores (adjusted for maternal age, breastfed, and socioeconomic status) were lower among AHEU versus HUU children at every time point, significant (P < 0.05) among Ugandan but not Malawian children. Similar patterns were seen for WAZ but not for WHZ or HCAZ scores. High stunting was observed in both countries, significantly higher in Malawi; and higher among AHEU versus HUU children through 48 months of age, significantly (P < 0.05) among Ugandan but not Malawian children. We found no differences in childhood growth trajectories with in-utero exposures to ZDV compared with cART. CONCLUSION AHEU versus HUU children had lower median LAZ and WAZ scores persisting through 60 months of age. However, proportions of children with stunting or underweight decreased after 24 months of age.
Collapse
Affiliation(s)
| | - Jim Aizire
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
| | - Alla Sikorskii
- Department of Statistics & Probability, Michigan State University
- Department of Psychiatry, Michigan State University
| | | | | | - Alex Mutebe
- Makerere U.-Johns Hopkins U. Research Collaboration
| | - Chaplain Katumbi
- College of Medicine – Johns Hopkins University Research Project, Blantyre, Malawi
| | - Limbika Maliwichi
- College of Medicine – Johns Hopkins University Research Project, Blantyre, Malawi
| | | | - Taha Taha
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
| | - Michael J. Boivin
- Department of Statistics & Probability, Michigan State University
- Department of Psychiatry, Michigan State University
- Department of Neurology & Ophthalmology, Michigan State University
| | | |
Collapse
|
32
|
Joseph Davey DL, Le Roux SM, Brittain K, Dovell K, Shoptaw S, Miller AP, Phillips TK, Zerbe A, Abrams EJ, Myer L. Alcohol use and intimate partner violence in HIV-uninfected pregnant women in Cape Town, South Africa. AIDS Care 2022; 34:214-219. [PMID: 34495777 PMCID: PMC8857014 DOI: 10.1080/09540121.2021.1975626] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 08/25/2021] [Indexed: 02/03/2023]
Abstract
In settings with a high burden of HIV, pregnant women often experience a cluster of risk factors, including alcohol use and intimate partner violence (IPV). These interrelated risks are poorly understood among pregnant women at risk of HIV in sub-Saharan Africa. We aim to determine cross-sectional associations between pregnant women's alcohol use and victimization due to IPV in the HIV-Unexposed-Uninfected Mother-Infant Cohort Study in Cape Town, South Africa. Women who tested HIV-negative at first antenatal care (ANC) visit were followed to delivery. Trained interviewers collected demographic and psychosocial information, including recent alcohol use and experiences of IPV victimization. We assess the prevalence of alcohol use and associations with IPV using multivariable logistic regression. In 406 HIV-uninfected pregnant women (mean age = 28 years; mean gestational age = 21 weeks), 41 (10%) reported alcohol consumption in the past 12 months; 30/41 (73%) of these at hazardous levels. Any and hazardous alcohol use were associated with greater odds of reporting past year IPV (adjusted odds ratio [aOR] for hazardous use: 3.24, 95% CI = 1.11, 7.56; aOR for any alcohol use: 2.97, 95% CI = 1.19, 7.45). These data suggest the occurrence of overlapping HIV risk factors among pregnant women and may help design improved health interventions in this population.
Collapse
Affiliation(s)
- Dvora L Joseph Davey
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Observatory, South Africa
| | - Stanzi M Le Roux
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Observatory, South Africa
| | - Kirsty Brittain
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Observatory, South Africa
| | - Kathryn Dovell
- Division of Infectious Diseases, Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Steve Shoptaw
- Department of Family Medicine, Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Psychiatry, University of Cape Town, Cape Town, South Africa
| | - Amanda P Miller
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Tamsin K Phillips
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Observatory, South Africa
| | - Allison Zerbe
- Mailman School of Public Health, ICAP at Columbia University, Columbia University, New York, NY, USA
| | - Elaine J Abrams
- Department of Psychiatry, University of Cape Town, Cape Town, South Africa
- Mailman School of Public Health, ICAP at Columbia University, Columbia University, New York, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Observatory, South Africa
| |
Collapse
|
33
|
Wedderburn CJ, Groenewold NA, Roos A, Yeung S, Fouche JP, Rehman AM, Gibb DM, Narr KL, Zar HJ, Stein DJ, Donald KA. Early structural brain development in infants exposed to HIV and antiretroviral therapy in utero in a South African birth cohort. J Int AIDS Soc 2022; 25:e25863. [PMID: 35041774 PMCID: PMC8765561 DOI: 10.1002/jia2.25863] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022] Open
Abstract
Introduction There is a growing population of children who are HIV‐exposed and uninfected (HEU) with the successful expansion of antiretroviral therapy (ART) use in pregnancy. Children who are HEU are at risk of delayed neurodevelopment; however, there is limited research on early brain growth and maturation. We aimed to investigate the effects of in utero exposure to HIV/ART on brain structure of infants who are HEU compared to HIV‐unexposed (HU). Methods Magnetic resonance imaging using a T2‐weighted sequence was undertaken in a subgroup of infants aged 2–6 weeks enrolled in the Drakenstein Child Health Study birth cohort, South Africa, between 2012 and 2015. Mother–child pairs received antenatal and postnatal HIV testing and ART per local guidelines. We compared subcortical and total grey matter volumes between HEU and HU groups using multivariable linear regression adjusting for infant age, sex, intracranial volume and socio‐economic variables. We further assessed associations between brain volumes with maternal CD4 cell count and ART exposure. Results One hundred forty‐six infants (40 HEU; 106 HU) with high‐resolution images were included in this analysis (mean age 3 weeks; 50.7% male). All infants who were HEU were exposed to ART (88% maternal triple ART). Infants who were HEU had smaller caudate volumes bilaterally (5.4% reduction, p < 0.05) compared to HU infants. There were no group differences in other subcortical volumes (all p > 0.2). Total grey matter volume was also reduced in infants who were HEU (2.1% reduction, p < 0.05). Exploratory analyses showed that low maternal CD4 cell count (<350 cells/mm3) was associated with decreased infant grey matter volumes. There was no relationship between timing of ART exposure and grey matter volumes. Conclusions Lower caudate and total grey matter volumes were found in infants who were HEU compared to HU in the first weeks of life, and maternal immunosuppression was associated with reduced volumes. These findings suggest that antenatal HIV exposure may impact early structural brain development and improved antenatal HIV management may have the potential to optimize neurodevelopmental outcomes of children who are HEU.
Collapse
Affiliation(s)
- Catherine J Wedderburn
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa.,Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK.,The Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Nynke A Groenewold
- The Neuroscience Institute, University of Cape Town, Cape Town, South Africa.,Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Annerine Roos
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa.,The Neuroscience Institute, University of Cape Town, Cape Town, South Africa.,SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | - Shunmay Yeung
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | - Jean-Paul Fouche
- The Neuroscience Institute, University of Cape Town, Cape Town, South Africa.,Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Andrea M Rehman
- MRC International Statistics & Epidemiology Group, London School of Hygiene & Tropical Medicine, London, UK
| | - Diana M Gibb
- MRC Clinical Trials Unit, University College London, London, UK
| | - Katherine L Narr
- Departments of Neurology, Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California, USA
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa.,SA MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- The Neuroscience Institute, University of Cape Town, Cape Town, South Africa.,Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.,SA MRC Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| | - Kirsten A Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa.,The Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
34
|
Nyemba DC, Kalk E, Vinikoor MJ, Madlala HP, Mubiana-Mbewe M, Mzumara M, Moore CB, Slogrove AL, Boulle A, Davies MA, Myer L, Powis K. Growth patterns of infants with in- utero HIV and ARV exposure in Cape Town, South Africa and Lusaka, Zambia. BMC Public Health 2022; 22:55. [PMID: 35000577 PMCID: PMC8744341 DOI: 10.1186/s12889-021-12476-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022] Open
Abstract
Background Infants born HIV-exposed yet remain uninfected (HEU) are at increased risk of poorer growth and health compared to infants born HIV-unexposed (HU). Whether maternal antiretroviral treatment (ART) in pregnancy ameliorates this risk of poorer growth is not well understood. Furthermore, whether risks are similar across high burden HIV settings has not been extensively explored. Methods We harmonized data from two prospective observational studies conducted in Cape Town, South Africa, and Lusaka, Zambia, to compare weight-for-age (WAZ), length-for-age (LAZ) and weight-for-length (WLZ) Z-scores between infants who were HEU and HU, converting infant anthropometric measures using World Health Organisation Growth Standards adjusted for age and sex. Linear mixed effects models were fit to identify risk factors for differences in anthropometrics at 6–10 weeks and 6 months by infant HIV exposures status and by timing of exposure to maternal ART, either from conception or later in gestation. Results Overall 773 mother-infant pairs were included across two countries: women living with HIV (WLHIV), 51% (n = 395) with 65% on ART at conception and 35% initiating treatment in pregnancy. In linear mixed effects models, WAZ and WLZ at 6–10 weeks were lower among infants who were HEU vs HU [β = − 0.29 (95% CI: − 0.46, − 0.12) and [β = − 0.42 (95% CI: − 0.68, − 0.16)] respectively after adjusting for maternal characteristics and infant feeding with a random intercept for country. At 6 months, LAZ was lower [β = − 0.28 CI: − 0.50, − 0.06)] among infants who were HEU, adjusting for the same variables, with no differences in WAZ and WLZ. Within cohort evaluations identified different results with higher LAZ among infants who were HEU from Zambia at 6–10 weeks, [β = + 0.34 CI: + 0.01, + 0.68)] and lower LAZ among infants who were HEU from South Africa [β = − 0.30 CI: − 0.59, − 0.01)] at 6 months, without other anthropometric differences at either site. Conclusion Infant growth trajectories differed by country, highlighting the importance of studying contextual influences on outcomes of infants who were HEU. Supplementary Information The online version contains supplementary material available at 10.1186/s12889-021-12476-z.
Collapse
Affiliation(s)
- Dorothy C Nyemba
- Division of Epidemiology & Biostatistics, Faculty of Health Sciences, School of Public Health and Family Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa. .,Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa.
| | - Emma Kalk
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Michael J Vinikoor
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia.,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hlengiwe P Madlala
- Division of Epidemiology & Biostatistics, Faculty of Health Sciences, School of Public Health and Family Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.,Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Maureen Mzumara
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Carolyn Bolton Moore
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia.,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amy L Slogrove
- Department of Paediatrics & Child Health, Faculty of Medicine & Health Sciences, Stellenbosch University, Worcester, South Africa.,Ukwanda Centre for Rural Health, Faculty of Medicine & Health Sciences, Stellenbosch University, Worcester, South Africa
| | - Andrew Boulle
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa.,Western Cape Government: Health, Cape Town, South Africa
| | - Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa.,Western Cape Government: Health, Cape Town, South Africa
| | - Landon Myer
- Division of Epidemiology & Biostatistics, Faculty of Health Sciences, School of Public Health and Family Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.,Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Kathleen Powis
- Department of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
35
|
Toledo G, Landes M, van Lettow M, Tippett Barr BA, Bailey H, Thorne C, Crichton S. No Difference in Growth Outcomes up to 24 Months of Age by Duration of Exposure to Maternal Antiretroviral Therapy Among Children Who Are HIV-Exposed and Uninfected in Malawi. Front Pediatr 2022; 10:882468. [PMID: 35795331 PMCID: PMC9251312 DOI: 10.3389/fped.2022.882468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/05/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND With the implementation of lifelong antiretroviral therapy (ART) for HIV treatment and prevention, the proportion of children exposed to ART in utero from conception is increasing. We estimated the effect of timing of ART exposure on growth of children HIV-exposed and uninfected (CHEU) up to Up to 24 months of age in Malawi. METHODS Data were collected from a prospective cohort of infants HIV-exposed aged 1-6 months (enrollment) and their mothers with HIV enrolled in the National Evaluation of Malawi's Prevention of Mother-to-Child Transmission of HIV Programme (2014-2018). Anthropometry was measured at enrollment, visit 1 (approximately 12 months), and visit 2 (approximately 24 months). Weight-for-age (WAZ) and length-for-age (LAZ) were calculated using the WHO Growth Standards. Multivariable mixed-effects models with linear splines for age were used to examine differences in growth by timing of ART exposure (from conception, first/second trimester, or third trimester/postpartum). Models were adjusted for confounders selected a priori guided by a conceptual framework. Hypothesized interactions and potential mediators were explored, and interactions with splines were included in final models if P < 0.1. RESULTS A total of 1,206 singleton CHEU and their mothers were enrolled and 563 completed the follow-up through 24 months of age. Moreover, 48% of CHEU were exposed to ART from conception, 40% from first/second trimester, and 12% from third trimester/postpartum. At enrollment, 12% of infants had low birthweight (LBW), 98% had been breastfed in past 7 days, and 57% were enrolled in an HIV care clinic. CHEU growth trajectories demonstrated cohort-wide growth faltering after the age of 12 months. Of 788 and 780 CHEU contributing to WAZ and LAZ multivariable models, respectively, there was no evidence of differences in mean WAZ or LAZ among those exposed from conception or first/second trimester vs. third trimester/postpartum and no evidence of a difference in WAZ or LAZ rate of change by timing of ART exposure (all interactions P > 1.0). CONCLUSION Reassuringly, ART exposure from conception was not associated with decreased WAZ or LAZ in CHEU Up 24 months of age. Overall growth trajectories suggest CHEU experience growth faltering after 12 months of age and may need support through and beyond the first 2 years of life.
Collapse
Affiliation(s)
- Gabriela Toledo
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Megan Landes
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Monique van Lettow
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | - Heather Bailey
- Institute for Global Health, University College London, London, United Kingdom
| | - Claire Thorne
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Siobhan Crichton
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| |
Collapse
|
36
|
Slogrove AL. It is a question of equity: time to talk about children who are HIV-exposed and "HIV-free". J Int AIDS Soc 2021; 24:e25850. [PMID: 34792840 PMCID: PMC8601192 DOI: 10.1002/jia2.25850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/02/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Amy L. Slogrove
- Department of Paediatrics & Child HealthFaculty of Medicine & Health SciencesStellenbosch UniversityWorcesterSouth Africa
| |
Collapse
|
37
|
Fourman LT, Mueller SB, Boutin A, Zheng I, Pan CS, Gerard ME, Stanley TL, Roberts DJ. Placental Vascular Abnormalities in Association With Prenatal and Long-Term Health Characteristics Among HIV-Exposed Uninfected Adolescents and Young Adults. J Acquir Immune Defic Syndr 2021; 88:103-109. [PMID: 34034303 PMCID: PMC8373807 DOI: 10.1097/qai.0000000000002734] [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: 02/06/2021] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND HIV-exposed uninfected (HEU) individuals are predisposed to adverse health outcomes, which in part may stem from the influence of an altered intrauterine milieu on fetal programming. The placenta serves as a readout for the effects of the maternal environment on the developing fetus and may itself contribute to the pathogenesis of disease. SETTING US academic health system. METHODS We leveraged a previously established registry-based cohort of HEU adolescents and young adults to identify 26 subjects for whom placental histopathology was available. We further obtained placental tissue from 29 HIV-unexposed pregnancies for comparison. We examined differences in placental histopathology between the groups and related villous vascularity in the HEU group to prenatal maternal characteristics and long-term health outcomes. RESULTS Placentas from HEU pregnancies demonstrated a higher blood vessel count per villus as compared with controls (5.9 ± 1.0 vs. 5.4 ± 0.8; P = 0.05), which was independent of maternal prenatal age, race, body mass index, smoking status, hemoglobin, and gestational age. Furthermore, within the HEU group, lower CD4+ T-cell count during pregnancy was associated with greater placental vascularity (r = -0.44; P = 0.03). No significant relationships were observed between placental blood vessel count per villus and body mass index z-score or reactive airway disease among HEU individuals later in life. CONCLUSIONS Placentas from HEU pregnancies demonstrated increased villous vascularity compared with HIV-unexposed controls in proportion to the severity of maternal immune dysfunction. Further studies are needed to examine intrauterine exposure to hypoxia as a potential mechanism of fetal programming in HIV.
Collapse
Affiliation(s)
- Lindsay T. Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Sarah B. Mueller
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Autumn Boutin
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Isabel Zheng
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Chelsea S. Pan
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Marisa E. Gerard
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Takara L. Stanley
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Pediatric Endocrinology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Drucilla J. Roberts
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
38
|
Stranix-Chibanda L, Tierney C, Pinilla M, George K, Aizire J, Chipoka G, Mallewa M, Naidoo M, Nematadzira T, Kusakara B, Violari A, Mbengeranwa T, Njau B, Fairlie L, Theron G, Mubiana-Mbewe M, Khadse S, Browning R, Fowler MG, Siberry GK. Effect on growth of exposure to maternal antiretroviral therapy in breastmilk versus extended infant nevirapine prophylaxis among HIV-exposed perinatally uninfected infants in the PROMISE randomized trial. PLoS One 2021; 16:e0255250. [PMID: 34415933 PMCID: PMC8378741 DOI: 10.1371/journal.pone.0255250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/04/2021] [Indexed: 11/25/2022] Open
Abstract
Background Malnutrition is highly prevalent in HIV-exposed perinatally uninfected infants (HEUs) increasing the risk of morbidity and mortality throughout the life course. We set out to compare the effect of postnatal exposure to maternal antiretroviral therapy (mART) in breastmilk versus infant Nevirapine prophylaxis (iNVP) on somatic growth of HEUs in the randomized PROMISE trial. Methods and findings We randomized 2431 mothers with HIV and their 2444 HEUs from six African countries and India 6–14 days after delivery to mART or iNVP for prevention of breastmilk HIV transmission. The mART regimen contained tenofovir/emtricitabine (99%) plus lopinavir/ritonavir. Infant growth parameters were compared at postnatal week 10, 26, 74 and 104 using World Health Organization (WHO) z-scores for length-for-age (LAZ), weight-for-age (WAZ), and head circumference-for-age (HCAZ). Week 26 LAZ was the primary endpoint measure. Student T-tests compared mean LAZ, WAZ, and HCAZ; estimated mean and 95% confidence interval (CI) are presented. Maternal and infant baseline characteristics were comparable between study arms. The estimated median breastfeeding duration was 70 weeks. After a mean follow-up of 88 weeks, mean LAZ and WAZ were below the WHO reference population mean at all timepoints, whereas mean HCAZ was not. The mART and iNVP arms did not differ for the primary outcome measure of LAZ at week 26 (p-value = 0.39; estimated mean difference (95%CI) of -0.05 (-0.18, 0.07)) or any of the other secondary growth outcome measures or timepoints (all p-values≥0.16). Secondary analyses of the primary outcome measure adjusting for week 0 LAZ and other covariates did not change these results (all p-values≥0.09). However, infants assigned to mART were more likely to have stunting compared to iNVP infants at week 26 (odds ratio (95% CI): 1.28 (1.05, 1.57)). Conclusions In HEUs, growth effects from postnatal exposure to mART compared to iNVP were comparable for measures on length, weight and head circumference with no clinically relevant differences between the groups. Despite breastfeeding into the second year of life, length and weight were below reference population means at all ages in both arms. Further investment is needed to optimize postnatal growth of infants born to women with HIV. Clinical trial registration ClinicalTrials.gov number NCT01061151.
Collapse
Affiliation(s)
- Lynda Stranix-Chibanda
- University of Zimbabwe Faculty of Medicine and Health Sciences, Child and Adolescent Health Unit, Harare, Zimbabwe
- University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
- * E-mail: ,
| | - Camlin Tierney
- Harvard T.H. Chan School of Public Health, Center for Biostatistics in AIDS Research in the Department of Biostatistics, Boston, MA, United States of America
| | - Mauricio Pinilla
- Harvard T.H. Chan School of Public Health, Center for Biostatistics in AIDS Research in the Department of Biostatistics, Boston, MA, United States of America
| | | | - Jim Aizire
- Makerere University—Johns Hopkins University Research Programme, Kampala, Uganda
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | | | | | - Megeshinee Naidoo
- University of KwaZulu-Natal, Centre Aids Prevention Research South Africa (CAPRISA), Durban, South Africa
| | | | - Bangani Kusakara
- University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
| | - Avy Violari
- Perinatal HIV Research Unit, Johannesburg, South Africa
| | - Tapiwa Mbengeranwa
- University of Zimbabwe Clinical Trials Research Centre, Harare, Zimbabwe
| | - Boniface Njau
- Kilimanjaro Christian Medical Center, Moshi, United Republic of Tanzania
| | - Lee Fairlie
- Wits Reproductive Health and HIV Institute, University of the Witwatersrand, Johannesburg, South Africa
| | - Gerard Theron
- Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town, South Africa
| | | | - Sandhya Khadse
- Department of Obstetrics and Gynaecology, BJ Government Medical College, Pune, India
| | - Renee Browning
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States of America
| | - Mary Glenn Fowler
- Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - George K. Siberry
- Office of HIV/AIDS, United States Agency for International Development, Washington, DC, United States of America
| | | |
Collapse
|
39
|
Madlala HP, Steyn NP, Kalk E, Davies MA, Nyemba D, Malaba TR, Mehta U, Petro G, Boulle A, Myer L. Association between food intake and obesity in pregnant women living with and without HIV in Cape Town, South Africa: a prospective cohort study. BMC Public Health 2021; 21:1504. [PMID: 34348683 PMCID: PMC8335890 DOI: 10.1186/s12889-021-11566-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/24/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although global nutrition/dietary transition resulting from industrialisation and urbanisation has been identified as a major contributor to widespread trends of obesity, there is limited data in pregnant women, including those living with HIV in South Africa. We examined food-based dietary intake in pregnant women with and without HIV at first antenatal care (ANC) visit, and associations with maternal overweight/obesity and gestational weight gain (GWG). METHODS In an urban South African community, consecutive women living with (n = 479) and without (n = 510) HIV were enrolled and prospectively followed to delivery. Interviewer-administered non-quantitative food frequency questionnaire was used to assess dietary intake (starch, protein, dairy, fruits, vegetables, legumes, oils/fats) at enrolment. Associations with maternal body mass index (BMI) and GWG were examined using logistic regression models. RESULTS Among women (median age 29 years, IQR 25-34), the prevalence of obesity (BMI ≥ 30 kg/m2) at first ANC was 43% and that of excessive GWG (per IOM guidelines) was 37% overall; HIV prevalence was 48%. In women without HIV, consumption of potato (any preparation) (aOR 1.98, 95% CI 1.02-3.84) and pumpkin/butternut (aOR 2.13, 95% CI 1.29-3.49) for 1-3 days a week increased the odds of overweight/obesity compared to not consuming any; milk in tea/coffee (aOR 6.04, 95% CI 1.37-26.50) increased the odds of excessive GWG. Consumption of eggs (any) (aOR 0.52, 95% CI 0.32-0.86) for 1-3 days a week reduced the odds of overweight/obesity while peanut and nuts consumption for 4-7 days a week reduced the odds (aOR 0.34, 95% CI 0.14-0.80) of excessive GWG. In women with HIV, consumption of milk/yoghurt/maas to drink/on cereals (aOR 0.35, 95% CI 0.18-0.68), tomato (raw/cooked) (aOR 0.50, 95% CI 0.30-0.84), green beans (aOR 0.41, 95% CI 0.20-0.86), mixed vegetables (aOR 0.49, 95% CI 0.29-0.84) and legumes e.g. baked beans, lentils (aOR 0.50, 95% CI 0.28-0.86) for 4-7 days a week reduced the odds of overweight/obesity; tomato (raw/cooked) (aOR 0.48, 95% CI 0.24-0.96) and mixed vegetables (aOR 0.38, 95% CI 0.18-0.78) also reduced the odds of excessive GWG. CONCLUSIONS Diet modification may promote healthy weight in pregnant women living with and without HIV.
Collapse
Affiliation(s)
- Hlengiwe P Madlala
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Falmouth Building, Anzio Road, Observatory, Cape Town, Western Cape, 7925, South Africa.
| | - Nelia P Steyn
- Division of Human Nutrition, Department of Human Biology, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Emma Kalk
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Mary-Anne Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, Western Cape, South Africa
- Health Impact Assessment Unit, Western Cape Department of Health, Cape Town, South Africa
| | - Dorothy Nyemba
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Falmouth Building, Anzio Road, Observatory, Cape Town, Western Cape, 7925, South Africa
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Thokozile R Malaba
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Falmouth Building, Anzio Road, Observatory, Cape Town, Western Cape, 7925, South Africa
| | - Ushma Mehta
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, Western Cape, South Africa
| | - Gregory Petro
- Department of Obstetrics and Gynaecology, University of Cape Town and New Somerset Hospital, Cape Town, Western Cape, South Africa
| | - Andrew Boulle
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, Western Cape, South Africa
- Health Impact Assessment Unit, Western Cape Department of Health, Cape Town, South Africa
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Falmouth Building, Anzio Road, Observatory, Cape Town, Western Cape, 7925, South Africa
| |
Collapse
|
40
|
Abstract
Advances in perinatal HIV management have averted a significant number of infections in neonates and have made the possibility of elimination of mother-to-child transmission a reality; however, significant gaps in implementation of early testing programs as well as the expansion of therapeutic strategies to neonates are hindering prevention efforts and access to safer, more effective and easier to administer treatment. This article provides insights on the current state of perinatal HIV, recent advances, and future needs.
Collapse
Affiliation(s)
- Andres F Camacho-Gonzalez
- Division of Pediatric Infectious Diseases, Children's Healthcare of Atlanta, Emory University School of Medicine, 2015 Uppergate Drive, Suite 500, Atlanta, GA 30322, USA.
| | - Paul Palumbo
- Section of Infectious Diseases and International Health, Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 03756, USA
| |
Collapse
|
41
|
Mitochondrial DNA Instability Is Common in HIV-Exposed Uninfected Newborns. J Clin Med 2021; 10:jcm10112399. [PMID: 34071681 PMCID: PMC8197798 DOI: 10.3390/jcm10112399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 01/13/2023] Open
Abstract
Worldwide, one million HIV-exposed uninfected (HEU) children are born yearly, and chronic health impairments have been reported in these children. Mitochondrial DNA (mtDNA) instability and altered mtDNA content have been evidenced in these children, but an exhaustive characterization of altered mitochondrial genomes has never been reported. We applied deep mtDNA sequencing coupled to the deletion identification algorithm eKLIPse to the blood of HEU neonates (n = 32), which was compared with healthy controls (n = 15). Dried blood spots (DBS) from African HEU children were collected seven days after birth between November 2009 and May 2012. DBS from French healthy controls were collected at birth (or <3 days of life) in 2012 and in 2019. In contrast to the absence of mtDNA instability observed at the nucleotide level, we identified significant amounts of heteroplasmic mtDNA deletions in 75% of HEU children and in none of controls. The heteroplasmy rate of the 62 mtDNA deletions identified varied from 0.01% to up to 50%, the highest rates being broadly compatible with bioenergetic defect and clinical expression. mtDNA integrity is commonly affected in HEU neonates. The nature of the deletions suggests a mechanism related to aging or tumor-associated mtDNA instability. This child population may be at risk of additional mtDNA genetic alterations considering that they will be exposed to other mitotoxic drugs including antiretroviral or anti-tuberculosis treatment.
Collapse
|
42
|
Nyemba DC, Kalk E, Madlala HP, Malaba TR, Slogrove AL, Davies MA, Boulle A, Myer L, Powis KM. Lower birth weight-for-age and length-for-age z-scores in infants with in-utero HIV and ART exposure: a prospective study in Cape Town, South Africa. BMC Pregnancy Childbirth 2021; 21:354. [PMID: 33947351 PMCID: PMC8097797 DOI: 10.1186/s12884-021-03836-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/27/2021] [Indexed: 12/03/2022] Open
Abstract
Background Successful scale-up of antiretroviral therapy (ART) during pregnancy has minimized infant HIV acquisition, and over 1 million infants are born HIV-exposed but uninfected (HEU), with an increasing proportion also exposed in utero to maternal ART. While benefits of ART in pregnancy outweigh risks, some studies have reported associations between in utero ART exposure and impaired fetal growth, highlighting the need to identify the safest ART regimens for use in pregnancy. Methods We compared birth anthropometrics of infants who were HEU with those HIV-unexposed (HU) in Cape Town, South Africa. Pregnant women had gestational age assessed by ultrasound at enrolment. Women living with HIV were on ART (predominately tenofovir-emtricitabine-efavirenz) either prior to conception or initiated during pregnancy. Birth weights and lengths were converted to weight-for-age (WAZ) and length-for-age (LAZ) z-scores using Intergrowth-21st software. Linear regression was used to compare mean z-scores adjusting for maternal and pregnancy characteristics. Results Among 888 infants, 49% (n = 431) were HEU and 51% (n = 457) HU. Of 431 HEU infants, 62% (n = 268) were exposed to HIV and antiretrovirals (ARVs) from conception and 38% (n = 163) were exposed to ARVs during gestation but after conception (median fetal ARV exposure of 21 weeks [IQR; 17–26]). In univariable analysis, infants who were HEU had lower mean WAZ compared with HU [β = − 0.15 (95% Confidence Interval (CI): − 0.28, − 0.020)]. After adjustment for maternal age, gravidity, alcohol use, marital and employment status the effect remained [adjusted β − 0.14 (95%CI: − 0.28, − 0.01]. Similar differences were noted for mean LAZ in univariable [β − 0.20 (95%CI: − 0.42, − 0.01] but not multivariable analyses [adjusted β − 0.18 (95%CI: − 0.41, + 0.04] after adjusting for the same variables. Mean WAZ and LAZ did not vary by in utero ARV exposure duration among infants who were HEU. Conclusion In a cohort with high prevalence of ART exposure in pregnancy, infants who were HEU had lower birth WAZ compared with those HU. Studies designed to identify the mechanisms and clinical significance of these disparities, and to establish the safest ART for use in pregnancy are urgently needed. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-021-03836-z.
Collapse
Affiliation(s)
- Dorothy C Nyemba
- Division of Epidemiology & Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa. .,Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa.
| | - Emma Kalk
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Hlengiwe P Madlala
- Division of Epidemiology & Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.,Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Thokozile R Malaba
- Division of Epidemiology & Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.,Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Amy L Slogrove
- Department of Paediatrics & Child Health, Faculty of Medicine & Health Sciences, Stellenbosch University, Worcester, South Africa.,Ukwanda Centre for Rural Health, Faculty of Medicine & Health Sciences, Stellenbosch University, Worcester, South Africa
| | - Mary-Ann Davies
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa.,Western Cape Government: Health, Cape Town, South Africa
| | - Andrew Boulle
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa.,Western Cape Government: Health, Cape Town, South Africa
| | - Landon Myer
- Division of Epidemiology & Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.,Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Kathleen M Powis
- Department of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| |
Collapse
|
43
|
Sirajee R, Conroy AL, Namasopo S, Opoka RO, Lavoie S, Forgie S, Salami BO, Hawkes MT. Growth Faltering and Developmental Delay in HIV-Exposed Uninfected Ugandan Infants: A Prospective Cohort Study. J Acquir Immune Defic Syndr 2021; 87:730-740. [PMID: 33819206 DOI: 10.1097/qai.0000000000002626] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/30/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND HIV-exposed but uninfected (HEU) infants are at increased risk of impaired early linear growth and cognitive development. We examined associations between prenatal and postnatal growth and subsequent neurodevelopment in Ugandan HEU infants, hypothesizing that early insults may explain alterations in both somatic growth and brain development. METHODS We prospectively followed a cohort of HEU infants from birth to 18 months of age, and measured length/height, weight, head, and arm circumference longitudinally. The Malawi Development Assessment Tool (MDAT, 12 and 18 months) and the Color Object Association Test (18 months) were used for developmental assessments. RESULTS Among 170 HEU infants, the prevalence of low-birth weight and failure to thrive was 7.6% and 37%, respectively. HEU infants had MDAT scores that were similar to the reference population. The mean (SD) score on the Color Object Association Test was 5.5 (3.1) compared with 6.9 (5.3) in developmentally normal children. Developmental ability at age 18 months showed strong cross-sectional correlation with weight-for-age (ρ = 0.36, P < 0.0001), length/height-for-age (ρ = 0.41, P < 0.0001), head circumference-for-age (ρ = 0.26, P = 0.0011), and mid-upper arm circumference-for-age (ρ = 0.34, P = 0.0014). There was a statistically significant correlation between birth weight and MDAT z-score at 18 months (ρ = 0.20, P = 0.010). Failure to thrive was associated with lower MDAT z-score [median -0.13 (IQR -0.75 to +0.14) versus +0.14 (IQR -0.44 to +0.63), P = 0.042]. CONCLUSION Growth faltering in HEU infants was associated with lower attainment of developmental milestones at age 18 months. Our findings point to a simple screening method for identifying HEU infants at risk for developmental intervention.
Collapse
Affiliation(s)
- Reshma Sirajee
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Andrea L Conroy
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN
| | - Sophie Namasopo
- Department of Paediatrics, Jinja Regional Referral Hospital, Jinja, Uganda
| | - Robert O Opoka
- Department of Paediatrics and Child Health, Mulago Hospital and Makerere University, Kampala, Uganda
| | - Stephanie Lavoie
- National Laboratory for HIV Reference Services (NLHRS), National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Sarah Forgie
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Bukola O Salami
- Women and Children's Health Research Institute; and
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
| | - Michael T Hawkes
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute; and
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- Department of Global Health, School of Public Health, University of Alberta, Edmonton, AB, Canada
- Distinguished Researcher, Stollery Science Lab
| |
Collapse
|
44
|
Slogrove AL, Powis KM. Widening the Lens to Ensure Children Who Are Human Immunodeficiency Virus (HIV) Exposed Are Alive, HIV Free, and Thriving. Clin Infect Dis 2021; 72:595-597. [PMID: 31974581 DOI: 10.1093/cid/ciaa079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 11/12/2022] Open
Affiliation(s)
- Amy L Slogrove
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Worcester, South Africa
- Ukwanda Centre for Rural Health, Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Worcester, South Africa
| | - Kathleen M Powis
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Departments of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| |
Collapse
|
45
|
Pillay L, Moodley D, Emel LM, Nkwanyana NM, Naidoo K. Growth patterns and clinical outcomes in association with breastfeeding duration in HIV exposed and unexposed infants: a cohort study in KwaZulu Natal, South Africa. BMC Pediatr 2021; 21:183. [PMID: 33874900 PMCID: PMC8054353 DOI: 10.1186/s12887-021-02662-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
Background Exclusive breastfeeding for 6 months and breastfeeding with complementary feeds until 12 months for HIV exposed and uninfected (HEU) infants or 24 months for HIV unexposed (HU) infants is the current World Health Organisation (WHO) recommendation for low and middle income countries (LMICs) to improve clinical outcomes and growth trajectories in infants. In a post-hoc evaluation of HEU and HU cohorts, we examine growth patterns and clinical outcomes in the first 9 months of infancy in association with breastfeeding duration. Methods Two cohorts of infants, HEU and HU from a low-socioeconomic township in South Africa, were evaluated from birth until 9 months of age. Clinical, anthropometric and infant feeding data were analysed. Standard descriptive statistics and regression analysis were performed to determine the effect of HIV exposure and breastfeeding duration on growth and clinical outcomes. Results Included in this secondary analysis were 123 HEU and 157 HU infants breastfed for a median of 26 and 14 weeks respectively. Median WLZ score was significantly (p < 0.001) lower in HEU than HU infants at 3, 6 and 9 months (− 0.19 vs 2.09; − 0.81 vs 0.28; 0.05 vs 0.97 respectively). The median LAZ score was significantly lower among HU infants at 3 and 6 months (− 1.63 vs 0.91, p < 0.001; − 0.37 vs 0.51, p < 0.01) and a significantly higher proportion of HU was classified as stunted (LAZ < -2SD) at 3 and 6 months (3.9% vs 44.9%, p < 0.001; 4.8% vs 20.9%, p < 0.001 respectively) independent of breastfeeding duration. A higher proportion of HEU infants experienced one or more episodes of skin rash (44.5% vs 12.8%) and upper respiratory tract infection (URTI) (30.1% vs 10.9%) (p < 0.0001). In a multivariable analysis, the odds of occurrence of wasting, skin rash, URTI or any clinical adverse event in HEU infants were 2.86, 7.06, 3.01 and 8.89 times higher than HU infants after adjusting for breastfeeding duration. Conclusion Our study has generated additional evidence that HEU infants are at substantial risk of infectious morbidity and decreased growth trajectories however we have further demonstrated that these adverse outcomes were independent of breastfeeding duration.
Collapse
Affiliation(s)
- Larisha Pillay
- Department of Paediatrics and Child Health, School of Clinical Medicine, University of KwaZulu-Natal, 719 Umbilo Road, Congella, 4013, South Africa
| | - Dhayendre Moodley
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, 719 Umbilo Road, Congella, 4013, South Africa.
| | - Lynda Marie Emel
- Biostatistics, Bioinformatics, and Epidemiology/VIDD, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Kimesh Naidoo
- Department of Paediatrics and Child Health, School of Clinical Medicine, University of KwaZulu-Natal, 719 Umbilo Road, Congella, 4013, South Africa
| |
Collapse
|
46
|
Evans C, Chasekwa B, Ntozini R, Majo FD, Mutasa K, Tavengwa N, Mutasa B, Mbuya MNN, Smith LE, Stoltzfus RJ, Moulton LH, Humphrey JH, Prendergast AJ. Mortality, Human Immunodeficiency Virus (HIV) Transmission, and Growth in Children Exposed to HIV in Rural Zimbabwe. Clin Infect Dis 2021; 72:586-594. [PMID: 31974572 PMCID: PMC7884806 DOI: 10.1093/cid/ciaa076] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 01/23/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Clinical outcomes of children who are human immunodeficiency virus (HIV)-exposed in sub-Saharan Africa remain uncertain. METHODS The Sanitation Hygiene Infant Nutrition Efficacy (SHINE) trial evaluated improved infant and young child feeding (IYCF) and/or improved water, sanitation, and hygiene in 2 rural Zimbabwean districts with 15% antenatal HIV prevalence and > 80% prevention of mother-to-child transmission (PMTCT) coverage. Children born between February 2013 and December 2015 had longitudinal HIV testing and anthropometry. We compared mortality and growth between children who were HIV-exposed and HIV-unexposed through 18 months. Children receiving IYCF were excluded from growth analyses. RESULTS Fifty-one of 738 (7%) children who were HIV-exposed and 198 of 3989 (5%) children who were HIV-unexposed (CHU) died (hazard ratio, 1.41 [95% confidence interval {CI}, 1.02-1.93]). Twenty-five (3%) children who were HIV-exposed tested HIV positive, 596 (81%) were HIV-exposed uninfected (CHEU), and 117 (16%) had unknown HIV status by 18 months; overall transmission estimates were 4.3%-7.7%. Mean length-for-age z score at 18 months was 0.38 (95% CI, .24-.51) standard deviations lower among CHEU compared to CHU. Among 367 children exposed to HIV in non-IYCF arms, 147 (40%) were alive, HIV-free, and nonstunted at 18 months, compared to 1169 of 1956 (60%) CHU (absolute difference, 20% [95% CI, 15%-26%]). CONCLUSIONS In rural Zimbabwe, mortality remains 40% higher among children exposed to HIV, vertical transmission exceeds elimination targets, and half of CHEU are stunted. We propose the composite outcome of "alive, HIV free, and thriving" as the long-term goal of PMTCT programs. CLINICAL TRIALS REGISTRATION NCT01824940.
Collapse
Affiliation(s)
- Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Batsirai Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Mduduzi N N Mbuya
- Global Alliance for Improved Nutrition, Washington, District of Columbia, USA
| | - Laura E Smith
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | | | - Lawrence H Moulton
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | |
Collapse
|
47
|
Gilmore JC, Serghides L, Bendayan R. Differential effects of antiretroviral drug toxicity in male versus female children who are HIV-exposed but uninfected. AIDS 2021; 35:1-14. [PMID: 33048885 DOI: 10.1097/qad.0000000000002707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
: In recent years, widespread use of antiretroviral therapy (ART) during pregnancy has been increasingly effective in reducing risk of vertical transmission of HIV, with over 80% of pregnant women living with HIV now accessing ART, and a 41% reduction in new infections in children between 2010 and 2018. Despite these strides, the developmental toxicity of widely administered antiretroviral drugs (ARVs) remains poorly described and existing literature often fails to account for fetal and infant sex as a variable. Recent reports have identified associations between in-utero exposure to commonly used antiretroviral regimens and alteration in neurodevelopment, growth, and metabolism amongst children who are HIV-exposed but uninfected, with findings of sex differences in the prevalence and severity of ARV toxicity. These differences are potentially explained by variable exposure to ARV drugs in utero or exacerbation of existing sex-linked risk factors. Fetal ARV exposure is mediated by placental and fetal drug transporters and metabolic enzymes, which may contribute to the manifestation of sex differences. Existing evidence of sex differences in ARV toxicity in fetal development is concerning, and demands further research to guide optimal treatment options for maternal health and prevention of vertical HIV transmission.
Collapse
Affiliation(s)
| | - Lena Serghides
- Toronto General Hospital Research Institute, University Health Network (UHN)
- Department of Immunology and Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, University of Toronto
| |
Collapse
|
48
|
Kapito-Tembo AP, Bauleni A, Wesevich A, Ongubo D, Hosseinipour MC, Dube Q, Mwale P, Corbett A, Mwapasa V, Phiri S. Growth and Neurodevelopment Outcomes in HIV-, Tenofovir-, and Efavirenz-Exposed Breastfed Infants in the PMTCT Option B+ Program in Malawi. J Acquir Immune Defic Syndr 2021; 86:81-90. [PMID: 33027153 DOI: 10.1097/qai.0000000000002515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pregnant and breastfeeding women in Option B+ in Malawi received antiretroviral drugs (ARVs) containing efavirenz (EFV) and tenofovir disoproxil fumarate (TDF). However, effects on growth, renal, bone metabolism, and neurodevelopment of long-term exposure to low doses of these drugs through breast milk in HIV-exposed infants are unclear. METHODS Prospective cohorts of TDF-and-EFV-exposed and TDF-and-EFV-unexposed breastfed infants of HIV-infected and HIV-uninfected mothers in Option B+ were recruited in 2:1 ratio, respectively, followed from birth to 18 months. Infants with low birth weight, premature birth, and congenital abnormalities were excluded. Anthropometrics were assessed at birth, 6 weeks, 3, 6, 12, and 18 months. Neurodevelopment assessments used the Bayley Scales of Infant and Toddler Development III from 6 weeks. Creatinine, alkaline phosphatase, and phosphorus were assessed at 3, 6, and 12 months. RESULTS Of 260 HIV-and-ARV-exposed and 125 HIV-and-ARV-unexposed infants enrolled at birth, 87% and 57%, 78% and 59%, 77% and 54%, 73% and 51%, and 65% and 43% completed 6-weeks, 3, 6, 12, and 18 months visits, respectively. There were no significant differences in the mean Z-scores for length-for-age, weight-for-age, weight-for-length, mid-upper arm circumference-for-age, and head circumference-for-age between groups except at 6-weeks for length-for-age. No bone fractures occurred. Neurodevelopment outcomes were similar between groups. Of creatinine, alkaline phosphatase, and serum phosphate measurements, 1.7%, 2.6%, and 3.3% reached any toxicity levels grades 1-4, respectively, with no differences between groups. CONCLUSION Long-term exposure to EFV and TDF through breastfeeding in infants of HIV-infected mothers does not seem to result in significant growth, neurodevelopment, renal, or bone adverse outcomes. Data support safety of breastfeeding through 18 months within the Option B+ program.
Collapse
Affiliation(s)
- Atupele P Kapito-Tembo
- MAC Communicable Diseases Action Centre, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Public Health, College of Medicine, School of Public Health and Family Medicine, University of Malawi, Malawi
| | - Andy Bauleni
- MAC Communicable Diseases Action Centre, University of Malawi College of Medicine, Blantyre, Malawi
| | | | | | - Mina C Hosseinipour
- UNC Project-Malawi, Lilongwe, Malawi
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Queen Dube
- Ministry of Health Queen Elizabeth Central Hospital, Blantyre, Malawi
| | - Patrick Mwale
- MAC Communicable Diseases Action Centre, University of Malawi College of Medicine, Blantyre, Malawi
| | - Amanda Corbett
- UNC Eshleman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Victor Mwapasa
- Department of Public Health, College of Medicine, School of Public Health and Family Medicine, University of Malawi, Malawi
| | - Sam Phiri
- Department of Public Health, College of Medicine, School of Public Health and Family Medicine, University of Malawi, Malawi
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Lighthouse Trust, Kamuzu Central Hospital, Lilongwe, Malawi ; and
- Department of Global Health, University of Washington, Seattle, WA
| |
Collapse
|
49
|
Bengtson AM, Phillips TK, le Roux SM, Brittain K, Zerbe A, Madlala HP, Malaba TR, Petro G, Abrams EJ, Myer L. High blood pressure at entry into antenatal care and birth outcomes among a cohort of HIV-uninfected women and women living with HIV initiating antiretroviral therapy in South Africa. Pregnancy Hypertens 2020; 23:79-86. [PMID: 33285444 DOI: 10.1016/j.preghy.2020.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To examine associations between high blood pressure (BP) when entering antenatal care (ANC) and birth outcomes in a cohort of pregnant HIV- and women living with HIV (WLHIV) initiating antiretroviral treatment (ART). STUDY DESIGN Prospective cohort study. MAIN OUTCOME MEASURES Cesarean delivery, preterm birth (<37 weeks' gestation), low birthweight (LBW, <2500 g), small-for-gestational age (SGA, <10th percentile), and large-for-gestational age (LGA, >10th percentile for GA). RESULTS Of 1116 women (median GA 20 weeks; WLHIV 53%), 48% (53% WLHIV; 43% HIV-) entered ANC with high BP, defined as elevated (120-129 or < 80 mmHg), stage 1 (>130-139 or 80-89) or stage 2 hypertension (≥140 / or ≥ 90). WLHIV were more likely to have high BP (RR 1.32; 95%CI 1.12-1.57), controlling for pre-pregnancy body mass index and additional confounders. In multivariable analysis, there was no evidence that high BP increased the risk of cesarean delivery (RR 1.10, 95% CI 0.92-1.30), preterm birth (RR 1.15, 95% CI 0.81-1.62), LBW (RR 1.16, 95% CI 0.84-1.60) or SGA (RR 1.02, 0.72-1.44), overall or when stratified by HIV-status. High BP was associated with an increased risk of LGA (RR 1.43; 95% CI 1.00-2.03). CONCLUSION In this setting, half of women had high BP at entry into ANC, with WLHIV at increased risk of high BP. There was no strong evidence that high BP increased the risk of adverse birth outcomes overall, or by HIV-status, with the exception of LGA. WLHIV may be at high risk of high BP during pregnancy and should be monitored closely.
Collapse
Affiliation(s)
- Angela M Bengtson
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA.
| | - Tamsin K Phillips
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa; Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Stanzi M le Roux
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Kirsty Brittain
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa; Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Allison Zerbe
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Hlengiwe P Madlala
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Thokozile R Malaba
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa; Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Gregory Petro
- Department of Obstetrics & Gynaecology, University of Cape Town and New Somerset Hospital, Cape Town, South Africa
| | - Elaine J Abrams
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, NY, USA; Department of Pediatrics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, United States
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa; Centre for Infectious Disease Epidemiology & Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
50
|
Sarkar A, Balogun K, Guzman Lenis MS, Acosta S, Mount HT, Serghides L. In utero exposure to protease inhibitor-based antiretroviral regimens delays growth and developmental milestones in mice. PLoS One 2020; 15:e0242513. [PMID: 33211746 PMCID: PMC7676697 DOI: 10.1371/journal.pone.0242513] [Citation(s) in RCA: 6] [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: 07/01/2020] [Accepted: 11/03/2020] [Indexed: 12/26/2022] Open
Abstract
Antiretroviral therapy (ART) in pregnancy has dramatically reduced HIV vertical transmission rates. Consequently, there is a growing number of children that are HIV exposed uninfected (CHEUs). Studies suggest that CHEUs exposed in utero to ART may experience developmental delays compared to their peers. We investigated the effects of in utero ART exposure on perinatal neurodevelopment in mice, through assessment of developmental milestones. Developmental milestone tests (parallel to reflex testing in human infants) are reflective of brain maturity and useful in predicting later behavioral outcomes. We hypothesized that ART in pregnancy alters the in utero environment and thereby alters developmental milestone outcomes in pups. Throughout pregnancy, dams were treated with boosted-atazanavir combined with either abacavir/lamivudine (ATV/r/ABC/3TC), or tenofovir/emtricitabine (ATV/r/TDF/FTC), or water as control. Pups were assessed daily for general somatic growth and on a battery of tests for primitive reflexes including surface-righting, negative-geotaxis, cliff-aversion, rooting, ear-twitch, auditory-reflex, forelimb-grasp, air-righting, behaviors in the neonatal open field, and olfactory test. In utero exposure to either ART regimen delayed somatic growth in offspring and evoked significant delays in the development of negative geotaxis, cliff-aversion, and ear-twitch reflexes. Exposure to ATV/r/ABC/3TC was also associated with olfactory deficits in male and forelimb grasp deficits in female pups. To explore whether delays persisted into adulthood we assessed performance in the open field test. We observed no significant differences between treatment arm for males. In females, ATV/r/TDF/FTC exposure was associated with lower total distance travelled and less ambulatory time in the centre, while ATV/r/ABC/3TC exposure was associated with higher resting times compared to controls. In utero PI-based ART exposure delays the appearance of primitive reflexes that involve vestibular and sensory-motor pathways in a mouse model. Our findings suggest that ART could be disrupting the normal progress/maturation of the underlying neurocircuits and encourage further investigation for underlying mechanisms.
Collapse
Affiliation(s)
- Ambalika Sarkar
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kayode Balogun
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Monica S. Guzman Lenis
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Sebastian Acosta
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Howard T. Mount
- Departments of Psychiatry & Physiology, Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Lena Serghides
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
- Women’s College Research Institute, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|