1
|
Zappulo E, Giaccone A, Schiano Moriello N, Gentile I. Pharmacological approaches to prevent vertical transmission of HIV and HBV. Expert Rev Clin Pharmacol 2022; 15:863-876. [PMID: 35876100 DOI: 10.1080/17512433.2022.2105202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Mother-to-child transmission (MTCT) is mainly responsible for the global pediatric HIV and HBV epidemic. Vertical transmission can be prevented and reduced through a series of interventions at the primary healthcare level, including extensive screening of pregnant women, administration of antivirals or immune-based treatments, counselling on type of delivery and breastfeeding. AREAS COVERED In this narrative review, approved therapeutic options for the treatment of pregnant women living with HIV or HBV are discussed with special focus on efficacy and safety profiles of each agent or drug class examined. The search was performed using Medline (via PubMed), Web of Science, and Google Scholar to identify studies assessing vertical transmission of both HIV and HBV. EXPERT OPINION Elimination of MTCT of both infections is firmly endorsed by major global commitments and the integration of tailored preventive interventions into maternal and newborn health services is of strategical importance to achieve this critical target. However, further research centered on antiviral-based and immunization trials among pregnant women is urgently needed to mitigate the risk of maternal and neonatal adverse outcomes, effectively prevent transmission to the offspring and finally eliminate the pediatric HIV and HBV epidemic, one of the key global health challenges of our time.
Collapse
Affiliation(s)
- Emanuela Zappulo
- Department of Clinical Medicine and Surgery, Infectious Diseases Unit, University of Naples Federico II, Naples, Italy
| | - Agnese Giaccone
- Department of Clinical Medicine and Surgery, Infectious Diseases Unit, University of Naples Federico II, Naples, Italy
| | - Nicola Schiano Moriello
- Department of Clinical Medicine and Surgery, Infectious Diseases Unit, University of Naples Federico II, Naples, Italy
| | - Ivan Gentile
- Department of Clinical Medicine and Surgery, Infectious Diseases Unit, University of Naples Federico II, Naples, Italy
| |
Collapse
|
2
|
Spencer DA, Shapiro MB, Haigwood NL, Hessell AJ. Advancing HIV Broadly Neutralizing Antibodies: From Discovery to the Clinic. Front Public Health 2021; 9:690017. [PMID: 34123998 PMCID: PMC8187619 DOI: 10.3389/fpubh.2021.690017] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
Despite substantial progress in confronting the global HIV-1 epidemic since its inception in the 1980s, better approaches for both treatment and prevention will be necessary to end the epidemic and remain a top public health priority. Antiretroviral therapy (ART) has been effective in extending lives, but at a cost of lifelong adherence to treatment. Broadly neutralizing antibodies (bNAbs) are directed to conserved regions of the HIV-1 envelope glycoprotein trimer (Env) and can block infection if present at the time of viral exposure. The therapeutic application of bNAbs holds great promise, and progress is being made toward their development for widespread clinical use. Compared to the current standard of care of small molecule-based ART, bNAbs offer: (1) reduced toxicity; (2) the advantages of extended half-lives that would bypass daily dosing requirements; and (3) the potential to incorporate a wider immune response through Fc signaling. Recent advances in discovery technology can enable system-wide mining of the immunoglobulin repertoire and will continue to accelerate isolation of next generation potent bNAbs. Passive transfer studies in pre-clinical models and clinical trials have demonstrated the utility of bNAbs in blocking or limiting transmission and achieving viral suppression. These studies have helped to define the window of opportunity for optimal intervention to achieve viral clearance, either using bNAbs alone or in combination with ART. None of these advances with bNAbs would be possible without technological advancements and expanding the cohorts of donor participation. Together these elements fueled the remarkable growth in bNAb development. Here, we review the development of bNAbs as therapies for HIV-1, exploring advances in discovery, insights from animal models and early clinical trials, and innovations to optimize their clinical potential through efforts to extend half-life, maximize the contribution of Fc effector functions, preclude escape through multiepitope targeting, and the potential for sustained delivery.
Collapse
Affiliation(s)
- David A. Spencer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Mariya B. Shapiro
- Molecular Microbiology & Immunology Department, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
- Molecular Microbiology & Immunology Department, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| |
Collapse
|
3
|
Mangold JF, Goswami R, Nelson AN, Martinez DR, Fouda GG, Permar SR. Maternal Intervention to Prevent Mother-to-Child Transmission of HIV: Moving Beyond Antiretroviral Therapy. Pediatr Infect Dis J 2021; 40:S5-S10. [PMID: 34042904 PMCID: PMC9215267 DOI: 10.1097/inf.0000000000002774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Widespread availability of antiretroviral therapy among pregnant women living with HIV has greatly reduced the rate of mother-to-child transmission (MTCT) of HIV across the globe. However, while Joint United Nations Programme on HIV/AIDS has set targets to reduce the annual number of new pediatric HIV infections to fewer than 40,000 in 2018 and fewer than 20,000 in 2020, progress towards these targets has plateaued at an unacceptably high global estimate of greater than 160,000 children newly infected with HIV in 2018. Moreover, it has become clear that expansion of maternal antiretroviral therapy alone will not be sufficient to close the remaining gap and eliminate MTCT of HIV. Additional strategies such as maternal or infant passive and/or active immunization that synergize with maternal antiretroviral therapy will be required to end the pediatric HIV epidemic. In this review, we outline the landscape of existing maternal interventions and emerging maternal immune-based approaches to prevent MTCT of HIV.
Collapse
Affiliation(s)
- Jesse F. Mangold
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Ria Goswami
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Ashley N. Nelson
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - David R. Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Global Public Health, Chapel Hill, NC, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
4
|
Kumar A, Giorgi EE, Tu JJ, Martinez DR, Eudailey J, Mengual M, Honnayakanahalli Marichannegowda M, Van Dyke R, Gao F, Permar SR. Mutations that confer resistance to broadly-neutralizing antibodies define HIV-1 variants of transmitting mothers from that of non-transmitting mothers. PLoS Pathog 2021; 17:e1009478. [PMID: 33798244 PMCID: PMC8055002 DOI: 10.1371/journal.ppat.1009478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/19/2021] [Accepted: 03/15/2021] [Indexed: 01/17/2023] Open
Abstract
Despite considerable reduction of mother-to-child transmission (MTCT) of HIV through use of maternal and infant antiretroviral therapy (ART), over 150,000 infants continue to become infected with HIV annually, falling far short of the World Health Organization goal of reaching <20,000 annual pediatric HIV cases worldwide by 2020. Prior to the widespread use of ART in the setting of pregnancy, over half of infants born to HIV-infected mothers were protected against HIV acquisition. Yet, the role of maternal immune factors in this protection against vertical transmission is still unclear, hampering the development of synergistic strategies to further reduce MTCT. It has been established that infant transmitted/founder (T/F) viruses are often resistant to maternal plasma, yet it is unknown if the neutralization resistance profile of circulating viruses predicts the maternal risk of transmission to her infant. In this study, we amplified HIV-1 envelope genes (env) by single genome amplification and produced representative Env variants from plasma of 19 non-transmitting mothers from the U.S. Women Infant Transmission Study (WITS), enrolled in the pre-ART era. Maternal HIV Env variants from non-transmitting mothers had similar sensitivity to autologous plasma as observed for non-transmitting variants from transmitting mothers. In contrast, infant variants were on average 30% less sensitive to paired plasma neutralization compared to non-transmitted maternal variants from both transmitting and non-transmitting mothers (p = 0.015). Importantly, a signature sequence analysis revealed that motifs enriched in env sequences from transmitting mothers were associated with broadly neutralizing antibody (bnAb) resistance. Altogether, our findings suggest that circulating maternal virus resistance to bnAb-mediated neutralization, but not autologous plasma neutralization, near the time of delivery, predicts increased MTCT risk. These results caution that enhancement of maternal plasma neutralization through passive or active vaccination during pregnancy may potentially drive the evolution of variants fit for vertical transmission. Despite widespread, effective use of ART among HIV infected pregnant women, new pediatric HIV infections increase by about 150,000 every year. Thus, alternative strategies will be required to reduce MTCT and eliminate pediatric HIV infections. Interestingly, in the absence of ART, less than half of HIV-infected pregnant women will transmit HIV, suggesting natural immune protection of infants from virus acquisition. To understand the impact of maternal plasma autologous virus neutralization responses on MTCT, we compared the plasma and bnAb neutralization sensitivity of the circulating viral population present at the time of delivery in untreated, HIV-infected transmitting and non-transmitting mothers. While there was no significant difference in the ability of transmitting and non-transmitting women to neutralize their own circulating virus strains, specific genetic motifs enriched in variants from transmitting mothers were associated with resistance to bnAbs, suggesting that acquired bnAb resistance is a common feature of vertically-transmitted variants. This work suggests that enhancement of plasma neutralization responses in HIV-infected mothers through passive or active vaccination could further drive selection of variants that could be vertically transmitted, and cautions the use of passive bnAbs for HIV-1 prophylaxis or therapy during pregnancy.
Collapse
Affiliation(s)
- Amit Kumar
- Duke Human Vaccine Institute, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Elena E. Giorgi
- Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Joshua J. Tu
- Duke Human Vaccine Institute, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - David R. Martinez
- Duke Human Vaccine Institute, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Joshua Eudailey
- Duke Human Vaccine Institute, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Michael Mengual
- Department of Medicine, Duke University Medical Centre, Durham, North Carolina, United States of America
| | | | - Russell Van Dyke
- Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Feng Gao
- Department of Medicine, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University Medical Centre, Durham, North Carolina, United States of America
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
5
|
Singh T, Otero CE, Li K, Valencia SM, Nelson AN, Permar SR. Vaccines for Perinatal and Congenital Infections-How Close Are We? Front Pediatr 2020; 8:569. [PMID: 33384972 PMCID: PMC7769834 DOI: 10.3389/fped.2020.00569] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022] Open
Abstract
Congenital and perinatal infections are transmitted from mother to infant during pregnancy across the placenta or during delivery. These infections not only cause pregnancy complications and still birth, but also result in an array of pediatric morbidities caused by physical deformities, neurodevelopmental delays, and impaired vision, mobility and hearing. Due to the burden of these conditions, congenital and perinatal infections may result in lifelong disability and profoundly impact an individual's ability to live to their fullest capacity. While there are vaccines to prevent congenital and perinatal rubella, varicella, and hepatitis B infections, many more are currently in development at various stages of progress. The spectrum of our efforts to understand and address these infections includes observational studies of natural history of disease, epidemiological evaluation of risk factors, immunogen design, preclinical research of protective immunity in animal models, and evaluation of promising candidates in vaccine trials. In this review we summarize this progress in vaccine development research for Cytomegalovirus, Group B Streptococcus, Herpes simplex virus, Human Immunodeficiency Virus, Toxoplasma, Syphilis, and Zika virus congenital and perinatal infections. We then synthesize this evidence to examine how close we are to developing a vaccine for these infections, and highlight areas where research is still needed.
Collapse
Affiliation(s)
- Tulika Singh
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Claire E. Otero
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Katherine Li
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Sarah M. Valencia
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Ashley N. Nelson
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
| | - Sallie R. Permar
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| |
Collapse
|
6
|
Liff I, Zash R, Mingochi D, Gaonakala FT, Diseko M, Mayondi G, Johnson K, James K, Makhema J, Shapiro R, Wylie BJ. Mid-trimester cervical length not associated with HIV status among pregnant women in Botswana. PLoS One 2020; 15:e0229500. [PMID: 32160214 PMCID: PMC7065819 DOI: 10.1371/journal.pone.0229500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/09/2020] [Indexed: 11/21/2022] Open
Abstract
Objective HIV-infected women on antiretroviral therapy have a higher risk of preterm birth than HIV-uninfected women in Botswana. To better understand the mechanism for preterm birth among HIV-infected women, we evaluated whether mid-trimester cervical length differed by HIV status as cervical shortening is associated with an increased risk for preterm birth. Methods We conducted a prospective cohort study among pregnant women receiving care at the Scottish Livingstone Hospital in Molepolole, Botswana. Consecutive women referred for routine obstetrical ultrasound were consented and enrolled if between 22w0d and 24w6d by ultrasound biometry. Blinded to maternal HIV status, an obstetrician measured transvaginal cervical length using standardized criteria. Cervical length, as well as the proportion of women with a short cervix (<25mm), were compared among HIV-infected and HIV-uninfected women. The acceptability of transvaginal ultrasound was also evaluated. Results Between April 2016 and April 2017, 853 women presenting for obstetric ultrasound were screened, 187 (22%) met eligibility criteria, and 179 (96%) were enrolled. Of those enrolled, 50 (28%) were HIV-infected (86% on antiretroviral therapy), 127 (71%) were HIV-uninfected, and 2 (1%) had unknown HIV status. There was no significant difference in mean cervical length between HIV-infected and HIV-uninfected women (32mm vs 31mm, p = 0.21), or in the proportion with a short cervix (10% vs 14%, p = 0.44). Acceptability data was available for 115 women who underwent a transvaginal ultrasound exam. Of these, 112 of 115 (97%) women deemed the transvaginal scan acceptable. Conclusions The increased risk of preterm birth observed among HIV-infected women receiving antiretroviral therapy in Botswana is unlikely associated with mid-trimester cervical shortening. Further research is needed to understand the underlying mechanism for preterm birth among HIV-infected women.
Collapse
Affiliation(s)
- Ingrid Liff
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, United States of America
- * E-mail:
| | - Rebecca Zash
- Department of Infectious Disease, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | | | | | - Modiegi Diseko
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Gloria Mayondi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Katherine Johnson
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Kaitlyn James
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Joseph Makhema
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Roger Shapiro
- Department of Infectious Disease, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Blair J. Wylie
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, United States of America
| |
Collapse
|
7
|
Voronin Y, Jani I, Graham BS, Cunningham CK, Mofenson LM, Musoke PM, Permar SR, Scarlatti G. Recent progress in immune-based interventions to prevent HIV-1 transmission to children. J Int AIDS Soc 2018; 20. [PMID: 29282882 PMCID: PMC5810316 DOI: 10.1002/jia2.25038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/04/2017] [Indexed: 11/30/2022] Open
Abstract
Globally, 150,000 new paediatric human immunodeficiency virus type 1 (HIV‐1) infections occurred in 2015. There remain complex challenges to the global elimination of paediatric HIV‐1 infection. Thus, for the global community to achieve elimination of new paediatric HIV‐1 infections, innovative approaches need to be explored. Immune‐based approaches to prevention of mother‐to‐child transmission (MTCT) may help fill some of the remaining gaps and provide new opportunities to achieve an AIDS‐free generation. Immune‐based interventions to prevent MTCT of HIV‐1 may include paediatric HIV vaccines and passive immunization approaches. Recent discoveries providing evidence of robust immune responses to HIV in infants open new and exciting prospects for paediatric HIV vaccines. Moreover, successful vaccination of infants has a different set of requirements than vaccination of adults and may be easier to achieve. Proof‐of‐concept has been established over the last two decades that passively administered HIV‐1 Env‐specific monoclonal antibody (mAbs) can prevent chimeric simian human immunodeficiency virus (SHIV) transmission to newborn nonhuman primates. There has been tremendous progress in isolating and characterizing broadly neutralizing antibodies to HIV, and clinical testing of these antibodies for treatment and prevention in both infants and adults is a major effort in the field. Immune‐based interventions need to be actively explored as they can provide critically important tools to address persistent challenges in MTCT prevention. It is a pivotal time for the field with active discussions on the best strategy to further reduce HIV infection of infants and accomplish the World Health Organization Fast‐Track 2030 goals to eliminate new paediatric HIV infections.
Collapse
Affiliation(s)
| | - Ilesh Jani
- Instituto Nacional de Saúde, Maputo, Mozambique
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Philippa M Musoke
- Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - Sallie R Permar
- Department of Pediatrics and Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
8
|
Gruell H, Klein F. Antibody-mediated prevention and treatment of HIV-1 infection. Retrovirology 2018; 15:73. [PMID: 30445968 PMCID: PMC6240265 DOI: 10.1186/s12977-018-0455-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/30/2018] [Indexed: 01/11/2023] Open
Abstract
Novel broadly neutralizing antibodies targeting HIV-1 hold promise for their use in the prevention and treatment of HIV-1 infection. Pre-clinical results have encouraged the evaluation of these antibodies in healthy and HIV-1-infected humans. In first clinical trials, highly potent broadly neutralizing antibodies have demonstrated their safety and significant antiviral activity by reducing viremia and delaying the time to viral rebound in individuals interrupting antiretroviral therapy. While emerging antibody-resistant viral variants have indicated limitations of antibody monotherapy, strategies to enhance the efficacy of broadly neutralizing antibodies in humans are under investigation. These include the use of antibody combinations to prevent viral escape, antibody modifications to increase the half-life and the co-administration of latency-reversing agents to target the cellular reservoir of HIV-1. We provide an overview of the results of pre-clinical and clinical studies of broadly HIV-1 neutralizing antibodies, discuss their implications and highlight approaches for the ongoing advancement into humans.
Collapse
Affiliation(s)
- Henning Gruell
- Laboratory of Experimental Immunology, Institute of Virology, University Hospital Cologne, Fürst-Pückler-Str. 56, 50935 Cologne, Germany
- German Center for Infection Research, Partner-Site Bonn-Cologne, Cologne, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, University Hospital Cologne, Fürst-Pückler-Str. 56, 50935 Cologne, Germany
- German Center for Infection Research, Partner-Site Bonn-Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
9
|
Carrillo J, Clotet B, Blanco J. Antibodies and Antibody Derivatives: New Partners in HIV Eradication Strategies. Front Immunol 2018; 9:2429. [PMID: 30405624 PMCID: PMC6205993 DOI: 10.3389/fimmu.2018.02429] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/02/2018] [Indexed: 12/25/2022] Open
Abstract
Promptly after primoinfection, HIV generates a pool of infected cells carrying transcriptionally silent integrated proviral DNA, the HIV-1 reservoir. These cells are not cleared by combined antiretroviral therapy (cART), and persist lifelong in treated HIV-infected individuals. Defining clinical strategies to eradicate the HIV reservoir and cure HIV-infected individuals is a major research field that requires a deep understanding of the mechanisms of seeding, maintenance and destruction of latently infected cells. Although CTL responses have been classically associated with the control of HIV replication, and hence with the size of HIV reservoir, broadly neutralizing antibodies (bNAbs) have emerged as new players in HIV cure strategies. Several reasons support this potential role: (i) over the last years a number of bNAbs with high potency and ability to cope with the extreme variability of HIV have been identified; (ii) antibodies not only block HIV replication but mediate effector functions that may contribute to the removal of infected cells and to boost immune responses against HIV; (iii) a series of new technologies have allowed for the in vitro design of improved antibodies with increased antiviral and effector functions. Recent studies in non-human primate models and in HIV-infected individuals have shown that treatment with recombinant bNAbs isolated from HIV-infected individuals is safe and may have a beneficial effect both on the seeding of the HIV reservoir and on the inhibition of HIV replication. These promising data and the development of antibody technology have paved the way for treating HIV infection with engineered monoclonal antibodies with high potency of neutralization, wide coverage of HIV diversity, extended plasma half-life in vivo and improved effector functions. The exciting effects of these newly designed antibodies in vivo, either alone or in combination with other cure strategies (latency reversing agents or therapeutic vaccines), open a new hope in HIV eradication.
Collapse
Affiliation(s)
- Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Institut de Recerca Germans Trias i Pujol, Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Institut de Recerca Germans Trias i Pujol, Badalona, Spain.,Chair in AIDS and Related Illnesses, Centre for Health and Social Care Research (CEES), Faculty of Medicine, Universitat de Vic - Universitat Central de Catalunya, Vic, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Institut de Recerca Germans Trias i Pujol, Badalona, Spain.,Chair in AIDS and Related Illnesses, Centre for Health and Social Care Research (CEES), Faculty of Medicine, Universitat de Vic - Universitat Central de Catalunya, Vic, Spain
| |
Collapse
|
10
|
Milligan C, Slyker JA, Overbaugh J. The Role of Immune Responses in HIV Mother-to-Child Transmission. Adv Virus Res 2017; 100:19-40. [PMID: 29551137 DOI: 10.1016/bs.aivir.2017.10.001] [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: 12/26/2022]
Abstract
HIV mother-to-child transmission (MTCT) represents a success story in the HIV/AIDS field given the significant reduction in number of transmission events with the scale-up of antiretroviral treatment and other prevention methods. Nevertheless, MTCT still occurs and better understanding of the basic biology and immunology of transmission will aid in future prevention and treatment efforts. MTCT is a unique setting given that the transmission pair is known and the infant receives passively transferred HIV-specific antibodies from the mother while in utero. Thus, infant exposure to HIV occurs in the face of HIV-specific antibodies, especially during delivery and breastfeeding. This review highlights the immune correlates of protection in HIV MTCT including humoral (neutralizing antibodies, antibody-dependent cellular cytotoxicity, and binding epitopes), cellular, and innate immune factors. We further discuss the future implications of this research as it pertains to opportunities for passive and active vaccination with the ultimate goal of eliminating HIV MTCT.
Collapse
Affiliation(s)
- Caitlin Milligan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, United States.
| | | | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
11
|
Bozzo J, Jorquera JI. Use of human immunoglobulins as an anti-infective treatment: the experience so far and their possible re-emerging role. Expert Rev Anti Infect Ther 2017; 15:585-604. [PMID: 28480779 DOI: 10.1080/14787210.2017.1328278] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Pooled human immunoglobulins (IGs) are prepared from plasma obtained from healthy donors as a concentrated antibody-containing solution. In addition, high-titer IGs (hyperimmune) against a specific pathogen can be obtained from vaccinated or convalescing donors. Currently, IGs can be used for the treatment of a variety of infections for which no specific therapy exists or that remain difficult to treat. Moreover, the recent pathogen outbreaks for which there is no approved treatment have renewed attention to the role of convalescent plasma and IGs. Areas covered: In this review, a historical perspective of the use of sera and IGs in humans as anti-infective agents (any viral, bacterial, parasitic infection), excluding immunodeficient patients, is presented from early development to the latest clinical studies. A Medline search was conducted to examine the peer-reviewed literature, with no date limits. Expert commentary: Human pooled plasma-derived IG products benefit from the polyclonal response of every individual donor and from the interindividual variability in such response. The trend to increased availability of vaccines for infectious diseases also opens new potential applications of hyperimmune IGs for emerging or re-emerging infectious diseases (e.g.: Ebola, Zika, Dengue), for the prevention and treatment in the general population, healthcare personnel and caregivers.
Collapse
Affiliation(s)
- Jordi Bozzo
- a Global Scientific & Medical Affairs , Grifols , Barcelona , Spain
| | - Juan I Jorquera
- b Bioscience Industrial Group, Research & Development , Grifols , Barcelona , Spain
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Although antiretroviral (ARV) prophylaxis can reduce mother-to-child transmission (MTCT) of HIV-1 to less than 2%, one-quarter of a million infants continue to be infected with HIV-1 annually. ARV prophylaxis alone will fail to eliminate infant HIV-1 infection because of issues of maternal adherence, toxicities, ARV-resistant virus strains, and acute maternal infection. Effective maternal and/or infant immunization will likely be required to achieve the goal of an HIV-free generation. RECENT FINDINGS This article describes recent studies of antibody responses that protect against vertical HIV-1 transmission. Studies have shown that maternal neutralization breadth is not a critical factor in MTCT, yet the ability of maternal plasma to neutralize autologous virus variants may be important in infant protection. There is also new evidence that infants mount robust and durable antibody responses to HIV-1 envelope following vaccination and can develop broad neutralization during infection. Finally, passive immunization of infants with highly potent and broad neutralizing antibodies may be an effective strategy to protect infants against infection with postnatally transmitted variants. SUMMARY Defining the characteristics of maternal and infant antibody responses that protect against MTCT will inform development of effective passive and active immunization strategies that will likely be required to eliminate pediatric HIV-1.
Collapse
|
13
|
Early weaning of HIV-exposed uninfected infants and risk of serious gastroenteritis: Findings from two perinatal HIV prevention trials in Kampala, Uganda. J Acquir Immune Defic Syndr 2015; 53:20-7. [PMID: 19779355 DOI: 10.1097/qai.0b013e3181bdf68e] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Objective: To assess serious gastroenteritis risk and mortality associated with early cessation of breastfeeding in infants enrolled in 2 prevention of maternal-to-child HIV-transmission trials in Uganda.Methods: We used hazard rates to evaluate serious gastroenteritis events by month of age and mortality among HIV-exposed uninfected infants enrolled in the HIV Network for Prevention Trials (HIVNET 012) (1997–2001) and HIV hyperimmune globulin (HIVIGLOB)/nevirapine (NVP) (2004–2007) trials. HIV-infected mothers were counseled using local infant feeding guidelines current at the time.Results: Breastfeeding cessation occurred earlier in HIVIGLOB/NVP compared with HIVNET 012 (median 4.0 versus 9.3 months,P,0.001). Rates of serious gastroenteritis were higher in HIVIGLOB/NVP (8.0/1000 child-months) than in HIVNET 012 (3.1/1000 child-months; P , 0.001). Serious gastroenteritis events also peaked earlier at 3–4 and 7–8 months (16.2/1000 and 15.0/1000 child-months,respectively) compared with HIVNET 012 at 9–10 months (20.8/1000 child-months). All cause infant mortality did not statistically differ between the HIVIGLOB/NVP and the HIVNET 012 trials [3.2/1000 versus 2.0/1000 child-months, respectively (P = 0.10)].Conclusions: Early breastfeeding cessation seen in the HIVIGLOB/NVP trial was associated with increased risk of serious gastroenteritis among HIV-exposed uninfected infants when compared with later breastfeeding cessation in the HIVNET 012 trial.Testing interventions, which could decrease HIV transmission through breastfeeding and allow safe
Collapse
|
14
|
US Public Health Service Task Force Recommendations for the Use of Antiretroviral Drugs in Pregnant Women Infected with HIV-1 for Maternal Health and for Reducing Perinatal HIV-1 Transmission in the United States, February 25, 2000, by the Perinatal. HIV CLINICAL TRIALS 2015. [DOI: 10.1310/3unn-lh5n-mcul-65gq] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
15
|
Voronin Y, Mofenson LM, Cunningham CK, Fowler MG, Kaleebu P, McFarland EJ, Safrit JT, Graham BS, Snow W. HIV monoclonal antibodies: a new opportunity to further reduce mother-to-child HIV transmission. PLoS Med 2014; 11:e1001616. [PMID: 24714363 PMCID: PMC3979646 DOI: 10.1371/journal.pmed.1001616] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Yegor Voronin and colleagues explore how monoclonal antibodies against HIV could provide a new opportunity to further reduce mother-to-child transmission of HIV and propose that new interventions should consider issues related to implementation, feasibility, and access. Please see later in the article for the Editors' Summary.
Collapse
Affiliation(s)
- Yegor Voronin
- Global HIV Vaccine Enterprise, New York, New York, United States of America
| | - Lynne M. Mofenson
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Rockville, Maryland, United States of America
| | - Coleen K. Cunningham
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - Mary G. Fowler
- Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Pontiano Kaleebu
- Medical Research Council/Uganda Virus Research Institute Uganda Research Unit on AIDS, Entebbe, Uganda
| | | | - Jeffrey T. Safrit
- Elizabeth Glaser Pediatric AIDS Foundation, Los Angeles, California, United States of America
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - William Snow
- Global HIV Vaccine Enterprise, New York, New York, United States of America
| |
Collapse
|
16
|
Abstract
Most infants born to human immunodeficiency virus (HIV)-infected women escape HIV infection. Infants evade infection despite an immature immune system and, in the case of breastfeeding, prolonged repetitive exposure. If infants become infected, the course of their infection and response to treatment differs dramatically depending upon the timing (in utero, intrapartum, or during breastfeeding) and potentially the route of their infection. Perinatally acquired HIV infection occurs during a critical window of immune development. HIV's perturbation of this dynamic process may account for the striking age-dependent differences in HIV disease progression. HIV infection also profoundly disrupts the maternal immune system upon which infants rely for protection and immune instruction. Therefore, it is not surprising that infants who escape HIV infection still suffer adverse effects. In this review, we highlight the unique aspects of pediatric HIV transmission and pathogenesis with a focus on mechanisms by which HIV infection during immune ontogeny may allow discovery of key elements for protection and control from HIV.
Collapse
|
17
|
Braibant M, Barin F. The role of neutralizing antibodies in prevention of HIV-1 infection: what can we learn from the mother-to-child transmission context? Retrovirology 2013; 10:103. [PMID: 24099103 PMCID: PMC3851888 DOI: 10.1186/1742-4690-10-103] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/10/2013] [Indexed: 01/12/2023] Open
Abstract
In most viral infections, protection through existing vaccines is linked to the presence of vaccine-induced neutralizing antibodies (NAbs). However, more than 30 years after the identification of AIDS, the design of an immunogen able to induce antibodies that would neutralize the highly diverse HIV-1 variants remains one of the most puzzling challenges of the human microbiology. The role of antibodies in protection against HIV-1 can be studied in a natural situation that is the mother-to-child transmission (MTCT) context. Indeed, at least at the end of pregnancy, maternal antibodies of the IgG class are passively transferred to the fetus protecting the neonate from new infections during the first weeks or months of life. During the last few years, strong data, presented in this review, have suggested that some NAbs might confer protection toward neonatal HIV-1 infection. In cases of transmission, it has been shown that the viral population that is transmitted from the mother to the infant is usually homogeneous, genetically restricted and resistant to the maternal HIV-1-specific antibodies. Although the breath of neutralization was not associated with protection, it has not been excluded that NAbs toward specific HIV-1 strains might be associated with a lower rate of MTCT. A better identification of the antibody specificities that could mediate protection toward MTCT of HIV-1 would provide important insights into the antibody responses that would be useful for vaccine development. The most convincing data suggesting that NAbs migh confer protection against HIV-1 infection have been obtained by experiments of passive immunization of newborn macaques with the first generation of human monoclonal broadly neutralizing antibodies (HuMoNAbs). However, these studies, which included only a few selected subtype B challenge viruses, provide data limited to protection against a very restricted number of isolates and therefore have limitations in addressing the hypervariability of HIV-1. The recent identification of highly potent second-generation cross-clade HuMoNAbs provides a new opportunity to evaluate the efficacy of passive immunization to prevent MTCT of HIV-1.
Collapse
Affiliation(s)
- Martine Braibant
- Université François-Rabelais, UFR Médecine, Inserm U966 10 bld Tonnellé, cedex, 37032 Tours, France.
| | | |
Collapse
|
18
|
Farzin A, Boyer P, Ank B, Nielsen-Saines K, Bryson Y. Amniotic fluid exhibits an innate inhibitory activity against HIV type 1 replication in vitro. AIDS Res Hum Retroviruses 2013; 29:77-83. [PMID: 22998428 DOI: 10.1089/aid.2011.0355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Indirect evidence suggests that amniotic fluid (AF) may play a role in the pathogenesis of in utero HIV-1 transmission. The purpose of this study was to evaluate the potential innate inhibitory role of AF on HIV replication, which may contribute to protection of the fetus against intrauterine transmission. AF was collected from term HIV-1-negative women undergoing scheduled cesarean section. The inhibitory effect of AF against HIV-1(BA-L) replication was tested in vitro with or without the addition of protease inhibitor cocktail (PIC) in PHA-stimulated PBMC cultures. Quantitative measurement of human neutrophil peptides 1-3 (HNP1-3) was performed on all AF samples, using an ELISA assay. AF exhibited a dose-dependent inhibitory activity against HIV-1(BA-L) replication, with all samples (n=12) reaching significant inhibitory effect using 50% AF. In vitro, this activity decreased over time, but was able to be sustained with the addition of PIC. The HNP1-3 concentration in AF samples (n=12) ranged from undetectable (<41 pg/ml, n=3) to >250,000 pg/ml with a median of 5,146 pg/ml. AF exhibited a significant and dose-dependent innate inhibitory activity against HIV-1 replication, which was present in all AF samples tested. This effect was prolonged in the presence of PIC, suggesting that the inhibitory factor was in the cell-free protein fraction. The HNP1-3 concentration in AF was in the subinhibitory range for HIV with no correlation between its concentration and the HIV-1 inhibitory activity. These data show the presence of a significant innate inhibitory activity against HIV in AF.
Collapse
Affiliation(s)
- Azadeh Farzin
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of California Los Angeles, Los Angeles, California
| | - Pamela Boyer
- Department of Obstetrics and Gynecology, University of California Los Angeles, Los Angeles, California
| | - Bonnie Ank
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of California Los Angeles, Los Angeles, California
| | - Karin Nielsen-Saines
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of California Los Angeles, Los Angeles, California
| | - Yvonne Bryson
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
19
|
Were the interests of the vulnerable truly served? The predictable failure of HIVIG. J Acquir Immune Defic Syndr 2012; 61:e8-10; author reply e10-2. [PMID: 22918129 DOI: 10.1097/qai.0b013e318253a5dc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Abstract
PURPOSE OF REVIEW The major target groups for an HIV vaccine include breastfeeding infants and adolescents. Differential immune maturity in these age groups may significantly impact vaccine efficacy, and should be taken into account when developing vaccines. Here we review these differences, with an emphasis on the immune response to vaccines for HIV and other pathogens. Recommendations for potential adaptation of current HIV vaccines are also made. RECENT FINDINGS An effective neonatal vaccine needs to be immunogenic in the presence of maternal antibody, and must induce cytotoxic T-lymphocyte responses, neutralizing antibody responses, both systemic and mucosal. There is renewed hope in the possibility of stimulating neutralizing antibodies with HIV vaccination. DNA vaccines are promising for neonates, but will need appropriate boosting. Certain adjuvants and vector delivery systems are more suitable for neonates. Adolescents may have stronger immune responses to HIV vaccines than adults, and will also require induction of mucosal neutralizing humoral and cellular immunity. SUMMARY Some current HIV vaccine strategies may need adaptation for neonates and suitable product development should be accelerated. Vaccines could induce better responses in adolescents and therefore should not be discarded prematurely. Development of vaccines that have potential for these age groups is an urgent global priority.
Collapse
|
21
|
Reply to “Were the Interests of the Vulnerable Truly Served? The Predictable Failure of HIV Immune Globulin”. J Acquir Immune Defic Syndr 2012. [DOI: 10.1097/qai.0b013e318263498c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
22
|
Safety and efficacy of HIV hyperimmune globulin for prevention of mother-to-child HIV transmission in HIV-1-infected pregnant women and their infants in Kampala, Uganda (HIVIGLOB/NVP STUDY). J Acquir Immune Defic Syndr 2011; 58:399-407. [PMID: 21826009 DOI: 10.1097/qai.0b013e31822f8914] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND This phase III, randomized, clinical trial compared single-dose nevirapine (sdNVP) plus HIV hyperimmune globulin (HIVIGLOB) with sdNVP alone for preventing maternal-to-child transmission of HIV. Primary objectives were to determine rates of HIV infection among infants and to assess the safety of HIVIGLOB in combination with sdNVP in HIV-infected Ugandan pregnant women and their infants. METHODS Mother-infant pairs were randomized to receive 200 mg of nevirapine to women in labor and 2 mg/kg NVP to newborns within 72 hours after birth (sdNVP arm) or to receive sdNVP plus a single intravenous 240-mL dose of HIVIGLOB given to women at 36- to 38-week gestation and a single intravenous 24-mL dose to newborns within 18 hours of birth (HIVIGLOB/sdNVP arm). Risk of HIV infection was determined using Kaplan-Meier and risk ratio estimates at birth, 2, 6, 14 weeks, 6, and 12 months of age. RESULTS Intent-to-treat analysis included 198 HIVIGLOB/sdNVP and 294 sdNVP mother-infant pairs. At 6 months of age, the primary endpoint, there was no statistically significant difference in HIV transmission in the HIVIGLOB/sdNVP arm vs. the sdNVP arm [18.7% vs. 15.0%; risk ratio = 1.240 (95% confidence interval: 0.833 to 1.846); P = 0.290]. Similarly, the proportion of serious adverse events in the HIVIGLOB/sdNVP and sdNVP arms, respectively, for mothers (18.9% vs. 19.3%; P = 0.91) and infants (62.6% vs. 59.5%; P = 0.51) was not significantly different. CONCLUSIONS Giving mother-infant pairs an infusion of peripartum HIV hyperimmune globulin in addition to sdNVP for preventing maternal-to-child transmission was as safe as sdNVP alone but was no more effective than sdNVP alone in preventing HIV transmission.
Collapse
|
23
|
Prevention of mother-to-child HIV-1 transmission--why we still need a preventive HIV immunization strategy. J Acquir Immune Defic Syndr 2011; 58:359-62. [PMID: 21909031 DOI: 10.1097/qai.0b013e318235517e] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
24
|
Rongkavilit C, Asmar BI. Advances in prevention of mother-to-child HIV transmission: the international perspectives. Indian J Pediatr 2011; 78:192-204. [PMID: 20953847 DOI: 10.1007/s12098-010-0258-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 09/27/2010] [Indexed: 10/18/2022]
Abstract
We have sufficient knowledge and unprecedented access to global resources to dramatically reduce the transmission of HIV-1 from mother to children worldwide. Most transmission occurs during delivery and after birth through breastfeeding. For this reason, efforts to interrupt transmission have focused on peripartum period and safe infant feeding. This includes the use of antiretroviral therapy, elective cesarean section, avoidance of breastfeeding, and exclusive breastfeeding. This review summarizes recent studies and new international development on the prevention of mother-to-child HIV transmission. Prevention of mother-to-child transmission of HIV should now be integrated as part of basic maternal and child health services.
Collapse
Affiliation(s)
- Chokechai Rongkavilit
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Children's Hospital of Michigan, Wayne State University School of Medicine, 3901 Beaubien Blvd., Detroit, MI 48201, USA.
| | | |
Collapse
|
25
|
Abstract
Identification of HIV-infected pregnant women by antenatal screening is crucial to prevent mother-to-child transmission. In Hong Kong, little is known about women's opinions about the screening test. This cross-sectional study examines the decisions of Hong Kong Chinese pregnant women to undergo antenatal HIV screening and the reasons for their choices. Among 191 pregnant women studied, 147 (77.0%) indicated that they would opt for antenatal HIV screening if offered. Of those, 113 (76.9%) would do so for 'the health of their newborns' and 93 (63.3%) for 'their own health'. Fourteen (7.3%) would decline the test, because they have 'one stable sexual partner' (n=9, 64.3%) and believe that 'HIV infection was not their concern' (n=8, 57.1%). Women with a lower educational attainment (adjusted OR=3.77; 95% C.I., 1.12-12.67, p=0.03) were likely to choose antenatal HIV screening. Results of this study are predictive of pregnant women's responses to Hong Kong's newly enacted antenatal HIV screening programme.
Collapse
Affiliation(s)
- C F Ho
- Department of Health, Hong Kong, PR China
| | | |
Collapse
|
26
|
Watts DH, Lu M, Thompson B, Tuomala RE, Meyer WA, Mendez H, Rich K, Hanson C, LaRussa P, Diaz C, Mofenson LM. Treatment interruption after pregnancy: effects on disease progression and laboratory findings. Infect Dis Obstet Gynecol 2009; 2009:456717. [PMID: 19893751 PMCID: PMC2772023 DOI: 10.1155/2009/456717] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 08/25/2009] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To assess clinical progression and inflammatory markers among women stopping or continuing antiretroviral therapy (ART) after pregnancy. METHODS ART-naïve women with CD4+ lymphocyte counts >350 cells/uL initiating ART during pregnancy had clinical events and laboratory markers compared over one year postpartum between those stopping (n = 59) or continuing (n = 147) ART. RESULTS Slopes in CD4 count and HIV RNA did not differ between groups overall and in subsets of ZDV or combination therapy. The hazard ratio (HR) of a new class B event was 2.09 (95% CI 0.79-5.58) among women stopping ART, 1.24 (0.31-4.95) in those stopping ZDV, and 2.93 (0.64-13.36) among those stopping combination therapy. Women stopping ART had increased immune activation. No significant differences were seen in C-reactive protein, lipids, leptin, or interleukin-6. CONCLUSIONS While changes in CD4 and HIV RNA levels over one year were similar between women stopping or continuing ART postpartum, higher immune activation among women stopping therapy requires further study.
Collapse
Affiliation(s)
- D H Watts
- Pediatric, Adolescent and Maternal AIDS Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Menezes EV, Yakoob MY, Soomro T, Haws RA, Darmstadt GL, Bhutta ZA. Reducing stillbirths: prevention and management of medical disorders and infections during pregnancy. BMC Pregnancy Childbirth 2009; 9 Suppl 1:S4. [PMID: 19426467 PMCID: PMC2679410 DOI: 10.1186/1471-2393-9-s1-s4] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND An estimated two-thirds of the world's 3.2 million stillbirths occur antenatally, prior to labour, and are often overlooked in policy and programs. Poorly recognised, untreated or inadequately treated maternal infections such as syphilis and malaria, and maternal conditions including hypertensive disorders, are known risk factors for stillbirth. METHODS We undertook a systematic review of the evidence for 16 antenatal interventions with the potential to prevent stillbirths. We searched a range of sources including PubMed and the Cochrane Library. For interventions with prior Cochrane reviews, we conducted additional meta-analyses including eligible newer randomised controlled trials following the Cochrane protocol. We focused on interventions deliverable at the community level in low-/middle-income countries, where the burden of stillbirths is greatest. RESULTS Few of the studies we included reported stillbirth as an outcome; most that did were underpowered to assess this outcome. While Cochrane reviews or meta-analyses were available for many interventions, few focused on stillbirth or perinatal mortality as outcomes, and evidence was frequently conflicting. Several interventions showed clear evidence of impact on stillbirths, including heparin therapy for certain maternal indications; syphilis screening and treatment; and insecticide-treated bed nets for prevention of malaria. Other interventions, such as management of obstetric intrahepatic cholestasis, maternal anti-helminthic treatment, and intermittent preventive treatment of malaria, showed promising impact on stillbirth rates but require confirmatory studies. Several interventions reduced known risk factors for stillbirth (e.g., anti-hypertensive drugs for chronic hypertension), yet failed to show statistically significant impact on stillbirth or perinatal mortality rates. Periodontal disease emerged as a clear risk factor for stillbirth but no interventions have reduced stillbirth rates. CONCLUSION Evidence for some newly recognised risk factors for stillbirth, including periodontal disease, suggests the need for large, appropriately designed randomised trials to test whether intervention can minimise these risks and prevent stillbirths. Existing evidence strongly supports infection control measures, including syphilis screening and treatment and malaria prophylaxis in endemic areas, for preventing antepartum stillbirths. These interventions should be incorporated into antenatal care programs based on attributable risks and burden of disease.
Collapse
MESH Headings
- Anthelmintics/therapeutic use
- Anti-Bacterial Agents/therapeutic use
- Anticoagulants/therapeutic use
- Antihypertensive Agents/therapeutic use
- Antioxidants/therapeutic use
- Antiviral Agents/therapeutic use
- Causality
- Cholestasis, Intrahepatic/epidemiology
- Cholestasis, Intrahepatic/prevention & control
- Comorbidity
- Dental Care/methods
- Dietary Supplements
- Evidence-Based Medicine
- Female
- Fetal Death/epidemiology
- Fetal Death/prevention & control
- Fetal Membranes, Premature Rupture/epidemiology
- Fetal Membranes, Premature Rupture/prevention & control
- Global Health
- HIV Infections/epidemiology
- HIV Infections/prevention & control
- Humans
- Hypertension/epidemiology
- Hypertension/prevention & control
- Hypertension, Pregnancy-Induced/epidemiology
- Hypertension, Pregnancy-Induced/prevention & control
- Infectious Disease Transmission, Vertical/prevention & control
- Platelet Aggregation Inhibitors/therapeutic use
- Pregnancy
- Pregnancy Complications, Cardiovascular/epidemiology
- Pregnancy Complications, Cardiovascular/prevention & control
- Pregnancy Complications, Infectious/epidemiology
- Pregnancy Complications, Infectious/prevention & control
- Pregnancy Complications, Infectious/therapy
- Prenatal Care/methods
- Risk Factors
- Stillbirth/epidemiology
- Venous Thromboembolism/epidemiology
- Venous Thromboembolism/prevention & control
Collapse
Affiliation(s)
- Esme V Menezes
- Division of Maternal and Child Health, The Aga Khan University, Karachi-74800, Pakistan
| | - Mohammad Yawar Yakoob
- Division of Maternal and Child Health, The Aga Khan University, Karachi-74800, Pakistan
| | - Tanya Soomro
- Division of Maternal and Child Health, The Aga Khan University, Karachi-74800, Pakistan
| | - Rachel A Haws
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gary L Darmstadt
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zulfiqar A Bhutta
- Division of Maternal and Child Health, The Aga Khan University, Karachi-74800, Pakistan
| |
Collapse
|
28
|
Jourdain G, Mary JY, Coeur SL, Ngo-Giang-Huong N, Yuthavisuthi P, Limtrakul A, Traisathit P, McIntosh K, Lallemant M. Risk factors for in utero or intrapartum mother-to-child transmission of human immunodeficiency virus type 1 in Thailand. J Infect Dis 2007; 196:1629-36. [PMID: 18008246 DOI: 10.1086/522009] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Accepted: 06/05/2007] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The identification of risk factors for in utero and intrapartum transmission of human immunodeficiency virus type 1 (HIV-1) is crucial to the design and understanding of preventive interventions. METHODS The randomized Perinatal HIV Prevention Trial-1 enrolled 1437 pregnant women and their non-breast-fed infants, to compare the efficacy of various durations of zidovudine prophylaxis. Using univariate and multivariate logistic regression analyses, we studied the role that factors known or occurring at various times during gestation or delivery play in in utero and intrapartum transmission. RESULTS Variables independently associated with in utero transmission were HIV-1 load >35,000 copies/mL (adjusted odds ratio [AOR], 4.2) and delayed initiation of maternal zidovudine prophylaxis until >31.4 weeks gestation (AOR, 3.0). Variables associated with intrapartum transmission were HIV-1 load >10,000 copies/mL (AOR, 3.8 for 10,000-35,000 copies/mL and 7.1 for >35,000 copies/mL), induction of labor (AOR, 2.6), and premature labor with tocolysis (AOR, 15.1). CONCLUSIONS With the exception of very high HIV-1 load, risk factors for in utero transmission were different from those for intrapartum transmission. Optimal prophylactic interventions must address each of the major risk factors, with appropriate timing.
Collapse
Affiliation(s)
- Gonzague Jourdain
- Institut de Recherche pour le Developpement, UMI 174 "Epidemiologie Clinique, Sante Maternelle et Infantile et Sida", Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Harris NS, Fowler MG, Sansom SL, Ruffo N, Lampe MA. Use of enhanced perinatal human immunodeficiency virus surveillance methods to assess antiretroviral use and perinatal human immunodeficiency virus transmission in the United States, 1999-2001. Am J Obstet Gynecol 2007; 197:S33-41. [PMID: 17825649 DOI: 10.1016/j.ajog.2007.03.081] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 03/20/2007] [Accepted: 03/27/2007] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Significant reductions in perinatal human immunodeficiency virus (HIV) transmission have been demonstrated in which the HIV-infected mothers and their HIV-exposed infants receive prenatal, intrapartum, and neonatal antiretroviral therapy. STUDY DESIGN We used data that were collected through the Enhanced Perinatal Surveillance system for HIV-exposed singleton births that occurred 1999-2001 in 24 sites. RESULTS The overall infant infection rate for the 3 years was 4.7%. Compared with zidovudine monotherapy, those patients who received zidovudine with other drugs that included a protease inhibitor and those who received zidovudine and other drugs with no protease inhibitor were less likely to have an infected infant (adjusted odds ratio, 0.4 [95% CI, 0.3-0.07]; adjusted odds ratio, 0.5 [95% CI, 0.3-0.8], respectively). CONCLUSION These data support the current treatment recommendations and show that infants were less likely to be infected when the mothers were given a prenatal antiretroviral therapy regimen that contained zidovudine with additional antiretroviral drugs with or without a protease inhibitor in addition to receiving antiretrovirals during delivery and neonatally.
Collapse
Affiliation(s)
- Norma S Harris
- Centers for Disease Control and Prevention, National Center for HIV, STD, and TB Prevention, Division of HIV/AIDS Prevention, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
30
|
Escobar PA, Olivero OA, Wade NA, Abrams EJ, Nesel CJ, Ness RB, Day RD, Day BW, Meng Q, O'Neill JP, Walker DM, Poirier MC, Walker VE, Bigbee WL. Genotoxicity assessed by the comet and GPA assays following in vitro exposure of human lymphoblastoid cells (H9) or perinatal exposure of mother-child pairs to AZT or AZT-3TC. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2007; 48:330-43. [PMID: 17358027 DOI: 10.1002/em.20285] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The genotoxicity of zidovudine (AZT) based treatments was investigated in human H9 lymphoblastoid cells in an in vitro study and in red blood cells (RBCs) from perinatally exposed HIV-1-infected mothers and their infants in an observational cohort study. Exposure of H9 cells for 24 hr to AZT produced dose-dependent increases in Comet assay tail moment (TM) when electrophoresed at pH 13.0, but not at pH 12.1 or pH 8.0, suggesting that DNA damage was via alkali-labile lesions and not double-stranded DNA strand breaks. The TM dose response at pH 13.0 correlated directly with AZT-DNA incorporation determined by AZT-radioimmunoassay. Levels of DNA damage in utero, measured by Comet assay TM, were similar in cord blood mononuclear cells of nucleoside analog-exposed newborns (n = 43) and unexposed controls (n = 40). In contrast, the glycophorin A (GPA) somatic cell mutation assay (which screens for large-scale DNA damage in RBCs) showed clear evidence that GPA N/N variants, arising from chromosome loss and duplication, somatic recombination, and gene conversion, were significantly elevated in mother-child pairs receiving prepartum AZT plus lamivudine (3TC). Cord blood from newborns exposed to AZT-3TC had GPA N/N variant frequencies of 4.7 +/- 0.7 (mean +/- SE) x 10(-6) RBCs (n = 26 infants) compared with 2.2 +/- 0.3 x 10(-6) RBCs for unexposed controls (n = 30 infants; P < 0.001). Elevations in GPA N/N variants generally persisted through 1 year of age in nucleoside analog-exposed children. Overall, the mutagenic effects found in mother-child pairs receiving AZT-based treatments justify their surveillance for long-term genotoxic consequences.
Collapse
Affiliation(s)
- Patricia A Escobar
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Watts DH, Li D, Handelsman E, Tilson H, Paul M, Foca M, Vajaranant M, Diaz C, Tuomala R, Thompson B. Assessment of Birth Defects According to Maternal Therapy Among Infants in the Women and Infants Transmission Study. J Acquir Immune Defic Syndr 2007; 44:299-305. [PMID: 17159659 DOI: 10.1097/qai.0b013e31802e2229] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND To evaluate rate and types of birth defects according to timing of antiretroviral exposure among babies born to HIV-infected women. METHODS Anomalies identified during the prenatal, neonatal, or follow-up period were classified using criteria of the Antiretroviral Pregnancy Registry. Antiretroviral use was classified as none, second or third trimester only, or first trimester. RESULTS From January 1, 1990 through June 30, 2004, 2527 live births (LBs) occurred to 2353 women. Defects were identified in 90 babies for a rate of 3.56 defects per 100 LBs. The rate of defects was 3.19 per 100 LBs (24 of 752 LBs) with first-trimester antiretroviral exposure, 3.54 per 100 LBs (41 of 1158 LBs) with exposure later in pregnancy, and 4.05 of 100 LBs (25 of 617 LBs) with no antiretroviral use. Only genital abnormalities, specifically hypospadias, were significantly increased among babies born to women with first-trimester exposure to antiretrovirals (7 of 382 male LBs) compared with the 2 other groups (2 of 892 male LBs; P = 0.007). On logistic regression, use of zidovudine in the first trimester was associated with hypospadias (adjusted odds ratio = 10.68, 95% confidence interval: 2.11 to 54.13; P = 0.004). CONCLUSIONS In general, data were reassuring, although the frequency of exposure to newer agents was limited. The increased risk of hypospadias after first-trimester exposure must be explored, because this association has not been detected previously.
Collapse
Affiliation(s)
- D Heather Watts
- Pediatric, Adolescent, and Maternal AIDS Branch, Center for Research on Mothers and Infants, National Institute of Child Health and Human Development/NIH, 6100 Executive Boulevard, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
The authors discuss a new study in PLoS Medicine that examines whether human anti-HIV monoclonal antibodies might be useful in South Africa for preventing mother-to-child transmission of subtype C HIV viruses.
Collapse
|
33
|
Lehtovirta P, Skogberg K, Salo E, Ammälä P, Ristola M, Suni J, Paavonen J, Heikinheimo O. Pregnancy outcome among HIV-infected women in the Helsinki metropolitan area. Acta Obstet Gynecol Scand 2005; 84:945-50. [PMID: 16167909 DOI: 10.1111/j.0001-6349.2005.00755.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Antiretroviral medication and good obstetric practice have greatly reduced the rate of vertical transmission of human immunodeficiency virus (HIV) infection. The incidence of HIV infection has remained low in Finland. Universal antenatal screening has been offered to all pregnant women since 1998. METHODS We analyzed the outcomes of 52 pregnancies among 45 HIV-infected mothers delivering at our department during 1993-2003. A multidisciplinary team planned the management strategy individually and supported the mother's adherence to treatment. RESULTS The incidence of HIV infection among women delivering in our hospital district increased from 0.6/10,000 (95% CI 0-1.6) to 4.8/10,000 (95% CI 1.4-8.2) between 1993 and 2002. HIV infection was diagnosed during pregnancy in 40% (18/45) of the mothers. Antiretroviral medication was used prior to pregnancy in 17 (33%) cases; in 34 (66%) cases of the pregnancies, medication was started during the pregnancy. A good virological response (i.e. HIV RNA load <1000/ml during the last trimester) to antiretroviral medication was achieved in 90% (36/40) of the patients; in 60% (24/40) of cases, the HIV RNA load was below the assay detection limit. One mother with advanced HIV infection succumbed to pneumonia shortly after delivery. Of the infants, 92% were born at term, and their mean (+/-SD) birth weight was 3350 +/- 395 g. The Caesarean section (CS) rate was 25% (13/52). All newborns were in good condition, and their mean umbilical pH was 7.23. All newborns received antiretroviral medication. One infant was infected with HIV, the mother was found HIV-positive postpartum. None of the infants born to mothers known to be HIV positive were infected. CONCLUSIONS A combination of universal antenatal screening and multidisciplinary management allows individualized treatment and prevents vertical transmission of HIV infection.
Collapse
Affiliation(s)
- Päivi Lehtovirta
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Lambert JS, Moye J, Plaeger SF, Stiehm ER, Bethel J, Mofenson LM, Mathieson B, Kagan J, Rosenblatt H, Paxton H, Suter H, Landay A. Association of selected phenotypic markers of lymphocyte activation and differentiation with perinatal human immunodeficiency virus transmission and infant infection. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 12:622-31. [PMID: 15879023 PMCID: PMC1112077 DOI: 10.1128/cdli.12.5.622-631.2005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This study of a subset of women and infants participating in National Institutes of Health Pediatric AIDS Clinical Trials Group protocol 185 evaluated lymphocyte phenotypic markers of immune activation and differentiation to determine their association with the likelihood of human immunodeficiency virus (HIV) transmission from the women to their infants and the potential for early identification and/or prognosis of infection in the infants. Lymphocytes from 215 human immunodeficiency virus type 1 (HIV)-infected women and 192 of their infants were analyzed by flow cytometry with an extended three-color panel of monoclonal antibodies. Women who did not transmit to their infants tended to have higher CD4+ T cells. Most notably, levels of total CD8+ T cells and CD8+ CD38+ cells made significant independent contributions to predicting the risk of mother-to-child transmission. Adjusting for HIV-1 RNA level at entry, a one percentage-point increase in these marker combinations was associated with a nine percent increase in the likelihood of maternal transmission. Total as well as naive CD4+ T cells were significantly higher in uninfected than infected infants. Total CD8+ cells, as well as CD8+ cells positive for HLA-DR+, CD45 RA+ HLA-DR+, and CD28+ HLA-DR+ were elevated in infected infants. Detailed immunophenotyping may be helpful in predicting which pregnant HIV-infected women are at increased risk of transmitting HIV to their infants. Increasing differences in lymphocyte subsets between infected and uninfected infants became apparent as early as six weeks of age. Detailed immunophenotyping may be useful in supporting the diagnosis of HIV infection in infants with perinatal HIV exposure.
Collapse
Affiliation(s)
- John S Lambert
- Institute of Human Virology, University of Maryland, Baltimore, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
El Beitune P, Duarte G, Machado AA, Quintana SM, Figueiró-Filho EA, Abduch R. Effect of antiretroviral drugs on maternal CD4 lymphocyte counts, HIV-1 RNA levels, and anthropometric parameters of their neonates. Clinics (Sao Paulo) 2005; 60:207-12. [PMID: 15962081 DOI: 10.1590/s1807-59322005000300005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To study the effect of antiretroviral drugs administered during pregnancy on CD4 lymphocyte counts and HIV-1 RNA levels of pregnant women and on the anthropometric parameters of their neonates. METHODS A prospective study was conducted on 57 pregnant women and their neonates divided into 3 groups: ZDV Group, HIV-infected mothers taking zidovudine (n=20); triple therapy (TT) Group, mothers taking zidovudine+lamivudine+nelfinavir (n=25), and Control Group, normal women (n=12). CD4 lymphocyte counts and HIV-1 RNA levels of pregnant women were analyzed during two periods of pregnancy. The perinatal prognosis took into account preterm rates, birth weight, intrauterine growth restriction, perinatal death, and vertical transmission of HIV-1. Data were analyzed statistically using the nonparametric chi-square, Mann-Whitney, Friedman, Kruskal-Wallis, and Wilcoxon matched pairs tests, with the level of significance set at P<.05. RESULTS The major maternal demographic and anthropometric data were homogeneous for the various groups. HIV-1 viral burden, which was initially elevated, median of 14,370 copies/mL, was significantly reduced in the TT group, reaching 40 copies/mL. With respect to T-CD4+ lymphocyte counts, there was a significant recovery in Group TT at the end of pregnancy, this value being significantly different from that for the ZDV group (P=0052). There was no difference between groups regarding gestation length, Apgar scores, or neonatal anthropometric classification. There was no case of vertical HIV-1 transmission. CONCLUSIONS The results obtained for the present series demonstrate the efficiency and suggest safety of the use of antiretroviral drugs during pregnancy as revealed by anthropometric parameters of the neonate.
Collapse
Affiliation(s)
- Patrícia El Beitune
- Department of Obstetrics and Gynecology, Medicine School of Ribeirão Preto, University of São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
36
|
Jaspan HB, Robinson JE, Amedee AM, Van Dyke RB, Garry RF. Amniotic fluid has higher relative levels of lentivirus-specific antibodies than plasma and can contain neutralizing antibodies. J Clin Virol 2004; 31:190-7. [PMID: 15465411 DOI: 10.1016/j.jcv.2004.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/04/2004] [Accepted: 03/08/2004] [Indexed: 11/23/2022]
Abstract
BACKGROUND The in utero transmission rate of HIV-1 is estimated to be 10-15% in the absence of interventions and breastfeeding. Natural protective mechanisms involving lentivirus-specific antibodies may therefore exist to limit in utero transmission of lentiviruses. OBJECTIVES HIV-1- and SIV-specific immunoglobulin G (IgG) levels in amniotic fluid samples from humans and rhesus macaques were assessed. STUDY DESIGN HIV-1- and SIV-specific immunoglobulin G levels, relative to total IgG concentrations in amniotic fluid samples from humans and rhesus macaques, were determined using a quantitative Western blotting procedure. Amniotic fluid from rhesus macaques was tested for the ability to neutralize SIV infection of CEMX174 cells. RESULTS The levels of HIV-1- and SIV-specific immunoglobulin G, relative to total IgG concentrations in amniotic fluid samples from humans and rhesus macaques, were approximately 3-10-fold higher than in plasma. The ability of antibodies in human amniotic fluid samples to neutralize viral infectivity could not be assessed, because zidovidine was present in the samples. Most amniotic fluid samples from rhesus macques not treated with antiretrovirals were able to neutralize SIV infectivity, except for a sample from a SIV positive rhesus whose infant was infected in utero. CONCLUSIONS Active immunity to HIV-1 resulting in virus-specific antibodies in amniotic fluid exists, and may be a natural barrier to in utero infection. This may provide hope for stimulating neutralizing antibody via vaccine design.
Collapse
Affiliation(s)
- Heather B Jaspan
- Interdisciplinary Program in Molecular and Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
37
|
Haigwood NL, Montefiori DC, Sutton WF, McClure J, Watson AJ, Voss G, Hirsch VM, Richardson BA, Letvin NL, Hu SL, Johnson PR. Passive immunotherapy in simian immunodeficiency virus-infected macaques accelerates the development of neutralizing antibodies. J Virol 2004; 78:5983-95. [PMID: 15140996 PMCID: PMC415787 DOI: 10.1128/jvi.78.11.5983-5995.2004] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Passively transferred neutralizing antibodies can block lentivirus infection, but their role in postexposure prophylaxis is poorly understood. In this nonhuman-primate study, the effects of short-term antibody therapy on 5-year disease progression, virus load, and host immunity were explored. We reported previously that postinfection passive treatment with polyclonal immune globulin with high neutralizing titers against SIVsmE660 (SIVIG) significantly improved the 67-week health of SIVsmE660-infected Macaca mulatta macaques. Four of six treated macaques maintained low or undetectable levels of virus in plasma, compared with one of ten controls, while two rapid progressors controlled viremia only as long as the SIVIG was present. SIVIG treatment delayed the de novo production of envelope (Env)-specific antibodies by 8 weeks (13). We show here that differences in disease progression were also significant at 5 years postinfection, excluding rapid progressors (P = 0.05). Macaques that maintained </=10(3) virus particles per ml of plasma and </=30 infectious virus particles per 10(6) mononuclear cells from peripheral blood and lymph nodes had delayed disease onset. All macaques that survived beyond 18 months had measurable Gag-specific CD8(+) cytotoxic T cells, regardless of treatment. Humoral immunity in survivors beyond 20 weeks was strikingly different in the SIVIG and control groups. Despite a delay in Env-specific binding antibodies, de novo production of neutralizing antibodies was significantly accelerated in SIVIG-treated macaques. Titers of de novo neutralizing antibodies at week 12 were comparable to levels achieved in controls only by week 32 or later. Acceleration of de novo simian immunodeficiency virus immunity in the presence of passively transferred neutralizing antibodies is a novel finding with implications for postexposure prophylaxis and vaccines.
Collapse
Affiliation(s)
- Nancy L Haigwood
- Seattle Biomedical Research Institute, 307 Westlake Ave. N., Suite 500, Seattle, WA 98109-5219, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Safrit JT, Ruprecht R, Ferrantelli F, Xu W, Kitabwalla M, Van Rompay K, Marthas M, Haigwood N, Mascola JR, Luzuriaga K, Jones SA, Mathieson BJ, Newell ML. Immunoprophylaxis to Prevent Mother-to-Child Transmission of HIV-1. J Acquir Immune Defic Syndr 2004; 35:169-77. [PMID: 14722451 DOI: 10.1097/00126334-200402010-00012] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Antiretroviral therapy can profoundly reduce the risk of mother-to-child transmission (MTCT) of HIV, but the drugs have a relatively short half-life and should thus be administered throughout breast-feeding to optimally prevent postnatal infection of the infant. The potential toxicities and the development of resistance may limit the long-term efficacy of antiretroviral prophylaxis, and a safe and effective active/passive immunoprophylaxis regimen, begun at birth, and potentially overlapping with interpartum or neonatal chemoprophylaxis, would pose an attractive alternative. This review draws on data presented at the Ghent Workshop on prevention of breast milk transmission and on selected issues from a workshop specifically relating to immunoprophylaxis held in Seattle in October 2002. This purpose of this review is to address the scientific rationale for the development of passive (antibody) and active (vaccine) immunization strategies for prevention of MTCT. Data regarding currently or imminently available passive and active immunoprophylaxis products are reviewed for their potential use in neonatal trials within the coming 1-2 years.
Collapse
Affiliation(s)
- Jeffrey T Safrit
- Elizabeth Glaser Pediatric AIDS Foundation, David Geffen School of Medicine, University of California, Los Angeles, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Farquhar C, John-Stewart G. The role of infant immune responses and genetic factors in preventing HIV-1 acquisition and disease progression. Clin Exp Immunol 2004; 134:367-77. [PMID: 14632739 PMCID: PMC1808883 DOI: 10.1111/j.1365-2249.2003.02292.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- C Farquhar
- Department of Medicine, University of Washington, Seattle 98104-2499, USA.
| | | |
Collapse
|
40
|
Lambert JS, Nogueira SA, Abreu T, Machado ES, Costa TP, Bondarovsky M, Andrade M, Halpern M, Barbosa R, Perez M. A pilot study to evaluate the safety and feasibility of the administration of AZT/3TC fixed dose combination to HIV infected pregnant women and their infants in Rio de Janeiro, Brazil. Sex Transm Infect 2004; 79:448-52. [PMID: 14663118 PMCID: PMC1744786 DOI: 10.1136/sti.79.6.448] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVES To evaluate the safety and feasibility of zidovudine and lamivudine (AZT/3TC) given to HIV infected pregnant women and their infants in Rio de Janeiro, Brazil. METHODS This open label phase II study enrolled 40 HIV infected antiretroviral naive women >or=20 weeks gestation, CD4 <500 cells x10(6)/l, from two public hospitals. TREATMENT fixed dose AZT 300 mg/3TC 150 mg by mouth every 12 hours until labour; AZT 300 mg by mouth every 3 hours until delivery; infants: AZT 4 mg/kg every 12 hours plus 3TC 2 mg/kg every 12 hours for 6 weeks. Blood haematology and chemistry were monitored; adherence evaluated by pills count; efficacy measured by changes in lymphocyte (CD4) and viral load, and by HIV RNA-PCR tests performed at birth, 6 and 12 weeks, to diagnose infant infection. No women breast fed. RESULTS PATIENT CHARACTERISTICS mean age 24.48 (SD 3.5) years; gestational age 24.5 (4.5) weeks; AZT/3TC duration 14.4 (4.4) weeks; vaginal delivery: 11/39; caesarean section: 28/39. Entry and pre-labour CD4: 310/486 cells x10(6)/l (p<0.001); entry and pre-labour viral load: 53 818/2616 copies/ml (p<0.001). Thirty nine women tolerated treatment with >80% adherence; one was lost to follow up. Five newborns were excluded from 3TC receipt. All 39 babies were uninfected. Haematological toxicity in newborns was common: anaemia in 27; neutropenia in five (two severe); platelets counts <100000 in two. All values recovered on study completion. CONCLUSIONS Fixed dose AZT/3TC is well accepted, gives improvements in CD4 and viral load; no infants were HIV infected. Haematological toxicity in infants needs careful monitoring.
Collapse
Affiliation(s)
- J S Lambert
- University of Maryland Institute of Human Virology, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lambert JS, Harris DR, Stiehm ER, Moye J, Fowler MG, Meyer WA, Bethel J, Mofenson LM. Performance characteristics of HIV-1 culture and HIV-1 DNA and RNA amplification assays for early diagnosis of perinatal HIV-1 infection. J Acquir Immune Defic Syndr 2004; 34:512-9. [PMID: 14657763 DOI: 10.1097/00126334-200312150-00011] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A plasma HIV-1 RNA amplification assay (RNA assay), a quantitative peripheral blood mononuclear cell (PBMC) microculture (culture), and a PBMC HIV-1 DNA amplification assay (DNA assay) were compared for diagnosis of HIV-1 infection in infants receiving zidovudine in Pediatric AIDS Clinical Trials Group protocol 185; assays were performed for all 24 infected and 100 uninfected infants. HIV-1 infection was defined as >or=2 positive cultures or positive antibody to HIV-1 at >or=18 months. Cultures were performed at birth and 6 and 24 weeks of age; DNA and RNA assays were performed on cryopreserved specimens. The sensitivity of culture and DNA and RNA assays at birth was 20.8%, 10.5%, and 26.7%, respectively. At older ages, sensitivity typically exceeded 80%, remaining highest for the RNA assay (>85%). Assay specificity was >99%. Positive predictive values exceeded 93% for each assay at each age; negative predictive values were highest (>90%) for the RNA assay. At birth (P < 0.005) and age 6 weeks (P < 0.001), a significantly larger proportion of infected infants were identified by means of the RNA assay than by the other assays. The diagnostic performance of the RNA assay matched or exceeded that of culture and the DNA assay. Given that RNA assays require less blood volume and yield rapid results, our study adds to existing data suggesting that RNA assays may be used for early diagnosis of HIV-1 infection in infants.
Collapse
Affiliation(s)
- John S Lambert
- Institute of Human Virology, University of Maryland, Baltimore, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Rongkavilit C, Asmar BI. Advances in prevention of mother-to-child HIV transmission. Indian J Pediatr 2004; 71:69-79. [PMID: 14979391 DOI: 10.1007/bf02725662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Advances have been made in the understanding of the pathogenesis of mother-to-child transmission of human immunodeficiency virus (HIV). Most transmission occurs during delivery and after birth throught breastfeeding. For this reason, efforts to interrupt transmission have focused on peripartum period and infant feeding. This includes the use of antiretroviral therapy, elective cesarean section and avoidance of breastfeeding. This review summarizes recent major studies and new development on the prevention of mother-to-child HIV transmission. The application and the impact of such interventions in developing world is discussed. Prevention of mother-to-child transmission of HIV should now be integrated as part of basic maternal and child health services in developing countries.
Collapse
Affiliation(s)
- Chokechai Rongkavilit
- Division of Pediatric Infectious Diseases, Children's Hospital of Michigan, Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| | | |
Collapse
|
43
|
Read JS. Human milk, breastfeeding, and transmission of human immunodeficiency virus type 1 in the United States. American Academy of Pediatrics Committee on Pediatric AIDS. Pediatrics 2003; 112:1196-205. [PMID: 14595069 DOI: 10.1542/peds.112.5.1196] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transmission of human immunodeficiency virus type 1 (HIV-1) through breastfeeding has been conclusively demonstrated. The risk of such transmission has been quantified, the timing has been clarified, and certain risk factors for breastfeeding transmission have been identified. In areas where infant formula is accessible, affordable, safe, and sustainable, avoidance of breastfeeding has represented one of the main components of mother-to-child HIV-1 transmission prevention efforts for many years. In areas where affordable and safe alternatives to breastfeeding may not be available, interventions to prevent breastfeeding transmission are being investigated. Complete avoidance of breastfeeding by HIV-1-infected women has been recommended by the American Academy of Pediatrics and the Centers for Disease Control and Prevention and remains the only means by which prevention of breastfeeding transmission of HIV-1 can be absolutely ensured. This technical report summarizes the information available regarding breastfeeding transmission of HIV-1.
Collapse
|
44
|
Kourtis AP, Duerr A. Prevention of perinatal HIV transmission: a review of novel strategies. Expert Opin Investig Drugs 2003; 12:1535-44. [PMID: 12943497 DOI: 10.1517/13543784.12.9.1535] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Significant progress has been made in preventing transmission of HIV-1 from mother to infant. With combination antiretroviral therapies, transmission rates lower than 2% have been achieved in clinical studies. Abbreviated regimens covering labour and the first few days of neonatal life have shown considerable promise in the developing world. Several questions and challenges remain, however. These include choice of the optimal antiretroviral agent(s) and duration of the regimens, availability of antiretroviral agents in developing countries, long-term safety of antiretrovirals during pregnancy and early neonatal life and the problem of breastfeeding transmission in countries where alternatives to breastfeeding are not available. A wider array of strategies for prevention of mother-to-child transmission of HIV-1 during breastfeeding, including passive and active immunisation, may offer much needed answers to the problem of continued HIV transmission from mother to infant.
Collapse
|
45
|
Watts DH, Lambert J, Stiehm ER, Harris DR, Bethel J, Mofenson L, Meyer WA, Mathieson B, Fowler MG, Nemo G. Progression of HIV disease among women following delivery. J Acquir Immune Defic Syndr 2003; 33:585-93. [PMID: 12902802 DOI: 10.1097/00126334-200308150-00006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To assess patterns of HIV disease progression among HIV-1-infected women following delivery. METHODS Four hundred ninety-seven women enrolled in PACTG 185, a phase 3 trial of passive immunoprophylaxis in addition to zidovudine (ZDV) for the prevention of perinatal transmission, were included. Visits occurred twice during pregnancy; at delivery; and at 12, 26, 48, and 78 weeks postpartum. Repeated-measures linear regression and proportional hazards models were applied. RESULTS Trial treatment (HIV hyperimmune globulin vs. immune globulin) was not related to postpartum disease progression. Longitudinal analysis of HIV-1 RNA demonstrated stable levels during pregnancy, significantly increased HIV-1 RNA by 12 weeks postpartum even on stable therapy, and a gradual increase thereafter. Changes in CD4+ lymphocyte percentage over 18 months of follow-up were similar for women continuing or stopping ZDV postpartum. Compared with those receiving no therapy, the hazard ratio for AIDS or death among women who received monotherapy postpartum was 0.52 (95% confidence interval [CI]: 0.25-1.04), 0.17 (CI: 0.06-0.49) for women who received combination therapy, and 0.24 (CI: 0.06-1.01) for women who received highly active antiretroviral therapy. CONCLUSIONS RNA levels increased significantly from delivery to 12 weeks postpartum. Changes in HIV-1 RNA and CD4+ lymphocyte percentage were similar among women continuing or stopping therapy after delivery, and response to antiretroviral therapy was as expected postpartum.
Collapse
Affiliation(s)
- D Heather Watts
- Pediatric, Adolescent, and Maternal AIDS Branch, National Institute of Child Health and Human Development, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bardeguez AD, Shapiro DE, Mofenson LM, Coombs R, Frenkel LM, Fowler MG, Huang S, Sperling RS, Cunningham B, Gandia J, Maupin R, Zorrilla CD, Jones T, O'Sullivan MJ. Effect of cessation of zidovudine prophylaxis to reduce vertical transmission on maternal HIV disease progression and survival. J Acquir Immune Defic Syndr 2003; 32:170-81. [PMID: 12571527 DOI: 10.1097/00126334-200302010-00009] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Zidovudine prophylaxis is recommended to reduce perinatal HIV-1 transmission, but there are limited data on long-term effects on women's health. Pediatrics AIDS Clinical Trials Group (PACTG) 288 was a prospective observational study among US women randomized to zidovudine or placebo in PACTG 076 that was designed to evaluate and compare postpartum clinical, immune, and viral parameters between randomized treatment arms. Forty-eight percent (226/474) of eligible women enrolled in the study (mean follow-up of 4.1 years). Progression and time to AIDS or death were similar in both groups, observed in 21 (19%) zidovudine group women and 29 (25%) placebo group women (RR = 0.73, 90% CI: 0.46-1.17). No significant differences in CD4 lymphocyte count or HIV RNA levels were detected. Genotypic zidovudine resistance was detected in 10% of 156 women (9% of zidovudine group women and 11% of placebo group women). Based on our data, ZDV monotherapy could be considered as chemoprophylaxis to reduce perinatal HIV transmission for minimally symptomatic HIV-infected pregnant women with a low viral load and normal CD4 cell count who do not want to receive highly active antiretroviral therapy because of concern about potential side effects or who wish to reduce fetal exposure to multiple drugs during pregnancy.
Collapse
Affiliation(s)
- Arlene D Bardeguez
- University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The presence of human immunodeficiency virus (HIV) in pregnant women puts infants at risk for exposure through placental infection and contact with contaminated maternal blood and genital secretions. Efforts to combat this inevitably fatal disease continue to focus on preventing transmission of the virus from a mother who has HIV to her newborn during the prenatal, intrapartum, and postnatal periods. Prophylaxis against transmission and vigilant assessment for indicators of infection are hallmarks of appropriate health care for infants exposed to HIV.
Collapse
Affiliation(s)
- Marisha E Meleski
- Frances Payne Bolton School of Nursing, Case Western Reserve University, Cleveland, OH, USA.
| | | |
Collapse
|
48
|
Cameron T. Mandatory HIV testing of newborns in New York State: what are the implications? JOURNAL OF HEALTH & SOCIAL POLICY 2002; 14:59-78. [PMID: 12086013 DOI: 10.1300/j045v14n03_04] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This paper presents an analysis of a public health law (Maternal-Pediatric HIV Prevention and Care Program) enacted by New York State in 1997 and how it strives to reduce the vertical transmission of AIDS. This policy is twofold. First, it strongly encourages voluntary HIV testing of all pregnant women. Secondly, it mandates HIV testing for all newborns. Those found to be positive are referred for medical care. This paper analyzes the State's implementation of this law to determine if it will: exert control over poor, mostly minority women; provide health care for infected woman and infants; protect public health; prevent the spread of HIV/AIDS; or sacrifice a woman's privacy. The potential benefits and shortfalls of this policy are discussed as other states may consider adopting similar legislation.
Collapse
|
49
|
Guay LA, Musoke P, Hom DL, Nakabiito C, Bagenda D, Fletcher CV, Marum LH, Fowler MG, Falksveden LG, Wahren B, Kataaha P, Wigzell H, Mmiro FA, Jackson JB. Phase I/II trial of HIV-1 hyperimmune globulin for the prevention of HIV-1 vertical transmission in Uganda. AIDS 2002; 16:1391-400. [PMID: 12131216 DOI: 10.1097/00002030-200207050-00011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To assess the safety, tolerance, pharmacokinetics, and virologic and immunologic changes associated with the use of Ugandan HIV hyperimmune globulin (HIVIGLOB) in HIV infected pregnant Ugandan women and their infants. DESIGN A prospective, phase I/II, three-arm dose escalation trial of HIVIGLOB. METHODS HIVIGLOB was prepared from discarded HIV infected units of blood collected from the National Blood Bank in Kampala. From June 1996 to April 1997, 31 HIV positive pregnant women were enrolled with HIVIGLOB infusions given at 37 weeks gestation and within 16 h of birth for infants. The first 10 mother-infant pairs were infused at a dose of 50 mg/kg, followed by 11 pairs at 200 mg/kg, and 10 pairs at 400 mg/kg. Study participants were followed for 30 months. RESULTS Thirty-one women and 29 infants were infused with HIVIGLOB. The infusions were safe and well tolerated by the women and their infants at all doses. There were no significant changes in virologic or immunologic parameters after HIVIGLOB infusion. Pharmacokinetic properties of this product were similar to other immune globulin products with a median half-life of 28 days in women and 30 days in infants. CONCLUSION An HIV immune globulin product derived from HIV infected Ugandan donors is safe, well tolerated, and has pharmacokinetic properties consistent with other immunoglobulin products. Data suggest that a 400 mg/kg dose of HIVIGLOB would be the most appropriate dose for a subsequent efficacy trial of HIVIGLOB for the prevention of mother to child HIV transmission.
Collapse
Affiliation(s)
- Laura A Guay
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tuomala RE, Shapiro DE, Mofenson LM, Bryson Y, Culnane M, Hughes MD, O'Sullivan MJ, Scott G, Stek AM, Wara D, Bulterys M. Antiretroviral therapy during pregnancy and the risk of an adverse outcome. N Engl J Med 2002; 346:1863-70. [PMID: 12063370 DOI: 10.1056/nejmoa991159] [Citation(s) in RCA: 249] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Some studies suggest that combination antiretroviral therapy in pregnant women with human immunodeficiency virus type 1 (HIV-1) infection increases the risk of premature birth and other adverse outcomes of pregnancy. METHODS We studied pregnant women with HIV-1 infection who were enrolled in seven clinical studies and delivered their infants from 1990 through 1998. The cohort comprised 2123 women who received antiretroviral therapy during pregnancy (monotherapy in 1590, combination therapy without protease inhibitors in 396, and combination therapy with protease inhibitors in 137) and 1143 women who did not receive antiretroviral therapy. RESULTS After standardization for the CD4+ cell count and use or nonuse of tobacco, alcohol, and illicit drugs, the rate of premature delivery (<37 weeks of gestation) was similar among the women who received antiretroviral therapy and those who did not (16 percent and 17 percent, respectively); the rate of low birth weight (<2500 g) was 16 percent among the infants born to both groups; and the rate of very low birth weight (<1500 g) was 2 percent for the group that received antiretroviral therapy and 1 percent for the group that did not. The rates of low Apgar scores (<7) and stillbirth were also similar or the same in the two groups. After adjustment for multiple risk factors, combination antiretroviral therapy was not associated with an increased risk of premature delivery as compared with monotherapy (odds ratio, 1.08; 95 percent confidence interval, 0.71 to 1.62) or delivery of an infant with low birth weight (odds ratio, 1.03; 95 percent confidence interval, 0.64 to 1.63). Seven of the women who received combination therapy with protease inhibitors (5 percent) had infants with very low birth weight, as compared with nine women who received combination therapy without protease inhibitors (2 percent) (adjusted odds ratio, 3.56; 95 percent confidence interval, 1.04 to 12.19). CONCLUSIONS As compared with no antiretroviral therapy or monotherapy, combination therapy for HIV-1 infection in pregnant women is not associated with increased rates of premature delivery or with low birth weight, low Apgar scores, or stillbirth in their infants. The association between combination therapy with protease inhibitors and an increased risk of very low birth weight requires confirmation.
Collapse
Affiliation(s)
- Ruth E Tuomala
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston 02115, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|