1
|
Taton M, Willems F, Widomski C, Georges D, Martin C, Jiang Y, Renard K, Konopnicki D, Cogan A, Necsoi C, Matagne A, De Wit S, Ackerman ME, Marchant A, Dauby N. HIV-related immune activation attenuates polyfunctional IgG and memory B-cell responses to Tdap immunization during pregnancy. EBioMedicine 2024; 104:105179. [PMID: 38848615 PMCID: PMC11192781 DOI: 10.1016/j.ebiom.2024.105179] [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: 04/14/2023] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Maternal pertussis vaccination with Tdap vaccine is recommended to protect newborns from severe postnatal infection. HIV-exposed uninfected (HEU) infants have a higher incidence of pertussis infection and may particularly benefit from maternal immunization. The impact of HIV infection on the quality of IgG and memory B cell (MBC) responses to Tdap vaccination in pregnant women (PW) living with HIV (PWH) is unknown. METHODS In this observational study, humoral immune responses to Tdap vaccination, including IgG levels, Fc-dependent effector functions, and MBC frequencies, were measured before and after vaccination in 40 PWH and 42 HIV-uninfected PW. Placental transfer of IgG and avidity were assessed in cord blood (CB). Soluble and cellular immune activation markers were quantified at baseline. FINDINGS One month after vaccination, PWH had lower frequencies of MBC compared with HIV-uninfected PW. At delivery, PWH had attenuated pertussis-specific IgG levels and Fc-dependent effector functions. Reduced levels of maternal vaccine polyfunctional IgG and IgG avidity were transferred to HEU as compared to HIV-unexposed newborns. After adjustment with ethnicity, maternal antibody levels and gestational age at vaccination, HIV infection was independently associated with decreased levels of PT specific-IgG in CB. Both maternal and neonatal pertussis-specific IgG responses as well as PT-specific IgG avidity were inversely correlated with maternal sCD14 levels before vaccination among PWH. INTERPRETATION Maternal HIV infection is associated with attenuated humoral immune responses to Tdap vaccination that correlate with sCD14. Suboptimal transfer of maternal immunity may further increase the risk of severe pertussis infection in HEU infants. FUNDING This work was supported by IRIS Fund managed by the Foundation Roi Baudouin [2017J1820690206902], Association Vésale pour la Recherche Médicale and the Medical Council of CHU Saint-Pierre and has been funded in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, US Department of Health and Human Services, under Award No. U19AI145825. N.D. is a clinical researcher and A.M. is Research Director at the Fonds de la Recherche Scientifique (F.R.S.-FNRS), Belgium. M.E.A. was partially supported by NIHNIAID1U19AI14825. This article is published with the support of the Fondation Universitaire of Belgium.
Collapse
Affiliation(s)
- Martin Taton
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fabienne Willems
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Cyprien Widomski
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium
| | - Daphnée Georges
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium; Faculty of Sciences, Université de Liège, Liège, Belgium
| | - Charlotte Martin
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Yiwei Jiang
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium
| | - Katty Renard
- Clinical Research Unit, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Deborah Konopnicki
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alexandra Cogan
- Department of Gynecology and Obstetrics, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Coca Necsoi
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - André Matagne
- Faculty of Sciences, Université de Liège, Liège, Belgium
| | - Stéphane De Wit
- Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université Libre de Bruxelles (ULB), , Brussels, Belgium
| | - Nicolas Dauby
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium; Department of Infectious Diseases, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
2
|
Nakabembe E, Cooper J, Amaral K, Tusubira V, Hsia Y, Abu-Raya B, Sekikubo M, Nakimuli A, Sadarangani M, Le Doare K. The safety and immunogenicity of vaccines administered to pregnant women living with HIV: a systematic review and meta-analysis. EClinicalMedicine 2024; 69:102448. [PMID: 38333366 PMCID: PMC10850112 DOI: 10.1016/j.eclinm.2024.102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Background Human Immunodeficiency Virus (HIV)-exposed uninfected (HEU) infants have a higher burden of infectious diseases related morbidity and mortality compared with HIV-unexposed uninfected (HUU). Immunization of pregnant women living with HIV (PWLWH) could reduce the severity and burden of infectious diseases for HEU in early infancy. Methods We conducted a systematic review of safety and immunogenicity of vaccines administered to PWLWH and meta-analyses to test the overall effect of immunogenicity comparing pregnant women without HIV (PWWH) to PWLWH. We searched MEDLINE, Embase, Web of Science, Virtual Health Library and Cochrane databases in accordance with PRISMA guidelines for randomized controlled trials and observational studies. Review articles, case series, conference abstracts, and animal studies were excluded. Studies were included from inception to 6th September 2023, with no language restrictions. Random effects meta-analyses were performed for immunogenicity using Review manager (RevMan) analysis software version 5.4.1, Geometric Mean Titer (GMT) values were transformed to obtain the mean and standard deviation within RevMan, the effect size was computed and reported as mean difference with respective 95% confidence intervals. The review was registered with PROSPERO CRD42021289081. Findings We included 12 articles, comprising 3744 pregnant women, 1714 were PWLWH given either influenza, pneumococcal or an investigational Group B streptococcal (GBS) vaccine. Five studies described safety outcomes, and no increase in adverse events was reported in PWLWH compared to PWWH. The GMT increase from baseline to 28-35 weeks post vaccination in HA units ranged from 12.4 (95% CI: 9.84-14.9) to 238.8 (95% CI: 0.35-477.9). Meta-analyses of influenza vaccines showed the pooled geometric mean difference in Hemagglutination Inhibition (HAI) titers post vaccination was 56.01 (95% CI: 45.01-67.01), p < 0.001. The increase was less in PWLWH when compared with PWWH: -141.76 (95% CI: -194.96, -88.55), p < 0.001. Interpretation There are limited data on the safety and immunogenicity of vaccines given to PWLWH making policy consideration in this group difficult when new vaccines are introduced. With new vaccines on the horizon, PWLWH need to be included in studies to promote vaccine confidence for this special population. Funding This work was funded by Medical Research Council Joint Clinical Trials Round 9 [MR/T004983/1].
Collapse
Affiliation(s)
- Eve Nakabembe
- Department of Obstetrics and Gynaecology, School of Medicine, Makerere University College of Health Sciences, Upper Mulago Hill Road, P.O. Box 7072, Kampala, Uganda
- Centre for Neonatal and Pediatric Infection, St George’s University of London, Cranmer Terrace, London SW170RE, United Kingdom
| | - Jo Cooper
- Centre for Neonatal and Pediatric Infection, St George’s University of London, Cranmer Terrace, London SW170RE, United Kingdom
| | - Kyle Amaral
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Valerie Tusubira
- Makerere University-Johns Hopkins Research Collaboration, Upper Mulago Hill Road, P.O. Box 23491, Kampala, Uganda
| | - Yingfen Hsia
- Centre for Neonatal and Pediatric Infection, St George’s University of London, Cranmer Terrace, London SW170RE, United Kingdom
| | - Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3V4, Canada
| | - Musa Sekikubo
- Department of Obstetrics and Gynaecology, School of Medicine, Makerere University College of Health Sciences, Upper Mulago Hill Road, P.O. Box 7072, Kampala, Uganda
| | - Annettee Nakimuli
- Department of Obstetrics and Gynaecology, School of Medicine, Makerere University College of Health Sciences, Upper Mulago Hill Road, P.O. Box 7072, Kampala, Uganda
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3V4, Canada
| | - Kirsty Le Doare
- Centre for Neonatal and Pediatric Infection, St George’s University of London, Cranmer Terrace, London SW170RE, United Kingdom
- Makerere University-Johns Hopkins Research Collaboration, Upper Mulago Hill Road, P.O. Box 23491, Kampala, Uganda
| |
Collapse
|
3
|
Martin C, Florence E, Domingo C, Delforge M, De Wit S, Dauby N. Seroconversion and antibody persistence after yellow fever vaccination in people living with HIV: impact of baseline HIV viral load and yellow fever seropositivity. J Travel Med 2022; 29:6548118. [PMID: 35285913 DOI: 10.1093/jtm/taac024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/01/2022] [Accepted: 11/14/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Data on seroconversion rates following yellow fever (YF) vaccine and effect of revaccination in people living with HIV (PLWH) are scarce. We aimed at determining key factors for seroconversion after YF vaccine in PLWH and the role of preexisting neutralizing antibodies (NAbs) at vaccination. METHODS A retrospective cross-sectional study at several timepoints in two Belgian AIDS Reference Center. For each individual, plasma samples from three timepoints were selected: Timepoint 0 (TP0) in the year before administration of the YF vaccine, Timepoint 1 (TP1) in the year following the YF vaccine, Timepoint 2 (TP2) >1 year after the YF vaccine. Plasma samples were analysed for YF NAbs by plaque reduction neutralization test. The primary endpoint was the number of patients with protective levels of NAbs ≥ 1/10. A boosted immune response was defined as a 4-fold increase in serologic titres following revaccination. RESULTS Of the 160 PLWH included, protective levels of NAbs were present in 36%, 87% and 72% of subjects at baseline, at a median of 12 months and a median of 96 months after YF vaccination, respectively. Among vaccine recipients negative for YF NAbs at baseline (n = 102), 83% seroconverted. PLWH with undetectable HIV viral load (VL) at baseline were more likely to seroconvert (P < 0·01). A booster response was observed in only 17% of subjects with baseline seropositivity (n = 10 out of 58). In multivariate analysis, undetectable HIV VL at vaccination and baseline YF seropositivity were associated with persistent levels of protective NAbs at a median of 8 years after YF vaccination. CONCLUSION Undetectable HIV VL at baseline is associated with high rates of seroconversion. YF seropositivity before revaccination is associated with low rates of booster effect but a higher chance of long term persistent NAbs response, suggesting a benefit of revaccination in PLWH.
Collapse
Affiliation(s)
- Charlotte Martin
- Infectious Diseases Department, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Eric Florence
- Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Cristina Domingo
- Robert Koch Institute, Centre for Biological Threats and Special Pathogens - Highly Pathogenic Viruses- ZBS-1, 13353 Berlin, Germany.,Robert Koch Institute, Centre for International Health Protection (ZIG) -ZIG-4 Public Health Laboratory Support, 13353 Berlin, Germany
| | - Marc Delforge
- Infectious Diseases Department, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Stéphane De Wit
- Infectious Diseases Department, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Nicolas Dauby
- Infectious Diseases Department, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium.,Institute for Medical Immunology, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.,School of Public Health, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
4
|
Sainz T, Casas I, González-Esguevillas M, Escosa-Garcia L, Muñoz-Fernández MÁ, Prieto L, Gosalbes MJ, Jiménez-Hernández N, Ramos JT, Navarro ML, Mellado MJ, Serrano-Villar S, Calvo C. Nutritional Supplementation to Increase Influenza Vaccine Response in Children Living With HIV: A Pilot Clinical Trial. Front Pediatr 2022; 10:919753. [PMID: 35928688 PMCID: PMC9343631 DOI: 10.3389/fped.2022.919753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Vaccine response is poor among children living with HIV. The gut microbiota has been identified as a potential target to improve vaccine immunogenicity, but data are scarce in the context of HIV infection. METHODS Pilot, double-blind, randomized placebo-controlled trial in which 24 HIV-infected children were randomized to receive a mixture of symbiotics, omega-3/6 fatty acids, and amino acids or placebo for 4 weeks, each in combination with ART, and were then immunized against influenza. Vaccine response and safety of the nutritional supplementation were the primary outcomes. RESULTS Eighteen HIV-infected children completed the follow-up period (mean age 11.5 ± 4.14 years, 61% female). The nutritional supplement was safe but did not enhance the response to the influenza vaccine. A 4-fold rise in antibody titers was obtained in only 37.5% of participants in the intervention arm vs. 40% in the placebo. No immunological or inflammatory predictors of vaccine response were identified. CONCLUSIONS In this exploratory study, a 4-week course of symbiotics did not increase influenza vaccine immunogenicity in HIV-infected children. Larger studies are warranted to address the potential of modulating the microbiome in children living with HIV.
Collapse
Affiliation(s)
- Talía Sainz
- Servicio de Pediatría, Hospital Universitario La Paz and IdiPAZ, Madrid, Spain.,Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Inmaculada Casas
- Respiratory Virus and Influenza Unit, Instituto de Salud Carlos III, Madrid, Spain.,Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | | | - Luis Escosa-Garcia
- Servicio de Pediatría, Hospital Universitario La Paz and IdiPAZ, Madrid, Spain.,Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - María Ángeles Muñoz-Fernández
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Laboratorio de InmunoBiología Molecular Hospital General Universitario Gregorio Marañón e IISHGM, Madrid, Spain
| | - Luis Prieto
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Hospital 12 de Octubre, Madrid, Spain.,Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - María José Gosalbes
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Área Genómica y Salud, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica (FISABIO), Valencia, Spain
| | - Nuria Jiménez-Hernández
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Área Genómica y Salud, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica (FISABIO), Valencia, Spain
| | - José Tomas Ramos
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.,Universidad Complutense de Madrid (UCM), Madrid, Spain.,Servicio de Pediatría, Hospital Clinico San Carlos and IdISSC, Madrid, Spain
| | - María Luisa Navarro
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.,Universidad Complutense de Madrid (UCM), Madrid, Spain.,Unidad de Investigación Materno-Infantil Familia Alonso (UDIMIFFA), IISGM, Servicio de Pediatría, Hospital General Universitario Gregorio Marañón e IISHGM, Madrid, Spain
| | - María José Mellado
- Servicio de Pediatría, Hospital Universitario La Paz and IdiPAZ, Madrid, Spain.,Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Serrano-Villar
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Enfermedades Infecciosas, Hospital Universitario Ramón y Cajal, and IRYCIS, Madrid, Spain
| | - Cristina Calvo
- Servicio de Pediatría, Hospital Universitario La Paz and IdiPAZ, Madrid, Spain.,Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Vazquez-Pagan A, Schultz-Cherry S. Serological Responses to Influenza Vaccination during Pregnancy. Microorganisms 2021; 9:microorganisms9112305. [PMID: 34835431 PMCID: PMC8619416 DOI: 10.3390/microorganisms9112305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/25/2021] [Accepted: 11/03/2021] [Indexed: 11/25/2022] Open
Abstract
Pregnant women, newborns, and infants under six months old are at the highest risk of developing severe and even fatal influenza. This risk is compounded by the inability to vaccinate infants under six months, highlighting the importance of vertically transferred immunity. This review identifies novel insights that have emerged from recent studies using animal models of pregnancy and vaccination. We also discuss the knowledge obtained using existing clinical trials that have evaluated influenza-specific serological responses in pregnant women and how these responses may impact early life immunity. We delineate the mechanisms involved in transferring specific maternal antibodies and discuss the consequences for early life immunity. Most importantly, we highlight the need for continued research using pregnant animal models and the inclusion of pregnant women, a commonly neglected population, when evaluating novel vaccine platforms to better serve and treat communicable diseases.
Collapse
Affiliation(s)
- Ana Vazquez-Pagan
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Stacey Schultz-Cherry
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Correspondence:
| |
Collapse
|
6
|
Swieboda D, Littauer EQ, Beaver JT, Mills LK, Bricker KM, Esser ES, Antao OQ, Williams DT, Skountzou I. Pregnancy Downregulates Plasmablast Metabolic Gene Expression Following Influenza Without Altering Long-Term Antibody Function. Front Immunol 2020; 11:1785. [PMID: 32922392 PMCID: PMC7457062 DOI: 10.3389/fimmu.2020.01785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
While the majority of influenza-infected individuals show no or mild symptomatology, pregnant women are at higher risk of complications and infection-associated mortality. Although enhanced lung pathology and dysregulated hormones are thought to underlie adverse pregnancy outcomes following influenza infection, how pregnancy confounds long-term maternal anti-influenza immunity remains to be elucidated. Previously, we linked seasonal influenza infection to clinical observations of adverse pregnancy outcomes, enhanced lung and placental histopathology, and reduced control of viral replication in lungs of infected pregnant mothers. Here, we expand on this work and demonstrate that lower infectious doses of the pandemic A/California/07/2009 influenza virus generated adverse gestational outcomes similar to higher doses of seasonal viruses. Mice infected during pregnancy demonstrated lower hemagglutination inhibition and neutralizing antibody titers than non-pregnant animals until 63 days post infection. These differences in humoral immunity suggest that pregnancy impacts antibody maturation mechanisms without alterations to B cell frequency or antibody secretion. This is further supported by transcriptional analysis of plasmablasts, which demonstrate downregulated B cell metabolism and post-translational modification systems only among pregnant animals. In sum, these findings corroborate a link between adverse pregnancy outcomes and severe pathology observed during pandemic influenza infection. Furthermore, our data propose that pregnancy directly confounds humoral responses following influenza infection which resolves post-partem. Additional studies are required to specify the involvement of plasmablast metabolism with early humoral immunity abnormalities to best guide vaccination strategies and improve our understanding of the immunological consequences of pregnancy.
Collapse
Affiliation(s)
- Dominika Swieboda
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Elizabeth Q Littauer
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Jacob T Beaver
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Lisa K Mills
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine M Bricker
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - E Stein Esser
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Olivia Q Antao
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Dahnide T Williams
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
7
|
Nunes MC, Cutland CL, Moultrie A, Jones S, Ortiz JR, Neuzil KM, Klugman KP, Simões EAF, Weinberg A, Madhi SA. Immunogenicity and safety of different dosing schedules of trivalent inactivated influenza vaccine in pregnant women with HIV: a randomised controlled trial. Lancet HIV 2020; 7:e91-e103. [PMID: 31911146 PMCID: PMC7167514 DOI: 10.1016/s2352-3018(19)30322-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 12/04/2022]
Abstract
BACKGROUND Standard-dose, seasonal, trivalent, inactivated influenza vaccine induces moderate-to-low haemagglutination-inhibition antibody responses in people living with HIV. This study assessed the immunogenicity and safety of different dosing schedules of inactivated influenza vaccine in pregnant women living with HIV in South Africa. METHODS In this double-blind, randomised, controlled trial, we recruited pregnant women with HIV from seven antenatal clinics in Soweto, South Africa. Pregnant women were eligible if they were aged 18-38 years, infected with HIV, and had an estimated gestational age of 12-36 weeks. Women were randomly assigned (1:1:1), using a computer-generated randomisation list, to receive inactivated influenza vaccine containing 15 μg of each of the three seasonal influenza strains for that year, as a single dose, a double dose, or two single doses 1 month apart. Participants and study personnel were masked to group allocation. Haemagglutination-inhibition antibody responses were measured for all groups in the mothers at enrolment and at 1 month after each vaccine dose, and in the single-dose and double-dose groups within 7 days of birth in the neonates. Immunogenicity analyses only included women with visits 28-35 days apart and infants who were born at least 28 days after maternal immunisation. The primary was seroconversion rate to each of the vaccine strains in the mothers 1 month after completion of the dosing schedule, and the primary safety outcomes were frequency of local and systemic reactions. Safety was assessed in mothers and infants until 24 weeks post partum and analysed in all participants who received at least one dose of vaccine. This study is registered with ClinicalTrials.gov, NCT01527825, and is closed to accrual. FINDINGS Between Feb 11, and June 6, 2013, 800 pregnant women living with HIV were enrolled and randomly assigned to the single-dose (n=266), double-dose (n=265), or two-single-doses (n=269) group. In the analysable population, seroconversion rates in mothers 1 month after the final vaccine dose were significantly higher in the double-dose group (n=230; ranging from 29% to 65% for the three vaccine strains) than in the single-dose group (n=230; ranging from 18% to 49%; p≤0·019 for the three vaccine strains), but were similar between the two-single-doses group (n=220; ranging from 23% to 52%) and the single-dose group (p≥0·20 for the three vaccine strains). Safety outcomes were similar in the three groups, except for more injection-site reactions in recipients in the double-dose group. INTERPRETATION A regimen of double-dose inactivated influenza vaccine gave slightly greater immunogenicity than did a single-dose regimen in pregnant women living with HIV. However, immunogenicity in the double-dose group was still lower than historical data from the same setting in pregnant women without HIV. More immunogenic vaccines are needed for pregnant women living with HIV to enhance transplacental transfer of vaccine-induced protective antibodies to their newborn infants. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Marta C Nunes
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation, SARCHI: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa.
| | - Clare L Cutland
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation, SARCHI: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Andrew Moultrie
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation, SARCHI: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephanie Jones
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation, SARCHI: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Justin R Ortiz
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Keith P Klugman
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, Johannesburg, South Africa
| | - Eric A F Simões
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, Johannesburg, South Africa; School of Public Health, Center for Global Health, University of Colorado, Aurora, CO, USA; Department of Pediatric Infectious Diseases, University of Colorado, Aurora, CO, USA
| | - Adriana Weinberg
- Department of Pediatrics, Medicine and Pathology, University of Colorado, Aurora, CO, USA
| | - Shabir A Madhi
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, Johannesburg, South Africa; Department of Science and Technology/National Research Foundation, SARCHI: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
8
|
Bengtson AM, Sanfilippo AM, Hughes BL, Savitz DA. Maternal immunisation to improve the health of HIV-exposed infants. THE LANCET. INFECTIOUS DISEASES 2018; 19:e120-e131. [PMID: 30529212 DOI: 10.1016/s1473-3099(18)30545-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/28/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022]
Abstract
HIV-exposed but uninfected (HEU) infants are at an increased risk of many infectious diseases that can contribute to the high mortality seen among HEU children. Maternal immunisation could be a promising strategy to reduce infections in HEU infants. However, very little research has explored the effect of HIV on the immunogenicity and effectiveness of vaccines given during pregnancy. We review the available evidence on maternal immunisation among women living with HIV (WLWH) for all vaccines recommended, considered, or being investigated for routine or risk-based use during pregnancy. Of the 11 vaccines included, only three have been investigated in WLWH. Available evidence suggests that maternal HIV infection limits the immunogenicity of several vaccines, leaving HEU infants more susceptible to infection during their first few months of life. Whether maternal immunisation reduces the infectious morbidity and mortality associated with infectious diseases in HEU children remains unknown. We conclude the Review by identifying future research priorities.
Collapse
Affiliation(s)
- Angela M Bengtson
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA.
| | - Alan M Sanfilippo
- Department of Pathology, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, USA
| | - Brenna L Hughes
- Division of Maternal Fetal Medicine, Duke University, Durham, NC, USA
| | - David A Savitz
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| |
Collapse
|
9
|
Dangor Z, Nunes MC, Kwatra G, Lala SG, Madhi SA. Vaccination of HIV-infected pregnant women: implications for protection of their young infants. TROPICAL DISEASES TRAVEL MEDICINE AND VACCINES 2017; 3:1. [PMID: 28883971 PMCID: PMC5530931 DOI: 10.1186/s40794-016-0044-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/18/2016] [Indexed: 12/02/2022]
Abstract
Background The prevention of mother to child transmission of HIV has resulted in reduced burden of pediatric HIV-infection, but the prevalence of maternal HIV infection remains high in sub-Saharan African countries. HIV-exposed-uninfected infants have an increased risk of morbidity and mortality due to infectious diseases than HIV-unexposed infants, particularly during the first six months of life, which in part might be due to lower levels of pathogen-specific protective antibodies acquired transplacentally from their mothers. This could be mitigated by vaccinating pregnant women to boost antibody levels; although vaccine responses among HIV-infected pregnant women might differ compared to HIV-uninfected women. We reviewed studies that compared natural and vaccine-induced antibody levels to different epitopes between HIV-infected and HIV-uninfected pregnant women. Findings Most studies reported lower baseline/pre-vaccination antibody levels in HIV-infected pregnant women, which may not be reversed by antiretroviral therapy during pregnancy. There were only few studies on vaccination of HIV-infected pregnant women, mainly on influenza virus and group B Streptococcus (GBS) vaccines. Immunogenicity studies on influenza vaccines indicated that HIV-infected pregnant women had lower vaccine induced hemagglutination inhibition antibody titers and a decreased likelihood of seroconversion compared to HIV-uninfected women; and while higher CD4+ T-lymphocyte levels were associated with better immune responses to vaccination, HIV viral load was not associated with responses. Furthermore, infants born to influenza vaccinated HIV-infected pregnant women also had lower antibody levels and a lower proportion of HIV-exposed infants had titers above the putative correlate of protection compared to HIV-unexposed infants. The immunogenicity of a CRM197-conjugated trivalent GBS vaccine was also lower in HIV-infected pregnant women compared to HIV-uninfected women, irrespective of CD4+ T-lymphocyte counts. Conclusions Poorer immunogenicity of vaccines reported in HIV-infected compared to HIV-uninfected pregnant women might compromise the potential benefits to their young infants. Alternate vaccination strategies, including vaccines with higher antigen concentration, adjuvanted vaccines or multiple doses schedules might be required in HIV-infected pregnant women to optimize antibody transferred to their fetuses.
Collapse
Affiliation(s)
- Ziyaad Dangor
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa.,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marta C Nunes
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Sanjay G Lala
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa.,National Institute for Communicable Diseases: a division of National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
10
|
Avelino-Silva VI, Miyaji KT, Hunt PW, Huang Y, Simoes M, Lima SB, Freire MS, Caiaffa-Filho HH, Hong MA, Costa DA, Dias JZC, Cerqueira NB, Nishiya AS, Sabino EC, Sartori AM, Kallas EG. CD4/CD8 Ratio and KT Ratio Predict Yellow Fever Vaccine Immunogenicity in HIV-Infected Patients. PLoS Negl Trop Dis 2016; 10:e0005219. [PMID: 27941965 PMCID: PMC5179051 DOI: 10.1371/journal.pntd.0005219] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/22/2016] [Accepted: 11/30/2016] [Indexed: 11/30/2022] Open
Abstract
Background HIV-infected individuals have deficient responses to Yellow Fever vaccine (YFV) and may be at higher risk for adverse events (AE). Chronic immune activation–characterized by low CD4/CD8 ratio or high indoleamine 2,3-dioxygenase-1 (IDO) activity—may influence vaccine response in this population. Methods We prospectively assessed AE, viremia by the YFV virus and YF-specific neutralizing antibodies (NAb) in HIV-infected (CD4>350) and -uninfected adults through 1 year after vaccination. The effect of HIV status on initial antibody response to YFV was measured during the first 3 months following vaccination, while the effect on persistence of antibody response was measured one year following vaccination. We explored CD4/CD8 ratio, IDO activity (plasma kynurenine/tryptophan [KT] ratio) and viremia by Human Pegivirus as potential predictors of NAb response to YFV among HIV-infected participants with linear mixed models. Results 12 HIV-infected and 45-uninfected participants were included in the final analysis. HIV was not significantly associated with AE, YFV viremia or NAb titers through the first 3 months following vaccination. However, HIV–infected participants had 0.32 times the NAb titers observed for HIV-uninfected participants at 1 year following YFV (95% CI 0.13 to 0.83, p = 0.021), independent of sex, age and prior vaccination. In HIV-infected participants, each 10% increase in CD4/CD8 ratio predicted a mean 21% higher post-baseline YFV Nab titer (p = 0.024). Similarly, each 10% increase in KT ratio predicted a mean 21% lower post-baseline YFV Nab titer (p = 0.009). Viremia by Human Pegivirus was not significantly associated with NAb titers. Conclusions HIV infection appears to decrease the durability of NAb responses to YFV, an effect that may be predicted by lower CD4/CD8 ratio or higher KT ratio. Yellow Fever (YF) vaccine is considered one of the most effective vaccines ever produced. However, previous studies suggest that HIV impairs YF vaccine response. In this study, we assessed if HIV infection impacts the risk of adverse events and could reduce antibody response to YF vaccine. We explored if laboratory markers of persistent inflammation, frequently present among HIV-infected patients, could predict antibody response to YF vaccine in this population. We found that HIV had no significant effect on adverse events or levels of antibodies through 3 months after vaccination, but this may be limited by the small sample size of 12 HIV-infected and 45-uninfected participants in the study. However, we were able to show that, compared to HIV-uninfected participants, HIV–infected patients had lower antibody titers 1 year following YF vaccine even after statistical adjustment for the potential effects of sex, age and prior vaccination. Persistent inflammation seems to reduce YF vaccine antibody response in HIV-infected participants. In conclusion, HIV-infected individuals have impaired antibody response to YFV due to a poorer persistence of antibodies, despite a seemingly normal initial response. HIV-infected patients at permanent or recurring risk of YF infection may benefit from a booster dose of YF vaccine.
Collapse
Affiliation(s)
- Vivian I. Avelino-Silva
- Department of Infectious and Parasitic Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
- * E-mail:
| | - Karina T. Miyaji
- Department of Infectious and Parasitic Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Peter W. Hunt
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States of America
| | - Yong Huang
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy, University of California, San Francisco, San Francisco, California, United States of America
| | | | | | | | - Helio H. Caiaffa-Filho
- Instituto Adolfo Lutz, São Paulo, Brazil
- Laboratory of Medical Investigation LIM-3, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Marisa A. Hong
- Instituto Adolfo Lutz, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Dayane Alves Costa
- Division of Clinical Immunology and Allergy, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Juliana Zanatta C. Dias
- Division of Clinical Immunology and Allergy, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Natalia B. Cerqueira
- Division of Clinical Immunology and Allergy, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Ester Cerdeira Sabino
- Department of Infectious and Parasitic Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Fundação Pro-Sangue Hemocentro de Sao Paulo, Sao Paulo, Brazil
| | - Ana M. Sartori
- Department of Infectious and Parasitic Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Esper G. Kallas
- Department of Infectious and Parasitic Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
11
|
Lelic A, Verschoor CP, Lau VWC, Parsons R, Evelegh C, Bowdish DM, Bramson JL, Loeb MB. Immunogenicity of Varicella Vaccine and Immunologic Predictors of Response in a Cohort of Elderly Nursing Home Residents. J Infect Dis 2016; 214:1905-1910. [PMID: 27707807 DOI: 10.1093/infdis/jiw462] [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] [Received: 07/24/2016] [Accepted: 09/23/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Little is known about the immunogenicity of live-attenuated Oka/Merck varicella zoster virus (VZV)-containing vaccine (hereafter, "varicella vaccine") in frail nursing homes residents nor about immune phenotypes associated with a response. METHODS A cohort of 190 frail nursing home residents aged 80-102 years and a cohort of 50 community-dwelling seniors aged 60-75 years (a comparison group) received varicella vaccine. Interferon γ (IFN-γ) enzyme-linked immunospot assays were performed before and 6 weeks after vaccination. Cellular markers of immunosenescence were measured in the nursing home elderly. RESULTS The average number of IFN-γ spot-forming cells at baseline was significantly lower in the elderly nursing home residents than in the community-dwelling seniors. However, following vaccination, the VZV immune response increased in both cohorts, and no difference was noted in the fold difference of the response between the 2 cohorts. Upon further examination of the elderly nursing home residents, we found that higher frequencies of regulatory T cells and cytomegalovirus-specific CD4+ T cells correlated negatively with the magnitude of VZV-specific responses. CONCLUSIONS The Oka/Merck varicella vaccine induces VZV immunity in elderly nursing home residents that is similar to that produced in community-dwelling seniors. CLINICAL TRIALS REGISTRATION NCT01328548.
Collapse
Affiliation(s)
- Alina Lelic
- Department of Pathology and Molecular Medicine
| | - Chris P Verschoor
- Department of Pathology and Molecular Medicine.,Canadian Longitudinal Study on Aging
| | | | | | | | - Dawn M Bowdish
- Department of Pathology and Molecular Medicine.,Institute of Infectious Diseases Research
| | | | - Mark B Loeb
- Department of Pathology and Molecular Medicine.,Institute of Infectious Diseases Research.,Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Canada
| |
Collapse
|
12
|
Abu-Raya B, Smolen KK, Willems F, Kollmann TR, Marchant A. Transfer of Maternal Antimicrobial Immunity to HIV-Exposed Uninfected Newborns. Front Immunol 2016; 7:338. [PMID: 27630640 PMCID: PMC5005931 DOI: 10.3389/fimmu.2016.00338] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/22/2016] [Indexed: 11/13/2022] Open
Abstract
The transfer of maternal immune factors to the newborn is critical for protection from infectious disease in early life. Maternally acquired passive immunity provides protection until the infant is beyond early life's increased susceptibility to severe infections or until active immunity is achieved following infant's primary immunization. However, as reviewed here, human immunodeficiency virus (HIV) infection alters the transfer of immune factors from HIV-infected mothers to the HIV-exposed newborns and young infants. This may relate to the immune activation in HIV-infected pregnant women, associated with the production of inflammatory cytokines at the maternofetal interface associated with inflammatory responses in the newborn. We also summarize mother-targeting interventions to improve the health of infants born to HIV-infected women, such as immunization during pregnancy and reduction of maternal inflammation. Maternal immunization offers the potential to compensate for the decreased transplacentally transferred maternal antibodies observed in HIV-exposed infants. Current data suggest reduced immunogenicity of vaccines in HIV-infected pregnant women, possibly reducing the protective impact of maternal immunization for HIV-exposed infants. Fortunately, levels of antibodies appear preserved in the breast milk of HIV-infected women, which supports the recommendation to breast-feed during antiretroviral treatment to protect HIV-exposed infants.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia , Vancouver, BC , Canada
| | - Kinga K Smolen
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| | - Fabienne Willems
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| | - Tobias R Kollmann
- Department of Pediatrics, Division of Infectious Diseases, University of British Columbia , Vancouver, BC , Canada
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles , Charleroi , Belgium
| |
Collapse
|
13
|
CD4/CD8 Ratio Predicts Yellow Fever Vaccine-Induced Antibody Titers in Virologically Suppressed HIV-Infected Patients. J Acquir Immune Defic Syndr 2016; 71:189-95. [PMID: 26361176 DOI: 10.1097/qai.0000000000000845] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Yellow fever vaccine (YFV) induces weaker immune responses in HIV-infected individuals. However, little is known about YFV responses among antiretroviral-treated patients and potential immunological predictors of YFV response in this population. METHODS We enrolled 34 antiretroviral therapy (ART)-treated HIV-infected and 58 HIV-uninfected adults who received a single YFV dose to evaluate antibody levels and predictors of immunity, focusing on CD4(+) T-cell count, CD4(+)/CD8(+) ratio, and Human Pegivirus (GBV-C) viremia. Participants with other immunosuppressive conditions were excluded. RESULTS Median time since YFV was nonsignificantly shorter in HIV-infected participants than in HIV-uninfected participants (42 and 69 months, respectively, P = 0.16). Mean neutralizing antibody (NAb) titers was lower in HIV-infected participants than HIV-uninfected participants (3.3 vs. 3.6 log10mIU/mL, P = 0.044), a difference that remained significant after adjustment for age, sex, and time since vaccination (P = 0.024). In HIV-infected participants, lower NAb titers were associated with longer time since YFV (rho: -0.38, P = 0.027) and lower CD4(+)/CD8(+) ratio (rho: 0.42, P = 0.014), but not CD4(+) T-cell count (P = 0.52). None of these factors were associated with NAb titers in HIV-uninfected participant. GBV-C viremia was not associated with difference in NAb titers overall or among HIV-infected participants. CONCLUSIONS ART-treated HIV-infected individuals seem to have impaired and/or less durable responses to YFV than HIV-uninfected individuals, which were associated with lower CD4(+)/CD8(+) ratio, but not with CD4(+) T-cell count. These results supports the notion that low CD4(+)/CD8(+) ratio, a marker linked to persistent immune activation, is a better indicator of functional immune disturbance than CD4(+) T-cell count in patients with successful ART.
Collapse
|
14
|
Phadke VK, Omer SB. Maternal vaccination for the prevention of influenza: current status and hopes for the future. Expert Rev Vaccines 2016; 15:1255-80. [PMID: 27070268 DOI: 10.1080/14760584.2016.1175304] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Influenza is an important cause of morbidity and mortality among pregnant women and young infants, and influenza infection during pregnancy has also been associated with adverse obstetric and birth outcomes. There is substantial evidence - from randomized trials and observational studies - that maternal influenza immunization can protect pregnant women and their infants from influenza disease. In addition, there is compelling observational evidence that prevention of influenza in pregnant women can also protect against certain adverse pregnancy outcomes, including stillbirth and preterm birth. In this article we will review and evaluate the literature on both the burden of influenza disease in pregnant women and infants, as well as the multiple potential benefits of maternal influenza immunization for mother, fetus, and infant. We will also review key clinical aspects of maternal influenza immunization, as well as identify remaining knowledge gaps, and discuss avenues for future investigation.
Collapse
Affiliation(s)
- Varun K Phadke
- a Division of Infectious Diseases, School of Medicine , Emory University , Atlanta , GA , USA
| | - Saad B Omer
- b Department of Pediatrics, School of Medicine , Emory University , Atlanta , GA , USA.,c Departments of Global Health and Epidemiology, Rollins School of Public Health , Emory University , Atlanta , GA , USA.,d Emory Vaccine Center , Emory University , Atlanta , GA , USA
| |
Collapse
|
15
|
Nunes MC, Cutland CL, Dighero B, Bate J, Jones S, Hugo A, van Niekerk N, Kuwanda L, Izu A, Weinberg A, Madhi SA. Kinetics of Hemagglutination-Inhibiting Antibodies Following Maternal Influenza Vaccination Among Mothers With and Those Without HIV Infection and Their Infants. J Infect Dis 2015; 212:1976-87. [PMID: 26080370 DOI: 10.1093/infdis/jiv339] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/09/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND We evaluated the immunogenicity of trivalent inactivated influenza vaccine (IIV3) in pregnant women with and those without human immunodeficiency virus (HIV) infection and the persistence of hemagglutination-inhibiting antibodies in mothers and infants. METHODS Antibodies were measured before vaccination, 1 month after vaccination, at delivery, and at postpartum week 24 in mothers and within 1 week of birth and at 8, 16, and 24 weeks of age in infants. RESULTS We enrolled 98 HIV-uninfected and 100 HIV-infected pregnant women, including 93% with a CD4(+) T-cell count of ≥ 200 cells/µL. Compared with HIV-uninfected women, HIV-infected women had lower seroconversion rates (ranging from 63%-92% vs 36%-40%), lower antibody titers through postpartum week 24, and overlapping antibody half-lives (ranging from 106-121 vs 87-153 days). Infant titers were lower than the maternal titers within 1 week of delivery, regardless of vaccine strain and HIV exposure status. Compared with HIV-unexposed infants, HIV-exposed infants had a similar transplacental influenza virus antibody transfer ratio, lower titers, and a lower frequency of titers ≥ 1:40 (ranging from 82%-95% vs 43%-79%) at birth and higher antibody half-lives (ranging from 43-45 vs 56-65 days). CONCLUSIONS Compared with HIV-uninfected pregnant women, HIV-infected pregnant women had lower antibody responses and persistence. Compared with HIV-unexposed infants, HIV-exposed infants had lower antibody levels at birth but similar antibody levels after 8 weeks of life. Early IIV3 administration during pregnancy did not decrease antibody titers among infants at birth.
Collapse
Affiliation(s)
- Marta C Nunes
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand
| | - Clare L Cutland
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand
| | - Bonnie Dighero
- Department of Pediatrics, University of Colorado, Aurora Department of Medicine, University of Colorado, Aurora Department of Pathology, University of Colorado, Aurora
| | - Janie Bate
- Department of Pediatrics, University of Colorado, Aurora Department of Medicine, University of Colorado, Aurora Department of Pathology, University of Colorado, Aurora
| | - Stephanie Jones
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand
| | - Andrea Hugo
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand
| | - Nadia van Niekerk
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand
| | - Locadiah Kuwanda
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand
| | - Alane Izu
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand
| | - Adriana Weinberg
- Department of Pediatrics, University of Colorado, Aurora Department of Medicine, University of Colorado, Aurora Department of Pathology, University of Colorado, Aurora
| | - Shabir A Madhi
- Vaccine Preventable Diseases Unit, Department of Science and Technology, National Research Foundation Respiratory and Meningeal Pathogens Research Unit, Medical Research Council, University of the Witwatersrand National Institute for Communicable Diseases, National Health Laboratory Service, Centre for Vaccines and Immunology, Johannesburg, South Africa
| |
Collapse
|
16
|
Weinberg A, Muresan P, Richardson KM, Fenton T, Dominguez T, Bloom A, Watts DH, Abzug MJ, Nachman SA, Levin MJ. Determinants of vaccine immunogenicity in HIV-infected pregnant women: analysis of B and T cell responses to pandemic H1N1 monovalent vaccine. PLoS One 2015; 10:e0122431. [PMID: 25874544 PMCID: PMC4395240 DOI: 10.1371/journal.pone.0122431] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/11/2015] [Indexed: 11/19/2022] Open
Abstract
Influenza infections have high frequency and morbidity in HIV-infected pregnant women, underscoring the importance of vaccine-conferred protection. To identify the factors that determine vaccine immunogenicity in this group, we characterized the relationship of B- and T-cell responses to pandemic H1N1 (pH1N1) vaccine with HIV-associated immunologic and virologic characteristics. pH1N1 and seasonal-H1N1 (sH1N1) antibodies were measured in 119 HIV-infected pregnant women after two double-strength pH1N1 vaccine doses. pH1N1-IgG and IgA B-cell FluoroSpot, pH1N1- and sH1N1-interferon γ (IFNγ) and granzyme B (GrB) T-cell FluoroSpot, and flow cytometric characterization of B- and T-cell subsets were performed in 57 subjects. pH1N1-antibodies increased after vaccination, but less than previously described in healthy adults. pH1N1-IgG memory B cells (Bmem) increased, IFNγ-effector T-cells (Teff) decreased, and IgA Bmem and GrB Teff did not change. pH1N1-antibodies and Teff were significantly correlated with each other and with sH1N1-HAI and Teff, respectively, before and after vaccination. pH1N1-antibody responses to the vaccine significantly increased with high proportions of CD4+, low CD8+ and low CD8+HLADR+CD38+ activated (Tact) cells. pH1N1-IgG Bmem responses increased with high proportions of CD19+CD27+CD21- activated B cells (Bact), high CD8+CD39+ regulatory T cells (Treg), and low CD19+CD27-CD21- exhausted B cells (Bexhaust). IFNγ-Teff responses increased with low HIV plasma RNA, CD8+HLADR+CD38+ Tact, CD4+FoxP3+ Treg and CD19+IL10+ Breg. In conclusion, pre-existing antibody and Teff responses to sH1N1 were associated with increased responses to pH1N1 vaccination in HIV-infected pregnant women suggesting an important role for heterosubtypic immunologic memory. High CD4+% T cells were associated with increased, whereas high HIV replication, Tact and Bexhaust were associated with decreased vaccine immunogenicity. High Treg increased antibody responses but decreased Teff responses to the vaccine. The proportions of immature and transitional B cells did not affect the responses to vaccine. Increased Bact were associated with high Bmem responses to the vaccine.
Collapse
Affiliation(s)
- Adriana Weinberg
- University of Colorado Anschutz Medical Center, Aurora, Colorado, United States of America
- * E-mail:
| | - Petronella Muresan
- Statistical and Data Analysis Center, Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Kelly M. Richardson
- University of Colorado Anschutz Medical Center, Aurora, Colorado, United States of America
| | - Terence Fenton
- Statistical and Data Analysis Center, Center for Biostatistics in AIDS Research, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Teresa Dominguez
- University of Colorado Anschutz Medical Center, Aurora, Colorado, United States of America
| | - Anthony Bloom
- Frontier Science and Technology Research Foundation, Buffalo, New York, United States of America
| | - D. Heather Watts
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Mark J. Abzug
- University of Colorado Anschutz Medical Center, Aurora, Colorado, United States of America
| | - Sharon A. Nachman
- State University of New York Health Science Center at Stony Brook, Stony Brook, New York, United States of America
| | - Myron J. Levin
- University of Colorado Anschutz Medical Center, Aurora, Colorado, United States of America
| | | |
Collapse
|
17
|
Characterization of functional antibody and memory B-cell responses to pH1N1 monovalent vaccine in HIV-infected children and youth. PLoS One 2015; 10:e0118567. [PMID: 25785995 PMCID: PMC4364897 DOI: 10.1371/journal.pone.0118567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 01/20/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES We investigated immune determinants of antibody responses and B-cell memory to pH1N1 vaccine in HIV-infected children. METHODS Ninety subjects 4 to <25 years of age received two double doses of pH1N1 vaccine. Serum and cells were frozen at baseline, after each vaccination, and at 28 weeks post-immunization. Hemagglutination inhibition (HAI) titers, avidity indices (AI), B-cell subsets, and pH1N1 IgG and IgA antigen secreting cells (ASC) were measured at baseline and after each vaccination. Neutralizing antibodies and pH1N1-specific Th1, Th2 and Tfh cytokines were measured at baseline and post-dose 1. RESULTS At entry, 26 (29%) subjects had pH1N1 protective HAI titers (≥1:40). pH1N1-specific HAI, neutralizing titers, AI, IgG ASC, IL-2 and IL-4 increased in response to vaccination (p<0.05), but IgA ASC, IL-5, IL-13, IL-21, IFNγ and B-cell subsets did not change. Subjects with baseline HAI ≥1:40 had significantly greater increases in IgG ASC and AI after immunization compared with those with HAI <1:40. Neutralizing titers and AI after vaccination increased with older age. High pH1N1 HAI responses were associated with increased IgG ASC, IFNγ, IL-2, microneutralizion titers, and AI. Microneutralization titers after vaccination increased with high IgG ASC and IL-2 responses. IgG ASC also increased with high IFNγ responses. CD4% and viral load did not predict the immune responses post-vaccination, but the B-cell distribution did. Notably, vaccine immunogenicity increased with high CD19+CD21+CD27+% resting memory, high CD19+CD10+CD27+% immature activated, low CD19+CD21-CD27-CD20-% tissue-like, low CD19+CD21-CD27-CD20-% transitional and low CD19+CD38+HLADR+% activated B-cell subsets. CONCLUSIONS HIV-infected children on HAART mount a broad B-cell memory response to pH1N1 vaccine, which was higher for subjects with baseline HAI≥1:40 and increased with age, presumably due to prior exposure to pH1N1 or to other influenza vaccination/infection. The response to the vaccine was dependent on B-cell subset distribution, but not on CD4 counts or viral load. TRIAL REGISTRATION ClinicalTrials.gov NCT00992836.
Collapse
|
18
|
Abzug MJ, Nachman SA, Muresan P, Handelsman E, Watts DH, Fenton T, Heckman B, Petzold E, Weinberg A, Levin MJ. Safety and immunogenicity of 2009 pH1N1 vaccination in HIV-infected pregnant women. Clin Infect Dis 2013; 56:1488-97. [PMID: 23378284 DOI: 10.1093/cid/cit057] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Pregnant women infected with human immunodeficiency virus (HIV) may have particular vulnerability to 2009 pandemic H1N1 influenza (pH1N1) infection. The safety and immunogenicity of pH1N1 vaccination in HIV-infected pregnant women are unknown. METHODS HIV-infected women 18-39 years of age and 14-34 weeks' gestation on antiretroviral therapy received two 30-μg doses of unadjuvanted, inactivated pH1N1 vaccine 21 days apart. Hemagglutination inhibition titers were measured at entry, 21 days after dose 1, and 10 and 21 days after dose 2, and, in mothers and infants, at delivery and 3 and 6 months postdelivery. RESULTS No severe vaccine-related adverse events were observed among 127 subjects. At entry, 21% had seroprotective (≥1:40) titers. Seroprotection and seroresponse (≥4-fold rise) occurred in 73% and 66% after dose 1 and 80% and 72% after dose 2, respectively. Of women lacking seroprotection at entry, 66% attained seroprotection after dose 1 and 75% after dose 2. Seroprotective titers were present in 67% of mothers and 65% of infants at delivery (median 66 days after dose 2), 60% of mothers and 26% of infants at 3 months postdelivery, and 59% of mothers and 12% of infants at 6 months postdelivery. CONCLUSIONS Two 30-μg doses were moderately immunogenic in HIV-infected pregnant women. No concerning vaccine-related safety signals were observed. Seroprotection persisted in most women postpartum. Efficient transplacental antibody transfer occurred, but seroprotection in infants waned rapidly. Vaccination to protect HIV-infected pregnant women and their newborns from new influenza strains is feasible, but more immunogenic platforms should be evaluated. Clinical Trials Registration. NCT00992017.
Collapse
Affiliation(s)
- Mark J Abzug
- Department of Pediatrics, Infectious Diseases, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zheng R, Qin X, Li Y, Yu X, Wang J, Tan M, Yang Z, Li W. Imbalanced anti-H1N1 immunoglobulin subclasses and dysregulated cytokines in hospitalized pregnant women with 2009 H1N1 influenza and pneumonia in Shenyang, China. Hum Immunol 2012; 73:906-11. [PMID: 22750537 DOI: 10.1016/j.humimm.2012.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 06/06/2012] [Accepted: 06/13/2012] [Indexed: 10/28/2022]
Abstract
Influenza virus can cause an acute respiratory illness of variable intensity. In our study, we describe the clinical and immunological characteristics of pregnant and nonpregnant women who were hospitalized with 2009 H1N1 influenza and pneumonia in Shenyang, China, from November 2009 to January 2010. Forty-two female patient infected with H1N1 were divided into groups according to pregnancy. Clinical data were collected. Cytokines and anti-H1N1 IgG subclasses were detected. We observed significant lymphopenia, hypoproteinemia, reduced CD4(+)T cell counts and CD4(+)/CD8(+) ratios, reduced anti-H1N1 IgG subclasses IgG2 and IgG3 constituent ratios, elevated C reactive protein and interleukin-10 levels with regard to nonpregnant H1N1 group. Compared with the healthy pregnant group, the pregnant H1N1 group showed elevated aspartate aminotransferase and glutamic alanine aminotransferase levels, an increased interferon-gamma and interleukin-10 levels and reduced anti-H1N1 IgG subclasses IgG2, IgG3 and IgG4 combination ratios. There was a statistically significant association between imbalanced anti-H1N1 immunoglobulin subclasses and dysregulated cytokines in pregnant women with H1N1 infection.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Nahmias AJ, Schollin J, Abramowsky C. Evolutionary-developmental perspectives on immune system interactions among the pregnant woman, placenta, and fetus, and responses to sexually transmitted infectious agents. Ann N Y Acad Sci 2011; 1230:25-47. [PMID: 21824164 DOI: 10.1111/j.1749-6632.2011.06137.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A balance has evolved over deep time between the various immune systems of the "triad" that is linked together for a short period: the pregnant woman, the fetus, and the placenta. This balance is affected by, and helps to determine, the immune responses to maternal infectious agents that may be transmitted to the fetus/infant transplacentally, intrapartum, or via breast milk. This review identifies newer evolutionary concepts and processes related particularly to the human placenta, innate and adaptive immune systems involved in tolerance, and in responses to sexually transmitted infectious (STI) agents that may be pathogenic to the fetus/infant at different gestational periods and in the first year of life. An evolutionary-developmental (EVO-DEVO) perspective has been applied to the complexities within, and among, the different actors and their beneficial or deleterious outcomes. Such a phylogenetic and ontogenic approach has helped to stimulate several basic questions and suggested possible explanations and novel practical interventions.
Collapse
|