1
|
Dadabhai S, Chou VB, Pinilla M, Chinula L, Owor M, Violari A, Moodley D, Stranix-Chibanda L, Matubu TA, Chareka GT, Theron G, Kinikar AA, Mubiana-Mbewe M, Fairlie L, Bobat R, Mmbaga BT, Flynn PM, Taha TE, McCarthy KS, Browning R, Mofenson LM, Brummel SS, Fowler MG. Effects of preterm birth, maternal ART and breastfeeding on 24-month infant HIV-free survival in a randomized trial. AIDS 2024; 38:1304-1313. [PMID: 38427596 PMCID: PMC11211053 DOI: 10.1097/qad.0000000000003878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND IMPAACT 1077BF/FF (PROMISE) compared the safety/efficacy of two HIV antiretroviral therapy (ART) regimens to zidovudine (ZDV) alone during pregnancy for HIV prevention. PROMISE found an increased risk of preterm delivery (<37 weeks) with antepartum triple ART (TDF/FTC/LPV+r or ZDV/3TC/LPV+r) compared with ZDV alone. We assessed the impact of preterm birth, breastfeeding, and antepartum ART regimen on 24-month infant survival. METHODS We compared HIV-free and overall survival at 24 months for liveborn infants by gestational age, time-varying breastfeeding status, and antepartum ART arm at 14 sites in Africa and India. Kaplan-Meier survival probabilities and Cox proportional hazards ratios were estimated. RESULTS Three thousand four hundred and eighty-two live-born infants [568 (16.3%) preterm and 2914 (83.7%) term] were included. Preterm birth was significantly associated with lower HIV-free survival [0.85; 95% confidence interval (CI) 0.82-0.88] and lower overall survival (0.89; 95% CI 0.86-0.91) versus term birth (0.96; 95% CI 0.95-0.96). Very preterm birth (<34 weeks) was associated with low HIV-free survival (0.65; 95% CI 0.54-0.73) and low overall survival (0.66; 95% CI 0.56-0.74). Risk of HIV infection or death at 24 months was higher with TDF-ART than ZDV-ART (adjusted hazard ratio 2.37; 95% CI 1.21-4.64). Breastfeeding initiated near birth decreased risk of infection or death at 24 months (adjusted hazard ratio 0.05; 95% CI 0.03-0.08) compared with not breastfeeding. CONCLUSION Preterm birth and antepartum TDF-ART were associated with lower 24-month HIV-free survival compared with term birth and ZDV-ART. Any breastfeeding strongly promoted HIV-free survival, especially if initiated close to birth. Reducing preterm birth and promoting infant feeding with breastmilk among HIV/antiretroviral drug-exposed infants remain global health priorities.
Collapse
Affiliation(s)
- Sufia Dadabhai
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
- Kamuzu University of Health Sciences-Johns Hopkins Research Project, Blantyre, Malawi
| | - Victoria B. Chou
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mauricio Pinilla
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Lameck Chinula
- Division of Global Women's Health, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- University of North Carolina Project Malawi, Tidziwe Centre, Lilongwe, Malawi
| | - Maxensia Owor
- MU-JHU Research Collaboration; Upper Mulago Hill Road, Kampala, Uganda
| | - Avy Violari
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Soweto
| | - Dhayendre Moodley
- Centre for the AIDS Programme of Research in South Africa and School of Clinical Medicine, University of KwaZulu Natal, Congella, South Africa
| | - Lynda Stranix-Chibanda
- Child, Adolescent and Women's Health Department, Faculty of Medicine and Health Sciences, University of Zimbabwe, Avondale
- University of Zimbabwe Clinical Trials Research Centre, Belgravia, Harare, Zimbabwe
| | - Taguma Allen Matubu
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | - Gerhard Theron
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | | | - Lee Fairlie
- Wits RHI, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg
| | - Raziya Bobat
- Department of Paediatrics and Child Health, University of KwaZulu-Natal, Durban, South Africa
| | - Blandina Theophil Mmbaga
- Kilimanjaro Christian Medical Centre, Kilimanjaro Clinical Research Institute and Kilimanjaro Christian Medical University College/Kilimanjaro CRS, Moshi, Tanzania
| | | | - Taha E. Taha
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
| | | | - Renee Browning
- National Institute of Allergy and Infectious Diseases/NIH, Rockville, MD
| | | | - Sean S. Brummel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Mary Glenn Fowler
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
2
|
Belete NK, Megersa ND, Hebo SH, Animut MD, Tariku EZ. Feeding modalities, HIV transmission and its predictors among HIV-exposed infants visited Gamo and Gofa zones public health facilities, Southern Ethiopia: a retrospective follow up study. BMC Pediatr 2024; 24:410. [PMID: 38926639 PMCID: PMC11202369 DOI: 10.1186/s12887-024-04894-w] [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: 06/24/2023] [Accepted: 06/19/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Despite the highest (88%) Prevention of Mother-To-Child Transmission (PMTCT) of HIV coverage in Eastern Africa, 50% of new HIV infections in children aged 0-14 years occur in the region. OBJECTIVE The aim of this study was to assess the feeding modalities, the rate of HIV transmission and its predictors among HIV exposed infants (HIV-EIs) visited Gamo and Gofa Zones public health facilities, Southern Ethiopia from January 2013 to February 2019. METHOD AND MATERIALS Institution-based retrospective follow up study was employed among 450 HIV-EIs having DNA/PCR test results. All infant-mother pair records in selected health facilities were reviewed using a standard data extraction tool from March to July 2019. HIV transmission probabilities were assessed by Kaplan-Meier time-to-event analysis method and log-rank tests were used to compare the risk among different groups. The Cox-proportional hazards model, adjusted on infant feeding modalities and other co-variants was used to identify predictors of HIV transmission, and statistical significance was declared at a p-value of < 0.05. RESULTS In total, 383 complete records were analyzed. In the study, 85.6% (95%CI: 81.6%, 89.1%) of HIV-EIs were exclusively breastfed in the first six months. The 18 months probability of infant HIV transmission was 64 (16.7%) (95%CI: 13.1%-20.8%). The risk of HIV-transmission was higher among infants who were delivered at the hospital than health centers/health posts (AHR = 3.07; 95%CI: 1.19, 7.95); discontinued Cotrimoxazole prophylaxis in at least one visit (AHR = 6.32; 95%CI: 3.35, 11.94); did not exclusively breastfeed (AHR = 3.07; 95%CI: 1.72, 5.47) and came from urban areas (AHR = 5.90; 95%CI: 1.40, 24.85). CONCLUSIONS The study showed that HIV-EIs had a greater rate of 18 months HIV transmission than the national pooled prevalence. The risk of transmission is higher among infants who do not breastfeed exclusively for the first 6 months, and the risk increases with the number of months spent by breastfeeding. Therefore, strengthening counselling on safer feeding options and Cotrimoxazole prophylaxis use; provision of quality PMTCT service with special focus in hospitals and urban residents were recommended.
Collapse
Affiliation(s)
| | | | | | | | - Eshetu Zerihun Tariku
- School of Public Health, Arba Minch University, Arba Minch, Ethiopia
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Cardenas MC, Farnan S, Hamel BL, Mejia Plazas MC, Sintim-Aboagye E, Littlefield DR, Behl S, Punia S, Enninga EAL, Johnson E, Temesgen Z, Theiler R, Gray CM, Chakraborty R. Prevention of the Vertical Transmission of HIV; A Recap of the Journey so Far. Viruses 2023; 15:849. [PMID: 37112830 PMCID: PMC10142818 DOI: 10.3390/v15040849] [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: 12/26/2022] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
In 1989, one in four (25%) infants born to women living with HIV were infected; by the age of 2 years, there was 25% mortality among them due to HIV. These and other pieces of data prompted the development of interventions to offset vertical transmission, including the landmark Pediatric AIDS Clinical Trial Group Study (PACTG 076) in 1994. This study reported a 67.5% reduction in perinatal HIV transmission with prophylactic antenatal, intrapartum, and postnatal zidovudine. Numerous studies since then have provided compelling evidence to further optimize interventions, such that annual transmission rates of 0% are now reported by many health departments in the US and elimination has been validated in several countries around the world. Despite this success, the elimination of HIV's vertical transmission on the global scale remains a work in progress, limited by socioeconomic factors such as the prohibitive cost of antiretrovirals. Here, we review some of the key trials underpinning the development of guidelines in the US as well as globally, and discuss the evidence through a historic lens.
Collapse
Affiliation(s)
- Maria Camila Cardenas
- Pediatric Residency Program, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Sheila Farnan
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Benjamin L. Hamel
- Pediatric Residency Program, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Maria Camila Mejia Plazas
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Pediatric Residency Program, Nicklaus Children’s Hospital, 3100 SW 62nd Ave, Miami, FL 33155, USA
| | - Elise Sintim-Aboagye
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Dawn R. Littlefield
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Supriya Behl
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Sohan Punia
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Elizabeth Ann L Enninga
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN 33155, USA
| | - Erica Johnson
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Zelalem Temesgen
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Regan Theiler
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN 33155, USA
| | - Clive M. Gray
- Division of Molecular Biology and Human Genetics, Biomedical Research Institute, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town 7600, South Africa
| | - Rana Chakraborty
- Division of Pediatric Infectious Diseases, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
4
|
Jones AJ, Eke UA, Eke AC. Prediction and prevention of preterm birth in pregnant women living with HIV on antiretroviral therapy. Expert Rev Anti Infect Ther 2022; 20:837-848. [PMID: 35196941 PMCID: PMC9133156 DOI: 10.1080/14787210.2022.2046463] [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: 10/10/2021] [Accepted: 02/22/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The rate of spontaneous preterm-birth among pregnant women living with HIV on antiretroviral therapy (ART) is 3- to 4-fold higher when compared to HIV-negative women. The pathophysiology of preterm-birth related to HIV or ART remains unknown, especially as women living with HIV are often excluded from preterm birth studies. AREAS COVERED This review discusses the currently available evidence on the prediction and prevention of preterm-birth in pregnant women living with HIV. A review of the literature was conducted of primary articles between 2005 and 2021 measuring the association or lack thereof between combination ART and preterm birth, as well as of other predisposing factors to preterm birth in women living with HIV, including cervical length, vaginal microbiome, and cervico-vaginal biomarkers. EXPERT OPINION Further research into the effect of ART exposure on preterm-birth risk is critical, and development of preterm-birth predictive tools in this population should be a priority. Vaginal progesterone supplementation deserves further investigation as a therapeutic option to prevent recurrent preterm birth in pregnant women living with HIV. The ProSPAR study, a multicenter randomized controlled trial studying progesterone supplementation in pregnant women on protease inhibitor-based regimens, has been designed but is not yet recruiting patients.
Collapse
Affiliation(s)
| | - Uzoamaka A Eke
- Division of Infectious Diseases and Institute of Human Virology, Department of Internal Medicine, University of Maryland School of Medicine, Baltimore, United States of America
| | - Ahizechukwu C Eke
- Division of Maternal Fetal Medicine, Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore
| |
Collapse
|
5
|
Knapp KM. Prevention of Mother-to-Child Human Immunodeficiency Virus Transmission in Resource-Limited Countries. Pediatr Clin North Am 2022; 69:1-18. [PMID: 34794668 DOI: 10.1016/j.pcl.2021.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The first pediatric AIDS cases were reported in 1982. A decade later, the World Health Organization estimated there were more than 500,000 pediatric AIDS cases resulting from mother-to-child transmission, 90% of which were in sub-Saharan Africa. Although the rate of new infections globally has been cut in half since the peak of the pandemic, human immunodeficiency virus (HIV) remains a public health threat, and rates of new infections continue to increase in some regions. Mother-to-child transmission of HIV has now been virtually eliminated in many parts of the world but remains an issue in resource-limited countries.
Collapse
Affiliation(s)
- Katherine M Knapp
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 600, Memphis, TN 38105, USA.
| |
Collapse
|
6
|
Mayondi GK, Mussa A, Zash R, Moyo S, Issacson A, Diseko M, Mabuta J, Mogomotsi G, Dintwa E, Makhema J, Mmalane M, Lockman S, Morroni C, Shapiro R. Lack of HIV RNA test result is a barrier to breastfeeding among women living with HIV in Botswana. Int Breastfeed J 2021; 16:81. [PMID: 34645461 PMCID: PMC8513305 DOI: 10.1186/s13006-021-00424-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Background Botswana updated its antiretroviral treatment (ART) guidelines in May 2016 to support breastfeeding for women living with HIV (WLHIV) on ART who have documented HIV RNA suppression during pregnancy. Methods From September 2016 to March 2019, we evaluated feeding method at discharge among WLHIV at eight government maternity wards in Botswana within the Tsepamo Study. We validated the recorded feeding method on the obstetric record using the prevention of mother-to-child transmission of HIV (PMTCT) counsellor report, infant formula dispensing log or through direct observation. Available HIV RNA results were recorded from the obstetric record, and from outpatient HIV records (starting February 2018). In a subset of participants, we used electronic laboratory records to verify whether an HIV RNA test had occurred. Univariable and multivariable logistic regression analyses were performed to identify factors associated with infant feeding choice. Results Among 13,354 WLHIV who had a validated feeding method at discharge, 5303 (39.7%) chose to breastfeed and 8051 (60.3%) chose to formula feed. Women who had a documented HIV RNA result in the obstetric record available to healthcare providers at delivery were more likely to breastfeed (50.8%) compared to women who did not have a documented HIV RNA result (35.4%) (aOR 0.59; 95% CI 0.54, 0.65). Among women with documented HIV RNA, 2711 (94.6%) were virally suppressed (< 400 copies/mL). Breastfeeding occurred in a substantial proportion of women who did not meet criteria, including 46 (30.1%) of 153 women with HIV RNA > 400 copies/mL, and 134 (27.4%) of 489 women with no reported ART use. A sub-analysis of electronic laboratory records among 150 women without a recorded result on the obstetric record revealed that 93 (62%) women had an HIV RNA test during pregnancy. Conclusions In a setting of long-standing use of suppressive ART, with majority of WLHIV on ART from the time of conception, requiring documentation of HIV RNA suppression in the obstetric record to inform infant feeding decisions is a barrier to breastfeeding but unlikely to prevent a substantial amount of HIV transmission.
Collapse
Affiliation(s)
| | - Aamirah Mussa
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Rebecca Zash
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Sikhulile Moyo
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Arielle Issacson
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard Medical School, Boston, MA, USA
| | - Modiegi Diseko
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Judith Mabuta
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana
| | | | - Eldah Dintwa
- Ministry of Health and Wellness, Gaborone, Botswana
| | - Joseph Makhema
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mompati Mmalane
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Shahin Lockman
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Infectious Disease, Brigham and Women's Hospital, Boston, MA, USA
| | - Chelsea Morroni
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Roger Shapiro
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
7
|
Li KMC, Li KYC, Bick D, Chang Y. Human immunodeficiency virus-positive women's perspectives on breastfeeding with antiretrovirals: A qualitative evidence synthesis. MATERNAL & CHILD NUTRITION 2021; 17:e13244. [PMID: 34258858 PMCID: PMC8476435 DOI: 10.1111/mcn.13244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 11/03/2022]
Abstract
Human immunodeficiency virus (HIV)-positive women can breastfeed with minimal risk of mother-to-child transmission if taking antiretrovirals. Guidelines surrounding infant feeding for HIV-positive women have evolved several times over the last two decades. Our review aimed to explore perspectives of breastfeeding with antiretrovirals from HIV-positive women since the World Health Organization (2010) infant feeding and antiretroviral guidelines. HIV-positive pregnant and postnatal women from all countries/settings were eligible. HIV-positive women were either on an antiretroviral regimen at the time of the study, previously on an antiretroviral regimen, not initiated on a regimen yet, or enrolled in prevention of mother-to-child transmission (PMTCT) care. Quality assessment of all included studies were conducted. Four databases (CINAHL, EMBASE, MEDLINE and PsycINFO) were searched for studies conducted from January 2010 to October 2020. Nine papers were included in the review, of which two presented findings from the same study. Five analytical themes were developed via thematic synthesis: (1) awareness of breastfeeding with antiretrovirals, (2) turmoil of emotions, (3) coping mechanisms, (4) the intertwining of secret, stigma and support and (5) support needed. Support from family and health care professionals and coping approaches were important to overcome stigma and the emotional challenges of breastfeeding with antiretrovirals. Health care professionals should be familiar with the most updated national and local guidance surrounding infant feeding and antiretrovirals. Further research into interventions to encourage HIV-positive women to adhere and commit to lifelong antiretroviral treatment (Option B+) for breastfeeding is required.
Collapse
Affiliation(s)
- Kan Man Carmen Li
- Guy's and St. Thomas' NHS Foundation TrustEvelina London Children's HospitalLondonUK
| | - Kan Yan Chloe Li
- Institute of Cardiovascular ScienceUniversity College LondonLondonUK
| | - Debra Bick
- Warwick Clinical Trials Unit, Warwick Medical SchoolUniversity of WarwickWarwickUK
| | - Yan‐Shing Chang
- Florence Nightingale Faculty of Nursing, Midwifery and Palliative CareKing's College LondonLondonUK
| |
Collapse
|
8
|
Gutin SA, Harper GW, Moshashane N, Bitsang C, Harries J, Ramogola-Masire D, Morroni C. "What if they are pre-conception? What should we do?": Knowledge, practices, and preferences for safer conception among women living with HIV and healthcare providers in Gaborone, Botswana. Front Glob Womens Health 2021; 1. [PMID: 33693437 PMCID: PMC7943178 DOI: 10.3389/fgwh.2020.582463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Safer conception interventions that address HIV care, treatment, and prevention for HIV-affected couples are increasingly available in sub-Saharan Africa. Botswana, an HIV endemic country, is yet to offer formal safer conception services although universal test-and-treat approaches mean that increasing numbers of young, sexually active people living with HIV will start treatment and likely desire childbearing. In order to advance the safer conception discussion in Botswana, it is necessary to understand the current safer conception knowledge, practices, and preferences of healthcare providers and women living with HIV (WLHIV). We conducted qualitative in-depth interviews with 10 HIV healthcare providers and 10 WLHIV in Gaborone. Interviews were analyzed using a phenomenological approach. Safer conception knowledge was limited and safer conception discussions were rare. Healthcare provider and WLHIV preferences were at odds, with providers preferring WLHIV to initiate safer conception discussions, and WLHIV desiring providers to initiate safer conception discussions. Quotes from women and providers highlight deeper issues about power dynamics, concerns about stigma among women, and provider fears about promoting pregnancy. Providers emphasized the need for guidelines and training in order to improve the provision of safer conception counseling. These findings point to areas where safer conception in Botswana can be improved. Both WLHIV and providers would benefit from having information about a range of safer conception methods and approaches. In addition, since WLHIV felt hesitant about initiating safer conception conversations and feared stigma, and because putting the onus for starting safer conception discussions on women is a reversal of normal roles and power structures, providers must take the lead and routinely initiate fertility desire and safer conception discussions. Assisting healthcare providers with clear safer conception guidelines and training would improve the provision of accurate safer conception counseling and facilitate reproductive choice.
Collapse
Affiliation(s)
- Sarah A Gutin
- Dept. of Health Behavior and Health Education, School of Public Health, University of Michigan, Ann Arbor, MI, USA.,Women's Health Research Unit, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Center for AIDS Prevention Studies, Division of Prevention Science, University of California San Francisco, San Francisco, CA, USA
| | - Gary W Harper
- Dept. of Health Behavior and Health Education, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | - Christina Bitsang
- Career and Counseling Services, University of Botswana, Gaborone, Botswana
| | - Jane Harries
- Women's Health Research Unit, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Doreen Ramogola-Masire
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Botswana, Gaborone, Botswana
| | - Chelsea Morroni
- Women's Health Research Unit, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Botswana U-Penn Partnership, Gaborone, Botswana.,Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| |
Collapse
|
9
|
Boucoiran I, Roy M, Poliquin V, Elwood C, Sheehan NL, Thibaudeau R, Ferreira E, Autmizguine J, Kakkar F, Boucher M, Money D, Tulloch K. Evaluation of cabergoline for lactation inhibition in women living with HIV. Int J STD AIDS 2021; 32:654-661. [PMID: 33612017 DOI: 10.1177/0956462420984694] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We wished to evaluate the efficacy, safety, and acceptability of cabergoline for lactation inhibition in women who live with HIV. In this multicenter prospective observational study, cabergoline was offered as a single oral dose of 1 mg within the first 48 h postpartum. Women were recruited if they delivered a live infant after 35 weeks of gestational age. Participants filled out a questionnaire regarding symptoms of lactation and cabergoline adverse effects on day 2 and day 14 postpartum. On day 14, they also completed a questionnaire about their satisfaction with cabergoline treatment. Prolactin serum level was measured on both visits. Among 68 participants, all but one received cabergoline. The overall effectiveness defined by partial or complete success at day 14 was 98.3% (confidence intervals: 89.5-99.9). At day 14, 67.4% of women who received cabergoline had prolactin serum levels <25 mcg/L (threshold necessary for galactopoiesis). Mild nonspecific adverse effects were experienced by 24 (29.9%) women on day 2 and 24 (41.4%) on day 14, and lasted 48 h or less. Overall, 96% of women were satisfied with cabergoline's ability to prevent postpartum lactation symptoms. In conclusion, cabergoline is an effective, well-accepted, and well-tolerated medication for lactation inhibition in WLWH.
Collapse
Affiliation(s)
- Isabelle Boucoiran
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, 5622Université de Montréal, Montreal, Canada.,Department of Social and Preventive Medicine, 5622École de Santé Publique de Université de Montréal, Montreal, QC, Canada.,Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
| | - Melissa Roy
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, 5622Université de Montréal, Montreal, Canada
| | - Vanessa Poliquin
- Department of Obstetrics and Gynecology, 8664University of Manitoba, Winnipeg, Canada
| | - Chelsea Elwood
- Department of Obstetrics and Gynecology, 12358University of British Columbia, Vancouver, Canada.,Women's Health Research Institute, Vancouver, Canada
| | - Nancy L Sheehan
- Québec Antiretroviral Therapeutic Drug Monitoring Program, 54473McGill University Health Centre, Montréal, Canada.,Department of Pharmacology and Physiology, 5622Université de Montréal, Montréal, Canada
| | - Rosie Thibaudeau
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, 5622Université de Montréal, Montreal, Canada
| | - Ema Ferreira
- Department of Pharmacology and Physiology, 5622Université de Montréal, Montréal, Canada.,Department of Pharmacy, CHU Sainte-Justine, Montreal, Canada
| | - Julie Autmizguine
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada.,Department of Pharmacology and Physiology, 5622Université de Montréal, Montréal, Canada.,Department of Pediatrics, Division of Infectious Diseases, CHU Sainte-Justine, 5622Université de Montréal, Montreal, Canada
| | - Fatima Kakkar
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada.,Department of Pediatrics, Division of Infectious Diseases, CHU Sainte-Justine, 5622Université de Montréal, Montreal, Canada
| | - Marc Boucher
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, 5622Université de Montréal, Montreal, Canada.,Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
| | - Deborah Money
- Department of Obstetrics and Gynecology, 12358University of British Columbia, Vancouver, Canada.,Women's Health Research Institute, Vancouver, Canada
| | - Karen Tulloch
- Department of Pharmacy, BC Women's Hospital and Health Centre, Vancouver, Canada
| |
Collapse
|
10
|
Aguti I, Kimbugwe C, Apai P, Munyaga S, Nyeko R. HIV-free survival among breastfed infants born to HIV-positive women in northern Uganda: a facility-based retrospective study. Pan Afr Med J 2020; 37:297. [PMID: 33654517 PMCID: PMC7881925 DOI: 10.11604/pamj.2020.37.297.22928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/01/2020] [Indexed: 11/11/2022] Open
Abstract
Introduction the HIV-free survival rate is the gold-standard measure of the effectiveness of interventions towards prevention of mother-to-child transmission of HIV in any setting. However, data on HIV-free survival among the HIV-exposed infants followed up in most low-resource settings are lacking. We determined the HIV-free survival among breastfed infants in two tertiary facilities in a resource-poor setting in northern Uganda. Methods we conducted a retrospective cohort study in May 2019 and retrospectively reviewed records of HIV-exposed infants registered in 2014 through 2016 at two tertiary facilities in northern Uganda. We analyzed data using SPSS v16 software package. The chi-square and Student t-tests were used to compare factors among infant groups. Multivariate logistic regression analysis was used to determine factors independently associated with HIV-free survival. P-value <0.05 was considered for statistical significance. Results majority of the infants were males 55.6% (203/365) and 98.6% (360/365) received nevirapine prophylaxis. A total of 345 (94.5%) infants were exclusively breastfed, only 100/345 (29.0%) of whom were exclusively breastfed for at least 6 months, while the breastfeeding status of 44/345 (12.8 %) infants could not be ascertained. The overall HIV-free survival rate was 93.7% (342/365), while 2.7% (10/365) were HIV-infected and 3.6% (13/365) died. Infants´ age at enrolment in care (aOR 5.20, p=0.008) and treatment facility (aOR 3.76, p=0.027) were the independent determinants of HIV-free survival. Conclusion the HIV-free survival rate among the breastfed infants in the study setting marginally falls short of the recommended standard, thus calling for more efforts to improve survival.
Collapse
Affiliation(s)
- Irene Aguti
- Faculty of Medicine, Gulu University, Gulu, Uganda
| | | | | | | | - Richard Nyeko
- Department of Paediatrics and Child Health, Faculty of Health Sciences, Lira University, Lira, Uganda
| |
Collapse
|
11
|
Tiam A, Gill MM, Machekano R, Tukei V, Mokone M, Viana S, Letsie M, Tsietso M, Seipati I, Khachane C, Nei M, Mohai F, Tylleskär T, Guay L. 18-24-month HIV-free survival as measurement of the effectiveness of prevention of mother-to-child transmission in the context of lifelong antiretroviral therapy: Results of a community-based survey. PLoS One 2020; 15:e0237409. [PMID: 33002002 PMCID: PMC7529246 DOI: 10.1371/journal.pone.0237409] [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: 05/29/2020] [Accepted: 07/24/2020] [Indexed: 11/21/2022] Open
Abstract
Introduction Population-based HIV-free survival at 18–24 months of age among HIV-exposed infants in high prevalence settings in the era of treatment for all is largely unknown. We conducted a community-based survey to determine outcomes of HIV-exposed infants at 18–24 months in Lesotho. Methods Between November 2015 and December 2016, we conducted a survey among households with a child born 18–24 months prior to data collection. Catchment areas from 25 health facilities in Butha-Buthe, Maseru, Mohale’s Hoek and Thaba-Tseka districts were randomly selected using probability proportional to size sampling. Consecutive households were visited and eligible consenting caregivers and children were enrolled. Rapid HIV antibody testing was performed on mothers of unknown HIV status (never tested or tested HIV-negative >3 months prior) and their children, and to children born to known HIV-positive mothers. Information on demographics, health-seeking behavior, HIV, and mortality were captured for mothers and children, including those who died. The difference in survival between subgroups was determined using the log-rank test. Results Of the 1,852 mothers/caregivers enrolled, 570 mothers were HIV-positive. The mother-to-child HIV transmission rate was 5.7% [95% CI: 4.0–8.0]. The mortality rate was 2.6% [95% CI: 1.6–4.2] among HIV-exposed children compared to 1.4% (95% CI: 0.9–2.3) among HIV-unexposed children. HIV-free survival was 91.8% [95% CI: 89.2–93.8] among HIV-exposed infants. Disclosure of mother’s HIV status (aOR = 4.9, 95% CI: 1.3–18.2) and initiation of cotrimoxazole prophylaxis in the child (aOR = 3.9, 95% CI: 1.2–12.6) were independently associated with increased HIV-free survival while child growth problems (aOR = 0.2, 95% CI: 0.09–0.5) were independently associated with reduced HIV-free survival. Conclusion Even in the context of lifelong antiretroviral therapy among pregnant and breastfeeding women, HIV has a significant effect on survival among HIV-exposed children compared to unexposed children. Lesotho has not reached elimination of HIV transmission from mother to child.
Collapse
Affiliation(s)
- Appolinaire Tiam
- Centre for International Health, University of Bergen, Bergen, Norway
- Elizabeth Glaser Pediatric AIDS Foundation, Washington D.C., United State of America
- * E-mail:
| | - Michelle M. Gill
- Centre for International Health, University of Bergen, Bergen, Norway
- Elizabeth Glaser Pediatric AIDS Foundation, Washington D.C., United State of America
| | - Rhoderick Machekano
- Centre for International Health, University of Bergen, Bergen, Norway
- Elizabeth Glaser Pediatric AIDS Foundation, Washington D.C., United State of America
| | - Vincent Tukei
- Elizabeth Glaser Pediatric AIDS Foundation, Maseru, Lesotho
- Ministry of Health, Maseru, Lesotho
| | | | - Shannon Viana
- Centre for International Health, University of Bergen, Bergen, Norway
- Elizabeth Glaser Pediatric AIDS Foundation, Washington D.C., United State of America
| | - Mosilinyane Letsie
- Milken Institute School of Public Health, The George Washington University, Washington D.C., United States of America
| | - Mots’oane Tsietso
- Milken Institute School of Public Health, The George Washington University, Washington D.C., United States of America
| | - Irene Seipati
- Milken Institute School of Public Health, The George Washington University, Washington D.C., United States of America
| | - Cecilia Khachane
- Milken Institute School of Public Health, The George Washington University, Washington D.C., United States of America
| | | | | | | | - Laura Guay
- Centre for International Health, University of Bergen, Bergen, Norway
- Elizabeth Glaser Pediatric AIDS Foundation, Washington D.C., United State of America
- Milken Institute School of Public Health, The George Washington University, Washington D.C., United States of America
| |
Collapse
|
12
|
Wood LF, Brown BP, Lennard K, Karaoz U, Havyarimana E, Passmore JAS, Hesseling AC, Edlefsen PT, Kuhn L, Mulder N, Brodie EL, Sodora DL, Jaspan HB. Feeding-Related Gut Microbial Composition Associates With Peripheral T-Cell Activation and Mucosal Gene Expression in African Infants. Clin Infect Dis 2019; 67:1237-1246. [PMID: 29659737 DOI: 10.1093/cid/ciy265] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 04/03/2018] [Indexed: 12/31/2022] Open
Abstract
Background Exclusive breastfeeding reduces the rate of postnatal human immunodeficiency virus (HIV) transmission compared to nonexclusive breastfeeding; however, the mechanisms of this protection are unknown. Our study aimed to interrogate the mechanisms underlying the protective effect of exclusive breastfeeding. Methods We performed a prospective, longitudinal study of infants from a high-HIV-prevalence, low-income setting in South Africa. We evaluated the role of any non-breast milk feeds, excluding prescribed medicines on stool microbial communities via 16S rRNA gene sequencing, peripheral T-cell activation via flow cytometry, and buccal mucosal gene expression via quantitative polymerase chain reaction assay. Results A total of 155 infants were recruited at birth with mean gestational age of 38.9 weeks and mean birth weight of 3.2 kg. All infants were exclusively breastfed (EBF) at birth, but only 43.5% and 20% remained EBF at 6 or 14 weeks of age, respectively. We observed lower stool microbial diversity and distinct microbial composition in exclusively breastfed infants. These microbial communities, and the relative abundance of key taxa, were correlated with peripheral CD4+ T-cell activation, which was lower in EBF infants. In the oral mucosa, gene expression of chemokine and chemokine receptors involved in recruitment of HIV target cells to tissues, as well as epithelial cytoskeletal proteins, was lower in EBF infants. Conclusions These data suggest that nonexclusive breastfeeding alters the gut microbiota, increasing T-cell activation and, potentially, mucosal recruitment of HIV target cells. Study findings highlight a biologically plausible mechanistic explanation for the reduced postnatal HIV transmission observed in EBF infants.
Collapse
Affiliation(s)
- Lianna F Wood
- University of Washington Schools of Medicine and Public Health, Seattle
| | - Bryan P Brown
- Duke University, Durham, North Carolina.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town Health Sciences Faculty, South Africa
| | - Katie Lennard
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town Health Sciences Faculty, South Africa
| | - Ulas Karaoz
- Earth and Environmental Science, Lawrence Berkeley National Laboratories, Berkeley.,University of California, Berkeley
| | - Enock Havyarimana
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town Health Sciences Faculty, South Africa
| | - Jo-Ann S Passmore
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town Health Sciences Faculty, South Africa.,National Health Laboratory Services, Cape Town, South Africa
| | - Anneke C Hesseling
- Desmond Tutu TB Centre, Stellenbosch University, Cape Town, South Africa
| | | | | | - Nicola Mulder
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town Health Sciences Faculty, South Africa
| | - Eoin L Brodie
- Earth and Environmental Science, Lawrence Berkeley National Laboratories, Berkeley.,University of California, Berkeley
| | | | - Heather B Jaspan
- University of Washington Schools of Medicine and Public Health, Seattle.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town Health Sciences Faculty, South Africa.,Seattle Children's Research Institute, Washington
| |
Collapse
|
13
|
Sania A, Sudfeld CR, Danaei G, Fink G, McCoy DC, Zhu Z, Fawzi MCS, Akman M, Arifeen SE, Barros AJD, Bellinger D, Black MM, Bogale A, Braun JM, van den Broek N, Carrara V, Duazo P, Duggan C, Fernald LCH, Gladstone M, Hamadani J, Handal AJ, Harlow S, Hidrobo M, Kuzawa C, Kvestad I, Locks L, Manji K, Masanja H, Matijasevich A, McDonald C, McGready R, Rizvi A, Santos D, Santos L, Save D, Shapiro R, Stoecker B, Strand TA, Taneja S, Tellez-Rojo MM, Tofail F, Yousafzai AK, Ezzati M, Fawzi W. Early life risk factors of motor, cognitive and language development: a pooled analysis of studies from low/middle-income countries. BMJ Open 2019; 9:e026449. [PMID: 31585969 PMCID: PMC6797384 DOI: 10.1136/bmjopen-2018-026449] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To determine the magnitude of relationships of early life factors with child development in low/middle-income countries (LMICs). DESIGN Meta-analyses of standardised mean differences (SMDs) estimated from published and unpublished data. DATA SOURCES We searched Medline, bibliographies of key articles and reviews, and grey literature to identify studies from LMICs that collected data on early life exposures and child development. The most recent search was done on 4 November 2014. We then invited the first authors of the publications and investigators of unpublished studies to participate in the study. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Studies that assessed at least one domain of child development in at least 100 children under 7 years of age and collected at least one early life factor of interest were included in the study. ANALYSES Linear regression models were used to assess SMDs in child development by parental and child factors within each study. We then produced pooled estimates across studies using random effects meta-analyses. RESULTS We retrieved data from 21 studies including 20 882 children across 13 LMICs, to assess the associations of exposure to 14 major risk factors with child development. Children of mothers with secondary schooling had 0.14 SD (95% CI 0.05 to 0.25) higher cognitive scores compared with children whose mothers had primary education. Preterm birth was associated with 0.14 SD (-0.24 to -0.05) and 0.23 SD (-0.42 to -0.03) reductions in cognitive and motor scores, respectively. Maternal short stature, anaemia in infancy and lack of access to clean water and sanitation had significant negative associations with cognitive and motor development with effects ranging from -0.18 to -0.10 SDs. CONCLUSIONS Differential parental, environmental and nutritional factors contribute to disparities in child development across LMICs. Targeting these factors from prepregnancy through childhood may improve health and development of children.
Collapse
Affiliation(s)
- Ayesha Sania
- ICAP and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York city, New York, USA
| | - Christopher R Sudfeld
- Department of Global Health and Population, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Goodarz Danaei
- Deaprtment of Global Health and Population, and Epidemiology, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Günther Fink
- Household Economics and Health System Research Unit, Schweizerisches Tropen- und Public Health-Institut, Basel, Switzerland
| | - Dana C McCoy
- Harvard Graduate School of Education, Harvard University, Cambridge, Massachusetts, USA
| | - Zhaozhong Zhu
- Departments of Epidemiology and Environmental Health, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Mary C Smith Fawzi
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Mehmet Akman
- Department of Family Medicine, Marmara University School of Medicine, Istanbul, Turkey
| | - Shams E Arifeen
- Maternal and Child Health Division, ICDDR,B, Dhaka, Bangladesh
| | - Aluisio J D Barros
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - David Bellinger
- Department of Neurology, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts, USA
| | - Maureen M Black
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alemtsehay Bogale
- Nutrition and Scientific Affairs, The Nature's Bounty Co, Ronkonkoma, New York, USA
| | - Joseph M Braun
- Brown University School of Public Health, Providence, Rhode Island, USA
| | - Nynke van den Broek
- Maternal and Newborn Health, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Verena Carrara
- Department of Maternal and Child Health, Shoklo Malaria Research Unit, Mae Sot, Thailand
| | - Paulita Duazo
- Office of Population Studies Foundation, Inc, University of San Carlos, Cebu City, Philippines
| | - Christopher Duggan
- Center for Nutrition, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lia C H Fernald
- Community Health Sciences, School of Public Health, University of California, Berkeley, California, USA
| | - Melissa Gladstone
- Women and Children's Health, University of Liverpool, Institute of Translational Medicine, Liverpool, UK
| | - Jena Hamadani
- Maternal and Child Health Division, ICDDR,B, Dhaka, Bangladesh
| | - Alexis J Handal
- College of Population Health, University of New Mexico, Albuquerque, New Mexico, USA
| | - Siobán Harlow
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Melissa Hidrobo
- Poverty Health and Nutrition Division, International Food Policy Research Institute, Washington, DC, USA
| | - Chris Kuzawa
- Department of Anthropology, Northwestern University, Evanston, Illinois, USA
| | - Ingrid Kvestad
- Regional Centre for Child and Youth Mental Health and Child Welfare, NORCE Norwegian Research Center, Bergen, Norway
| | - Lindsey Locks
- Department of Nutrition, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Karim Manji
- Department of Pediatrics and Child Health, Muhibili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Alicia Matijasevich
- Departamento de Medicina Preventiva, Faculdade de Medicina FMUSP, Universidade de São Paulo, Sao Paulo, Brasil
| | - Christine McDonald
- Children's Hospital Oakland Research Institute, UCSF Benioff Children's Hospital, Oakland, California, USA
| | - Rose McGready
- Faculty of Tropical Medicine, Mahidol University, Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Arjumand Rizvi
- Pediatrics and Child Health, Aga Khan Medical University, Karachi, Pakistan
| | - Darci Santos
- Department of Collective Health, Universidade Federal da Bahia, Salvador, Brazil
| | - Leticia Santos
- Department of Collective Health, Universidade Federal da Bahia, Salvador, Brazil
| | - Dilsad Save
- Department of Public Health, Marmara University School of Medicine, Istanbul, Turkey
| | - Roger Shapiro
- Department of Immunology and Infectious Disease, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Barbara Stoecker
- Department of Nutritional Sciences, Oklahoma State University College of Human Environmental Sciences, Stillwater, Oklahoma, USA
| | - Tor A Strand
- Department of Laboratory Medicine, Sykehuset Innlandet Helseforetaket, Brumunddal, Norway
| | - Sunita Taneja
- Centre for Health Research and Development, Society for Applied Studies, New Delhi, India
| | | | - Fahmida Tofail
- Nutrition and Clinical Services Division, ICDDR,B, Dhaka, Bangladesh
| | - Aisha K Yousafzai
- Department of Global Health and Population, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Majid Ezzati
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Wafaie Fawzi
- Deaprtment of Global Health and Population, Epidemiology, and Nutrition, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Brown BP, Jaspan HB. Compositional analyses reveal correlations between taxon-level gut bacterial abundance and peripheral T cell marker expression in African infants. Gut Microbes 2019; 11:237-244. [PMID: 31347944 PMCID: PMC7053881 DOI: 10.1080/19490976.2019.1643673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Although exclusive breastfeeding has been linked to lower rates of postnatal HIV transmission compared to nonexclusive breastfeeding, mechanisms underlying this are unclear. Across a longitudinally sampled cohort of South African infants, we showed that exclusively breastfed (EBF) infants had altered gut bacterial communities when compared to nonexclusively breastfed (NEBF) infants, as well as reduced peripheral CD4 + T cell activation and lowered chemokine and chemokine receptor expression in the oral mucosa. We further demonstrated that the relative abundance of key taxa was correlated with peripheral CD4 + T cell activation. Here, we supplement those findings by using compositional data analyses to identify shifts in the abundance of several Bifidobacteria strains relative to select strains of Escherichia, Bacteroides, and others that are associated with the transition to NEBF. We illustrate that the abundance ratio of these taxa is tightly correlated with feeding modality and is a strong predictor of peripheral T cell activation. More broadly, we discuss our study in the context of novel developments and explore future directions for the field.
Collapse
Affiliation(s)
- Bryan P. Brown
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, DC, USA,Departments of Pediatrics and Global Health, Schools of Medicine and Public Health, University of WA, Seattle, WA, USA,CONTACT Heather Jaspan Seattle Children’s Research Institute, Seattle, Washington, 98101 USA
| | - Heather B. Jaspan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, DC, USA,Departments of Pediatrics and Global Health, Schools of Medicine and Public Health, University of WA, Seattle, WA, USA,Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
15
|
Adeniyi OV, Obi CL, Goon DT, Iweriebor B, Ajayi AI, Lambert J, Okoh A. HIV-1 drug resistance surveillance among parturient women on anti-retroviral therapy in the Eastern Cape, South Africa: Implications for elimination of mother-to-child transmission. J Clin Virol 2019; 117:89-95. [PMID: 31255794 DOI: 10.1016/j.jcv.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/20/2019] [Accepted: 06/13/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The emergence of HIV drug resistance poses a significant threat to achieving the goal of elimination of mother-to-child transmission. OBJECTIVES We assessed the extent and patterns of HIV-1 drug resistance mutations (DRMs) within the context of the public sector prevention of mother-to-child transmission (PMTCT) programme in the Eastern Cape, South Africa. STUDY DESIGN We conducted analysis of the Pol sub-genomic sequence of RNA extracted from plasma samples of women with probable virological failure at delivery between January and May 2018 from two large maternity centres in the Eastern Cape using standard protocols. Partial pol gene covering 1030bp were amplified and sequenced according to previously reported protocol. DRMs were determined by submitting the generated partial pol sequences to the Stanford drug resistance database for query on mutations associated with drug resistance in HIV viruses. We examined the correlates of DRMs using bivariate analysis. RESULTS The age of parturient women ranged from 16 to 43 years. The majority of the parturient women were currently on Efavirenz-based regimen (first line ART) (82.5%) and had been on ART for more than 12 months (65.0%). The prevalence of DRMs was 72.5% (n = 58). The CD4 count demonstrated a negative linear association with the DRMs (p = 0.002). The predominant DRMs were K103 N (n = 43; 74.1%), M184 V (n = 28; 48.3%) and K65R (n = 11; 19%). Among the parturient women on EFV-based regimen treatment; 79.1% already had K103 N while nine patients on protease inhibitor-based regimen still harboured K103 N. The majority of the M184 V mutations were observed in parturient women on first line regimen (n = 23; 82.1%). CONCLUSIONS We found a high prevalence of DRMs in women delivering their index babies at high viral loads in the study settings. Drug resistance surveillance using point-of-care reverse transcriptase-PCR strategies for the screening of pregnant women on ART could be a game-changer in the resource-constrained settings.
Collapse
Affiliation(s)
- Oladele Vincent Adeniyi
- Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Fort Hare, Alice, South Africa; SAMRC Microbial Water Quality Monitoring Centre, Faculty of Science and Agriculture, University of Fort Hare, Alice, South Africa.
| | - Chikwelu Larry Obi
- School of Science and Technology, Sefako Makgatho Health Sciences University, Ga-Rankuwa, Pretoria, South Africa.
| | - Daniel Ter Goon
- Department of Public Health, Faculty of Health Sciences, University of Fort Hare, East London, South Africa.
| | - Benson Iweriebor
- School of Science and Technology, Sefako Makgatho Health Sciences University, Ga-Rankuwa, Pretoria, South Africa.
| | - Anthony Idowu Ajayi
- Population Dynamics and Sexual and Reproductive Health, African Population and Health Research Center, Nairobi, Kenya.
| | - John Lambert
- University College Dublin/Mater Misericordiae University Hospital, Catherine McAuley Education & Research Centre, Dublin, Ireland.
| | - Anthony Okoh
- Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Fort Hare, Alice, South Africa; SAMRC Microbial Water Quality Monitoring Centre, Faculty of Science and Agriculture, University of Fort Hare, Alice, South Africa.
| |
Collapse
|
16
|
Hoffman RM, Angelidou KN, Brummel SS, Saidi F, Violari A, Dula D, Mave V, Fairlie L, Theron G, Kamateeka M, Chipato T, Chi BH, Stranix-Chibanda L, Nematadzira T, Moodley D, Bhattacharya D, Gupta A, Coletti A, McIntyre JA, Klingman KL, Chakhtoura N, Shapiro DE, Fowler MG, Currier JS. Maternal health outcomes among HIV-infected breastfeeding women with high CD4 counts: results of a treatment strategy trial. HIV CLINICAL TRIALS 2018; 19:209-224. [PMID: 30890061 PMCID: PMC6428202 DOI: 10.1080/15284336.2018.1537327] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/28/2018] [Accepted: 10/12/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND IMPAACT PROMISE 1077BF/FF was a randomized study of antiretroviral therapy (ART) strategies for pregnant and postpartum women with high CD4+ T-cell counts. We describe postpartum outcomes for women in the study who were randomized to continue or discontinue ART after delivery. METHODS Women with pre-ART CD4+ cell counts ≥350 cells/mm3 who started ART during pregnancy were randomized postpartum to continue or discontinue treatment. Women were enrolled from India, Malawi, South Africa, Tanzania, Uganda, Zambia, and Zimbabwe. The primary outcome was a composite of progression to AIDS-defining illness or death. Log-rank tests and Cox regression models assessed treatment effects. Incidence rates were calculated per 100 person-years. A post hoc analysis evaluated WHO Stage 2/3 events. All analyses were intent-to-treat. FINDINGS 1611 women were enrolled (June 2011-October 2014) and 95% were breastfeeding. Median age at entry was 27 years, CD4+ count 728 cells/mm3 and the majority of women were Black African (97%). After a median follow-up of 1.6 years, progression to AIDS-defining illness or death was rare and there was no significant difference between arms (HR: 0·55; 95%CI 0·14, 2·08, p = 0.37). WHO Stage 2/3 events were reduced with continued ART (HR: 0·60; 95%CI 0·39, 0·90, p = 0.01). The arms did not differ with respect to the rate of grade 2, 3, or 4 safety events (p = 0.61). INTERPRETATION Serious clinical events were rare among predominately breastfeeding women with high CD4+ cell counts over 18 months after delivery. ART had significant benefit in reducing WHO 2/3 events in this population.
Collapse
Affiliation(s)
- Risa M Hoffman
- a Division of Infectious Diseases, Department of Medicine , David Geffen School of Medicine at the University of California, Los Angeles , Los Angeles , CA , USA
| | - Konstantia Nadia Angelidou
- b Center for Biostatistics in AIDS Research , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Sean S Brummel
- b Center for Biostatistics in AIDS Research , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Friday Saidi
- c University of North Carolina Project-Malawi , Lilongwe , Malawi
| | - Avy Violari
- d Perinatal HIV Research Unit , Chris Hani Baragwanath Hospital , Soweto , South Africa
| | - Dingase Dula
- e Malawi College of Medicine , Johns Hopkins Project , Chichiri , Malawi
| | - Vidya Mave
- f BJGMC Clinical Trials Unit , Pune , India
- g Division of Infectious Diseases , Johns Hopkins School of Medicine , Baltimore , MD , USA
| | - Lee Fairlie
- h Wits Reproductive Health and HIV Institute , Johannesburg , South Africa
| | | | - Moreen Kamateeka
- j Makerere University, Johns Hopkins University Research Collaboration , Mulago Kampala , Uganda
| | - Tsungai Chipato
- k Department of Obstetrics and Gynecology , University of Zimbabwe , Harare , Zimbabwe
| | - Benjamin H Chi
- l Department of Obstetrics and Gynecology , University of North Carolina School of Medicine , Chapel Hill , NC , USA
| | | | - Teacler Nematadzira
- n University of Zimbabwe College of Health Sciences Clinical Trials Research Centre , Harare , Zimbabwe
| | - Dhayendre Moodley
- o Centre for the AIDS Programme of Research in South Africa and School of Clinical Medicine , University of KwaZulu Natal , Durban , South Africa
| | - Debika Bhattacharya
- a Division of Infectious Diseases, Department of Medicine , David Geffen School of Medicine at the University of California, Los Angeles , Los Angeles , CA , USA
| | - Amita Gupta
- f BJGMC Clinical Trials Unit , Pune , India
- g Division of Infectious Diseases , Johns Hopkins School of Medicine , Baltimore , MD , USA
| | - Anne Coletti
- p Family Health International 360 , Durham , NC , USA
| | - James A McIntyre
- q Anova Health Institute , Johannesburg , South Africa
- r School of Public Health & Family Medicine , University of Cape Town , Cape Town , South Africa
| | - Karin L Klingman
- s Division of AIDS , National Institute of Allergy and Infectious Diseases National Institutes of Health , Bethesda , MD , USA
| | - Nahida Chakhtoura
- t Eunice Kennedy Shriver National Institute of Child Health and Human Development , National Institutes of Health , Bethesda , MD , USA
| | - David E Shapiro
- b Center for Biostatistics in AIDS Research , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| | - Mary Glenn Fowler
- u Department of Pathology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Judith S Currier
- a Division of Infectious Diseases, Department of Medicine , David Geffen School of Medicine at the University of California, Los Angeles , Los Angeles , CA , USA
| |
Collapse
|
17
|
Mutsaerts EA, Nunes MC, van Rijswijk MN, Klipstein-Grobusch K, Grobbee DE, Madhi SA. Safety and Immunogenicity of Measles Vaccination in HIV-Infected and HIV-Exposed Uninfected Children: A Systematic Review and Meta-Analysis. EClinicalMedicine 2018; 1:28-42. [PMID: 31193646 PMCID: PMC6537570 DOI: 10.1016/j.eclinm.2018.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/08/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND HIV-infected and HIV-exposed uninfected (HEU) children have an increased risk of measles that may be due to altered immune responses or suboptimal timing of measles vaccination. We aimed to evaluate the safety and immunogenicity of measles vaccination in HIV-infected and HEU children. METHODS For this systematic review and meta-analysis, we searched PubMed, Embase, Cochrane Library, CINAHL, Global Health Library and IndMED on May 9, 2018. Studies were included if they reported on safety or seroresponse (either seroprotection/seropositivity/seroconversion) after measles vaccination in HIV-infected or HEU children. We calculated pooled estimates to compare immunogenicity outcomes between HIV-infected, HEU and HIV-unexposed children, using risk ratios [RRs] (with 95%CIs). PROSPERO registration number: CRD42017057411. FINDINGS Seventy-one studies met the inclusion criteria (15,363 children). Twenty-eight studies reported on safety; vaccine-associated adverse events and deaths were uncommon. Sixty-two studies reported on immunogenicity, 27 were included in the meta-analysis. HIV-infected children had lower seroresponse rates after primary vaccination compared with HIV-unexposed (RR 0.74; 95%CI: 0.61-0.90, I 2 = 85.9%) and HEU children (0.78; 0.69-0.88, I 2 = 77.1%), which was mitigated by antiretroviral therapy and time interval between vaccination and serology. HEU and HIV-unexposed children had similar seroresponses. Vaccination at 6-months resulted in similar proportions of HIV-infected children having seroresponse compared with HIV-unexposed (0.96; 0.77-1.19) and HEU children (1.00; 0.73-1.37, I 2 = 63.7%). INTERPRETATION Primary measles vaccination at 6-months of age may provide protection against measles during early infancy in settings with high prevalence of maternal HIV-infection, however, further studies are needed to evaluate this strategy in HEU children and HIV-infected children receiving antiretroviral therapy. FUNDING South African Research Chairs Initiative of the Department of Science and Technology and National Research Foundation in Vaccine Preventable Diseases; Medical Research Council: Respiratory and Meningeal Pathogens Research Unit.
Collapse
Affiliation(s)
- Eleonora A.M.L. Mutsaerts
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Julius Global Health, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marta C. Nunes
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Martijn N. van Rijswijk
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Julius Global Health, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Kerstin Klipstein-Grobusch
- Division of Epidemiology and Biostatistics, School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Julius Global Health, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Diederick E. Grobbee
- Julius Global Health, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Clinical Epidemiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Shabir A. Madhi
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
18
|
Arikawa S, Rollins N, Jourdain G, Humphrey J, Kourtis AP, Hoffman I, Essex M, Farley T, Coovadia HM, Gray G, Kuhn L, Shapiro R, Leroy V, Bollinger RC, Onyango-Makumbi C, Lockman S, Marquez C, Doherty T, Dabis F, Mandelbrot L, Le Coeur S, Rolland M, Joly P, Newell ML, Becquet R. Contribution of Maternal Antiretroviral Therapy and Breastfeeding to 24-Month Survival in Human Immunodeficiency Virus-Exposed Uninfected Children: An Individual Pooled Analysis of African and Asian Studies. Clin Infect Dis 2018; 66:1668-1677. [PMID: 29272387 PMCID: PMC5961296 DOI: 10.1093/cid/cix1102] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/19/2017] [Indexed: 11/14/2022] Open
Abstract
Background Human immunodeficiency virus (HIV)-infected pregnant women increasingly receive antiretroviral therapy (ART) to prevent mother-to-child transmission (PMTCT). Studies suggest HIV-exposed uninfected (HEU) children face higher mortality than HIV-unexposed children, but most evidence relates to the pre-ART era, breastfeeding of limited duration, and considerable maternal mortality. Maternal ART and prolonged breastfeeding while on ART may improve survival, although this has not been reliably quantified. Methods Individual data on 19 219 HEU children from 21 PMTCT trials/cohorts undertaken from 1995 to 2015 in Africa and Asia were pooled to estimate the association between 24-month mortality and maternal/infant factors, using random-effects Cox proportional hazards models. Adjusted attributable fractions of risks computed using the predict function in the R package "frailtypack" were used to estimate the relative contribution of risk factors to overall mortality. Results Cumulative incidence of death was 5.5% (95% confidence interval, 5.1-5.9) by age 24 months. Low birth weight (LBW <2500 g, adjusted hazard ratio (aHR, 2.9), no breastfeeding (aHR, 2.5), and maternal death (aHR, 11.1) were significantly associated with increased mortality. Maternal ART (aHR, 0.5) was significantly associated with lower mortality. At the population level, LBW accounted for 16.2% of 24-month mortality, never breastfeeding for 10.8%, mother not receiving ART for 45.6%, and maternal death for 4.3%; combined, these factors explained 63.6% of deaths by age 24 months. Conclusions Survival of HEU children could be substantially improved if public health practices provided all HIV-infected mothers with ART and supported optimal infant feeding and care for LBW neonates.
Collapse
Affiliation(s)
- Shino Arikawa
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team IDLIC, France
| | - Nigel Rollins
- Department of Maternal, Newborn, Child and Adolescent Health, World Health Organization, Geneva, Switzerland
| | - Gonzague Jourdain
- Institut de recherche pour le développement UMI 174-PHPT, Marseille, France
- Faculty of Associated Medical Sciences, Chiang Mai University, Thailand
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jean Humphrey
- Department of International Health, Center for Global Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Athena P Kourtis
- Women’s Health and Fertility Branch, Division of Reproductive Health, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention
- Emory University School of Medicine and Eastern Virginia Medical School, Atlanta, Georgia
| | - Irving Hoffman
- Division of Infectious Diseases, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill
| | - Max Essex
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Hoosen M Coovadia
- Maternal Adolescent and Child Health, University of the Witwatersrand, Johannesburg
| | - Glenda Gray
- South African Medical Research Council, Cape Town
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Roger Shapiro
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Valériane Leroy
- Inserm, Centre de recherche Inserm U1027, Université Paul Sabatier Toulouse 3, France
| | - Robert C Bollinger
- Center for Clinical Global Health Education, Johns Hopkins University, Baltimore, Maryland
| | - Carolyne Onyango-Makumbi
- Makerere University–Johns Hopkins University Research Collaboration/MU-JHU CARE LTD, Kampala, Uganda
| | - Shahin Lockman
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Carina Marquez
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, and Zuckerberg San Francisco General Hospital
| | | | - François Dabis
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team IDLIC, France
| | | | - Sophie Le Coeur
- Institut de recherche pour le développement UMI 174-PHPT, Marseille, France
- Faculty of Associated Medical Sciences, Chiang Mai University, Thailand
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Institut National d’Etudes Démographiques (Ined), Paris
| | - Matthieu Rolland
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team IDLIC, France
| | - Pierre Joly
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team Biostatistics, France
| | - Marie-Louise Newell
- Institute for Developmental Science and Global Health Research Institute, Faculty of Medicine, University of Southampton, United Kingdom
| | - Renaud Becquet
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team IDLIC, France
| |
Collapse
|
19
|
HIV-1 transmission and survival according to feeding options in infants born to HIV-infected women in Yaoundé, Cameroon. BMC Pediatr 2018; 18:69. [PMID: 29458337 PMCID: PMC5817808 DOI: 10.1186/s12887-018-1049-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/06/2018] [Indexed: 11/17/2022] Open
Abstract
Background Evidence of 24-months survival in the frame of prevention of mother-to-child transmission (PMTCT) cascade-care is scare from routine programs in sub-Saharan African (SSA) settings. Specifically, data on infant outcomes according to feeding options remain largely unknown by month-24, thus limiting its breath for public-health recommendations toward eliminating new pediatric HIV-1 infections and improving care. We sought to evaluate HIV-1 vertical transmission and infant survival rates according to feeding options. Methods A retrospective cohort-study conducted in Yaounde from April 2008 through December 2013 among 1086 infants born to HIV-infected women and followed-up throughout the PMTCT cascade-care until 24-months. Infants with documented feeding option during their first 3 months of life (408 on Exclusive Breastfeeding [EBF], 663 Exclusive Replacement feeding [ERF], 15 mixed feeding [MF]) and known HIV-status were enrolled. HIV-1 vertical transmission, survival and feeding options were analyzed using Kaplan Meier Survival Estimate, Cox model and Schoenfeld residuals tests, at 5% statistical significance. Results Overall HIV-1 vertical transmission was 3.59% (39), and varied by feeding options: EBF (2.70%), ERF (3.77%), MF (20%), p = 0.002; without significance between EBF and ERF (p = 0.34). As expected, HIV-1 transmission also varied with PMTCT-interventions: 1.7% (10/566) from ART-group, 1.9% (8/411) from AZT-group, and 19.2% (21/109) from ARV-naïve group, p < 0.0001. Overall mortality was 2.58% (28), higher in HIV-infected (10.25%) vs. uninfected (2.29%) infants (p = 0.016); with a survival cumulative probability of 89.3% [79.9%–99.8%] vs. 96.4% [94.8%–97.9% respectively], p = 0.024. Mortality also varied by feeding option: ERF (2.41%), EBF (2.45%), MF (13.33%), p = 0.03; with a survival cumulative probability of 96% [94%–98%] in ERF, 96.4% [94.1%–98.8%] in EBF, and 86.67% [71.06%–100%] in MF, p = 0.04. Using Schoenfeld residuals test, only HIV status was a predictor of survival at 24 months (hazard ratio 0.23 [0.072–0.72], p = 0.01). Conclusion Besides using ART for PMTCT-interventions, practice of MF also drives HIV-1 vertical transmission and mortality among HIV-infected children. Thus, throughout PMTCT option B+ cascade-care, continuous counseling on safer feeding options would to further eliminating new MTCT, optimizing response to care, and improving the life expectancy of these children in high-priority countries.
Collapse
|
20
|
Gill MM, Hoffman HJ, Ndatimana D, Mugwaneza P, Guay L, Ndayisaba GF, Bobrow EA, Asiimwe A, Mofenson LM. 24-month HIV-free survival among infants born to HIV-positive women enrolled in Option B+ program in Kigali, Rwanda: The Kabeho Study. Medicine (Baltimore) 2017; 96:e9445. [PMID: 29390577 PMCID: PMC5758279 DOI: 10.1097/md.0000000000009445] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Lifelong antiretroviral therapy (ART) provision to all pregnant HIV-positive women ("Option B+") has been recommended by the World Health Organization since 2013, but there remain limited data on the effects of Option B+ on long-term HIV-free survival in breastfeeding HIV-exposed infants. The Kigali Antiretroviral and Breastfeeding Assessment for the Elimination of HIV (Kabeho) study enrolled HIV-positive women from the third trimester of pregnancy to 2 weeks postpartum in 14 heath facilities implementing Option B+ in Kigali, Rwanda. Mother-child pairs in the longitudinal observational cohort were followed until 24 months postpartum, with HIV diagnostic testing at 6 weeks, and 9, 18 and 24 months. The Kaplan-Meier method was used to estimate HIV transmission, survival, and HIV-free survival through 24 months. We enrolled 608 HIV-positive women in 2013-2014; birth outcome data were available for 600 women and 597 live-born infants. By 6 weeks, 11 infants had died and 3 infants had confirmed HIV infection (0.5% transmission; 95% confidence interval [CI] 0.2-1.6). At 9 months, there were 9 additional deaths and 2 new infections (cumulative transmission 0.9%, 95% CI 0.4-2.2). At 18 months, there were 6 additional deaths and no new infant infections. At 24 months, there were no additional child deaths and 1 new infection (cumulative 2.2%, 95% CI 0.7-7.0), for an overall 24-month HIV-free survival of 93.2% (95% CI 89.5-95.6). Low transmission rates and high HIV-free survival at 24 months were achieved in breastfeeding infants of HIV-positive mothers receiving universal ART in urban health facilities in Rwanda, though vigilance on maintaining viral suppression for ART-experienced women is needed.
Collapse
Affiliation(s)
| | - Heather J. Hoffman
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, The George Washington University, Washington, DC
| | | | | | - Laura Guay
- Elizabeth Glaser Pediatric AIDS Foundation
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, The George Washington University, Washington, DC
| | | | | | - Anita Asiimwe
- Rwanda University Teaching Hospitals (CHU), Kigali, Rwanda
| | | |
Collapse
|
21
|
Moretton MA, Bertera F, Lagomarsino E, Riedel J, Chiappetta DA, Höcht C. Advances in therapy for the prevention of HIV transmission from mother to child. Expert Opin Pharmacother 2017; 18:657-666. [PMID: 28398096 DOI: 10.1080/14656566.2017.1316369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Actually, ~17.8 million women and 1.8 million children (<15 years) are currently infected with the Human Immunodeficiency Virus (HIV)/Acquired Immunodeficiency Syndrome (AIDS). Particularly, the majority of pediatric infections (>90%) resulted from 'HIV mother-to-child transmission' (MTCT), both in pregnancy, labour, delivery and later by breastfeeding. Due to its high pediatric incidence, MTCT represents a public health concern. Areas covered: In this review, we focus on available treatments and antiretroviral drugs recommended by the World Health Organization, and the main clinical investigations in antiretroviral pharmacotherapy to prevent the MTCT. Expert opinion: The MTCT has been improved dramatically in the last few years mainly due to prophylactic perinatal antiretroviral therapy for pregnant women living with HIV. However, there is still a milestone to reach since HIV MTCT remains as a public health challenge associated with MTCT though breastfeeding (post-natal transmission). In this context, different strategies could be employed as an attempt to reduce pediatric HIV infections. One of them involves the improvement of patient adherence to the HIV therapy. One possible solution is the development of novel long-acting formulations for prophylaxis of mothers and children, and a second possible solution is increase the inclusion of mothers and infants in care programs to more effectively prevent the vertical transmission.
Collapse
Affiliation(s)
- Marcela A Moretton
- a Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Cátedra de Tecnología Farmacéutica I ., Universidad de Buenos Aires , Buenos Aires , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Buenos Aires , Argentina
| | - Facundo Bertera
- c Departamento de Farmacología , Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología , Buenos Aires , Argentina
| | - Eduardo Lagomarsino
- d Departamento de Farmacología, Universidad de Buenos Aires , Facultad de Farmacia y Bioquímica, Cátedra de Farmacia Clínica , Buenos Aires , Argentina
| | - Jennifer Riedel
- a Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Cátedra de Tecnología Farmacéutica I ., Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Diego A Chiappetta
- a Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Cátedra de Tecnología Farmacéutica I ., Universidad de Buenos Aires , Buenos Aires , Argentina.,b Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Buenos Aires , Argentina
| | - Christian Höcht
- c Departamento de Farmacología , Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología , Buenos Aires , Argentina
| |
Collapse
|
22
|
Chikhungu LC, Newell ML, Rollins N. Under-five mortality according to maternal survival: a systematic review and meta-analysis. Bull World Health Organ 2017; 95:281-287. [PMID: 28479623 PMCID: PMC5407247 DOI: 10.2471/blt.15.157149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To investigate, within so-called general populations, the relationship between maternal survival and mortality of children younger than five years. METHODS We conducted a systematic review of literature published between January 1990 and November 2016 that reported maternal vital status and the corresponding mortality of children younger than five years. Seven studies were included in a qualitative analysis and four in a random-effects meta-analysis. Summary estimates of the odds of dying by maternal survival were obtained and statistical heterogeneity estimated. Quality of the included studies and evidence was assessed using a Cochrane tool for assessing risk of bias and the Grading of Recommendations Assessment, Development and Evaluation criteria, respectively. FINDINGS Among children younger than five years, those whose mother had died were found to be 4.09 times (95% confidence interval, CI: 2.40-6.98) more likely to die than those with surviving mothers. Due to heterogeneity (I2: 83%), further pooled estimates were not possible. For children that were motherless as a result of maternal mortality, the increased odds of dying ranged from 1.40 (95% CI: 0.47-4.21) to 2.92 (95% CI: 1.21-7.04) among those aged between two and four years, 6.1 (95% CI: 2.27-16.77) to 33.78 (95% CI: 24.21-47.14) for those younger than one year and 4.39 (95% CI: 3.34-5.78) to 51.68 (95% CI: 20.26-131.80) for those younger than six months. CONCLUSION The loss of a mother was associated with increased mortality among children, especially when maternal death occurred in the first year of the child's life.
Collapse
Affiliation(s)
- Lana Clara Chikhungu
- School of Languages and Area Studies, University of Portsmouth, Park Building, King Henry 1 Street, Portsmouth, PO1 2DZ, England
| | - Marie-Louise Newell
- Academic Unit of Health and Development, University of Southampton, Southampton, England
| | - Nigel Rollins
- Department of Maternal, Newborn, Child and Adolescent Health, World Health Organization, Geneva, Switzerland
| |
Collapse
|
23
|
Zash RM, Souda S, Leidner J, Binda K, Hick C, Powis K, Makhema J, Mmalane M, Essex M, Lockman S, Shapiro RL. High Proportion of Deaths Attributable to HIV Among Postpartum Women in Botswana Despite Widespread Uptake of Antiretroviral Therapy. AIDS Patient Care STDS 2017; 31:14-19. [PMID: 28051898 DOI: 10.1089/apc.2016.0154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mortality in the postpartum period may be impacted by antiretroviral therapy (ART) received in pregnancy, and whether ART is continued in the postpartum period. HIV-infected and HIV-uninfected mothers were enrolled within 48 h of delivery at five public hospital maternity wards throughout Botswana and followed for 24 months. Maternal deaths were reported by one of the approved contacts given by the mother at enrollment. Detailed information on the cause of death was not available. Risk factors for 24-month mortality were assessed using Cox proportional hazard models. From February 2012 to March 2013, 3000 mothers (1499 HIV infected and 1501 HIV uninfected) were enrolled, and 2985 (99.5%) were followed to 24 months or death, or until the death of their child. There were 26 total maternal deaths through 24 months postpartum [439 per 100,000 person-years (p-y)], 22 among HIV-infected women (758 per 100,000 p-y) and 4 among HIV-uninfected women (132 per 100,000 p-y). Maternal HIV-infection (aHR 5.0, 95% CI 1.6-15.2) and infant birth injury (aHR 3.8, 95% CI 1.3-11.4) were independent risk factors for maternal death. Universal ART in pregnancy became the standard-of-care after June 2012, and 978 (65%) women received ART in pregnancy; by 24 months postpartum or end of follow-up, 1148 (79%) had started ART overall. There was no significant difference in 24-month mortality among HIV-infected women who took ART in pregnancy and continued throughout the follow-up period compared with HIV-infected women who took ART or zidovudine in pregnancy and stopped postpartum (aHR 0.6, 95% CI 0.2-1.7). Despite high uptake of ART in pregnancy and postpartum, women with HIV infection in Botswana are five times more likely to die than HIV-uninfected women in the 24 months postpartum.
Collapse
Affiliation(s)
- Rebecca Marie Zash
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Immunology and Infectious Diseases, TH Chan Harvard School of Public Health, Boston, Massachusetts
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Sajini Souda
- Department of Pathology, Faculty of Medicine, University of Botswana, Gaborone, Botswana
| | | | | | - Chazha Hick
- Care Quality Commission, London, United Kingdom
| | - Kathleen Powis
- Immunology and Infectious Diseases, TH Chan Harvard School of Public Health, Boston, Massachusetts
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts
| | - Joseph Makhema
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Mompati Mmalane
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Max Essex
- Immunology and Infectious Diseases, TH Chan Harvard School of Public Health, Boston, Massachusetts
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Shahin Lockman
- Immunology and Infectious Diseases, TH Chan Harvard School of Public Health, Boston, Massachusetts
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts
| | - Roger L. Shapiro
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Immunology and Infectious Diseases, TH Chan Harvard School of Public Health, Boston, Massachusetts
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| |
Collapse
|
24
|
Dryden-Peterson S, Bvochora-Nsingo M, Suneja G, Efstathiou JA, Grover S, Chiyapo S, Ramogola-Masire D, Kebabonye-Pusoentsi M, Clayman R, Mapes AC, Tapela N, Asmelash A, Medhin H, Viswanathan AN, Russell AH, Lin LL, Kayembe MK, Mmalane M, Randall TC, Chabner B, Lockman S. HIV Infection and Survival Among Women With Cervical Cancer. J Clin Oncol 2016; 34:3749-3757. [PMID: 27573661 PMCID: PMC5477924 DOI: 10.1200/jco.2016.67.9613] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Purpose Cervical cancer is the leading cause of cancer death among the 20 million women with HIV worldwide. We sought to determine whether HIV infection affected survival in women with invasive cervical cancer. Patients and Methods We enrolled sequential patients with cervical cancer in Botswana from 2010 to 2015. Standard treatment included external beam radiation and brachytherapy with concurrent cisplatin chemotherapy. The effect of HIV on survival was estimated by using an inverse probability weighted marginal Cox model. Results A total of 348 women with cervical cancer were enrolled, including 231 (66.4%) with HIV and 96 (27.6%) without HIV. The majority (189 [81.8%]) of women with HIV received antiretroviral therapy before cancer diagnosis. The median CD4 cell count for women with HIV was 397 (interquartile range, 264 to 555). After a median follow-up of 19.7 months, 117 (50.7%) women with HIV and 40 (41.7%) without HIV died. One death was attributed to HIV and the remaining to cancer. Three-year survival for the women with HIV was 35% (95% CI, 27% to 44%) and 48% (95% CI, 35% to 60%) for those without HIV. In an adjusted analysis, HIV infection significantly increased the risk for death among all women (hazard ratio, 1.95; 95% CI, 1.20 to 3.17) and in the subset that received guideline-concordant curative treatment (hazard ratio, 2.63; 95% CI, 1.05 to 6.55). The adverse effect of HIV on survival was greater for women with a more-limited stage cancer ( P = .035), those treated with curative intent ( P = .003), and those with a lower CD4 cell count ( P = .036). Advanced stage and poor treatment completion contributed to high mortality overall. Conclusion In the context of good access to and use of antiretroviral treatment in Botswana, HIV infection significantly decreases cervical cancer survival.
Collapse
Affiliation(s)
- Scott Dryden-Peterson
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Memory Bvochora-Nsingo
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Gita Suneja
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Jason A. Efstathiou
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Surbhi Grover
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Sebathu Chiyapo
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Doreen Ramogola-Masire
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Malebogo Kebabonye-Pusoentsi
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Rebecca Clayman
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Abigail C. Mapes
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Neo Tapela
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Aida Asmelash
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Heluf Medhin
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Akila N. Viswanathan
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Anthony H. Russell
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Lilie L. Lin
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Mukendi K.A. Kayembe
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Mompati Mmalane
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Thomas C. Randall
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Bruce Chabner
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| | - Shahin Lockman
- Scott Dryden-Peterson, Akila N. Viswanathan, and Shahin Lockman, Brigham and Women’s Hospital; Scott Dryden-Peterson and Shahin Lockman, Harvard T.H. Chan School of Public Health; Scott Dryden-Peterson, Jason A. Efstathiou, Akila N. Viswanathan, Anthony H. Russell, Thomas C. Randall, Bruce Chabner, and Shahin Lockman, Harvard Medical School; Jason A. Efstathiou, Rebecca Clayman, Anthony H. Russell, Thomas C. Randall, and Bruce Chabner, Massachusetts General Hospital; Akila N. Viswanathan, Dana-Farber Cancer Institute, Boston, MA; Scott Dryden-Peterson, Abigail C. Mapes, Neo Tapela, Aida Asmelash, Mompati Mmalane, and Shahin Lockman, Botswana Harvard AIDS Institute Partnership; Memory Bvochora-Nsingo, Gaborone Private Hospital; Sebathu Chiyapo, Princess Marina Hospital; Doreen Ramogola-Masire, Botswana-University of Pennsylvania Partnership; Malebogo Kebabonye-Pusoentsi, Neo Tapela, Heluf Medhin, and Mukendi K.A. Kayembe, Botswana Ministry of Health, Gaborone, Botswana; Gita Suneja, University of Utah School of Medicine, Salt Lake City, UT; and Surbhi Grover and Lilie L. Lin, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
25
|
Ogwu A, Moyo S, Powis K, Asmelash A, Lockman S, Moffat C, Leidner J, Makhema J, Essex M, Shapiro R. Predictors of early breastfeeding cessation among HIV-infected women in Botswana. Trop Med Int Health 2016; 21:1013-1018. [PMID: 27224454 DOI: 10.1111/tmi.12729] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Infants born to HIV-infected women receiving antiretroviral treatment (ART) can be breastfed through at least 6 months with very low risk of HIV acquisition. We aimed to identify demographic and cultural factors that may influence mothers' willingness to breastfeed for the recommended duration. METHODS We evaluated factors associated with early cessation of breastfeeding (i.e. before 5 months post-partum) in a randomized clinical trial evaluating different ART regimens used for prevention of mother-to-child transmission during breastfeeding in Botswana. Univariate and multivariable Cox regressions were used to describe predictors of early exclusive BF cessation. RESULTS Among 677 women who started breastfeeding, the median time to breastfeeding cessation was 178 days (IQR 150-181) and 25.1% weaned early. In multivariable analysis, urban location (aHR = 1.86 95%CI 1.27-2.73; P = 0.002), salaried employment or being a student (aHR = 2.78 95% CI 1.63-4.75; P < 0.001) and infant hospitalisation before weaning (aHR = 2.04 95% CI 1.21-3.45; P = 0.008) were independently and significantly associated with early BF cessation. CONCLUSIONS Improved support for breastfeeding among employed mothers, especially in urban settings, may allow HIV-infected women who are receiving ART prophylaxis to breastfeed longer.
Collapse
Affiliation(s)
- Anthony Ogwu
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Trinity Medical Centre, Piccadilly, WA, Australia
| | - Sikhulile Moyo
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Kathleen Powis
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Aida Asmelash
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shahin Lockman
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Brigham and Women's Hospital, Boston, MA, USA
| | - Claire Moffat
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Greater Manchester West Mental Health NHS Foundation Trust, Prestwich, UK
| | | | - Joseph Makhema
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Max Essex
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Roger Shapiro
- Botswana-Harvard AIDS Institute Partnership, Gaborone, Botswana.,Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
26
|
Chikhungu LC, Bispo S, Rollins N, Siegfried N, Newell ML. HIV-free survival at 12-24 months in breastfed infants of HIV-infected women on antiretroviral treatment. Trop Med Int Health 2016; 21:820-8. [PMID: 27120500 PMCID: PMC5096069 DOI: 10.1111/tmi.12710] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Objective To provide estimates of HIV‐free survival at 12–24 months in breastfed children by maternal ART (6 months or lifelong) to inform WHO HIV and Infant Feeding guidelines. Methods Eighteen studies published 2005–2015 were included in a systematic literature review (1295 papers identified, 156 abstracts screened, 55 full texts); papers were analysed by narrative synthesis and meta‐analysis of HIV‐free survival by maternal ART regimen in a random effects model. We also grouped studies by feeding modality. Study quality was assessed using a modified Newcastle–Ottawa Scale (NOS) and GRADE. Results The pooled estimates for 12‐month HIV‐free survival were 89.8% (95% confidence interval, CI: 86.5%, 93.2%) for infants of mothers on ART for 6 months post‐natally (six studies) and 91.4% (95% CI 87.5%, 95.4%) for infants of mothers on lifelong ART (three studies). Eighteen‐month HIV‐free survival estimates were 89.0% (95% CI 83.9%, 94.2%) with 6 months ART (five studies) and 96.1% (95% CI 92.8%, 99.0%) with lifelong ART (three studies). Twenty‐four‐month HIV‐free survival for infants whose mothers were on ART to 6 months post‐natally (two studies) was 89.2% (95% CI 79.9%, 98.5%). Heterogeneity was considerable throughout. In four studies, HIV‐free survival in breastfed infants ranged from 87% (95% CI 78%, 92%) to 96% (95% CI 91%, 98%) and in formula‐fed infants from 67% (95% CI 35.5%, 87.9%) to 97.6% (95% CI 93.0%, 98.2%). Conclusion Our results highlight the importance of breastfeeding for infant survival and of ART in reducing the risk of mother‐to‐child HIV transmission and support the WHO recommendation to initiate ART for life immediately after HIV diagnosis.
Collapse
Affiliation(s)
| | - Stephanie Bispo
- Department of Social Statistics and Demography, University of Southampton, Southampton, UK
| | - Nigel Rollins
- Department of Maternal, New-born, Child and Adolescent Health, World Health Organization, Geneva, Switzerland
| | | | - Marie-Louise Newell
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
27
|
le Roux SM, Abrams EJ, Nguyen K, Myer L. Clinical outcomes of HIV-exposed, HIV-uninfected children in sub-Saharan Africa. Trop Med Int Health 2016; 21:829-45. [PMID: 27125333 DOI: 10.1111/tmi.12716] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE HIV-exposed but HIV-uninfected (HEU) children are widely considered at increased risk of mortality and morbidity. Recent advances in prevention of mother-to-child HIV transmission (PMTCT) strategies, incorporating life-long universal maternal antiretroviral therapy (ART, "Option B+") with extended breastfeeding, may improve HEU child health substantially. We critically reviewed reports of mortality/morbidity among HEU and HIV-unexposed (HU) children in sub-Saharan Africa. METHODS We searched Medline, EMBASE, CINAHL, PsycINFO, Academic Search Premier, Global Health & Psychosocial Instruments databases, conference abstracts, and reference lists for longitudinal studies from sub-Saharan Africa reporting mortality and clinical morbidity among HIV-uninfected children aged ≤10 years, by maternal HIV status. Studies were appraised by Newcastle-Ottawa Scale and ACROBAT-NRSI. Due to substantial heterogeneity of study designs, populations and results (I(2) = 75%), data were not synthesised. RESULTS We included 37 reports (28 studies, 11 164 HEU children); methodological and reporting quality were variable. Most reports came from settings without universal access to maternal ART (n = 35). Results were conflicting, with some studies indicating increased risk of mortality, hospitalisation and/or under-nutrition among HEU children, while others found no evidence of increased risk. In subanalyses, improved maternal health, ART use and breastfeeding were strongly protective for all outcomes. Only 39% (11/28) of studies adjusted for major confounders. Reports from settings using universal maternal ART with breastfeeding (n = 2) found no differences in growth or development but did not report mortality or infectious morbidity. CONCLUSIONS The existing literature provides little insight into HEU child health under recently adopted PMTCT strategies. There is a need for robust comparative data on HEU and HIV-unexposed child health outcomes under Option B+; optimising breastfeeding practices and increasing maternal use of ART should be urgent public health priorities.
Collapse
Affiliation(s)
- Stanzi M le Roux
- Division of Epidemiology & Biostatistics, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa.,Centre for Infectious Diseases & Epidemiology Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Elaine J Abrams
- ICAP, Mailman School of Public Health, Columbia University, New York, NY, USA.,College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Kelly Nguyen
- Centre for Infectious Diseases & Epidemiology Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Landon Myer
- Division of Epidemiology & Biostatistics, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa.,Centre for Infectious Diseases & Epidemiology Research, School of Public Health & Family Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
28
|
Abstract
OBJECTIVE To describe the pharmacokinetics of abacavir 600 mg once daily (q.d.) in HIV-1-positive women during pregnancy and postpartum. DESIGN A nonrandomized, open-label, multicentre, phase-IV study. METHODS HIV-positive pregnant women receiving abacavir 600 mg q.d. as part of clinical care were included. Intensive 24-h pharmacokinetic sampling was performed during the third trimester and at least 2 weeks after delivery. Pharmacokinetic parameters were calculated by noncompartmental analysis. Paired cord blood and maternal blood samples were taken at delivery when feasible. RESULTS A total of 14 women were included in the analysis. Geometric mean ratios (90% confidence intervals) of third trimester versus postpartum were 1.05 (0.92-1.19) for AUC0-24h and 1.00 (0.83-1.21) for Cmax. The median (range) ratio of abacavir cord plasma to maternal plasma was 1.0 (0.7-1.0, n = 3). Viral load at the third trimester visit was less than 50 copies/ml in 13 participants (93%; one unknown). In total, 13 (93%; one unknown) children were tested HIV-negative. CONCLUSION The pharmacokinetics of abacavir 600 mg q.d. during pregnancy are equivalent to postpartum. No dose adjustments are required during pregnancy and similar antiviral activity is expected.
Collapse
|
29
|
Evans C, Jones CE, Prendergast AJ. HIV-exposed, uninfected infants: new global challenges in the era of paediatric HIV elimination. THE LANCET. INFECTIOUS DISEASES 2016; 16:e92-e107. [PMID: 27049574 DOI: 10.1016/s1473-3099(16)00055-4] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 01/01/2016] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
The number of infants infected with HIV is declining with the rise in interventions for the elimination of paediatric HIV infection, but the number of uninfected infants exposed to HIV through their HIV-infected mothers is increasing. Interest in the health outcomes of HIV-exposed, uninfected infants has grown in the past decade, with several studies suggesting that these infants have increased mortality rates, increased infectious morbidity, and impaired growth compared with HIV-unexposed infants. However, heterogeneous results might reflect the inherent challenges in studies of HIV-exposed, uninfected infants, which need large populations with appropriate, contemporaneous comparison groups and repeated HIV testing throughout the period of breastfeeding. We review the effects of HIV exposure on mortality, morbidity, and growth, discuss the immunological abnormalities identified so far, and provide an overview of interventions that could be effective in this susceptible population. As the number of infants infected with HIV declines, the health needs of HIV-exposed, uninfected infants should be prioritised further, to ensure that post-2015 Sustainable Development Goals are achieved.
Collapse
Affiliation(s)
- Ceri Evans
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Christine E Jones
- Paediatric Infectious Diseases Research Group, Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
30
|
In-utero triple antiretroviral exposure associated with decreased growth among HIV-exposed uninfected infants in Botswana. AIDS 2016; 30:211-20. [PMID: 26684818 DOI: 10.1097/qad.0000000000000895] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To assess associations between in-utero triple antiretrovirals (cART) versus zidovudine (ZDV) monotherapy exposure and growth among HIV-uninfected children of HIV-infected women in Botswana. DESIGN Secondary retrospective data analysis from two randomized intervention trials of mother-to-child HIV transmission prevention. METHODS The Mashi and Mma Bana studies enrolled HIV-infected pregnant women, following their children through 24 months of age. This analysis includes singleton, full-term, HIV-exposed uninfected children. Mothers received cART or ZDV at least 2 weeks predelivery, and breastfed up to 6 months. Weight-for-age (WAZ), length-for-age (LAZ) and weight-for-length (WLZ) z-scores were derived. Mean z-scores were compared by exposure group at 24 months (t-test, linear regression). RESULTS Of 819 children, 303 were ZDV- and 516 cART-exposed in utero. Maternal median enrolment CD4 was higher among ZDV versus cART-treated mothers (393 versus 324 cells/μl; P < 0.0001). Median duration of antepartum antiretroviral use was shorter among ZDV-treated women (5.7 versus 12.0 weeks; P < 0.0001). Median months breastfed were similar (5.9 and 6.0; P = 0.43). At 24 months, mean LAZ and WAZ were significantly lower among cART-exposed children (LAZ -1.01 versus -0.74; P = 0.003) (WAZ -0.53 versus -0.30; P = 0.002) in unadjusted analyses. Adjusting for maternal CD4, viral load, enrolment site and maternal anthropometric measures, cART-exposed children had significantly lower LAZ and WAZ at 24 months (P = 0.0004 for both). CONCLUSION At 24 months, in-utero cART-exposed children had significantly lower LAZ and WAZ. Poor growth impacts childhood and adult mortality. These findings raise concerns for potential lasting health impacts among HIV-exposed uninfected children with in-utero cART exposure.
Collapse
|
31
|
NELSON JAE, FOKAR A, HUDGENS MG, COMPLIMENT KJ, HAWKINS JT, TEGHA G, KAMWENDO DD, KAYIRA D, MOFOLO IA, KOURTIS AP, JAMIESON DJ, VAN DER HORST CM, FISCUS SA. Frequent nevirapine resistance in infants infected by HIV-1 via breastfeeding while on nevirapine prophylaxis. AIDS 2015; 29:2131-8. [PMID: 26186128 PMCID: PMC4715989 DOI: 10.1097/qad.0000000000000814] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The objective of this study is to assess nevirapine (NVP) resistance in infants who became infected in the three arms of the Breastfeeding, Antiretrovirals and Nutrition (BAN) study: daily infant NVP prophylaxis, triple maternal antiretrovirals or no extra intervention for 28 weeks of breastfeeding. DESIGN A prospective cohort study. METHODS The latest available plasma or dried blood spot specimen was tested from infants who became HIV-positive between 3 and 48 weeks of age. Population sequencing was used to detect mutations associated with reverse transcriptase inhibitor resistance. Sequences were obtained from 22 out of 25 transmissions in the infant-NVP arm, 23 out of 30 transmissions in the maternal-antiretroviral arm and 33 out of 38 transmissions in the control arm. RESULTS HIV-infected infants in the infant-NVP arm were significantly more likely to have NVP resistance than infected infants in the other two arms of the trial, especially during breastfeeding through 28 weeks of age (56% in infant-NVP arm vs. 6% in maternal-antiretroviral arm and 11% in control arm, P¼0.004). There was a nonsignificant trend, suggesting that infants with NVP resistance tended to be infected earlier and exposed to NVP while infected for a greater duration than infants without resistance. CONCLUSION Infants on NVP prophylaxis during breastfeeding are at a reduced risk of acquiring HIV, but are at an increased risk of NVP resistance if they do become infected. These findings point to the need for frequent HIV testing of infants while on NVP prophylaxis, and for the availability of antiretroviral regimens excluding NVP for treating infants who become infected while on such a prophylactic regimen.
Collapse
Affiliation(s)
- Julie A. E. NELSON
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ali FOKAR
- Department of Medicine, School of Medicine; University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael G. HUDGENS
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, Gillings School of Global Public Health; University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kara J. COMPLIMENT
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J. Tyler HAWKINS
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gerald TEGHA
- University of North Carolina Project, Lilongwe, Malawi
| | | | | | | | - Athena P. KOURTIS
- Division of Reproductive Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Denise J. JAMIESON
- Division of Reproductive Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Charles M. VAN DER HORST
- Department of Medicine, School of Medicine; University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan A. FISCUS
- UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
32
|
Thorne C, Idele P, Chamla D, Romano S, Luo C, Newell ML. Morbidity and mortality in HIV-exposed uninfected children. Future Virol 2015. [DOI: 10.2217/fvl.15.70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Approximately 1.5 million HIV-positive women become pregnant annually. Antiretroviral therapy (ART) is central to prevention of mother-to-child transmission and maternal ART continued postpartum allows breastfeeding for at least 1 year of life, with important benefits for the child. In the pre-ART era, it was suggested that HIV-exposed uninfected (HEU) children may be at higher morbidity and mortality risk than children of HIV-negative mothers, associated with maternal illness and death and the lack, or limited duration, of breastfeeding as recommended for preventing mother-to-child transmission at that time. This review summarizes the evidence on morbidity and mortality risk in HEU children compared with HIV-unexposed children, and assesses the likely impact of roll-out of ART, which prolongs maternal survival and allows breastfeeding.
Collapse
Affiliation(s)
- Claire Thorne
- Population, Policy & Practice Programme, UCL Institute of Child Health, University College London, London, WC1E 6BT, UK
| | - Priscilla Idele
- Data & Analytics Section, UNICEF New York, New York, NY, USA
| | - Dick Chamla
- Health Section, UNICEF New York, New York, NY, USA
| | | | - Chewe Luo
- HIV/AIDS Section, UNICEF New York, New York, NY, USA
| | - Marie-Louise Newell
- Faculty of Medicine/Faculty of Social & Human Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
33
|
Holtz SA, Thetard R, Konopka SN, Albertini J, Amzel A, Fogg KP. A Systematic Review of Interventions to Reduce Maternal Mortality among HIV-Infected Pregnant and Postpartum Women. Int J MCH AIDS 2015; 4:11-24. [PMID: 27622004 PMCID: PMC4948129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND In high-prevalence populations, HIV-related maternal mortality is high with increased mortality found among HIV-infected pregnant and postpartum women compared to their uninfected peers. The scale-up of HIV-related treatment options and broader reach of programming for HIV-infected pregnant and postpartum women is likely to have decreased maternal mortality. This systematic review synthesized evidence on interventions that have directly reduced mortality among this population. METHODS Studies published between January 1, 2003 and November 30, 2014 were searched using PubMed. Of the 1,373 records screened, 19 were included in the analysis. RESULTS Interventions identified through the review include antiretroviral therapy (ART), micronutrients (multivitamins, vitamin A, and selenium), and antibiotics. ART during pregnancy was shown to reduce mortality. Timing of ART initiation, duration of treatment, HIV disease status, and ART discontinuation after pregnancy influence mortality reduction. Incident pregnancy in women already on ART for their health appears not to have adverse consequences for the mother. Multivitamin use was shown to reduce disease progression while other micronutrients and antibiotics had no beneficial effect on maternal mortality. CONCLUSIONS ART was the only intervention identified that decreased death in HIV-infected pregnant and postpartum women. The findings support global trends in encouraging initiation of lifelong ART for all HIV-infected pregnant and breastfeeding women (Option B+), regardless of their CD4+ count, as an important step in ensuring appropriate care and treatment. GLOBAL HEALTH IMPLICATIONS Maternal mortality is a rare event that highlights challenges in measuring the impact of interventions on mortality. Developing effective patient-centered interventions to reduce maternal morbidity and mortality, as well as corresponding evaluation measures of their impact, requires further attention by policy makers, program managers, and researchers.
Collapse
Affiliation(s)
- Sara A Holtz
- Health Programs Group, 4301 North Fairfax Drive, Arlington, Virginia 22203, USA
| | - Rudi Thetard
- African Strategies for Health at Management Sciences for Health, 4301 North Fairfax Drive, Arlington, Virginia 22203, USA
| | - Sarah N Konopka
- African Strategies for Health at Management Sciences for Health, 4301 North Fairfax Drive, Arlington, Virginia 22203, USA
| | - Jennifer Albertini
- United States Agency for International Development (USAID)/Bureau for Africa/Office of Sustainable Development, 1201 Pennsylvania Ave NW, Washington, DC 20004 USA
| | - Anouk Amzel
- USAID/Bureau for Global Health (BGH)/Office of HIV/AIDS, 2100 Crystal Drive, Arlington, VA 22202 USA
| | - Karen P Fogg
- USAID/Bureau for Global Health, 2100 Crystal Drive, Arlington, VA 22202 USA
| |
Collapse
|
34
|
A public health perspective on HIV/AIDS in Africa: Victories and unmet challenges. PATHOPHYSIOLOGY 2014; 21:237-56. [DOI: 10.1016/j.pathophys.2014.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/15/2014] [Indexed: 01/05/2023] Open
|