1
|
Hampanda K, Grubbs H, Castillo-Mancilla J, Anderson PL, Thorne J, Helova A, Turan JM, Onono M, Abuogi LL. Antiretroviral therapy adherence among peripartum women with HIV in Kenya: an explanatory mixed methods study using dry blood spot measures and narrative interviews. AIDS Care 2024:1-12. [PMID: 39106970 DOI: 10.1080/09540121.2024.2383885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 07/18/2024] [Indexed: 08/09/2024]
Abstract
ABSTRACTAdherence to antiretroviral therapy (ART) remains sub-optimal among pregnant and postpartum women with HIV (PPWH) in high HIV prevalence low resource settings with few effective behavioral interventions. A large body of qualitative literature has established general barriers and facilitators to ART adherence in PPWH at various levels (individual, interpersonal, structural). However, research exploring the underlying behavioral mechanisms of ART adherence in PPWH with objectively verified adherence biomarkers is extremely limited. We conducted 24 in-depth interviews with postpartum women in western Kenya who had linked ART drug concentrations obtained from three dried blood spot samples across the peripartum period. Among PPWH with a low drug concentration (n = 13) compared to those with continuously high drug concentrations (n = 11), distinct themes emerged related to HIV status disclosure, social support, interactions with the health system, and health beliefs. By combining ART biomarkers with patient reported challenges, there is the potential for real-time interventions to support sustained ART adherence among PPWH and improve maternal and infant health outcomes.
Collapse
Affiliation(s)
- K Hampanda
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, USA
- Center for Global Health, University of Colorado, Aurora, CO, USA
| | - H Grubbs
- Department of Surgery, University of Oklahoma, Oklahoma City, OK, USA
| | - J Castillo-Mancilla
- Division of Infectious Diseases, Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - P L Anderson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - J Thorne
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, USA
- Center for Global Health, University of Colorado, Aurora, CO, USA
| | - A Helova
- Department of Health Policy and Organization, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
- Sparkman Center for Global Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J M Turan
- Department of Health Policy and Organization, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
- Sparkman Center for Global Health, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Public Health, School of Medicine, Koc University, Istanbul, Turkey
| | - M Onono
- Centre for Microbiology Research, Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - L L Abuogi
- Center for Global Health, University of Colorado, Aurora, CO, USA
- Department of Pediatrics, University of Colorado, Aurora, CO, USA
| |
Collapse
|
2
|
Xu F, Xiong Y, Gu M, Wan L, Wang Y. Interventions to prevent mother-to-child transmission in breastfeeding mothers with HIV: a systematic review and meta-analysis of randomized controlled trials. Rev Inst Med Trop Sao Paulo 2024; 66:e45. [PMID: 39082484 PMCID: PMC11295290 DOI: 10.1590/s1678-9946202466045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 08/04/2024] Open
Abstract
This study aimed to systematically review interventions to prevent mother-to-child transmission of HIV during breastfeeding. We conducted a systematic review and meta-analysis using specific criteria to identify randomized controlled trials that focused on pregnant and breastfeeding women living with HIV and their children from birth to 2 years of age. We extensively searched electronic databases, including Web of Science, Scopus, PubMed, MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, and Google Scholar up to October 24, 2023. After screening 3,110 titles and abstracts, we reviewed 306 full texts. Of these, we assessed the quality and risk of bias of fifty-five articles, ultimately identifying seven studies. Four of these studies, which focused on antiretroviral therapy (ART), were included in the meta-analysis. There was little heterogeneity in study methodology and pooled estimates. The postnatal HIV transmission rate was found to be 0.01 (95%CI: 0.00 - 0.02). Therefore, the risk of mother-to-child transmission among breastfeeding mothers with HIV was significantly lower in the intervention groups than in the placebo groups. Analysis of funnel plots and Egger's test (p = 0.589) showed no evidence of publication bias. In addition to the four articles, two studies compared different ART regimens and one study compared the administration of high-dose vitamin A to the mother or the child. The results suggest that the use of ART significantly reduces the risk of postnatal HIV transmission compared with placebo. However, the effectiveness of different ART regimens or other therapies, including high-dose vitamin A, is unclear.
Collapse
Affiliation(s)
- Fangping Xu
- Jiangxi Maternal and Child Health Hospital, Obstetrical Department, Jiangxi, Nanchang, China
| | - Ying Xiong
- Jiangxi Maternal and Child Health Hospital, Obstetrical Department, Jiangxi, Nanchang, China
| | - Min Gu
- Jiangxi Maternal and Child Health Hospital, Obstetrical Department, Jiangxi, Nanchang, China
| | - Lingling Wan
- Jiangxi Maternal and Child Health Hospital, Obstetrical Department, Jiangxi, Nanchang, China
| | - Yun Wang
- Jiangxi Maternal and Child Health Hospital, Obstetrical Department, Jiangxi, Nanchang, China
| |
Collapse
|
3
|
Bierke S, Häner M, Karpinski K, Hees T, Petersen W. No increased rate of cyclops lesions and extension deficits after remnant-preserving ACL reconstruction using the sparing technique. J Orthop Surg Res 2022; 17:463. [PMID: 36271418 PMCID: PMC9587551 DOI: 10.1186/s13018-022-03356-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Remnant-preserving anterior cruciate ligament reconstruction (ACLR) should have advantages for postoperative remodeling and proprioception. However, it has been suggested that the larger diameter of the graft tends to lead to impingement phenomena with a higher rate of cyclops lesions. The aim of this work was to find out whether the remnant-preserving ACLR actually leads to an increased rate of range of motion restraints compared to the remnant-sacrificing technique. METHODS Patients, who fulfilled the inclusion criteria, were followed up for one year after surgery. The primary endpoint was arthrolysis due to extension deficit or cyclops syndrome. Secondary outcome measures were pain (NRS), knee function (KOOS), patient satisfaction and return to sports rate. RESULTS One hundred and sixty-four patients were included in the study, 60 of whom received the "remnant augmentation" procedure (group 1). In the remnant augmentation group, one cyclops resection was performed, whereas in the non-remnant augmentation group three cyclops lesion resections had to be performed (odds ratio 0.6). There was no difference between the groups in pain (NRS) and knee function (KOOS) and patient satisfaction. The return to sports rate after one year was higher in the remnant augmentation group. CONCLUSIONS Patients who have undergone the sparing "remnant augmentation" ACLR have no increased risk of cyclops lesion formation or extension deficit in the first year after surgery. An improvement of the proprioceptive abilities by remnant augmentation ACLR should be investigated in further studies. LEVEL OF EVIDENCE III (prospective cohort study).
Collapse
Affiliation(s)
- Sebastian Bierke
- Department of Orthopedic and Trauma Surgery, Martin Luther Hospital, Caspar Theys Strasse 27-31, 14 193, Berlin, Germany
| | - Martin Häner
- Department of Orthopedic and Trauma Surgery, Martin Luther Hospital, Caspar Theys Strasse 27-31, 14 193, Berlin, Germany
| | - Katrin Karpinski
- Department of Orthopedic and Trauma Surgery, Martin Luther Hospital, Caspar Theys Strasse 27-31, 14 193, Berlin, Germany
| | - Tilman Hees
- Department of Orthopedic and Trauma Surgery, Martin Luther Hospital, Caspar Theys Strasse 27-31, 14 193, Berlin, Germany
| | - Wolf Petersen
- Department of Orthopedic and Trauma Surgery, Martin Luther Hospital, Caspar Theys Strasse 27-31, 14 193, Berlin, Germany.
| |
Collapse
|
4
|
Li X, Lin J, Zhang L, Liu Y. Effects of gonadotropin-releasing hormone agonist pretreatment on frozen embryo transfer outcomes in artificial cycles: a meta-analysis. Arch Gynecol Obstet 2022:10.1007/s00404-022-06823-7. [PMID: 36266549 DOI: 10.1007/s00404-022-06823-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Gonadotropin-releasing hormone agonist (GnRHa) before artificial cycle (AC) is expected to improve pregnancy outcomes in frozen-thawed embryo transfer (FET). Many studies have explored the impact of GnRHa pretreatment of AC in FET, but the results were inconsistent. This meta-analysis was performed to systematically evaluate the effect of GnRHa pretreatment on AC in FET. METHODS The last search was January 31, 2022. Randomized controlled trials and cohort studies aiming to assess the effect of GnRHa as the pretreatment of AC for endometrial preparation in FET were included. GnRHa was used before AC in the treatment group. In the control group, no pretreatment was used before AC. The eligible studies included at least one of the following outcomes: implantation, clinical pregnancy, and live birth. We calculated the odds ratio (OR) or mean difference (MD) and 95% confidence interval (CI) for each study and used a random-effects or fixed model to estimate the results. RESULTS 27 articles (10 RCTs and 17 non-RCTs) and 14152 patients were included. AC + GnRHa improved the implantation rate (OR = 1.31, 95% CI 1.03-1.66, I2 = 79%), clinical pregnancy rate (OR = 1.27, 95% CI 1.10-1.45, I2 = 53%), and live birth rate (OR = 1.16, 95% CI 1.05-1.29, I2 = 39%). We also found that AC + GnRHa increased the implantation rate (OR = 1.35, 95% CI 1.07-1.69, I2 = 53%) and clinical pregnancy rate (OR = 1.50, 95% CI 1.12-2.01, I2 = 50%) in repeated implantation failure. In addition, AC + GnRHa was positively associated with preterm birth (OR = 1.5, 95% CI 1.15-1.94, I2 = 0%). CONCLUSIONS GnRHa pretreatment in FET can improve implantation, clinical pregnancy, and live birth rates, especially in patients with repeated implantation failure. GnRHa pretreatment seems to improve FET outcomes, though with a higher preterm birth rate.
Collapse
Affiliation(s)
- Xueying Li
- Department of Obstetrics and Gynecology, Centre for Reproductive Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China.
| | - Jinli Lin
- Reproductive and Infertility Department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Linhao Zhang
- West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yao Liu
- Department of Gynecology, Zunhua People's Hospital, Zunhua, Hebei, China
| |
Collapse
|
5
|
From Undetectable Equals Untransmittable (U=U) to Breastfeeding: Is the Jump Short? Infect Dis Rep 2022; 14:220-227. [PMID: 35447879 PMCID: PMC9030015 DOI: 10.3390/idr14020027] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 01/27/2023] Open
Abstract
Background: Vertical transmission of HIV infection may occur during pregnancy, at childbirth or through breastfeeding. Recommendations on the safety of breastfeeding of HIV-infected women on effective antiretroviral treatment are not univocal among international guidelines (WHO 2010, EACS 2017, DHHS 2017), leaving space for variability at the patient’s level. Methods: We collected clinical, laboratory and outcome data from 13 HIV-infected pregnant women who, between March 2017 and June 2021, elected to breastfeed their children against specific medical advice. All mothers were on antiretroviral therapy with darunavir or raltegravir plus emtricitabine/tenofovir disoproxil and remained HIV-RNA undetectable and >400 cells/mmc CD4+ lymphocytes during pregnancy and breastfeeding. Prophylactic antiretroviral therapy (zidovudine for 4 weeks) was started immediately after birth in all newborns. The mean duration of breastfeeding was 5.4 months. Newborns were tested for HIV-RNA multiple times: at birth, 1, 3, and 6 months after birth, and 1, 3 and 6 months after the end of breastfeeding. Results: None of the infants were infected by HIV. Conclusions: Our experience, gathered in the setting of freedom of choice on the patient’s side, while insufficient to address the eventual safety of breastfeeding in HIV-infected mothers since the represented cohort is numerically irrelevant, supports the extension of the U=U (Undetectable Equals Untransmittable) paradigm to this setting. Since breastfeeding is often requested by women with HIV planning pregnancy, more extensive comparative studies should be performed.
Collapse
|
6
|
Banerjee R, Erridge S, Salazar O, Mangal N, Couch D, Pacchetti B, Sodergren MH. Real World Evidence in Medical Cannabis Research. Ther Innov Regul Sci 2021; 56:8-14. [PMID: 34748204 PMCID: PMC8688379 DOI: 10.1007/s43441-021-00346-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/12/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Whilst access to cannabis-based medicinal products (CBMPs) has increased globally subject to relaxation of scheduling laws globally, one of the main barriers to appropriate patient access remains a paucity of high-quality evidence surrounding their clinical effects. DISCUSSION Whilst randomised controlled trials (RCTs) remain the gold-standard for clinical evaluation, there are notable barriers to their implementation. Development of CBMPs requires novel approaches of evidence collection to address these challenges. Real world evidence (RWE) presents a solution to not only both provide immediate impact on clinical care, but also inform well-conducted RCTs. RWE is defined as evidence derived from health data sourced from non-interventional studies, registries, electronic health records and insurance data. Currently it is used mostly to monitor post-approval safety requirements allowing for long-term pharmacovigilance. However, RWE has the potential to be used in conjunction or as an extension to RCTs to both broaden and streamline the process of evidence generation. CONCLUSION Novel approaches of data collection and analysis will be integral to improving clinical evidence on CBMPs. RWE can be used in conjunction or as an extension to RCTs to increase the speed of evidence generation, as well as reduce costs. Currently, there is an abundance of potential data however, whilst a number of platforms now exist to capture real world data it is important the right tools and analysis are utilised to unlock potential insights from these.
Collapse
Affiliation(s)
- Rishi Banerjee
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Simon Erridge
- Department of Surgery and Cancer, Imperial College London, London, UK
- Sapphire Medical Clinics, UK Medical Cannabis Registry, London, UK
| | - Oliver Salazar
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Nagina Mangal
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Daniel Couch
- The Centre for Medicinal Cannabis, 18 Hanway Street, London, W1T 1UF, UK
| | | | - Mikael Hans Sodergren
- Department of Surgery and Cancer, Imperial College London, London, UK.
- Sapphire Medical Clinics, UK Medical Cannabis Registry, London, UK.
- Curaleaf International, London, UK.
- Division of Surgery, Department of Surgery & Cancer, Imperial College London, St Mary's Hospital, Academic Surgical Unit, 10th Floor QEQM, South Wharf Road, London, W2 1NY, UK.
| |
Collapse
|
7
|
Human Immunodeficiency Virus (HIV) and Human Cytomegalovirus (HCMV) Coinfection of Infant Tonsil Epithelium May Synergistically Promote both HIV-1 and HCMV Spread and Infection. J Virol 2021; 95:e0092121. [PMID: 34232730 DOI: 10.1128/jvi.00921-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mother-to-child transmission (MTCT) of human immunodeficiency virus type 1 (HIV-1) and human cytomegalovirus (HCMV) may occur during pregnancy, labor, or breastfeeding. These viruses from amniotic fluid, cervicovaginal secretions, and breast milk may simultaneously interact with oropharyngeal and tonsil epithelia; however, the molecular mechanism of HIV-1 and HCMV cotransmission through the oral mucosa and its role in MTCT are poorly understood. To study the molecular mechanism of HIV-1 and HCMV MTCT via oral epithelium, we established polarized infant tonsil epithelial cells and polarized-oriented ex vivo tonsil tissue explants. Using these models, we showed that cell-free HIV-1 and its proteins gp120 and tat induce the disruption of tonsil epithelial tight junctions and increase paracellular permeability, which facilitates HCMV spread within the tonsil mucosa. Inhibition of HIV-1 gp120-induced upregulation of mitogen-activated protein kinase (MAPK) and NF-κB signaling in tonsil epithelial cells, reduces HCMV infection, indicating that HIV-1-activated MAPK and NF-κB signaling may play a critical role in HCMV infection of tonsil epithelium. HCMV infection of tonsil epithelial cells also leads to the disruption of tight junctions and increases paracellular permeability, facilitating HIV-1 paracellular spread into tonsil mucosa. HCMV-promoted paracellular spread of HIV-1 increases its accessibility to tonsil CD4 T lymphocytes, macrophages, and dendritic cells. HIV-1-enhanced HCMV paracellular spread and infection of epithelial cells subsequently leads to the spread of HCMV to tonsil macrophages and dendritic cells. Our findings revealed that HIV-1- and HCMV-induced disruption of infant tonsil epithelial tight junctions promotes MTCT of these viruses through tonsil mucosal epithelium, and therapeutic intervention for both HIV-1 and HCMV infection may substantially reduce their MTCT. IMPORTANCE Most HIV-1 and HCMV MTCT occurs in infancy, and the cotransmission of these viruses may occur via infant oropharyngeal and tonsil epithelia, which are the first biological barriers for viral pathogens. We have shown that HIV-1 and HCMV disrupt epithelial junctions, reducing the barrier functions of epithelia and thus allowing paracellular penetration of both viruses via mucosal epithelia. Subsequently, HCMV infects epithelial cells, macrophages, and dendritic cells, and HIV-1 infects CD4+ lymphocytes, macrophages, and dendritic cells. Infection of these cells in HCMV- and HIV-1-coinfected tonsil tissues is much higher than that by HCMV or HIV-1 infection alone, promoting their MTCT at its initial stages via infant oropharyngeal and tonsil epithelia.
Collapse
|
8
|
Hanson NA, Lavallee MB, Thiele RH. Apophenia and anesthesia: how we sometimes change our practice prematurely. Can J Anaesth 2021; 68:1185-1196. [PMID: 33963519 PMCID: PMC8104920 DOI: 10.1007/s12630-021-02005-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 12/21/2022] Open
Abstract
Human beings are predisposed to identifying false patterns in statistical noise, a likely survival advantage during our evolutionary development. Moreover, humans seem to prefer "positive" results over "negative" ones. These two cognitive features lay a framework for premature adoption of falsely positive studies. Added to this predisposition is the tendency of journals to "overbid" for exciting or newsworthy manuscripts, incentives in both the academic and publishing industries that value change over truth and scientific rigour, and a growing dependence on complex statistical techniques that some reviewers do not understand. The purpose of this article is to describe the underlying causes of premature adoption and provide recommendations that may improve the quality of published science.
Collapse
Affiliation(s)
- Neil A Hanson
- Department of Anesthesiology, University of Virginia Health System, PO Box 800710, ville, VA, 22908-0710, USA.
| | - Matthew B Lavallee
- Department of Anesthesiology, University of Virginia Health System, PO Box 800710, ville, VA, 22908-0710, USA
| | - Robert H Thiele
- Department of Anesthesiology, University of Virginia Health System, PO Box 800710, ville, VA, 22908-0710, USA
| |
Collapse
|
9
|
Eastment MC, Kinuthia J, Wang L, Wanje G, Wilson K, Kaggiah A, Simoni JM, Mandaliya K, Poole DN, Richardson BA, Jaoko W, John-Stewart G, McClelland RS. Late antiretroviral refills and condomless sex in a cohort of HIV-seropositive pregnant and postpartum Kenyan women. PLoS One 2021; 16:e0254767. [PMID: 34280229 PMCID: PMC8289061 DOI: 10.1371/journal.pone.0254767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 07/02/2021] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION The postpartum period can be challenging for women living with HIV. Understanding how the postpartum period impacts ART adherence and condomless sex could inform the development of comprehensive sexual and reproductive health and HIV services tailored to the needs of women living with HIV during this critical interval. METHODS In a longitudinal cohort study of HIV-seropositive Kenyan women, late ART refills and self-reported condomless sex were compared between the woman's pregnancy and the postpartum period. Analyses were conducted using generalized estimating equations and adjusted for alcohol use, depressive symptoms, intimate partner violence (IPV), and having a recent regular partner. Effect modification was explored for selected variables. RESULTS AND DISCUSSION 151 women contributed visits. Late ART refills occurred at 7% (32/439) of pregnancy visits compared to 18% (178/1016) during the postpartum period (adjusted relative risk [aRR] 2.44, 95% confidence interval [CI] 1.62-3.67). This association differed by women's education level. Women with ≥8 years of education had late ART refills more during the postpartum period than pregnancy (aRR 3.00, 95%CI 1.95-4.62). In contrast, in women with <8 years of education, late ART refills occurred similarly during pregnancy and the postpartum period (aRR 0.88, 95%CI 0.18-4.35). Women reported condomless sex at 10% (60/600) of pregnancy visits compared to 7% (72/1081) of postpartum visits (aRR 0.76, 95%CI 0.45-1.27). This association differed by whether women had experienced recent IPV. Women without recent IPV had a significant decline in condomless sex from pregnancy to postpartum (aRR 0.53, 95%CI 0.30-0.95) while women with recent IPV had no significant change in condomless sex from pregnancy to postpartum (aRR 1.76, 95%CI 0.87-3.55). CONCLUSION Improved support for ART adherence during the postpartum period and addressing IPV to limit condomless sex could improve HIV treatment and prevention outcomes for HIV-seropositive women as well as their infants and sexual partners.
Collapse
Affiliation(s)
- McKenna C. Eastment
- Departments of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - John Kinuthia
- Global Health, University of Washington, Seattle, Washington, United States of America
- Kenyatta National Hospital, Nairobi, Kenya
| | - Lei Wang
- Global Health, University of Washington, Seattle, Washington, United States of America
| | - George Wanje
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Katherine Wilson
- Global Health, University of Washington, Seattle, Washington, United States of America
| | | | - Jane M. Simoni
- Global Health, University of Washington, Seattle, Washington, United States of America
- Psychology, University of Washington, Seattle, Washington, United States of America
| | | | - Danielle N. Poole
- Department of Geography, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Barbra A. Richardson
- Global Health, University of Washington, Seattle, Washington, United States of America
- Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Walter Jaoko
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
| | - Grace John-Stewart
- Departments of Medicine, University of Washington, Seattle, Washington, United States of America
- Global Health, University of Washington, Seattle, Washington, United States of America
- Epidemiology, University of Washington, Seattle, Washington, United States of America
- Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - R. Scott McClelland
- Departments of Medicine, University of Washington, Seattle, Washington, United States of America
- Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Medical Microbiology, University of Nairobi, Nairobi, Kenya
- Epidemiology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
10
|
Besnier E, Thomson K, Stonkute D, Mohammad T, Akhter N, Todd A, Rom Jensen M, Kilvik A, Bambra C. Which public health interventions are effective in reducing morbidity, mortality and health inequalities from infectious diseases amongst children in low- and middle-income countries (LMICs): An umbrella review. PLoS One 2021; 16:e0251905. [PMID: 34111134 PMCID: PMC8191901 DOI: 10.1371/journal.pone.0251905] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 05/04/2021] [Indexed: 01/08/2023] Open
Abstract
Despite significant progress in the last few decades, infectious diseases remain a major threat to child health in low- and middle-income countries (LMICs)-particularly amongst more disadvantaged groups. It is imperative to understand the best available evidence concerning which public health interventions reduce morbidity, mortality and health inequalities in children aged under five years. To address this gap, we carried out an umbrella review (a systematic reviews of reviews) to identify evidence on the effects of public health interventions (promotion, protection, prevention) on morbidity, mortality and/or health inequalities due to infectious diseases amongst children in LMICs. Ten databases were searched for records published between 2014-2021 alongside a manual search of gray literature. Articles were quality-assessed using the Assessment of Multiple Systematic Reviews tool (AMSTAR 2). A narrative synthesis was conducted. We identified 60 systematic reviews synthesizing 453 individual primary studies. A majority of the reviews reported on preventive interventions (n = 48), with a minority on promotion (n = 17) and almost no reviews covering health protection interventions (n = 2). Effective interventions for improving child health across the whole population, as well as the most disadvantaged included communication, education and social mobilization for specific preventive services or tools, such as immunization or bed nets. For all other interventions, the effects were either unclear, unknown or detrimental, either at the overall population level or regarding health inequalities. We found few reviews reporting health inequalities information and the quality of the evidence base was generally low. Our umbrella review identified some prevention interventions that might be useful in reducing under five mortality from infectious diseases in LMICs, particularly amongst the most disadvantaged groups.
Collapse
Affiliation(s)
- Elodie Besnier
- Department of Sociology and Political Science, Centre for Global Health Inequalities Research (CHAIN), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Katie Thomson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Donata Stonkute
- CHAIN, Department of Public Health and Nursing, NTNU, Trondheim, Norway
| | - Talal Mohammad
- CHAIN, Department of Public Health and Nursing, NTNU, Trondheim, Norway
| | - Nasima Akhter
- Department of Anthropology, Durham University, Durham, United Kingdom
| | - Adam Todd
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Magnus Rom Jensen
- Library Section for Humanities, Education and Social Sciences, NTNU, Trondheim, Norway
| | - Astrid Kilvik
- Medicine and Health Library, NTNU, Trondheim, Norway
| | - Clare Bambra
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
11
|
Mantziari A, Salminen S, Szajewska H, Malagón-Rojas JN. Postbiotics against Pathogens Commonly Involved in Pediatric Infectious Diseases. Microorganisms 2020; 8:E1510. [PMID: 33008065 PMCID: PMC7601467 DOI: 10.3390/microorganisms8101510] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
The Sustainable Development goals for 2020 included reducing all causes associated with infant and perinatal mortality in their priorities. The use of compounds with bioactive properties has been proposed as a therapeutic strategy due to their stimulating effect on the host's immune system. Additionally, biotherapeutic products such as postbiotics, tentatively defined as compounds produced during a fermentation process that support health and well-being, promote intestinal barrier integrity without posing considerable risks to children's health. Although this is a concept in development, there are increasing studies in the field of nutrition, chemistry, and health that aim to understand how postbiotics can help prevent different types of infections in priority populations such as minors under the age of five. The present review aims to describe the main mechanisms of action of postbiotics. In addition, it presents the available current evidence regarding the effects of postbiotics against pathogens commonly involved in pediatric infections. Postbiotics may constitute a safe alternative capable of modulating the cellular response and stimulating the host's humoral response.
Collapse
Affiliation(s)
- Anastasia Mantziari
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland;
| | - Hania Szajewska
- Department of Paediatrics at the Medical University of Warsaw, 02091 Warsaw, Poland;
| | - Jeadran Nevardo Malagón-Rojas
- Facultad de Medicina, Universidad El Bosque, 110121 Bogotá, Colombia;
- Instituto Nacional de Salud de Colombia, 111321 Bogotá, Colombia
| |
Collapse
|
12
|
Bhattacharjee NV, Schaeffer LE, Marczak LB, Ross JM, Swartz SJ, Albright J, Gardner WM, Shields C, Sligar A, Schipp MF, Pickering BV, Henry NJ, Johnson KB, Louie C, Cork MA, Steuben KM, Lazzar-Atwood A, Lu D, Kinyoki DK, Osgood-Zimmerman A, Earl L, Mosser JF, Deshpande A, Burstein R, Woyczynski LP, Wilson KF, VanderHeide JD, Wiens KE, Reiner RC, Piwoz EG, Rawat R, Sartorius B, Davis Weaver N, Nixon MR, Smith DL, Kassebaum NJ, Gakidou E, Lim SS, Mokdad AH, Murray CJL, Dwyer-Lindgren L, Hay SI. Mapping exclusive breastfeeding in Africa between 2000 and 2017. Nat Med 2019; 25:1205-1212. [PMID: 31332393 PMCID: PMC6749549 DOI: 10.1038/s41591-019-0525-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/17/2019] [Indexed: 11/30/2022]
Abstract
Exclusive breastfeeding (EBF)-giving infants only breast-milk (and medications, oral rehydration salts and vitamins as needed) with no additional food or drink for their first six months of life-is one of the most effective strategies for preventing child mortality1-4. Despite these advantages, only 37% of infants under 6 months of age in Africa were exclusively breastfed in 20175, and the practice of EBF varies by population. Here, we present a fine-scale geospatial analysis of EBF prevalence and trends in 49 African countries from 2000-2017, providing policy-relevant administrative- and national-level estimates. Previous national-level analyses found that most countries will not meet the World Health Organization's Global Nutrition Target of 50% EBF prevalence by 20256. Our analyses show that even fewer will achieve this ambition in all subnational areas. Our estimates provide the ability to visualize subnational EBF variability and identify populations in need of additional breastfeeding support.
Collapse
Affiliation(s)
| | - Lauren E Schaeffer
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Laurie B Marczak
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Jennifer M Ross
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Scott J Swartz
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - James Albright
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - William M Gardner
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Chloe Shields
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Amber Sligar
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Megan F Schipp
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Brandon V Pickering
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Nathaniel J Henry
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Kimberly B Johnson
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Celia Louie
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Michael A Cork
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Krista M Steuben
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Alice Lazzar-Atwood
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Dan Lu
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Damaris K Kinyoki
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | | | - Lucas Earl
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Jonathan F Mosser
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Aniruddha Deshpande
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Roy Burstein
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Lauren P Woyczynski
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Katherine F Wilson
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - John D VanderHeide
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Kirsten E Wiens
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Robert C Reiner
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | | | - Rahul Rawat
- Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Benn Sartorius
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Nicole Davis Weaver
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Molly R Nixon
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - David L Smith
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Nicholas J Kassebaum
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Emmanuela Gakidou
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Stephen S Lim
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Ali H Mokdad
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Christopher J L Murray
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Laura Dwyer-Lindgren
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA
| | - Simon I Hay
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA.
- Department of Health Metrics Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
13
|
A Systematic Review and Meta-analysis of Antiretroviral Therapy (ART) Adherence Interventions for Women Living with HIV. AIDS Behav 2019; 23:1998-2013. [PMID: 30443806 DOI: 10.1007/s10461-018-2341-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A systematic review and meta-analysis was conducted to determine the efficacy of women-focused ART adherence interventions. Included studies (a) reported on a behavioral ART adherence intervention for cis-women living with HIV, (b) measured ART adherence as an outcome, and (c) employed a randomized controlled trial design. Thirteen studies were included in the meta-analysis. Overall, interventions significantly improved ART adherence compared to control conditions (random-effects d = 0.82, 95% CI [0.18, 1.45], p = 0.01), however, this was largely driven by two studies that had effect sizes greater than 3 standard errors above the mean effect size. Key moderators were location, recruitment method, group-based intervention, and alteration of the healthcare system. Innovative behavioral interventions that focus on young women and adolescents, target the critical periods of pregnancy and postpartum and test the integration of multiple levels of intervention to create lasting effects on ART adherence are needed.
Collapse
|
14
|
Mallampati D, MacLean RL, Shapiro R, Dabis F, Engelsmann B, Freedberg KA, Leroy V, Lockman S, Walensky R, Rollins N, Ciaranello A. Optimal breastfeeding durations for HIV-exposed infants: the impact of maternal ART use, infant mortality and replacement feeding risk. J Int AIDS Soc 2019; 21:e25107. [PMID: 29667336 PMCID: PMC5904528 DOI: 10.1002/jia2.25107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 03/12/2018] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION In 2010, the WHO recommended women living with HIV breastfeed for 12 months while taking antiretroviral therapy (ART) to balance breastfeeding benefits against HIV transmission risks. To inform the 2016 WHO guidelines, we updated prior research on the impact of breastfeeding duration on HIV-free infant survival (HFS) by incorporating maternal ART duration, infant/child mortality and mother-to-child transmission data. METHODS Using the Cost-Effectiveness of Preventing AIDS Complications (CEPAC)-Infant model, we simulated the impact of breastfeeding duration on 24-month HFS among HIV-exposed, uninfected infants. We defined "optimal" breastfeeding durations as those maximizing 24-month HFS. We varied maternal ART duration, mortality rates among breastfed infants/children, and relative risk of mortality associated with replacement feeding ("RRRF"), modelled as a multiplier on all-cause mortality for replacement-fed infants/children (range: 1 [no additional risk] to 6). The base-case simulated RRRF = 3, median infant mortality, and 24-month maternal ART duration. RESULTS In the base-case, HFS ranged from 83.1% (no breastfeeding) to 90.2% (12-months breastfeeding). Optimal breastfeeding durations increased with higher RRRF values and longer maternal ART durations, but did not change substantially with variation in infant mortality rates. Optimal breastfeeding durations often exceeded the previous WHO recommendation of 12 months. CONCLUSIONS In settings with high RRRF and long maternal ART durations, HFS is maximized when mothers breastfeed longer than the previously-recommended 12 months. In settings with low RRRF or short maternal ART durations, shorter breastfeeding durations optimize HFS. If mothers are supported to use ART for longer periods of time, it is possible to reduce transmission risks and gain the benefits of longer breastfeeding durations.
Collapse
Affiliation(s)
- Divya Mallampati
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA
| | - Rachel L MacLean
- Medical Practice Evaluation Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Divisions of General Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roger Shapiro
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T.H, Chan School of Public Health, Boston, MA, USA.,The Botswana-Harvard School of Public Health AIDS Institute Partnership for HIV Research and Education, Gaborone, Botswana
| | - Francois Dabis
- Université Bordeaux, Institut de Santé Publique, d'Epidémiologie et de Dévelopement (ISPED), Centre INSERM, U1219-Bordeaux Population Health, Bordeaux, France
| | - Barbara Engelsmann
- Organization for Public Health Interventions and Development, Harare, Zimbabwe
| | - Kenneth A Freedberg
- Medical Practice Evaluation Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Divisions of General Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Harvard University Center for AIDS Research Boston, MA, USA
| | | | - Shahin Lockman
- Department of Immunology and Infectious Diseases, Harvard T.H, Chan School of Public Health, Boston, MA, USA.,The Botswana-Harvard School of Public Health AIDS Institute Partnership for HIV Research and Education, Gaborone, Botswana.,Division of Infectious Disease, Brigham and Women's Hospital, Boston, MA, USA
| | - Rochelle Walensky
- Medical Practice Evaluation Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Divisions of General Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Harvard University Center for AIDS Research Boston, MA, USA.,Division of Infectious Disease, Brigham and Women's Hospital, Boston, MA, USA.,Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nigel Rollins
- Department of Maternal Newborn, Child and Adolescent Health World Health Organization, Geneva, Switzerland
| | - Andrea Ciaranello
- Medical Practice Evaluation Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Twenty-Five Years of Lamivudine: Current and Future Use for the Treatment of HIV-1 Infection. J Acquir Immune Defic Syndr 2019; 78:125-135. [PMID: 29474268 PMCID: PMC5959256 DOI: 10.1097/qai.0000000000001660] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Supplemental Digital Content is Available in the Text. Innovation in medicine is a dynamic, complex, and continuous process that cannot be isolated to a single moment in time. Anniversaries offer opportunities to commemorate crucial discoveries of modern medicine, such as penicillin (1928), polio vaccination (inactivated, 1955; oral, 1961), the surface antigen of the hepatitis B virus (1967), monoclonal antibodies (1975), and the first HIV antiretroviral drugs (zidovudine, 1987). The advent of antiretroviral drugs has had a profound effect on the progress of the epidemiology of HIV infection, transforming a terminal, irreversible disease that caused a global health crisis into a treatable but chronic disease. This result has been driven by the success of antiretroviral drug combinations that include nucleoside reverse transcriptase inhibitors such as lamivudine. Lamivudine, an L-enantiomeric analog of cytosine, potently affects HIV replication by inhibiting viral reverse transcriptase enzymes at concentrations without toxicity against human polymerases. Although lamivudine was approved more than 2 decades ago, it remains a key component of first-line therapy for HIV because of its virological efficacy and ability to be partnered with other antiretroviral agents in traditional and novel combination therapies. The prominence of lamivudine in HIV therapy is highlighted by its incorporation in recent innovative treatment strategies, such as single-tablet regimens that address challenges associated with regimen complexity and treatment adherence and 2-drug regimens being developed to mitigate cumulative drug exposure and toxicities. This review summarizes how the pharmacologic and virologic properties of lamivudine have solidified its role in contemporary HIV therapy and continue to support its use in emerging therapies.
Collapse
|
16
|
Eke AC, McCormack SA, Best BM, Stek AM, Wang J, Kreitchmann R, Shapiro D, Smith E, Mofenson LM, Capparelli EV, Mirochnick M. Pharmacokinetics of Increased Nelfinavir Plasma Concentrations in Women During Pregnancy and Postpartum. J Clin Pharmacol 2018; 59:386-393. [PMID: 30358179 DOI: 10.1002/jcph.1331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/02/2018] [Indexed: 11/05/2022]
Abstract
This study aims to evaluate the safety, acceptability, and pharmacokinetics (PK) of an increased dose of nelfinavir (NFV) during the third trimester of pregnancy. The study was registered as part of the International Maternal Pediatric Adolescent AIDS Clinical Trials network (IMPAACT-P1026s), an ongoing multicenter prospective cohort study of antiretroviral PK during pregnancy (NCT00042289). NFV intensive PK evaluations were performed at steady state during the third trimester of pregnancy and 2-3 weeks postpartum. Plasma concentrations of NFV and its active metabolite, hydroxyl-tert-butylamide (M8) were measured using high-performance liquid chromatography with ultraviolet detection. A total of 18 women are included in the analysis. NFV area under the concentration-time curve (AUC) with the increased dose during the third trimester was nearly identical to the standard dose postpartum, with a geometric mean ratio for third trimester to postpartum AUC of 0.98 (90%CI 0.71-1.35). Despite the increased dose, M8 AUC was lower during the third trimester compared to postpartum (0.53, IQR [0.38-0.75]), as was the M8/NFV AUC ratio (0.51, IQR [0.42-0.63]). NFV AUC0-12 was above target in 15 of 18 (83%) of participants during the third trimester compared to 14 of 16 (88%) postpartum. No major safety concerns were noted. Increasing the NFV dose to 1875 mg twice daily during the third trimester achieved similar concentrations postpartum compared to standard dosing (1250 mg twice daily). Increased NFV dose regimens may still have some benefit to human immunodeficiency virus (HIV)-positive pregnant women living in countries where novel protease inhibitors are currently unavailable or in individuals who are intolerant to ritonavir-boosted HIV medications.
Collapse
Affiliation(s)
- Ahizechukwu C Eke
- Division of Maternal Fetal Medicine, Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Brookie M Best
- University of California San Diego School of Medicine, San Diego, CA, USA.,University of California San Diego Skaggs School of Pharmacy and Pharmaceutical Sciences, San Diego, CA, USA
| | - Alice M Stek
- University of Southern California School of Medicine, Los Angeles, CA, USA
| | - Jiajia Wang
- Harvard School of Public Health, Center for Biostatistics in AIDS Research, Boston, MA, USA
| | - Regis Kreitchmann
- Irmandade da Santa Casa de Misericórdia de Porto Alegre, HIV/AIDS Research Department, Porto Alegre, Rio Grande do Sul, Brazil
| | - David Shapiro
- Harvard School of Public Health, Center for Biostatistics in AIDS Research, Boston, MA, USA
| | - Elizabeth Smith
- National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Lynne M Mofenson
- National Institute of Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD, USA
| | - Edmund V Capparelli
- University of California San Diego School of Medicine, San Diego, CA, USA.,University of California San Diego Skaggs School of Pharmacy and Pharmaceutical Sciences, San Diego, CA, USA
| | | | -
- Division of Maternal Fetal Medicine, Department of Gynecology & Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Maraolo AE, Gentile I, Buonomo AR, Pinchera B, Borgia G. Current evidence on the management of hepatitis B in pregnancy. World J Hepatol 2018; 10:585-594. [PMID: 30310536 PMCID: PMC6177570 DOI: 10.4254/wjh.v10.i9.585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/26/2018] [Accepted: 06/09/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) infection is one of the main public health problems across the globe, since almost one third of the world population presents serological markers of contact with the virus. A profound impact on the epidemiology has been exerted by universal vaccination programmes in many countries, nevertheless the infection is still widespread also in its active form. In the areas of high endemicity (prevalence of hepatitis B surface antigen positivity > 7%), mother-to-child transmission represents the main modality of infection spread. That makes the correct management of HBV in pregnancy a matter of utmost importance. Furthermore, the infection in pregnancy needs to be carefully assessed and handled not only with respect to the risk of vertical transmission but also with respect to gravid women health. Each therapeutic or preventive choice deserves to be weighed upon attentively. On many aspects evidence is scarce or controversial. This review will highlight the latest insights into the paramount steps in managing HBV in pregnancy, with particular attention to recommendations from recent guidelines and data from up-do-date research syntheses.
Collapse
Affiliation(s)
- Alberto Enrico Maraolo
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Italy
| | - Ivan Gentile
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Italy
| | - Antonio Riccardo Buonomo
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Italy
| | - Biagio Pinchera
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Italy
| | - Guglielmo Borgia
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Italy
| |
Collapse
|
18
|
HIV-1 transmission and survival according to feeding options in infants born to HIV-infected women in Yaoundé, Cameroon. BMC Pediatr 2018; 18:69. [PMID: 29458337 PMCID: PMC5817808 DOI: 10.1186/s12887-018-1049-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/06/2018] [Indexed: 11/17/2022] Open
Abstract
Background Evidence of 24-months survival in the frame of prevention of mother-to-child transmission (PMTCT) cascade-care is scare from routine programs in sub-Saharan African (SSA) settings. Specifically, data on infant outcomes according to feeding options remain largely unknown by month-24, thus limiting its breath for public-health recommendations toward eliminating new pediatric HIV-1 infections and improving care. We sought to evaluate HIV-1 vertical transmission and infant survival rates according to feeding options. Methods A retrospective cohort-study conducted in Yaounde from April 2008 through December 2013 among 1086 infants born to HIV-infected women and followed-up throughout the PMTCT cascade-care until 24-months. Infants with documented feeding option during their first 3 months of life (408 on Exclusive Breastfeeding [EBF], 663 Exclusive Replacement feeding [ERF], 15 mixed feeding [MF]) and known HIV-status were enrolled. HIV-1 vertical transmission, survival and feeding options were analyzed using Kaplan Meier Survival Estimate, Cox model and Schoenfeld residuals tests, at 5% statistical significance. Results Overall HIV-1 vertical transmission was 3.59% (39), and varied by feeding options: EBF (2.70%), ERF (3.77%), MF (20%), p = 0.002; without significance between EBF and ERF (p = 0.34). As expected, HIV-1 transmission also varied with PMTCT-interventions: 1.7% (10/566) from ART-group, 1.9% (8/411) from AZT-group, and 19.2% (21/109) from ARV-naïve group, p < 0.0001. Overall mortality was 2.58% (28), higher in HIV-infected (10.25%) vs. uninfected (2.29%) infants (p = 0.016); with a survival cumulative probability of 89.3% [79.9%–99.8%] vs. 96.4% [94.8%–97.9% respectively], p = 0.024. Mortality also varied by feeding option: ERF (2.41%), EBF (2.45%), MF (13.33%), p = 0.03; with a survival cumulative probability of 96% [94%–98%] in ERF, 96.4% [94.1%–98.8%] in EBF, and 86.67% [71.06%–100%] in MF, p = 0.04. Using Schoenfeld residuals test, only HIV status was a predictor of survival at 24 months (hazard ratio 0.23 [0.072–0.72], p = 0.01). Conclusion Besides using ART for PMTCT-interventions, practice of MF also drives HIV-1 vertical transmission and mortality among HIV-infected children. Thus, throughout PMTCT option B+ cascade-care, continuous counseling on safer feeding options would to further eliminating new MTCT, optimizing response to care, and improving the life expectancy of these children in high-priority countries.
Collapse
|
19
|
Ramírez-Ramírez A, Sánchez-Serrano E, Loaiza-Flores G, Plazola-Camacho N, Rodríguez-Delgado RG, Figueroa-Damián R, Domínguez-Castro M, López-Martínez M, Flores-García Z, Hernández-Pineda J. Simultaneous quantification of four antiretroviral drugs in breast milk samples from HIV-positive women by an ultra-high performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method. PLoS One 2018; 13:e0191236. [PMID: 29351333 PMCID: PMC5774716 DOI: 10.1371/journal.pone.0191236] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/29/2017] [Indexed: 01/24/2023] Open
Abstract
The primary strategy to avoid mother-to-child transmission of human immunodeficiency virus (HIV) through breastfeeding is administration of highly active antiretroviral therapy (HAART) to HIV-positive pregnant women. Because significant changes in the pharmacokinetics of antiretroviral (ARV) drugs occur during pregnancy, quantifying HAART and the viral load in breast milk in this population is essential. Here, we developed an analytical assay for the simultaneous quantification of four ARV drugs in breast milk using ultra-performance liquid chromatography coupled to tandem mass spectrometry. We validated this method following Mexican and international guidelines. ARV drugs. We extracted the ARV drugs from 200 μL samples of breast milk and detected these drugs in a triple quadrupole mass spectrometer with positive electrospray ionization. The validated concentration ranges (ng/mL) for zidovudine, lamivudine, lopinavir, and ritonavir were 12.5–750, 50–2500, 100–5000 and 5 to 250, respectively. Additionally, the absolute recovery percentages (and matrix effects) were 91.4 (8.39), 88.78 (28.75), 91.38 (11.77) and 89.78 (12.37), respectively. We determined that ARV drugs are stable for 24 h at 8°C and 24°C for 15 days at –80°C. This methodology had the capacity for simultaneous detection; separation; and accurate, precise quantification of ARV drugs in human breast milk samples according to Mexican standard laws and United States Food and Drug Administration guidelines.
Collapse
Affiliation(s)
- Alicia Ramírez-Ramírez
- Departement of Infectology and Immunology, National Institute of Perinatology, Mexico City, Mexico
| | | | | | - Noemí Plazola-Camacho
- Departement of Infectology and Immunology, National Institute of Perinatology, Mexico City, Mexico
| | | | - Ricardo Figueroa-Damián
- Departement of Infectology and Immunology, National Institute of Perinatology, Mexico City, Mexico
| | - Mauricio Domínguez-Castro
- Department of Physiology and Cellular Development, National Institute of Perinatology, Mexico City, Mexico
| | - Margarita López-Martínez
- Department of Physiology and Cellular Development, National Institute of Perinatology, Mexico City, Mexico
| | - Zayra Flores-García
- Faculty of Higher Education, National Autonomous University of Mexico, Cuautitlan Izcalli, Mexico State, Mexico
| | - Jessica Hernández-Pineda
- Departement of Infectology and Immunology, National Institute of Perinatology, Mexico City, Mexico
- * E-mail:
| |
Collapse
|
20
|
Are Prophylactic and Therapeutic Target Concentrations Different?: the Case of Lopinavir-Ritonavir or Lamivudine Administered to Infants for Prevention of Mother-to-Child HIV-1 Transmission during Breastfeeding. Antimicrob Agents Chemother 2017; 61:AAC.01869-16. [PMID: 27895016 DOI: 10.1128/aac.01869-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/18/2016] [Indexed: 11/20/2022] Open
Abstract
The ANRS 12174 trial assessed the efficacy and tolerance of lopinavir (LPV)-ritonavir (LPV/r) prophylaxis versus those of lamivudine (3TC) prophylaxis administered to breastfed infants whose HIV-infected mothers were not on antiretroviral therapy. In this substudy, we assessed LPV/r and 3TC pharmacokinetics to evaluate the percentage of infants with therapeutic plasma concentrations and to discuss these data in the context of a prophylactic treatment. Infants from the South African trial site underwent blood sampling for pharmacokinetic study at weeks 6, 26, and 38 of life. We applied a Bayesian approach to derive the 3TC and LPV pharmacokinetic parameters on the basis of previously published pharmacokinetic models for HIV-infected children. We analyzed 114 LPV and 180 3TC plasma concentrations from 69 infants and 92 infants, respectively. A total of 30 LPV and 20 3TC observations were considered missing doses and discarded from the Bayesian analysis. The overall population analysis showed that 30 to 40% of the infants did not reach therapeutic targets, regardless of treatment group. The median LPV trough concentrations at weeks 6, 26, and 38 were 2.8 mg/liter (interquartile range [IQR], 1.7 to 4.4 mg/liter), 5.6 mg/liter (IQR, 3.2 to 7.7 mg/liter), and 3.4 mg/liter (IQR, 2.3 to 7.3 mg/liter), respectively. The median 3TC area under the curve from 0 to 12 h after the last drug intake were 5.6 mg · h/liter (IQR, 4.1 to 7.8 mg · h/liter), 5.9 mg · h/liter (IQR, 5.1 to 7.5 mg · h/liter), and 7.3 mg · h/liter (IQR, 4.9 to 8.5 mg · h/liter) at weeks 6, 26, and 38, respectively. Use of the therapeutic doses recommended by the WHO would have resulted in a higher proportion of infants achieving the targets. However, no HIV-1 infection was reported among these infants. These results suggest that the prophylactic targets for both 3TC and LPV may be lower than the therapeutic ones. For treatment, the WHO dosing guidelines should be suitable to maintain values above the therapeutic pharmacokinetic targets in most infants. (This study has been registered at ClinicalTrials.gov under identifier NCT00640263.).
Collapse
|
21
|
Peters MD, McArthur A, Munn Z. Safe management of expressed breast milk: A systematic review. Women Birth 2016; 29:473-481. [DOI: 10.1016/j.wombi.2016.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 05/26/2016] [Accepted: 05/29/2016] [Indexed: 10/21/2022]
|
22
|
Yudin MH, Kennedy VL, MacGillivray SJ. HIV and infant feeding in resource-rich settings: considering the clinical significance of a complicated dilemma. AIDS Care 2016; 28:1023-6. [PMID: 26881474 DOI: 10.1080/09540121.2016.1140885] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
With advances in the care of HIV-positive pregnant women, the likelihood of perinatal transmission is now less than 1%. In resource-rich settings women are instructed to abstain from breastfeeding, as studies have shown that breastfeeding increases the likelihood of infant acquisition of HIV. As practitioners caring for HIV-positive parents, we are now facing growing tension about the complex issues that inform decisions about infant feeding. In the face of changing guidelines and global immigration patterns, simply telling women that breastfeeding is contraindicated may no longer be good enough. We must fully open the lines of communication regarding this important and evolving issue. This commentary will review the clinical, social and cultural considerations that impact decisions regarding infant feeding in the context of HIV.
Collapse
Affiliation(s)
- Mark H Yudin
- a Department of Obstetrics and Gynecology , St. Michael's Hospital , Toronto , Canada.,b Department of Obstetrics and Gynecology , University of Toronto , Toronto , Canada
| | | | - S Jay MacGillivray
- a Department of Obstetrics and Gynecology , St. Michael's Hospital , Toronto , Canada
| |
Collapse
|