1
|
Makan-Murphy N, Madhi SA, Dangor Z. Safety, Efficacy, and Effectiveness of Maternal Vaccination against Respiratory Infections in Young Infants. Semin Respir Crit Care Med 2024. [PMID: 39708836 DOI: 10.1055/a-2471-6906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2024]
Abstract
Lower respiratory tract infection (LRTI) is a major cause of neonatal morbidity and mortality worldwide. Maternal vaccination is an effective strategy in protecting young infants from LRTI, particularly in the first few months after birth when infant is most vulnerable, and most primary childhood vaccinations have not been administered. Additionally, maternal vaccination protects the mother from illness during pregnancy and the postnatal period, and the developing fetus from adverse outcomes such as stillbirth and prematurity. In this paper, we review the safety, efficacy, and effectiveness of maternal vaccines against LRTIs, such as pertussis, influenza, coronavirus disease 2019, and respiratory syncytial virus.
Collapse
Affiliation(s)
- Nisha Makan-Murphy
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ziyaad Dangor
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
2
|
Vijayan K. K. V, De Paris K. Nonhuman primate models of pediatric viral diseases. Front Cell Infect Microbiol 2024; 14:1493885. [PMID: 39691699 PMCID: PMC11649651 DOI: 10.3389/fcimb.2024.1493885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious diseases are the leading cause of death in infants and children under 5 years of age. In utero exposure to viruses can lead to spontaneous abortion, preterm birth, congenital abnormalities or other developmental defects, often resulting in lifelong health sequalae. The underlying biological mechanisms are difficult to study in humans due to ethical concerns and limited sample access. Nonhuman primates (NHP) are closely related to humans, and pregnancy and immune ontogeny in infants are very similar to humans. Therefore, NHP are a highly relevant model for understanding fetal and postnatal virus-host interactions and to define immune mechanisms associated with increased morbidity and mortality in infants. We will discuss NHP models of viruses causing congenital infections, respiratory diseases in early life, and HIV. Cytomegalovirus (CMV) remains the most common cause of congenital defects worldwide. Measles is a vaccine-preventable disease, yet measles cases are resurging. Zika is an example of an emerging arbovirus with devastating consequences for the developing fetus and the surviving infant. Among the respiratory viruses, we will discuss influenza and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We will finish with HIV as an example of a lifelong infection without a cure or vaccine. The review will highlight (i) the impact of viral infections on fetal and infant immune development, (ii) how differences in infant and adult immune responses to infection alter disease outcome, and emphasize the invaluable contribution of pediatric NHP infection models to the design of effective treatment and prevention strategies, including vaccines, for human infants.
Collapse
Affiliation(s)
- Vidya Vijayan K. K.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Center for AIDS Research, University of North Carolina, Chapel Hill, NC, United States
- Children’s Research Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
3
|
Ayouni I, Mbangiwa T, Amponsah-Dacosta E, Noll S, Kagina BM, Muloiwa R. Acceptance and uptake of vaccines against tetanus, influenza, pertussis, and COVID-19 among pregnant and postpartum women in low- and middle-income countries: a systematic review and meta-analysis protocol. Syst Rev 2024; 13:227. [PMID: 39237969 PMCID: PMC11375862 DOI: 10.1186/s13643-024-02645-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 08/22/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Pregnant women, fetuses, and neonates are particularly vulnerable to vaccine-preventable diseases (VPDs). These VPDs are associated with high morbidity and mortality among expectant mothers and their fetuses and neonates. Vaccination during pregnancy can protect the expectant mother from VPDs to which she may be especially vulnerable while pregnant. In addition, the passive transfer of maternal neutralizing immunoglobulin G (IgG) and secretory immunoglobulin A (IgA) also protects the fetus against congenital infections and may further protect the neonate from infection during the first few months of life. Despite this, coverage of recommended maternal vaccines remains suboptimal globally, especially in resource-constrained settings. Determinants of vaccine acceptance and uptake are frequently understudied in low- and middle-income countries (LMICs) and among specific groups such as pregnant and postpartum women. This proposed systematic review will assess the acceptance and uptake of vaccines against tetanus, influenza, pertussis, and COVID-19 among pregnant and postpartum women in LMICs. METHODS A Boolean search strategy employing common and medical subject heading (MeSH) terms for tetanus, influenza, pertussis, and COVID-19 vaccines, as well as vaccine acceptance, hesitancy, together with uptake, pregnancy, and postpartum, will be used to search electronic databases for relevant literature published between 2009 and 2024. Only studies conducted in LMICs that investigated determinants of acceptance, hesitancy, and uptake of tetanus, influenza, pertussis, and COVID-19 vaccines among pregnant and postpartum women will be eligible for inclusion in the review. The quality and the risk of bias of all eligible full-text articles will be assessed using the Joanna Briggs Institute's (JBI) critical appraisal tools. DISCUSSION This protocol proposes a systematic review and meta-analysis that aims to assess the uptake of maternal vaccines and to systematically appraise and quantify determinants of the acceptance and uptake of recommended vaccines during pregnancy and postpartum in LMICs. A better understanding of these factors and how they influence maternal vaccine decision-making will enable public health practitioners as well as global and national policymakers to design more effective interventions as we look towards expanding the scope and reach of maternal immunization programs.
Collapse
Affiliation(s)
- Imen Ayouni
- Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa.
- Vaccines for Africa Initiative, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Tshepiso Mbangiwa
- Vaccines for Africa Initiative, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Edina Amponsah-Dacosta
- Vaccines for Africa Initiative, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Susanne Noll
- Vaccines for Africa Initiative, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Benjamin M Kagina
- Vaccines for Africa Initiative, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rudzani Muloiwa
- Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
- Vaccines for Africa Initiative, School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
4
|
Morrocchi E, van Haren S, Palma P, Levy O. Modeling human immune responses to vaccination in vitro. Trends Immunol 2024; 45:32-47. [PMID: 38135599 PMCID: PMC11688643 DOI: 10.1016/j.it.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
The human immune system is a complex network of coordinated components that are crucial for health and disease. Animal models, commonly used to study immunomodulatory agents, are limited by species-specific differences, low throughput, and ethical concerns. In contrast, in vitro modeling of human immune responses can enable species- and population-specific mechanistic studies and translational development within the same study participant. Translational accuracy of in vitro models is enhanced by accounting for genetic, epigenetic, and demographic features such as age, sex, and comorbidity. This review explores various human in vitro immune models, considers evidence that they may resemble human in vivo responses, and assesses their potential to accelerate and de-risk vaccine discovery and development.
Collapse
Affiliation(s)
- Elena Morrocchi
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Simon van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy.
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
5
|
Wimmers F, Burrell AR, Feng Y, Zheng H, Arunachalam PS, Hu M, Spranger S, Nyhoff LE, Joshi D, Trisal M, Awasthi M, Bellusci L, Ashraf U, Kowli S, Konvinse KC, Yang E, Blanco M, Pellegrini K, Tharp G, Hagan T, Chinthrajah RS, Nguyen TT, Grifoni A, Sette A, Nadeau KC, Haslam DB, Bosinger SE, Wrammert J, Maecker HT, Utz PJ, Wang TT, Khurana S, Khatri P, Staat MA, Pulendran B. Multi-omics analysis of mucosal and systemic immunity to SARS-CoV-2 after birth. Cell 2023; 186:4632-4651.e23. [PMID: 37776858 PMCID: PMC10724861 DOI: 10.1016/j.cell.2023.08.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/18/2023] [Accepted: 08/31/2023] [Indexed: 10/02/2023]
Abstract
The dynamics of immunity to infection in infants remain obscure. Here, we used a multi-omics approach to perform a longitudinal analysis of immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in infants and young children by analyzing blood samples and weekly nasal swabs collected before, during, and after infection with Omicron and non-Omicron variants. Infection stimulated robust antibody titers that, unlike in adults, showed no sign of decay for up to 300 days. Infants mounted a robust mucosal immune response characterized by inflammatory cytokines, interferon (IFN) α, and T helper (Th) 17 and neutrophil markers (interleukin [IL]-17, IL-8, and CXCL1). The immune response in blood was characterized by upregulation of activation markers on innate cells, no inflammatory cytokines, but several chemokines and IFNα. The latter correlated with viral load and expression of interferon-stimulated genes (ISGs) in myeloid cells measured by single-cell multi-omics. Together, these data provide a snapshot of immunity to infection during the initial weeks and months of life.
Collapse
Affiliation(s)
- Florian Wimmers
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, University of Tuebingen, 72076 Tuebingen, Baden-Wuerttemberg, Germany; DFG Cluster of Excellence 2180 "Image-guided and Functional Instructed Tumor Therapy" (iFIT), University of Tuebingen, 72076 Tuebingen, Baden-Wuerttemberg, Germany; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Baden-Wuerttemberg, Germany
| | - Allison R Burrell
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Environmental and Public Health Sciences, Division of Epidemiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - Hong Zheng
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - Sara Spranger
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lindsay E Nyhoff
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Devyani Joshi
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Meera Trisal
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - Mayanka Awasthi
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Lorenza Bellusci
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Usama Ashraf
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Sangeeta Kowli
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Katherine C Konvinse
- Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Emily Yang
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Blanco
- Stanford Genomics Service Center, Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Gregory Tharp
- Yerkes National Primate Research Center, Atlanta, GA 30024, USA
| | - Thomas Hagan
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - R Sharon Chinthrajah
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA 94305, USA
| | - Tran T Nguyen
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Kari C Nadeau
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA 94305, USA
| | - David B Haslam
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Steven E Bosinger
- Yerkes National Primate Research Center, Atlanta, GA 30024, USA; Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jens Wrammert
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taia T Wang
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mary A Staat
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
Baloh CH, Venturi GM, Fischer BM, Sadder LS, Kim-Chang JJ, Chan C, De Paris K, Yin L, Aldrovandi GM, Goodenow MM, Sleasman JW. Biomarkers detected in cord blood predict vaccine responses in young infants. Front Immunol 2023; 14:1152538. [PMID: 37251388 PMCID: PMC10213698 DOI: 10.3389/fimmu.2023.1152538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Factors influencing vaccine immune priming in the first year of life involve both innate and adaptive immunity but there are gaps in understanding how these factors sustain vaccine antibody levels in healthy infants. The hypothesis was that bioprofiles associated with B cell survival best predict sustained vaccine IgG levels at one year. Methods Longitudinal study of plasma bioprofiles in 82 term, healthy infants, who received standard recommended immunizations in the United States, with changes in 15 plasma biomarker concentrations and B cell subsets associated with germinal center development monitored at birth, soon after completion of the initial vaccine series at 6 months, and prior to the 12-month vaccinations. Post vaccination antibody IgG levels to Bordetella pertussis, tetanus toxoid, and conjugated Haemophilus influenzae type B (HiB) were outcome measures. Results Using a least absolute shrinkage and selection operator (lasso) regression model, cord blood (CB) plasma IL-2, IL-17A, IL-31, and soluble CD14 (sCD14) were positively associated with pertussis IgG levels at 12 months, while CB plasma concentrations of APRIL and IL-33 were negatively associated. In contrast, CB concentrations of sCD14 and APRIL were positively associated with sustained tetanus IgG levels. A separate cross-sectional analysis of 18 mother/newborn pairs indicated that CB biomarkers were not due to transplacental transfer, but rather due to immune activation at the fetal/maternal interface. Elevated percentages of cord blood switched memory B cells were positively associated with 12-month HiB IgG levels. BAFF concentrations at 6 and 12 months were positively associated with pertussis and HiB IgG levels respectively. Discussion Sustained B cell immunity is highly influenced by early life immune dynamics beginning prior to birth. The findings provide important insights into how germinal center development shapes vaccine responses in healthy infants and provide a foundation for studies of conditions that impair infant immune development.
Collapse
Affiliation(s)
- Carolyn H. Baloh
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Guglielmo M. Venturi
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Bernard M. Fischer
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Liane S. Sadder
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Julie J. Kim-Chang
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Kristina De Paris
- Institute of Global Health and Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Li Yin
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Grace M. Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, University of California, Los Angeles, CA, United States
| | - Maureen M. Goodenow
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - John W. Sleasman
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
7
|
Vono M, Mastelic-Gavillet B, Mohr E, Östensson M, Persson J, Olafsdottir TA, Lemeille S, Pejoski D, Hartley O, Christensen D, Andersen P, Didierlaurent AM, Harandi AM, Lambert PH, Siegrist CA. C-type lectin receptor agonists elicit functional IL21-expressing Tfh cells and induce primary B cell responses in neonates. Front Immunol 2023; 14:1155200. [PMID: 37063899 PMCID: PMC10102809 DOI: 10.3389/fimmu.2023.1155200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
IntroductionC-type lectin receptor (CLR) agonists emerged as superior inducers of primary B cell responses in early life compared with Toll-like receptor (TLR) agonists, while both types of adjuvants are potent in adults.MethodsHere, we explored the mechanisms accounting for the differences in neonatal adjuvanticity between a CLR-based (CAF®01) and a TLR4-based (GLA-SE) adjuvant administered with influenza hemagglutinin (HA) in neonatal mice, by using transcriptomics and systems biology analyses.ResultsOn day 7 after immunization, HA/CAF01 increased IL6 and IL21 levels in the draining lymph nodes, while HA/GLA-SE increased IL10. CAF01 induced mixed Th1/Th17 neonatal responses while T cell responses induced by GLA-SE had a more pronounced Th2-profile. Only CAF01 induced T follicular helper (Tfh) cells expressing high levels of IL21 similar to levels induced in adult mice, which is essential for germinal center (GC) formation. Accordingly, only CAF01- induced neonatal Tfh cells activated adoptively transferred hen egg lysozyme (HEL)-specific B cells to form HEL+ GC B cells in neonatal mice upon vaccination with HEL-OVA.DiscussionCollectively, the data show that CLR-based adjuvants are promising neonatal and infant adjuvants due to their ability to harness Tfh responses in early life.
Collapse
Affiliation(s)
- Maria Vono
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- *Correspondence: Maria Vono,
| | - Beatris Mastelic-Gavillet
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Elodie Mohr
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Malin Östensson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Josefine Persson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | | | - Sylvain Lemeille
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - David Pejoski
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Oliver Hartley
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Dennis Christensen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Arnaud M. Didierlaurent
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Ali M. Harandi
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Center, British Columbia (BC) Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Paul-Henri Lambert
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Center for Vaccine Immunology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
8
|
Wimmers F, Burrell AR, Feng Y, Zheng H, Arunachalam PS, Hu M, Spranger S, Nyhoff L, Joshi D, Trisal M, Awasthi M, Bellusci L, Ashraf U, Kowli S, Konvinse KC, Yang E, Blanco M, Pellegrini K, Tharp G, Hagan T, Chinthrajah RS, Grifoni A, Sette A, Nadeau KC, Haslam DB, Bosinger SE, Wrammert J, Maecker HT, Utz PJ, Wang TT, Khurana S, Khatri P, Staat MA, Pulendran B. Systems biological assessment of the temporal dynamics of immunity to a viral infection in the first weeks and months of life. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.28.23285133. [PMID: 36778389 PMCID: PMC9915811 DOI: 10.1101/2023.01.28.23285133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The dynamics of innate and adaptive immunity to infection in infants remain obscure. Here, we used a multi-omics approach to perform a longitudinal analysis of immunity to SARS-CoV-2 infection in infants and young children in the first weeks and months of life by analyzing blood samples collected before, during, and after infection with Omicron and Non-Omicron variants. Infection stimulated robust antibody titers that, unlike in adults, were stably maintained for >300 days. Antigen-specific memory B cell (MCB) responses were durable for 150 days but waned thereafter. Somatic hypermutation of V-genes in MCB accumulated progressively over 9 months. The innate response was characterized by upregulation of activation markers on blood innate cells, and a plasma cytokine profile distinct from that seen in adults, with no inflammatory cytokines, but an early and transient accumulation of chemokines (CXCL10, IL8, IL-18R1, CSF-1, CX3CL1), and type I IFN. The latter was strongly correlated with viral load, and expression of interferon-stimulated genes (ISGs) in myeloid cells measured by single-cell transcriptomics. Consistent with this, single-cell ATAC-seq revealed enhanced accessibility of chromatic loci targeted by interferon regulatory factors (IRFs) and reduced accessibility of AP-1 targeted loci, as well as traces of epigenetic imprinting in monocytes, during convalescence. Together, these data provide the first snapshot of immunity to infection during the initial weeks and months of life.
Collapse
Affiliation(s)
- Florian Wimmers
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Molecular Medicine, Interfaculty Institute for Biochemistry, University of Tuebingen, Tuebingen, Germany
- DFG Cluster of Excellence 2180 ‘Image-guided and Functional Instructed Tumor Therapy’ (iFIT), University of Tuebingen, Tuebingen, Germany
- German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Allison R. Burrell
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Environmental and Public Health Sciences, Division of Epidemiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Hong Zheng
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Prabhu S. Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Sara Spranger
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Lindsay Nyhoff
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine
| | - Devyani Joshi
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine
| | - Meera Trisal
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Mayanka Awasthi
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Lorenza Bellusci
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Usama Ashraf
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Sangeeta Kowli
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Katherine C. Konvinse
- Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Emily Yang
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Blanco
- Stanford Genomics Service Center, Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Gregory Tharp
- Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Thomas Hagan
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - R. Sharon Chinthrajah
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA 94305, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Kari C. Nadeau
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA 94305, USA
| | - David B. Haslam
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Steven E. Bosinger
- Yerkes National Primate Research Center, Atlanta, GA, USA
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jens Wrammert
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine
| | - Holden T. Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Paul J. Utz
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Taia T. Wang
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Mary A. Staat
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
9
|
Conti MG, Terreri S, Terrin G, Natale F, Pietrasanta C, Salvatori G, Brunelli R, Midulla F, Papaevangelou V, Carsetti R, Angelidou A. Severe Acute Respiratory Syndrome Coronavirus 2 Infection Versus Vaccination in Pregnancy: Implications for Maternal and Infant Immunity. Clin Infect Dis 2022; 75:S37-S45. [PMID: 35535796 PMCID: PMC9129222 DOI: 10.1093/cid/ciac359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/22/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been associated with adverse maternal and neonatal outcomes, yet uptake of SARS-CoV-2 vaccines during pregnancy and lactation has been slow. As a result, millions of pregnant and lactating women and their infants remain susceptible to the virus. METHODS We measured spike-specific immunoglobulin G (anti-S IgG) and immunoglobulin A (anti-S IgA) in serum and breastmilk (BM) samples from 3 prospective mother-infant cohorts recruited in 2 academic medical centers. The primary aim was to determine the impact of maternal SARS-CoV-2 immunization vs infection and their timing on systemic and mucosal immunity. RESULTS The study included 28 mothers infected with SARS-CoV-2 in late pregnancy (INF), 11 uninfected mothers who received 2 doses of the BNT162b2 vaccine in the latter half of pregnancy (VAX-P), and 12 uninfected mothers who received 2 doses of BNT162b2 during lactation. VAX dyads had significantly higher serum anti-S IgG compared to INF dyads (P < .0001), whereas INF mothers had higher BM:serum anti-S IgA ratios compared to VAX mothers (P = .0001). Median IgG placental transfer ratios were significantly higher in VAX-P compared to INF mothers (P < .0001). There was a significant positive correlation between maternal and neonatal serum anti-S IgG after vaccination (r = 0.68, P = .013), but not infection. CONCLUSIONS BNT161b2 vaccination in late pregnancy or lactation enhances systemic immunity through serum anti-S immunoglobulin, while SARS-CoV-2 infection induces mucosal over systemic immunity more efficiently through BM immunoglobulin production. Next-generation vaccines boosting mucosal immunity could provide additional protection to the mother-infant dyad. Future studies should focus on identifying the optimal timing of primary and/or booster maternal vaccination for maximal benefit.
Collapse
Affiliation(s)
- Maria Giulia Conti
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Sara Terreri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianluca Terrin
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Fabio Natale
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Carlo Pietrasanta
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Roberto Brunelli
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Fabio Midulla
- Department of Maternal and Child Health, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Vassiliki Papaevangelou
- Third Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Rita Carsetti
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy,Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children's Hospital, IRCCS; Piazza Sant’Onofrio, 4, 00165, Rome, Italy
| | - Asimenia Angelidou
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, United States,Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Harvard Medical School,Corresponding author: Asimenia Angelidou, MD PhD Instructor in Pediatrics, Harvard Medical School 4 Blackfan Circle, HIM Building, Rm 836, Boston MA 02115
| |
Collapse
|
10
|
Diray-Arce J, Angelidou A, Jensen KJ, Conti MG, Kelly RS, Pettengill MA, Liu M, van Haren SD, McCulloch SD, Michelloti G, Idoko O, Kollmann TR, Kampmann B, Steen H, Ozonoff A, Lasky-Su J, Benn CS, Levy O. Bacille Calmette-Guérin vaccine reprograms human neonatal lipid metabolism in vivo and in vitro. Cell Rep 2022; 39:110772. [PMID: 35508141 PMCID: PMC9157458 DOI: 10.1016/j.celrep.2022.110772] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/26/2021] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
Vaccines have generally been developed with limited insight into their molecular impact. While systems vaccinology enables characterization of mechanisms of action, these tools have yet to be applied to infants, who are at high risk of infection and receive the most vaccines. Bacille Calmette-Guérin (BCG) protects infants against disseminated tuberculosis (TB) and TB-unrelated infections via incompletely understood mechanisms. We employ mass-spectrometry-based metabolomics of blood plasma to profile BCG-induced infant responses in Guinea-Bissau in vivo and the US in vitro. BCG-induced lysophosphatidylcholines (LPCs) correlate with both TLR-agonist- and purified protein derivative (PPD, mycobacterial antigen)-induced blood cytokine production in vitro, raising the possibility that LPCs contribute to BCG immunogenicity. Analysis of an independent newborn cohort from The Gambia demonstrates shared vaccine-induced metabolites, such as phospholipids and sphingolipids. BCG-induced changes to the plasma lipidome and LPCs may contribute to its immunogenicity and inform the development of early life vaccines.
Collapse
Affiliation(s)
- Joann Diray-Arce
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Asimenia Angelidou
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Kristoffer Jarlov Jensen
- Research Center for Vitamins and Vaccines (CVIVA), Bandim Health Project, University of Southern Denmark, 2300 Copenhagen, Denmark; Bandim Health Project, Department of Clinical Research, University of Southern Denmark, 1455 Copenhagen K, Denmark; Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark, 2800 Kgs, Lyngby, Denmark
| | - Maria Giulia Conti
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Maternal and Child Health, Sapienza University of Rome, 00185 Rome, Italy
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Matthew A Pettengill
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark Liu
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Simon D van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Olubukola Idoko
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Tobias R Kollmann
- Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Hanno Steen
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Christine S Benn
- Research Center for Vitamins and Vaccines (CVIVA), Bandim Health Project, University of Southern Denmark, 2300 Copenhagen, Denmark; Bandim Health Project, Department of Clinical Research, University of Southern Denmark, 1455 Copenhagen K, Denmark; Danish Institute for Advanced Study, University of Southern Denmark, 5230 Odense M, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
11
|
Pulendran B, S Arunachalam P, O'Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov 2021; 20:454-475. [PMID: 33824489 PMCID: PMC8023785 DOI: 10.1038/s41573-021-00163-y] [Citation(s) in RCA: 754] [Impact Index Per Article: 188.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Adjuvants are vaccine components that enhance the magnitude, breadth and durability of the immune response. Following its introduction in the 1920s, alum remained the only adjuvant licensed for human use for the next 70 years. Since the 1990s, a further five adjuvants have been included in licensed vaccines, but the molecular mechanisms by which these adjuvants work remain only partially understood. However, a revolution in our understanding of the activation of the innate immune system through pattern recognition receptors (PRRs) is improving the mechanistic understanding of adjuvants, and recent conceptual advances highlight the notion that tissue damage, different forms of cell death, and metabolic and nutrient sensors can all modulate the innate immune system to activate adaptive immunity. Furthermore, recent advances in the use of systems biology to probe the molecular networks driving immune response to vaccines ('systems vaccinology') are revealing mechanistic insights and providing a new paradigm for the vaccine discovery and development process. Here, we review the 'known knowns' and 'known unknowns' of adjuvants, discuss these emerging concepts and highlight how our expanding knowledge about innate immunity and systems vaccinology are revitalizing the science and development of novel adjuvants for use in vaccines against COVID-19 and future pandemics.
Collapse
Affiliation(s)
- Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
- Chemistry, Engineering & Medicine for Human Health, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
12
|
Andrianov AK, Langer R. Polyphosphazene immunoadjuvants: Historical perspective and recent advances. J Control Release 2021; 329:299-315. [PMID: 33285104 PMCID: PMC7904599 DOI: 10.1016/j.jconrel.2020.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
The development of successful vaccines has been increasingly reliant on the use of immunoadjuvants - additives, which can enhance and modulate immune responses to vaccine antigens. Immunoadjuvants of the polyphosphazene family encompass synthetic biodegradable macromolecules, which attain in vivo activity via antigen delivery and immunostimulation mechanisms. Over the last decades, the technology has witnessed evolvement of next generation members, expansion to include various antigens and routes of administration, and progression to clinical phase. This was accompanied by gaining important insights into the mechanism of action and the development of a novel class of virus-mimicking nano-assemblies for antigen delivery. The present review evaluates in vitro and in vivo data generated to date in the context of latest advances in understanding the primary function and biophysical behavior of these macromolecules. It also provides an overview of relevant synthetic and characterization methods, macromolecular biodegradation pathways, and polyphosphazene-based multi-component, nanoparticulate, and microfabricated formulations.
Collapse
Affiliation(s)
- Alexander K Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA.
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
13
|
Soni D, Bobbala S, Li S, Scott EA, Dowling DJ. The sixth revolution in pediatric vaccinology: immunoengineering and delivery systems. Pediatr Res 2021; 89:1364-1372. [PMID: 32927471 PMCID: PMC7511675 DOI: 10.1038/s41390-020-01112-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 05/08/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
Abstract
Infection is the predominant cause of mortality in early life, and immunization is the most promising biomedical intervention to reduce this burden. However, very young infants fail to respond optimally to most vaccines currently in use, especially neonates. In 2005, Stanley Plotkin proposed that new delivery systems would spur a new revolution in pediatric vaccinology, just as attenuation, inactivation, cell culture of viruses, genetic engineering, and adjuvantation had done in preceding decades. Recent advances in the field of immunoengineering, which is evolving alongside vaccinology, have begun to increasingly influence vaccine formulation design. Historically, the particulate nature of materials used in many vaccine formulations was empiric, often because of the need to stabilize antigens or reduce endotoxin levels. However, present vaccine delivery systems are rationally engineered to mimic the size, shape, and surface chemistry of pathogens, and are therefore often referred to as "pathogen-like particles". More than a decade from his original assessment, we re-assess Plotkin's prediction. In addition, we highlight how immunoengineering and advanced delivery systems may be uniquely capable of enhancing vaccine responses in vulnerable populations, such as infants. IMPACT: Immunoengineering and advanced delivery systems are leading to new developments in pediatric vaccinology. Summarizes delivery systems currently in use and development, and prospects for the future. Broad overview of immunoengineering's impact on vaccinology, catering to Pediatric Clinicians and Immunologists.
Collapse
Affiliation(s)
- Dheeraj Soni
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Sharan Bobbala
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - Sophia Li
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - Evan A. Scott
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - David J. Dowling
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| |
Collapse
|
14
|
Kenney LL, Carter EP, Gil A, Selin LK. T cells in the brain enhance neonatal mortality during peripheral LCMV infection. PLoS Pathog 2021; 17:e1009066. [PMID: 33400715 PMCID: PMC7785120 DOI: 10.1371/journal.ppat.1009066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/14/2020] [Indexed: 11/18/2022] Open
Abstract
In adult mice the severity of disease from viral infections is determined by the balance between the efficiency of the immune response and the magnitude of viral load. Here, the impact of this dynamic is examined in neonates. Newborns are highly susceptible to infections due to poor innate responses, lower numbers of T cells and Th2-prone immune responses. Eighty-percent of 7-day old mice, immunologically equivalent to human neonates, succumbed to extremely low doses (5 PFU) of the essentially non-lethal lymphocytic choriomeningitis virus (LCMV-Armstrong) given intraperitoneally. This increased lethality was determined to be dependent upon poor early viral control, as well as, T cells and perforin as assessed in knockout mice. By day 3, these neonatal mice had 400-fold higher viral loads as compared to adults receiving a 10,000-fold (5X104 PFU) higher dose of LCMV. The high viral load in combination with the subsequent immunological defect of partial CD8 T cell clonal exhaustion in the periphery led to viral entry and replication in the brain. Within the brain, CD8 T cells were protected from exhaustion, and thus were able to mediate lethal immunopathology. To further delineate the role of early viral control, neonatal mice were infected with Pichinde virus, a less virulent arenavirus, or LCMV was given to pups of LCMV-immune mothers. In both cases, peak viral load was at least 29-fold lower, leading to functional CD8 T cell responses and 100% survival.
Collapse
Affiliation(s)
- Laurie L. Kenney
- University of Massachusetts Medical School, Department of Pathology, Worcester, Massachusetts, United States of America
| | - Erik P. Carter
- University of Massachusetts Medical School, Department of Pathology, Worcester, Massachusetts, United States of America
| | - Anna Gil
- University of Massachusetts Medical School, Department of Pathology, Worcester, Massachusetts, United States of America
| | - Liisa K. Selin
- University of Massachusetts Medical School, Department of Pathology, Worcester, Massachusetts, United States of America
| |
Collapse
|
15
|
Beijnen EMS, van Haren SD. Vaccine-Induced CD8 + T Cell Responses in Children: A Review of Age-Specific Molecular Determinants Contributing to Antigen Cross-Presentation. Front Immunol 2020; 11:607977. [PMID: 33424857 PMCID: PMC7786054 DOI: 10.3389/fimmu.2020.607977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Infections are most common and most severe at the extremes of age, the young and the elderly. Vaccination can be a key approach to enhance immunogenicity and protection against pathogens in these vulnerable populations, who have a functionally distinct immune system compared to other age groups. More than 50% of the vaccine market is for pediatric use, yet to date vaccine development is often empiric and not tailored to molecular distinctions in innate and adaptive immune activation in early life. With modern vaccine development shifting from whole-cell based vaccines to subunit vaccines also comes the need for formulations that can elicit a CD8+ T cell response when needed, for example, by promoting antigen cross-presentation. While our group and others have identified many cellular and molecular determinants of successful activation of antigen-presenting cells, B cells and CD4+ T cells in early life, much less is known about the ontogeny of CD8+ T cell induction. In this review, we summarize the literature pertaining to the frequency and phenotype of newborn and infant CD8+ T cells, and any evidence of induction of CD8+ T cells by currently licensed pediatric vaccine formulations. In addition, we review the molecular determinants of antigen cross-presentation on MHC I and successful CD8+ T cell induction and discuss potential distinctions that can be made in children. Finally, we discuss recent advances in development of novel adjuvants and provide future directions for basic and translational research in this area.
Collapse
Affiliation(s)
- Elisabeth M. S. Beijnen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Simon D. van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Abstract
Newborns, especially those born preterm, are at high risk for infection. Preterm birth rates appear to be increasing in most countries, with ∼15 million infants born preterm globally each year, corresponding to ∼11% of all deliveries. Importantly, the vulnerability of preterm infants to infection continues beyond the perinatal period, following them throughout childhood and adolescence, highlighting the long-lasting effects of infection on overall health and well-being. Other than access to clean drinking water and proper sewage systems, immunization is the most effective biomedical intervention to reduce early life infection. Nevertheless, a significant proportion of infants discharged on or after 2 months of age from the NICU remains unimmunized or underimmunized at that time. Despite being safe and effective, protective responses to immunization in early life are different from those in older individuals, in part because of the distinct immune system of newborns and young infants. The paradigms of the Bacille Calmette-Guérin, hepatitis B, and polio vaccines, the only immunizations currently routinely administered in the neonatal period, provide evidence that it is feasible to successfully administer vaccines via different routes of delivery; thus, production of sufficient vaccine-induced immunity leads to disease prevention in the newborn. Strategies such as maternal immunization, adjuvantation systems, leveraging trained immunity, and counseling caregivers can be used to enhance vaccine-induced specific and heterologous protection from infection and boost adherence to the recommended immunization schedule.
Collapse
Affiliation(s)
- Asimenia Angelidou
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA.,Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA.,Broad Institute of MIT & Harvard, Cambridge, MA
| |
Collapse
|
17
|
Sanchez-Schmitz G, Morrocchi E, Cooney M, Soni D, Khatun R, Palma P, Dowling DJ, Levy O. Neonatal monocytes demonstrate impaired homeostatic extravasation into a microphysiological human vascular model. Sci Rep 2020; 10:17836. [PMID: 33082466 PMCID: PMC7576166 DOI: 10.1038/s41598-020-74639-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022] Open
Abstract
Infections are most frequent at the extremes of life, especially among newborns, reflecting age-specific differences in immunity. Monocytes maintain tissue-homeostasis and defence-readiness by escaping circulation in the absence of inflammation to become tissue-resident antigen presenting cells in vivo. Despite equivalent circulating levels, neonates demonstrate lower presence of monocytes inside peripheral tissues as compared to adults. To study the ability of monocytes to undergo autonomous transendothelial extravasation under biologically accurate circumstances we engineered a three-dimensional human vascular-interstitial model including collagen, fibronectin, primary endothelial cells and autologous untreated plasma. This microphysiological tissue construct enabled age-specific autonomous extravasation of monocytes through a confluent human endothelium in the absence of exogenous chemokines and activation. Both CD16- and CD16+ newborn monocytes demonstrated lower adherence and extravasation as compared to adults. In contrast, pre-activated tissue constructs were colonized by newborn monocytes at the same frequency than adult monocytes, suggesting that neonatal monocytes are capable of colonizing inflamed tissues. The presence of autologous plasma neither improved newborn homeostatic extravasation nor shaped age-specific differences in endothelial cytokines that could account for this impairment. Newborn monocytes demonstrated significantly lower surface expression of CD31 and CD11b, and mechanistic experiments using blocking antibodies confirmed a functional role for CD31 and CD54 in neonatal homeostatic extravasation. Our data suggests that newborn monocytes are intrinsically impaired in extravasation through quiescent endothelia, a phenomenon that could contribute to the divergent immune responsiveness to vaccines and susceptibility to infection observed during early life.
Collapse
Affiliation(s)
- Guzman Sanchez-Schmitz
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA.
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Harvard University, Boston, MA, USA.
| | - Elena Morrocchi
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Academic Department of Paediatrics (DPUO), Research Unit of Congenital and Perinatal Infections, Children's Hospital Bambino Gesù, Rome, Italy
| | - Mitchell Cooney
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Dheeraj Soni
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Rahima Khatun
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Paolo Palma
- Academic Department of Paediatrics (DPUO), Research Unit of Congenital and Perinatal Infections, Children's Hospital Bambino Gesù, Rome, Italy
- Chair of Paediatrics, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - David J Dowling
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Ofer Levy
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| |
Collapse
|
18
|
Ramos L, Lunney JK, Gonzalez-Juarrero M. Neonatal and infant immunity for tuberculosis vaccine development: importance of age-matched animal models. Dis Model Mech 2020; 13:dmm045740. [PMID: 32988990 PMCID: PMC7520460 DOI: 10.1242/dmm.045740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neonatal and infant immunity differs from that of adults in both the innate and adaptive arms, which are critical contributors to immune-mediated clearance of infection and memory responses elicited during vaccination. The tuberculosis (TB) research community has openly admitted to a vacuum of knowledge about neonatal and infant immune responses to Mycobacterium tuberculosis (Mtb) infection, especially in the functional and phenotypic attributes of memory T cell responses elicited by the only available vaccine for TB, the Bacillus Calmette-Guérin (BCG) vaccine. Although BCG vaccination has variable efficacy in preventing pulmonary TB during adolescence and adulthood, 80% of endemic TB countries still administer BCG at birth because it has a good safety profile and protects children from severe forms of TB. As such, new vaccines must work in conjunction with BCG at birth and, thus, it is essential to understand how BCG shapes the immune system during the first months of life. However, many aspects of the neonatal and infant immune response elicited by vaccination with BCG remain unknown, as only a handful of studies have followed BCG responses in infants. Furthermore, most animal models currently used to study TB vaccine candidates rely on adult-aged animals. This presents unique challenges when transitioning to human trials in neonates or infants. In this Review, we focus on vaccine development in the field of TB and compare the relative utility of animal models used thus far to study neonatal and infant immunity. We encourage the development of neonatal animal models for TB, especially the use of pigs.
Collapse
Affiliation(s)
- Laylaa Ramos
- Mycobacteria Research Laboratories, Microbiology Immunology and Pathology Department, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA Building 1040, Room 103, Beltsville, MD 20705, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Microbiology Immunology and Pathology Department, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| |
Collapse
|
19
|
Willis E, Pardi N, Parkhouse K, Mui BL, Tam YK, Weissman D, Hensley SE. Nucleoside-modified mRNA vaccination partially overcomes maternal antibody inhibition of de novo immune responses in mice. Sci Transl Med 2020; 12:eaav5701. [PMID: 31915303 PMCID: PMC7339908 DOI: 10.1126/scitranslmed.aav5701] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 06/21/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022]
Abstract
Maternal antibodies provide short-term protection to infants against many infections. However, they can inhibit de novo antibody responses in infants elicited by infections or vaccination, leading to increased long-term susceptibility to infectious diseases. Thus, there is a need to develop vaccines that are able to elicit protective immune responses in the presence of antigen-specific maternal antibodies. Here, we used a mouse model to demonstrate that influenza virus-specific maternal antibodies inhibited de novo antibody responses in mouse pups elicited by influenza virus infection or administration of conventional influenza vaccines. We found that a recently developed influenza vaccine, nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP), partially overcame this inhibition by maternal antibodies. The mRNA-LNP influenza vaccine established long-lived germinal centers in the mouse pups and elicited stronger antibody responses than did a conventional influenza vaccine approved for use in humans. Vaccination with mRNA-LNP vaccines may offer a promising strategy for generating robust immune responses in infants in the presence of maternal antibodies.
Collapse
Affiliation(s)
- Elinor Willis
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kaela Parkhouse
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Okala SG, Darboe MK, Sosseh F, Sonko B, Faye-Joof T, Prentice AM, Moore SE. Impact of nutritional supplementation during pregnancy on antibody responses to diphtheria-tetanus-pertussis vaccination in infants: A randomised trial in The Gambia. PLoS Med 2019; 16:e1002854. [PMID: 31386660 PMCID: PMC6684039 DOI: 10.1371/journal.pmed.1002854] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/27/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Exposure to a nutritionally deficient environment during fetal life and early infancy may adversely alter the ontogeny of the immune system and affect an infant's ability to mount an optimal immune response to vaccination. We examined the effects of maternal nutritional supplementation during pregnancy on infants' antibody responses to the diphtheria-tetanus-pertussis (DTP) vaccine included in the Expanded Programme on Immunisation (EPI). METHODS AND FINDINGS The Early Nutrition and Immune Development (ENID) trial was a randomised, partially blinded trial conducted between April 2010 and February 2015 in the rural West Kiang region of The Gambia, a resource-poor region affected by chronic undernutrition. Pregnant women (<20 weeks' gestation) with a singleton pregnancy (n = 875) were randomised to receive one of four supplements: iron-folic acid (FeFol; standard of care), multiple micronutrient (MMN), protein-energy (PE), or PE + MMN daily from enrolment (mean [SD] 13.7 [3.3] weeks' gestation) until delivery. Infants were administered the DTP vaccine at 8, 12, and 16 weeks of age according to the Gambian Government protocol. Results for the primary outcome of the trial (infant thymic size) were described previously; here, we report on a secondary outcome, infant antibody response to vaccination. The effects of supplementation on mean DTP antibody titres measured in blood samples collected from infants at 12 weeks (n = 710) and 24 weeks (n = 662) were analysed with adjustment for confounders including maternal age, compliance to supplement, and infant sex and season. At 12 weeks, following a single dose of the vaccine, compared with FeFol (mean 95% confidence interval [CI]; 0.11 IU/mL, 0.09-0.12), antenatal supplementation with MMN or MMN + PE resulted in 42.4% (95% CI 20.1-64.6; p < 0.001) and 29.4% (6.4-52.5; p = 0.012) higher mean anti-diphtheria titres, respectively. Mean anti-tetanus titres were higher by 9.0% (5.5-12.5), 7.8% (4.3-11.4), and 7.3% (4.0-10.7) in MMN, PE, and PE + MMN groups (all, p < 0.001), respectively, than in the FeFol group (0.55 IU/mL, 0.52-0.58). Mean anti-pertussis titres were not significantly different in the FeFol, MMN, and PE + MNN groups but were all higher than in the PE group (all, p < 0.001). At 24 weeks, following all three doses, no significant differences in mean anti-diphtheria titres were detected across the supplement groups. Mean anti-tetanus titres were 3.4% (0.19-6.5; p = 0.038) higher in the PE + MMN group than in the FeFol group (3.47 IU/mL, 3.29-3.66). Mean anti-pertussis titres were higher by 9.4% (3.3-15.5; p = 0.004) and 15.4% (9.6-21.2; p < 0.001) in PE and PE + MMN groups, compared with the FeFol group (74.9 IU/mL, 67.8-82.8). Limitations of the study included the lack of maternal antibody status (breast milk or plasma) or prevaccination antibody measurements in the infants. CONCLUSION According to our results from rural Gambia, maternal supplementation with MMN combined with PE during pregnancy enhanced antibody responses to the DTP vaccine in early infancy. Provision of nutritional supplements to pregnant women in food insecure settings may improve infant immune development and responses to EPI vaccines. TRIAL REGISTRATION ISRCTN49285450.
Collapse
Affiliation(s)
- Sandra G. Okala
- Kings’ College London, Department of Women and Children’s Health, St Thomas’ Hospital, London, United Kingdom
| | - Momodou K. Darboe
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Fatou Sosseh
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Bakary Sonko
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Tisbeh Faye-Joof
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Andrew M. Prentice
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Sophie E. Moore
- Kings’ College London, Department of Women and Children’s Health, St Thomas’ Hospital, London, United Kingdom
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
- * E-mail:
| |
Collapse
|
21
|
Scavone C, di Mauro G, Mascolo A, Berrino L, Rossi F, Capuano A. The New Paradigms in Clinical Research: From Early Access Programs to the Novel Therapeutic Approaches for Unmet Medical Needs. Front Pharmacol 2019; 10:111. [PMID: 30814951 PMCID: PMC6381027 DOI: 10.3389/fphar.2019.00111] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/28/2019] [Indexed: 12/17/2022] Open
Abstract
Despite several innovative medicines gaining worldwide approval in recent years, there are still therapeutic areas for which unsatisfied therapeutic needs persist. For example, high unmet clinical need was observed in patients diagnosed with type 2 diabetes mellitus and hemophilia, as well as in specific age groups, such as the pediatric population. Given the urgent need to improve the therapy of clinical conditions for which unmet clinical need is established, clinical testing, and approval of new medicines are increasingly being carried out through accelerated authorization procedures. Starting from 1992, the Food and Drug Administration and the European Medicines Agency have supported the so-called Early Access Programs (EAPs). Such procedures, which can be based on incomplete clinical data, allow an accelerated marketing authorization for innovative medicines. The growth in pharmaceutical research has also resulted in the development of novel therapeutic approaches, such as biotech drugs and advanced therapy medicinal products, including new monoclonal antibodies for the treatment of asthma, antisense oligonucleotides for the treatment of Duchenne muscular dystrophy and spinal muscular atrophy, and new anticancer drugs that act on genetic biomarkers rather than any specific type of cancer. Even though EAPs and novel therapeutic approaches have brought huge benefits for public health, their implementation is limited by several challenges, including the high risk of safety-related label changes for medicines authorized through the accelerated procedure, the high costs, and the reimbursement and access concerns. In this context, regulatory agencies should provide the best conditions for the implementation of the described new tools.
Collapse
Affiliation(s)
- Cristina Scavone
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gabriella di Mauro
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annamaria Mascolo
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Liberato Berrino
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Rossi
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annalisa Capuano
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
22
|
Sun L, Liu S, Wang J, Wang L. Analysis of Risk Factors for Multiantibiotic-Resistant Infections Among Surgical Patients at a Children's Hospital. Microb Drug Resist 2019; 25:297-303. [PMID: 30676248 PMCID: PMC6441314 DOI: 10.1089/mdr.2018.0279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND To identify the potential risk factors for multiantibiotic-resistant infections and provide sufficient evidence for multiantibiotic resistance prevention and control. MATERIALS AND METHODS We conducted a retrospective study of all patients in pediatric orthopedics, pediatric heart surgery, and pediatric general surgery at a level 3, grade A children's hospital from January to December 2016. The clinical laboratory information monitoring system and the medical record system were used to collect patient information regarding age, surgery type, preoperative length of stay, admission season, incision type, preoperative infection, intraoperative blood loss, postoperative use of invasive equipment, duration of catheter drainage, and timepoint of intraoperative prophylactic antibiotics administration. We used logistic univariate and multivariate regression analysis to analyze the potential risk factors for multiantibiotic-resistant infections among pediatric surgical patients. SPSS 21.0 and Excel software packages were used for the statistical analysis. RESULTS In total, 2,973 patients met the inclusion criteria: 1,247 patients in pediatric orthopedics, 1,089 patients in pediatric heart surgery, and 637 patients in pediatric general surgery. At the end of the study, 113 patients were multiantibiotic-resistant infection cases; the rate of multiantibiotic-resistant infections was 3.80%, and the detection rate was 84.79%. Multivariate analysis indicated that the multiantibiotic-resistant infection cases were influenced by age, department, admission season, incision type, preoperative infection, and duration of catheter drainage. CONCLUSIONS Age, department, admission season, incision type, preoperative infection, and duration of catheter drainage may provide possible evidence for prevention and control strategies of multiantibiotic-resistant infections.
Collapse
Affiliation(s)
- Lixin Sun
- Management of Hospital Infection Control, Children's Hospital of Hebei Province Affiliated to Hebei Medical University, Shijiazhuang, China
| | - Suzhe Liu
- Management of Hospital Infection Control, Children's Hospital of Hebei Province Affiliated to Hebei Medical University, Shijiazhuang, China
| | - Jingming Wang
- Management of Hospital Infection Control, Children's Hospital of Hebei Province Affiliated to Hebei Medical University, Shijiazhuang, China
| | - Liqun Wang
- Management of Hospital Infection Control, Children's Hospital of Hebei Province Affiliated to Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
23
|
Pietrasanta C, Pugni L, Ronchi A, Bottino I, Ghirardi B, Sanchez-Schmitz G, Borriello F, Mosca F, Levy O. Vascular Endothelium in Neonatal Sepsis: Basic Mechanisms and Translational Opportunities. Front Pediatr 2019; 7:340. [PMID: 31456998 PMCID: PMC6700367 DOI: 10.3389/fped.2019.00340] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022] Open
Abstract
Neonatal sepsis remains a major health issue worldwide, especially for low-birth weight and premature infants, with a high risk of death and devastating sequelae. Apart from antibiotics and supportive care, there is an unmet need for adjunctive treatments to improve the outcomes of neonatal sepsis. Strong and long-standing research on adult patients has shown that vascular endothelium is a key player in the pathophysiology of sepsis and sepsis-associated organ failure, through a direct interaction with pathogens, leukocytes, platelets, and the effect of soluble circulating mediators, in part produced by endothelial cells themselves. Despite abundant evidence that the neonatal immune response to sepsis is distinct from that of adults, comparable knowledge on neonatal vascular endothelium is much more limited. Neonatal endothelial cells express lower amounts of adhesion molecules compared to adult ones, and present a reduced capacity to neutralize reactive oxygen species. Conversely, available evidence on biomarkers of endothelial damage in neonates is not as robust as in adult patients, and endothelium-targeted therapeutic opportunities for neonatal sepsis are almost unexplored. Here, we summarize current knowledge on the structure of neonatal vascular endothelium, its interactions with neonatal immune system and possible endothelium-targeted diagnostic and therapeutic tools for neonatal sepsis. Furthermore, we outline areas of basic and translational research worthy of further study, to shed light on the role of vascular endothelium in the context of neonatal sepsis.
Collapse
Affiliation(s)
- Carlo Pietrasanta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Lorenza Pugni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Andrea Ronchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Ilaria Bottino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Beatrice Ghirardi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organisation Center of Excellence, Naples, Italy
| | - Fabio Mosca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
24
|
Sanchez-Schmitz G, Stevens CR, Bettencourt IA, Flynn PJ, Schmitz-Abe K, Metser G, Hamm D, Jensen KJ, Benn C, Levy O. Microphysiologic Human Tissue Constructs Reproduce Autologous Age-Specific BCG and HBV Primary Immunization in vitro. Front Immunol 2018; 9:2634. [PMID: 30524426 PMCID: PMC6256288 DOI: 10.3389/fimmu.2018.02634] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/25/2018] [Indexed: 11/13/2022] Open
Abstract
Current vaccine development disregards human immune ontogeny, relying on animal models to select vaccine candidates targeting human infants, who are at greatest risk of infection worldwide, and receive the largest number of vaccines. To help accelerate and de-risk development of early-life effective immunization, we engineered a human age-specific microphysiologic vascular-interstitial interphase, suitable for pre-clinical modeling of distinct age-targeted immunity in vitro. Our Tissue Constructs (TCs) enable autonomous extravasation of monocytes that undergo rapid self-directed differentiation into migratory Dendritic Cells (DCs) in response to adjuvants and licensed vaccines such as Bacille Calmette-Guérin (BCG) or Hepatitis B virus Vaccine (HBV). TCs contain a confluent human endothelium grown atop a tri-dimensional human extracellular matrix substrate, employ human age-specific monocytes and autologous non heat-treated plasma, and avoid the use of xenogenic materials and exogenous cytokines. Vaccine-pulsed TCs autonomously generated DCs that induced single-antigen recall responses from autologous naïve and memory CD4+ T lymphocytes, matching study participant immune-status, including BCG responses paralleling donor PPD status, BCG-induced adenosine deaminase (ADA) activity paralleling infant cohorts in vivo, and multi-dose HBV antigen-specific responses as demonstrated by lymphoproliferation and TCR sequencing. Overall, our microphysiologic culture method reproduced age- and antigen-specific recall responses to BCG and HBV immunization, closely resembling those observed after a birth immunization of human cohorts in vivo, offering for the first time a new approach to early pre-clinical selection of effective age-targeted vaccine candidates.
Collapse
Affiliation(s)
- Guzman Sanchez-Schmitz
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Harvard University, Boston, MA, United States
| | - Chad R Stevens
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Ian A Bettencourt
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Peter J Flynn
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Klaus Schmitz-Abe
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Harvard University, Boston, MA, United States.,Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States.,Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Gil Metser
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - David Hamm
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Kristoffer J Jensen
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark.,Bandim Health Project, Indepth Network, Bissau, Guinea-Bissau.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Christine Benn
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark.,Bandim Health Project, Indepth Network, Bissau, Guinea-Bissau
| | - Ofer Levy
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Precision Vaccines Program, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Harvard University, Boston, MA, United States.,Broad Institute of Harvard and MIT, Cambridge, MA, United States
| |
Collapse
|
25
|
Perez Cuevas MB, Kodani M, Choi Y, Joyce J, O'Connor SM, Kamili S, Prausnitz MR. Hepatitis B vaccination using a dissolvable microneedle patch is immunogenic in mice and rhesus macaques. Bioeng Transl Med 2018; 3:186-196. [PMID: 30377659 PMCID: PMC6195907 DOI: 10.1002/btm2.10098] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/29/2018] [Accepted: 06/01/2018] [Indexed: 01/18/2023] Open
Abstract
Chronic Hepatitis B virus infection remains a major global public health problem, accounting for about 887,000 deaths in 2015. Perinatal and early childhood infections are strongly associated with developing chronic hepatitis B. Adding a birth dose of the hepatitis B vaccine (HepB BD) to routine childhood vaccination can prevent over 85% of these infections. However, HepB BD coverage remains low in many global regions, with shortages of birth attendants trained to vaccinate and limited HepB BD supply at birth. To address the challenges, we developed coated metal microneedle patches (cMNPs) and dissolvable microneedle patches (dMNPs) that deliver adjuvant‐free hepatitis B vaccine to the skin in a simple‐to‐administer manner. The dMNP contains micron‐scale, solid needles encapsulating vaccine antigen and dissolve in the skin, generating no sharps waste. We delivered HepB BD via cMNP to BALB/c mice and via dMNP to both mice and rhesus macaques. Both cMNP and dMNP were immunogenic, generating hepatitis B surface antibody levels similar to human seroprotection. Biomechanical analysis showed that at high forces the microneedles failed mechanically by yielding but microneedles partially blunted by axial compression were still able to penetrate skin. Overall, this study indicates that with further development, dMNPs could offer a method of vaccination to increase HepB BD access and reduce needle waste in developing countries.
Collapse
Affiliation(s)
- Monica B Perez Cuevas
- School of Chemical and Biomolecular Engineering Georgia Institute of Technology Atlanta GA 30332
| | - Maja Kodani
- Division of Viral Hepatitis, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention Centers for Disease Control and Prevention Atlanta GA 30329
| | - Youkyung Choi
- Division of Viral Hepatitis, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention Centers for Disease Control and Prevention Atlanta GA 30329
| | - Jessica Joyce
- Wallace Coulter Department of Biomedical Engineering at Georgia Tech and Emory University Georgia Institute of Technology Atlanta GA 30332
| | - Siobhan M O'Connor
- Division of Viral Hepatitis, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention Centers for Disease Control and Prevention Atlanta GA 30329
| | - Saleem Kamili
- Division of Viral Hepatitis, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention Centers for Disease Control and Prevention Atlanta GA 30329
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering Georgia Institute of Technology Atlanta GA 30332.,Wallace Coulter Department of Biomedical Engineering at Georgia Tech and Emory University Georgia Institute of Technology Atlanta GA 30332
| |
Collapse
|
26
|
Borriello F, van Haren SD, Levy O. First International Precision Vaccines Conference: Multidisciplinary Approaches to Next-Generation Vaccines. mSphere 2018; 3:e00214-18. [PMID: 30068557 PMCID: PMC6070736 DOI: 10.1128/msphere.00214-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vaccines represent a remarkable success in the history of medicine since they have prevented and, in some instances, eradicated a range of infectious diseases. However, for many existing vaccines, immunogenicity is limited, requiring multiple booster doses, and we are still unable to target many pathogens due to intrinsic features of the microorganism, such as genetic/antigenic variability between strains, and our limited understanding of the variables that regulate vaccine responsiveness, including age- and sex-specific differences. Moreover, the traditional approach to vaccine development is often empirical, relying on inactivation of microorganisms or purification of their components, which are usually less immunogenic than the whole microorganism from which they derive. This approach has yielded multiple important vaccines but has failed to consistently generate vaccines that are sufficiently immunogenic in populations with limited immune responsiveness such as newborns and elderly individuals. In an effort to trigger impactful collaborations, a community of scientists gathered in Boston in the United States for the first biennial International Precision Vaccines Conference, sponsored by the Boston Children's Hospital Precision Vaccines Program, to discuss innovation in vaccinology. Recent advancements in the field of systems biology that can identify vaccine immunogenicity biomarkers for target populations, in human in vitro models, and in novel adjuvant and formulation strategies offer unprecedented opportunities to dissect the human immune response to vaccines and inform dramatic improvements in vaccine efficacy. These approaches are poised to have a major scientific and translational impact in vaccinology.
Collapse
Affiliation(s)
- Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- WAO Center of Excellence, Naples, Italy
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
27
|
Cheminay C, Körner J, Bernig C, Brückel M, Feigl M, Schletz M, Suter M, Chaplin P, Volkmann A. A single vaccination with non-replicating MVA at birth induces both immediate and long-term protective immune responses. Vaccine 2018; 36:2427-2434. [PMID: 29599088 DOI: 10.1016/j.vaccine.2018.03.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/17/2017] [Accepted: 03/16/2018] [Indexed: 11/16/2022]
Abstract
Newborns are considered difficult to protect against infections shortly after birth, due to their ineffective immune system that shows quantitative and qualitative differences compared to adults. However, here we show that a single vaccination of mice at birth with a replication-deficient live vaccine Modified Vaccinia Ankara [MVA] efficiently induces antigen-specific B- and T-cells that fully protect against a lethal Ectromelia virus challenge. Protection was induced within 2 weeks and using genetically modified mice we show that this protection was mainly T-cell dependent. Persisting immunological T-cell memory and neutralizing antibodies were obtained with the single vaccination. Thus, MVA administered as early as at birth induced immediate and long-term protection against an otherwise fatal disease and appears attractive as a new generation smallpox vaccine that is effective also in children. Moreover, it may have the potential to serve as platform for childhood vaccines as indicated by measles specific T- and B-cell responses induced in newborn mice vaccinated with recombinant MVA expressing measles antigens.
Collapse
Affiliation(s)
- Cédric Cheminay
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Jana Körner
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Constanze Bernig
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Michael Brückel
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Markus Feigl
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Martin Schletz
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Mark Suter
- University of Zürich, Dekanat Vetsuisse-Fakultät Immunology, Winterthurerstrasse 204, CH-8057 Zürich, Switzerland
| | - Paul Chaplin
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany
| | - Ariane Volkmann
- Bavarian Nordic GmbH, Fraunhoferstrasse 13, D-82152 Martinsried, Germany.
| |
Collapse
|
28
|
Whittaker E, Goldblatt D, McIntyre P, Levy O. Neonatal Immunization: Rationale, Current State, and Future Prospects. Front Immunol 2018; 9:532. [PMID: 29670610 PMCID: PMC5893894 DOI: 10.3389/fimmu.2018.00532] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/28/2018] [Indexed: 12/11/2022] Open
Abstract
Infections take their greatest toll in early life necessitating robust approaches to protect the very young. Here, we review the rationale, current state, and future research directions for one such approach: neonatal immunization. Challenges to neonatal immunization include natural concern about safety as well as a distinct neonatal immune system that is generally polarized against Th1 responses to many stimuli such that some vaccines that are effective in adults are not in newborns. Nevertheless, neonatal immunization could result in high-population penetration as birth is a reliable point of healthcare contact, and offers an opportunity for early protection of the young, including preterm newborns who are deficient in maternal antibodies. Despite distinct immunity and reduced responses to some vaccines, several vaccines have proven safe and effective at birth. While some vaccines such as polysaccharide vaccines have little effectiveness at birth, hepatitis B vaccine can prime at birth and requires multiple doses to achieve protection, whereas the live-attenuated Bacille Calmette-Guérin (BCG), may offer single shot protection, potentially in part via heterologous ("non-specific") beneficial effects. Additional vaccines have been studied at birth including those directed against pertussis, pneumococcus, Haemophilus influenza type B and rotavirus providing important lessons. Current areas of research in neonatal vaccinology include characterization of early life immune ontogeny, heterogeneity in and heterologous effects of BCG vaccine formulations, applying systems biology and systems serology, in vitro platforms that model age-specific human immunity and discovery and development of novel age-specific adjuvantation systems. These approaches may inform, de-risk, and accelerate development of novel vaccines for use in early life. Key stakeholders, including the general public, should be engaged in assessing the opportunities and challenges inherent to neonatal immunization.
Collapse
Affiliation(s)
- Elizabeth Whittaker
- Centre for International Child Health, Department of Paediatrics, Imperial College London, London, United Kingdom
| | - David Goldblatt
- Immunobiology Section, UCL Great Ormond Street Institute of Child Health (ICH), London, United Kingdom
| | - Peter McIntyre
- National Centre for Immunisation Research and Surveillance, Kids Research, Sydney Children’s Hospital Network and University of Sydney, Sydney, NSW, Australia
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
29
|
Cui X, Li Y, Yang L, You L, Wang X, Shi C, Ji C, Guo X. Peptidome analysis of human milk from women delivering macrosomic fetuses reveals multiple means of protection for infants. Oncotarget 2018; 7:63514-63525. [PMID: 27566575 PMCID: PMC5325381 DOI: 10.18632/oncotarget.11532] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/13/2016] [Indexed: 01/11/2023] Open
Abstract
Breastfeeding is associated with a lower incidence of obesity, diabetes, and cardiovascular disease later in life. While macrosomic infants have a higher risk of developing obesity and other metabolic disorders. Breast milk may contain special nutrients to meet the different growth needs of different infants. Whether mothers make breast milk different to meet the requirement of macrosomic infants is still unknown. Here, we conducted a comparison between mothers delivering macrosomic and non-macrosomic infants in colostrum endogenous peptides. More than 400 peptides, originating from at least 34 protein precursors, were identified by Liquid Chromatography/Mass Spectrometry (LC/MS). Out of these, 29 peptides found to be significant differently expressed (|fold change| ≥ 3, P < 0.01). Blastp analysis revealed 41 peptides may have established biological activities, which exhibit immunomodulating, antibacterial action, antioxidation, opioid agonist and antihypertensive activity. Furthermore, we found that peptide located at β-Casein 24-38 AA has antimicrobial effect against E. coli, Y. enterocolitica and S. aureus. While, κ-Casein 89-109 AA-derived peptide plays as a regulator of preadipocyte proliferation. The profile of endogenous peptides from macrosomic term infants is different from non-macrosomic terms. This different peptide expression potentially has specific physiological function to benefit macrosomic infants. Finally, we believe that our research is a meaningfull finding which may add to the understanding of milk peptide physiological action.
Collapse
Affiliation(s)
- Xianwei Cui
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| | - Yun Li
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| | - Lei Yang
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| | - Lianghui You
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| | - Xing Wang
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| | - Chunmei Shi
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| | - Chenbo Ji
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| | - Xirong Guo
- From Nanjing Maternal and Child Health Medical Institute, Nanjing Medical University Affiliated Nanjing Maternal and Child Health Hospital
| |
Collapse
|
30
|
Scheid A, Borriello F, Pietrasanta C, Christou H, Diray-Arce J, Pettengill MA, Joshi S, Li N, Bergelson I, Kollmann T, Dowling DJ, Levy O. Adjuvant Effect of Bacille Calmette-Guérin on Hepatitis B Vaccine Immunogenicity in the Preterm and Term Newborn. Front Immunol 2018; 9:29. [PMID: 29416539 PMCID: PMC5787546 DOI: 10.3389/fimmu.2018.00029] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 01/04/2018] [Indexed: 01/21/2023] Open
Abstract
Immunization is key to protecting term and preterm infants from a heightened risk of infection. However, preterm immunity is distinct from that of the term, limiting its ability to effectively respond to vaccines routinely given at birth, such as hepatitis B vaccine (HBV). As part of the Expanded Program on Immunization, HBV is often given together with the live-attenuated vaccine Bacille Calmette-Guérin (BCG), known to activate multiple pattern-recognition receptors. Of note, some clinical studies suggest BCG can enhance efficacy of other vaccines in term newborns. However, little is known about whether BCG can shape Th-polarizing cytokine responses to HBV nor the age-dependency of such effects, including whether they may extend to the preterm. To characterize the effects of BCG on HBV immunogenicity, we studied individual and combined administration of these vaccines to cord newborn and adult human whole blood and mononuclear cells in vitro and to neonatal and adult mice in vivo. Compared to either BCG or HBV alone, (BCG + HBV) synergistically enhanced in vitro whole blood production of IL-1β, while (BCG + HBV) also promoted production of several cytokines/chemokines in all age groups, age-specific enhancement included IL-12p70 in the preterm and GM-CSF in the preterm and term. In human mononuclear cells, (BCG + HBV) enhanced mRNA expression of several genes including CSF2, which contributed to clustering of genes by vaccine treatment via principle component analysis. To assess the impact of BCG on HBV immunization, mice of three different age groups were immunized subcutaneously with, BCG, HBV, (BCG + HBV) into the same site; or BCG and HBV injected into separate sites. Whether injected into a separate site or at the same site, co-administration of BCG with HBV significantly enhanced anti-HBV IgG titers in mice immunized on day of life-0 or -7, respectively, but not in adult mice. In summary, our data demonstrate that innate and adaptive vaccine responses of preterm and term newborns are immunologically distinct. Furthermore, BCG or "BCG-like" adjuvants should be further studied as a promising adjuvantation approach to enhance immunogenicity of vaccines to protect these vulnerable populations.
Collapse
Affiliation(s)
- Annette Scheid
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Francesco Borriello
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- WAO Center of Excellence, Naples, Italy
| | - Carlo Pietrasanta
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Joann Diray-Arce
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Matthew A. Pettengill
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| | - Sweta Joshi
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Ning Li
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Medical Eli Lilly, Shanghai, China
| | - Ilana Bergelson
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Tobias Kollmann
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, British Columbia Children’s Hospital, Vancouver, BC, Canada
| | - David J. Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
| |
Collapse
|
31
|
Abstract
Immunisation of the newborn represents a key global strategy in overcoming morbidity and mortality due to infection in early life. Potential limitations, however, include poor immunogenicity, safety concerns and the development of tolerogenicity or hypo-responsiveness to either the same antigen and/or concomitant antigens administered at birth or in the subsequent months. Furthermore, the neonatal immunological milieu is polarised towards Th2-type immunity with dampening of Th1-type responses and impaired humoral immunity, resulting in qualitatively and quantitatively poorer antibody responses compared to older infants. Innate immunity also shows functional deficiency in antigen-presenting cells: the expression and signalling of Toll-like receptors undergo maturational changes associated with distinct functional responses. Nevertheless, the effectiveness of BCG, hepatitis B and oral polio vaccines, the only immunisations currently in use in the neonatal period, is proof of concept that vaccines can be successfully administered to the newborn via different routes of delivery to induce a range of protective mechanisms for three different diseases. In this review paper, we discuss the rationale for and challenges to neonatal immunisation, summarising progress made in the field, including lessons learnt from newborn vaccines in the pipeline. Furthermore, we explore important maternal, infant and environmental co-factors that may impede the success of current and future neonatal immunisation strategies. A variety of approaches have been proposed to overcome the inherent regulatory constraints of the newborn innate and adaptive immune system, including alternative routes of delivery, novel vaccine configurations, improved innate receptor agonists and optimised antigen-adjuvant combinations. Crucially, a dual strategy may be employed whereby immunisation at birth is used to prime the immune system in order to improve immunogenicity to subsequent homologous or heterologous boosters in later infancy. Similarly, potent non-specific immunomodulatory effects may be elicited when challenged with unrelated antigens, with the potential to reduce the overall risk of infection and allergic disease in early life.
Collapse
Affiliation(s)
- Anja Saso
- Centre of International Child Health, Department of Paediatrics, Imperial College London, W2 1NY, London, UK
| | - Beate Kampmann
- Centre of International Child Health, Department of Paediatrics, Imperial College London, W2 1NY, London, UK.
- Vaccines and Immunity Theme, MRC Unit The Gambia, Fajara, The Gambia.
| |
Collapse
|
32
|
Raymond SL, Rincon JC, Wynn JL, Moldawer LL, Larson SD. Impact of Early-Life Exposures to Infections, Antibiotics, and Vaccines on Perinatal and Long-term Health and Disease. Front Immunol 2017; 8:729. [PMID: 28690615 PMCID: PMC5481313 DOI: 10.3389/fimmu.2017.00729] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/08/2017] [Indexed: 12/21/2022] Open
Abstract
Essentially, all neonates are exposed to infections, antibiotics, or vaccines early in their lives. This is especially true for those neonates born underweight or premature. In contrast to septic adults and children who are at an increased risk for subsequent infections, exposure to infection during the neonatal period is not associated with an increased risk of subsequent infection and may be paradoxically associated with reductions in late-onset sepsis (LOS) in the most premature infants. Perinatal inflammation is also associated with a decreased incidence of asthma and atopy later in life. Conversely, septic neonates are at increased risk of impaired long-term neurodevelopment. While the positive effects of antibiotics in the setting of infection are irrefutable, prolonged administration of broad-spectrum, empiric antibiotics in neonates without documented infection is associated with increased risk of LOS, necrotizing enterocolitis, or death. Vaccines provide a unique opportunity to prevent infection-associated disease; unfortunately, vaccinations have been largely unsuccessful when administered in the first month of life with the exception of vaccines against hepatitis B and tuberculosis. Future vaccines will require the use of novel adjuvants to overcome this challenge. This review describes the influence of infections, antibiotics, and vaccines during the first days of life, as well as the influence on future health and disease. We will also discuss potential immunomodulating therapies, which may serve to train the preterm immune system and reduce subsequent infectious burden without subjecting neonates to the risks accompanied by virulent pathogens.
Collapse
Affiliation(s)
- Steven L Raymond
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jaimar C Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - James L Wynn
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Shawn D Larson
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
33
|
Protective Role of Passively Transferred Maternal Cytokines against Bordetella pertussis Infection in Newborn Piglets. Infect Immun 2017; 85:IAI.01063-16. [PMID: 28167667 DOI: 10.1128/iai.01063-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/27/2017] [Indexed: 11/20/2022] Open
Abstract
Maternal vaccination represents a potential strategy to protect both the mother and the offspring against life-threatening infections. This protective role has mainly been associated with antibodies, but the role of cell-mediated immunity, in particular passively transferred cytokines, is not well understood. Here, using a pertussis model, we have demonstrated that immunization of pregnant sows with heat-inactivated bacteria leads to induction of a wide range of cytokines (e.g., tumor necrosis factor alpha [TNF-α], gamma interferon [IFN-γ], interleukin-6 [IL-6], IL-8, and IL-12/IL-23p40) in addition to pertussis-specific antibodies. These cytokines can be detected in the sera and colostrum/milk of vaccinated sows and subsequently were detected at significant levels in the serum and bronchoalveolar lavage fluid of piglets born to vaccinated sows together with pertussis-specific antibodies. In contrast, active vaccination of newborn piglets with heat-inactivated bacteria induced high levels of specific IgG and IgA but no cytokines. Although the levels of antibodies in vaccinated piglets were comparable to those of passively transferred antibodies, no protection against Bordetella pertussis infection was observed. Thus, our results demonstrate that a combination of passively transferred cytokines and antibodies is crucial for disease protection. The presence of passively transferred cytokines/antibodies influences the cytokine secretion ability of splenocytes in the neonate, which provides novel evidence that maternal immunization can influence the newborn's cytokine milieu and may impact immune cell differentiation (e.g., Th1/Th2 phenotype). Therefore, these maternally derived cytokines may play an essential role both as mediators of early defense against infections and possibly as modulators of the immune repertoire of the offspring.
Collapse
|
34
|
Dowling DJ, Scott EA, Scheid A, Bergelson I, Joshi S, Pietrasanta C, Brightman S, Sanchez-Schmitz G, Van Haren SD, Ninković J, Kats D, Guiducci C, de Titta A, Bonner DK, Hirosue S, Swartz MA, Hubbell JA, Levy O. Toll-like receptor 8 agonist nanoparticles mimic immunomodulating effects of the live BCG vaccine and enhance neonatal innate and adaptive immune responses. J Allergy Clin Immunol 2017; 140:1339-1350. [PMID: 28343701 PMCID: PMC5667586 DOI: 10.1016/j.jaci.2016.12.985] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/15/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022]
Abstract
Background Newborns display distinct immune responses, leaving them vulnerable to infections and impairing immunization. Targeting newborn dendritic cells (DCs), which integrate vaccine signals into adaptive immune responses, might enable development of age-specific vaccine formulations to overcome suboptimal immunization. Objective Small-molecule imidazoquinoline Toll-like receptor (TLR) 8 agonists robustly activate newborn DCs but can result in reactogenicity when delivered in soluble form. We used rational engineering and age- and species-specific modeling to construct and characterize polymer nanocarriers encapsulating a TLR8 agonist, allowing direct intracellular release after selective uptake by DCs. Methods Chemically similar but morphologically distinct nanocarriers comprised of amphiphilic block copolymers were engineered for targeted uptake by murine DCs in vivo, and a range of TLR8 agonist–encapsulating polymersome formulations were then synthesized. Novel 96-well in vitro assays using neonatal human monocyte-derived DCs and humanized TLR8 mouse bone marrow–derived DCs enabled benchmarking of the TLR8 agonist–encapsulating polymersome formulations against conventional adjuvants and licensed vaccines, including live attenuated BCG vaccine. Immunogenicity of the TLR8 agonist adjuvanted antigen 85B (Ag85B)/peptide 25–loaded BCG-mimicking nanoparticle formulation was evaluated in vivo by using humanized TLR8 neonatal mice. Results Although alum-adjuvanted vaccines induced modest costimulatory molecule expression, limited TH-polarizing cytokine production, and significant cell death, BCG induced a robust adult-like maturation profile of neonatal DCs. Remarkably, TLR8 agonist polymersomes induced not only newborn DC maturation profiles similar to those induced by BCG but also stronger IL-12p70 production. On subcutaneous injection to neonatal mice, the TLR8 agonist–adjuvanted Ag85B peptide 25 formulation was comparable with BCG in inducing Ag85B-specific CD4+ T-cell numbers. Conclusion TLR8 agonist–encapsulating polymersomes hold substantial potential for early-life immunization against intracellular pathogens. Overall, our study represents a novel approach for rational design of early-life vaccines.
Collapse
Affiliation(s)
- David J Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, Ill.
| | - Annette Scheid
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Division of Newborn Medicine, Floating Hospital for Children, Tufts Medical Center, Boston, Mass; Precision Vaccine Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass
| | - Ilana Bergelson
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass
| | - Sweta Joshi
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Precision Vaccine Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass
| | - Carlo Pietrasanta
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy; Precision Vaccine Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass
| | - Spencer Brightman
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Precision Vaccine Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass
| | - Guzman Sanchez-Schmitz
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Precision Vaccine Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass
| | - Simon D Van Haren
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Precision Vaccine Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass
| | - Jana Ninković
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Dina Kats
- Department of Biomedical Engineering, Northwestern University, Evanston, Ill
| | | | - Alexandre de Titta
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Daniel K Bonner
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sachiko Hirosue
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Melody A Swartz
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, Chicago, Ill
| | - Jeffrey A Hubbell
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, Chicago, Ill
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass; Precision Vaccine Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Mass.
| |
Collapse
|
35
|
Dowling DJ, van Haren SD, Scheid A, Bergelson I, Kim D, Mancuso CJ, Foppen W, Ozonoff A, Fresh L, Theriot TB, Lackner AA, Fichorova RN, Smirnov D, Vasilakos JP, Beaurline JM, Tomai MA, Midkiff CC, Alvarez X, Blanchard JL, Gilbert MH, Aye PP, Levy O. TLR7/8 adjuvant overcomes newborn hyporesponsiveness to pneumococcal conjugate vaccine at birth. JCI Insight 2017; 2:e91020. [PMID: 28352660 DOI: 10.1172/jci.insight.91020] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Infection is the most common cause of mortality in early life, and immunization is the most promising biomedical intervention to reduce this burden. However, newborns fail to respond optimally to most vaccines. Adjuvantation is a key approach to enhancing vaccine immunogenicity, but responses of human newborn leukocytes to most candidate adjuvants, including most TLR agonists, are functionally distinct. Herein, we demonstrate that 3M-052 is a locally acting lipidated imidazoquinoline TLR7/8 agonist adjuvant in mice, which, when properly formulated, can induce robust Th1 cytokine production by human newborn leukocytes in vitro, both alone and in synergy with the alum-adjuvanted pneumococcal conjugate vaccine 13 (PCV13). When admixed with PCV13 and administered i.m. on the first day of life to rhesus macaques, 3M-052 dramatically enhanced generation of Th1 CRM-197-specific neonatal CD4+ cells, activation of newborn and infant Streptococcus pneumoniae polysaccharide-specific (PnPS-specific) B cells as well as serotype-specific antibody titers, and opsonophagocytic killing. Remarkably, a single dose at birth of PCV13 plus 0.1 mg/kg 3M-052 induced PnPS-specific IgG responses that were approximately 10-100 times greater than a single birth dose of PCV13 alone, rapidly exceeding the serologic correlate of protection, as early as 28 days of life. This potent immunization strategy, potentially effective with one birth dose, could represent a new paradigm in early life vaccine development.
Collapse
Affiliation(s)
- David J Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Simon D van Haren
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Precision Vaccines Program, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Annette Scheid
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Precision Vaccines Program, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA.,Division of Newborn Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Ilana Bergelson
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Dhohyung Kim
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Christy J Mancuso
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Willemina Foppen
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Al Ozonoff
- Harvard Medical School, Boston, Massachusetts, USA.,Precision Vaccines Program, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA.,Center for Patient Safety and Quality Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lynn Fresh
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Terese B Theriot
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Andrew A Lackner
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Raina N Fichorova
- Harvard Medical School, Boston, Massachusetts, USA.,Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | | | - Mark A Tomai
- 3M Drug Delivery Systems, Saint Paul, Minnesota, USA
| | - Cecily C Midkiff
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Xavier Alvarez
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - James L Blanchard
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Margaret H Gilbert
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Pyone Pyone Aye
- Tulane National Primate Research Center (TNPRC), Covington, Louisiana, USA
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Precision Vaccines Program, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Fattah MA, Omer AFA, Asaif S, Manlulu R, Karar T, Ahmed A, Aljada A, Saleh AM, Qureshi S, Nasr A. Utility of cytokine, adhesion molecule and acute phase proteins in early diagnosis of neonatal sepsis. J Nat Sci Biol Med 2017; 8:32-39. [PMID: 28250672 PMCID: PMC5320820 DOI: 10.4103/0976-9668.198362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background and Aim: Neonatal infection, including bacterial sepsis, is a major health care issue with an annual global mortality in excess of one million lives. Therefore, this study aimed to evaluate the potential diagnostic value of C-reactive protein (CRP), E-selectin, procalcitonin (PCT), interleukins-6 (IL-6), and tumor necrosis factor-α (TNF-α) both independently and in combination for the diagnosis of neonatal sepsis in its earliest stages. Materials and Methods: A total of 320 subjects were included in this study. A prospective cross-sectional study was conducted among neonates admitted to Neonatal Intensive Care Unit at King Abdulaziz Medical City, Riyadh, KSA during January 2013 to August 2015, the study based on three study groups categorized according to clinical symptoms and blood culture result. Study groups include healthy control neonates (n = 80), clinical sepsis (CS) group (n = 80) with clinical signs of sepsis but their blood culture was negative, and sepsis group with clinical signs of sepsis and their blood culture was positive. Results: The study observed significant difference in plasma levels of CRP, IL-6, TNF-α, E-selectin, and PCT in patients group when compared with control group (P < 0.001). Furthermore, the levels are significantly different between patient groups including CS and neonatal sepsis group. Moreover, result observed significant difference in CRP and IL-6 in early onset sepsis (EOS) when compared with late onset sepsis (LOS) neonates (P < 0.001 and 0.01), respectively, while there were no significant difference in TNF-α, E-selectin, and PCT between EOS and LOS (P = 0.44, 0.27 and 0.24), respectively. Regarding biomarkers accuracy, the result showed that CRP has the best diagnostic accuracy with cutoff value of 3.6 ng/ml (sensitivity 78% and specificity of 70%). The best combination is shown with CRP and IL-6 in which sensitivity increased to 89% and specificity to 79%. Conclusion: It was concluded that infected new-born babies have a higher E-selectin, PCT, IL-6, TNF-α, and CRP compared with the neonates with CS and control. IL-6, TNF-α, and CRP should be measured in combination for mare diagnostic accuracy in neonatal sepsis. Likewise, PCT should be investigated as a part of sepsis screening for all suspected neonates.
Collapse
Affiliation(s)
- M A Fattah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University, Riyadh, Saudi Arabia; College of Graduate Studies, Sudan University of Science and Technology, Khartoum, Sudan
| | - Al Fadhil A Omer
- Department of Medical Laboratory Sciences, Al Neelain University, Khartoum, Sudana
| | - S Asaif
- Department of Pediatrics, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center (KAIMRC) National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - R Manlulu
- Department of Pediatrics, King Fahad National Guard Hospital, National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - T Karar
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University, Riyadh, Saudi Arabia
| | - A Ahmed
- Department of Epidemiology and Biostatistics, College of Public Health and Health Informatics, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - A Aljada
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center (KAIMRC) National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Ayman M Saleh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, KSAU-HS, Jeddah, Kingdom of Saudi Arabia; King Abdullah International Medical Research Centre (KAIMRC), National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Shoeb Qureshi
- Department of Research, College of Applied Medical Sciences, King Saud Bin Abdulaziz University, Riyadh, Kingdom of Saudi Arabia
| | - A Nasr
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center (KAIMRC) National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
37
|
Saadatian-Elahi M, Aaby P, Shann F, Netea MG, Levy O, Louis J, Picot V, Greenberg M, Warren W. Heterologous vaccine effects. Vaccine 2016; 34:3923-30. [PMID: 27312214 DOI: 10.1016/j.vaccine.2016.06.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/25/2016] [Accepted: 06/03/2016] [Indexed: 11/18/2022]
Abstract
The heterologous or non-specific effects (NSEs) of vaccines, at times defined as "off-target effects" suggest that they can affect the immune response to organisms other than their pathogen-specific intended purpose. These NSEs have been the subject of clinical, immunological and epidemiological studies and are increasingly recognized as an important biological process by a growing group of immunologists and epidemiologists. Much remain to be learned about the extent and underlying mechanisms for these effects. The conference "Off-target effects of vaccination" held in Annecy-France (June 8-10 2015) intended to take a holistic approach drawing from the fields of immunology, systems biology, epidemiology, bioinformatics, public health and regulatory science to address fundamental questions of immunological mechanisms, as well as translational questions about vaccines NSEs. NSE observations were examined using case-studies on live attenuated vaccines and non-live vaccines followed by discussion of studies of possible biological mechanisms. Some possible pathways forward in the study of vaccines NSE were identified and discussed by the expert group.
Collapse
Affiliation(s)
- Mitra Saadatian-Elahi
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, 5 Place d'Arsonval, 69437 Lyon cedex 03, France.
| | - Peter Aaby
- Bandim Health Project, INDEPTH Network, CP861 Bissau, Guinea-Bissau
| | - Frank Shann
- Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital & Havard Medical School, Boston, MA 02115, USA
| | - Jacques Louis
- Fondation Mérieux, 17 rue Bourgelat, 69002 Lyon, France
| | | | | | | |
Collapse
|
38
|
In vitro cytokine induction by TLR-activating vaccine adjuvants in human blood varies by age and adjuvant. Cytokine 2016; 83:99-109. [PMID: 27081760 DOI: 10.1016/j.cyto.2016.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/29/2016] [Accepted: 04/02/2016] [Indexed: 12/24/2022]
Abstract
Most infections occur in early life, prompting development of novel adjuvanted vaccines to protect newborns and infants. Several Toll-like receptor (TLR) agonists (TLRAs) are components of licensed vaccine formulations or are in development as candidate adjuvants. However, the type and magnitude of immune responses to TLRAs may vary with the TLR activated as well as age and geographic location. Most notably, in newborns, as compared to adults, the immune response to TLRAs is polarized with lower Th1 cytokine production and robust Th2 and anti-inflammatory cytokine production. The ontogeny of TLR-mediated cytokine responses in international cohorts has been reported, but no study has compared cytokine responses to TLRAs between U.S. neonates and infants at the age of 6months. Both are critical age groups for the currently pediatric vaccine schedule. In this study, we report quantitative differences in the production of a panel of 14 cytokines and chemokines after in vitro stimulation of newborn cord blood and infant and adult peripheral blood with agonists of TLR4, including monophosphoryl lipid A (MPLA) and glucopyranosyl lipid Adjuvant aqueous formulation (GLA-AF), as well as agonists of TLR7/8 (R848) and TLR9 (CpG). Both TLR4 agonists, MPLA and GLA-AF, induced greater concentrations of Th1 cytokines CXCL10, TNF and Interleukin (IL)-12p70 in infant and adult blood compared to newborn blood. All the tested TLRAs induced greater infant IFN-α2 production compared to newborn and adult blood. In contrast, CpG induced greater IFN-γ, IL-1β, IL-4, IL-12p40, IL-10 and CXCL8 in newborn than in infant and adult blood. Overall, to the extent that these in vitro studies mirror responses in vivo, our study demonstrates distinct age-specific effects of TLRAs that may inform their development as candidate adjuvants for early life vaccines.
Collapse
|
39
|
Oh DY, Dowling DJ, Ahmed S, Choi H, Brightman S, Bergelson I, Berger ST, Sauld JF, Pettengill M, Kho AT, Pollack HJ, Steen H, Levy O. Adjuvant-induced Human Monocyte Secretome Profiles Reveal Adjuvant- and Age-specific Protein Signatures. Mol Cell Proteomics 2016; 15:1877-94. [PMID: 26933193 PMCID: PMC5083103 DOI: 10.1074/mcp.m115.055541] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Indexed: 12/16/2022] Open
Abstract
Adjuvants boost vaccine responses, enhancing protective immunity against infections that are most common among the very young. Many adjuvants activate innate immunity, some via Toll-Like Receptors (TLRs), whose activities varies with age. Accordingly, characterization of age-specific adjuvant-induced immune responses may inform rational adjuvant design targeting vulnerable populations. In this study, we employed proteomics to characterize the adjuvant-induced changes of secretomes from human newborn and adult monocytes in response to Alum, the most commonly used adjuvant in licensed vaccines; Monophosphoryl Lipid A (MPLA), a TLR4-activating adjuvant component of a licensed Human Papilloma Virus vaccine; and R848 an imidazoquinoline TLR7/8 agonist that is a candidate adjuvant for early life vaccines. Monocytes were incubated in vitro for 24 h with vehicle, Alum, MPLA, or R848 and supernatants collected for proteomic analysis employing liquid chromatography-mass spectrometry (LC-MS) (data available via ProteomeXchange, ID PXD003534). 1894 non-redundant proteins were identified, of which ∼30 - 40% were common to all treatment conditions and ∼5% were treatment-specific. Adjuvant-stimulated secretome profiles, as identified by cluster analyses of over-represented proteins, varied with age and adjuvant type. Adjuvants, especially Alum, activated multiple innate immune pathways as assessed by functional enrichment analyses. Release of lactoferrin, pentraxin 3, and matrix metalloproteinase-9 was confirmed in newborn and adult whole blood and blood monocytes stimulated with adjuvants alone or adjuvanted licensed vaccines with distinct clinical reactogenicity profiles. MPLA-induced adult monocyte secretome profiles correlated in silico with transcriptome profiles induced in adults immunized with the MPLA-adjuvanted RTS,S malaria vaccine (Mosquirix™). Overall, adjuvants such as Alum, MPLA and R848 give rise to distinct and age-specific monocyte secretome profiles, paralleling responses to adjuvant-containing vaccines in vivo. Age-specific in vitro modeling coupled with proteomics may provide fresh insight into the ontogeny of adjuvant action thereby informing targeted adjuvanted vaccine development for distinct age groups.
Collapse
Affiliation(s)
- Djin-Ye Oh
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts; ¶Division of Pediatric Infectious Diseases, New York University Medical School, New York; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| | - David J Dowling
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts
| | - Saima Ahmed
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hyungwon Choi
- **Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Spencer Brightman
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and
| | - Ilana Bergelson
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and
| | - Sebastian T Berger
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - John F Sauld
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Matthew Pettengill
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| | - Alvin T Kho
- ‡‡Children's Hospital Informatics Program, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Henry J Pollack
- ¶Division of Pediatric Infectious Diseases, New York University Medical School, New York
| | - Hanno Steen
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| | - Ofer Levy
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
40
|
Abstract
Pregnancy and early infancy are periods of relative immune suppression and increased vulnerability to infection. In these circumstances infections are associated with high morbidity and mortality. In particular, infants have high rates of invasive disease, higher than at any other stage of life with rates of 100 per 100 000 population. The concept of maternal vaccination is that maternal levels of pathogen-specific antibody are boosted and provide protection to the infant until the infant is able to mount an effective immune response to immunization. However, an important concern for women and healthcare providers is the safety of receiving vaccines during pregnancy. There are challenges associated with assessing safety in pregnant women. This review discusses the rationale for maternal vaccination, the concepts and mechanisms used. An assessment is made of the safety of vaccination during pregnancy, and the challenges associated with this are considered. In general terms, it is considered that the risk from disease far outweighs the small risk associated with vaccination during pregnancy and that they offer a new platform for preventing significant and serious infections in mothers and young infants.
Collapse
Affiliation(s)
- Chrissie Jones
- a Paediatric Infectious Diseases Research Group (PIDRG); St George's, University of London; London, UK
| | | |
Collapse
|
41
|
Gil A, Kenney LL, Mishra R, Watkin LB, Aslan N, Selin LK. Vaccination and heterologous immunity: educating the immune system. Trans R Soc Trop Med Hyg 2015; 109:62-9. [PMID: 25573110 DOI: 10.1093/trstmh/tru198] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This review discusses three inter-related topics: (1) the immaturity of the neonatal and infant immune response; (2) heterologous immunity, where prior infection history with unrelated pathogens alters disease outcome resulting in either enhanced protective immunity or increased immunopathology to new infections, and (3) epidemiological human vaccine studies that demonstrate vaccines can have beneficial or detrimental effects on subsequent unrelated infections. The results from the epidemiological and heterologous immunity studies suggest that the immune system has tremendous plasticity and that each new infection or vaccine that an individual is exposed to during a lifetime will potentially alter the dynamics of their immune system. It also suggests that each new infection or vaccine that an infant receives is not only perturbing the immune system but is educating the immune system and laying down the foundation for all subsequent responses. This leads to the question, is there an optimum way to educate the immune system? Should this be taken into consideration in our vaccination protocols?
Collapse
Affiliation(s)
- Anna Gil
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Laurie L Kenney
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Rabinarayan Mishra
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Levi B Watkin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Nuray Aslan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Liisa K Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
42
|
Ganapathi L, Van Haren S, Dowling DJ, Bergelson I, Shukla NM, Malladi SS, Balakrishna R, Tanji H, Ohto U, Shimizu T, David SA, Levy O. The Imidazoquinoline Toll-Like Receptor-7/8 Agonist Hybrid-2 Potently Induces Cytokine Production by Human Newborn and Adult Leukocytes. PLoS One 2015; 10:e0134640. [PMID: 26274907 PMCID: PMC4537157 DOI: 10.1371/journal.pone.0134640] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/11/2015] [Indexed: 11/30/2022] Open
Abstract
Background Newborns and young infants are at higher risk for infections than adults, and manifest suboptimal vaccine responses, motivating a search for novel immunomodulators and/or vaccine adjuvants effective in early life. In contrast to most TLR agonists (TLRA), TLR8 agonists such as imidazoquinolines (IMQs) induce adult-level Th1-polarizing cytokine production from human neonatal cord blood monocytes and are candidate early life adjuvants. We assessed whether TLR8-activating IMQ congeners may differ in potency and efficacy in inducing neonatal cytokine production in vitro, comparing the novel TLR7/8-activating IMQ analogues Hybrid-2, Meta-amine, and Para-amine to the benchmark IMQ resiquimod (R848). Methods TLRA-induced NF-κB activation was measured in TLR-transfected HEK cells. Cytokine production in human newborn cord and adult peripheral blood and in monocyte-derived dendritic cell cultures were measured by ELISA and multiplex assays. X-ray crystallography characterized the interaction of human TLR8 with Hybrid-2. Results Hybrid-2 selectively activated both TLR7 and 8 and was more potent than R848 in inducing adult-like levels of TNF-α, and IL-1β. Consistent with its relatively high in vitro activity, crystallographic studies suggest that absence in Hybrid-2 of an ether oxygen of the C2-ethoxymethyl substituent, which can engage in unfavorable electrostatic and/or dipolar interactions with the carbonyl oxygen of Gly572 in human TLR8, may confer greater efficacy and potency compared to R848. Conclusions Hybrid-2 is a selective and potent TLR7/8 agonist that is a candidate adjuvant for early life immunization.
Collapse
Affiliation(s)
- Lakshmi Ganapathi
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Simon Van Haren
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - David J. Dowling
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Ilana Bergelson
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
| | - Nikunj M. Shukla
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Subbalakshmi S. Malladi
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Rajalakshmi Balakrishna
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Hiromi Tanji
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Umeharu Ohto
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Toshiyuki Shimizu
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Sunil A. David
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Ofer Levy
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
43
|
Qian L, Weng XW, Chen W, Sun CH, Wu J. TREM-1 as a potential therapeutic target in neonatal sepsis. Int J Clin Exp Med 2014; 7:1650-1658. [PMID: 25126161 PMCID: PMC4132125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/28/2014] [Indexed: 06/03/2023]
Abstract
OBJECTIVE Bacterial sepsis in neonates is associated with elevated morbidity and mortality. A role for the pro-inflammatory Triggering Receptor Expressed on Myeloid cells-1 (TREM-1) is suspected in the innate immune response to bacteria, but little is known about its activities in infants. To begin exploring the feasibility of treating neonatal sepsis by blocking leukocyte TREM-1, we compared TREM-1 membrane expression and mRNA in newborns without clinical or microbiological evidence of infection, to that of healthy adults. The functionality of pro-inflammatory reactions in leukocyte TREM-1 of newborns was also evaluated. METHODS Twenty term newborns were enrolled in this study and cord blood samples were collected at birth. For comparison, peripheral blood specimens were collected from 20 healthy adults (control adult, CA). The expression of TREM-1 protein and mRNA in leukocytes was detected with flow cytometry and real-time qPCR, respectively. Whole cord blood was also stimulated by Escherichia coli or blocked by the TREM-1-specific peptide LP17 to identify changes in the secretion of pro-inflammatory cytokines interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α, as well as soluble TREM-1 (sTREM-1) using enzyme linked immunosorbent assay (ELISA). RESULTS Mean fluorescence intensity (MFI) of TREM-1 on leukocytes of newborns appeared comparable to healthy adults [monocytes: 37.5 ± 6.7 vs. 37.6 ± 8.7; polymorphonuclear cells (PMNs): 32.9 ± 6.6 vs. 33.6 ± 5.8]. However, the percentage of PMNs positive for TREM-1 was lower in newborns than in healthy adults (82.3 ± 7.1 vs. 98.6 ± 4.8; P < 0.01); the percentage of TREM-1-positive CD14-positive monocytes was comparable to that of healthy adults (97.1 ± 8.3 vs. 97.5 ± 7.4). Exposure of cord blood to E. coli resulted in increased secretion of IL-6, IL-8, TNF-α, and sTREM-1. In contrast, the concentrations of IL-6, IL-8, and TNF-α decreased by a minimum of 15% when TREM-1 was blocked by LP17 then exposed to E. coli, versus E. coli alone. In addition, the concentration of sTREM-1 was positively correlated with the levels of TNF-α (r = 0.519, P < 0.05), IL-6 (r = 0.507, P < 0.05), and IL-8 (r = 0.538, P < 0.05). CONCLUSIONS Healthy newborns exhibit expression of TREM-1 on monocytes similar to that in healthy adults, and most PMNs express TREM-1 at the newborn stage. Detection of sTREM-1 in neonatal peripheral blood should be further investigated as a potential method for the diagnosis of neonatal infection. Finally, blocking the TREM-1 signal transduction pathway may reduce inflammatory responses of neonate leukocytes and thereby provide a new strategy for treatment of neonatal infection.
Collapse
Affiliation(s)
- Lei Qian
- Department of Laboratory Medicine, Binghai County People’s HospitalJiangsu Province, China
| | - Xiao-Wen Weng
- Department of Laboratory Medicine, Binghai County People’s HospitalJiangsu Province, China
| | - Wei Chen
- Department of Laboratory Medicine, Binghai County People’s HospitalJiangsu Province, China
| | - Chang-Hong Sun
- Department of Laboratory Medicine, Binghai County People’s HospitalJiangsu Province, China
| | - Jian Wu
- Department of Laboratory Medicine, Yancheng People’s First HospitalJiangsu Province, China
| |
Collapse
|
44
|
Palmer CD, Ninković J, Prokopowicz ZM, Mancuso CJ, Marin A, Andrianov AK, Dowling DJ, Levy O. The effect of stable macromolecular complexes of ionic polyphosphazene on HIV Gag antigen and on activation of human dendritic cells and presentation to T-cells. Biomaterials 2014; 35:8876-8886. [PMID: 25023392 DOI: 10.1016/j.biomaterials.2014.06.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/22/2014] [Indexed: 12/19/2022]
Abstract
Neonates and infants are susceptible to infection due to distinct immune responses in early life. Therefore, development of vaccine formulation and delivery systems capable of activating human newborn leukocytes is of global health importance. Poly[di(carboxylatophenoxy)phosphazene] (PCPP) belongs to a family of ionic synthetic polyphosphazene polyelectrolyte compounds that can form non-covalent interactions with protein antigens and demonstrate adjuvant activity in animals and in human clinical trials. However, little is known about their ability to activate human immune cells. In this study, we characterized the effects of PCPP alone or in combination with a model antigen (recombinant HIV-Gag (Gag)), on the maturation, activation and antigen presentation by human adult and newborn dendritic cells (DCs) in vitro. PCPP treatment induced DC activation as assessed by upregulation of co-stimulatory molecules and cytokine production. Studies benchmarking PCPP to Alum, the most commonly used vaccine adjuvant, demonstrated that both triggered cell death and release of danger signals in adult and newborn DCs. When complexed with Gag antigen, PCPP maintained its immunostimulatory characteristics while permitting internalization and presentation of Gag by DCs to HIV-Gag-specific CD4(+) T cell clones. The PCPP vaccine formulation outlined here has intrinsic adjuvant activity, can facilitate effective delivery of antigen to DCs, and may be advantageous for induction of beneficial T cell-mediated immunity. Moreover, polyphosphazenes can further reduce cost of vaccine production and distribution through their dose-sparing and antigen-stabilizing properties, thus potentially eliminating the need for cold chain distribution.
Collapse
Affiliation(s)
- Christine D Palmer
- Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jana Ninković
- Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Zofia M Prokopowicz
- Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | | | | | - David J Dowling
- Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
45
|
Dowling DJ, Levy O. Ontogeny of early life immunity. Trends Immunol 2014; 35:299-310. [PMID: 24880460 DOI: 10.1016/j.it.2014.04.007] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/29/2014] [Accepted: 04/30/2014] [Indexed: 12/18/2022]
Abstract
The human immune system comprises cellular and molecular components designed to coordinately prevent infection while avoiding potentially harmful inflammation and autoimmunity. Immunity varies with age, reflecting unique age-dependent challenges including fetal gestation, the neonatal phase, and infancy. Here, we review novel mechanistic insights into early life immunity, with an emphasis on emerging models of human immune ontogeny, which may inform age-specific translational development of novel anti-infectives, immunomodulators, and vaccines.
Collapse
Affiliation(s)
- David J Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Abstract
Most infants born to human immunodeficiency virus (HIV)-infected women escape HIV infection. Infants evade infection despite an immature immune system and, in the case of breastfeeding, prolonged repetitive exposure. If infants become infected, the course of their infection and response to treatment differs dramatically depending upon the timing (in utero, intrapartum, or during breastfeeding) and potentially the route of their infection. Perinatally acquired HIV infection occurs during a critical window of immune development. HIV's perturbation of this dynamic process may account for the striking age-dependent differences in HIV disease progression. HIV infection also profoundly disrupts the maternal immune system upon which infants rely for protection and immune instruction. Therefore, it is not surprising that infants who escape HIV infection still suffer adverse effects. In this review, we highlight the unique aspects of pediatric HIV transmission and pathogenesis with a focus on mechanisms by which HIV infection during immune ontogeny may allow discovery of key elements for protection and control from HIV.
Collapse
|
47
|
Levy O, Wynn JL. A prime time for trained immunity: innate immune memory in newborns and infants. Neonatology 2013; 105:136-41. [PMID: 24356292 PMCID: PMC3946366 DOI: 10.1159/000356035] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 09/23/2013] [Indexed: 01/10/2023]
Abstract
The newborn and infant periods of early life are associated with heightened vulnerability to infection. Limited antigen exposure and distinct adaptive immune function compared to the adult places a greater burden on innate immunity for host defense to microbial challenge during this time. Trained immunity describes the phenomenon of augmented innate immune function following a stimulus that is not specific to the original stimulus. We review the concept of trained immunity in the context of the newborn's unique innate immune system function, the preclinical and clinical evidence that supports the tenet of innate immune memory in early life, and potential consequences of altered innate immune host responses.
Collapse
Affiliation(s)
- Ofer Levy
- Staff Physician, Department of Medicine, Division of Infectious Diseases, Children’s Hospital Boston, and Associate Professor, Harvard Medical School; Boston, Massachusetts
| | - James L. Wynn
- Assistant Professor of Pediatrics, Department of Pediatrics, Division of Neonatology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
48
|
Wynn JL, Hansen NI, Das A, Cotten CM, Goldberg RN, Sánchez PJ, Bell EF, Van Meurs KP, Carlo WA, Laptook AR, Higgins RD, Benjamin DK, Stoll BJ. Early sepsis does not increase the risk of late sepsis in very low birth weight neonates. J Pediatr 2013; 162:942-8.e1-3. [PMID: 23295144 PMCID: PMC3622770 DOI: 10.1016/j.jpeds.2012.11.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 10/05/2012] [Accepted: 11/07/2012] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To examine whether preterm very low birth weight (VLBW) infants have an increased risk of late-onset sepsis (LOS) following early-onset sepsis (EOS). STUDY DESIGN Retrospective analysis of VLBW infants (401-1500 g) born September 1998 through December 2009 who survived >72 hours and were cared for within the National Institute of Child Health and Human Development Neonatal Research Network. Sepsis was defined by growth of bacteria or fungi in a blood culture obtained ≤ 72 hours of birth (EOS) or >72 hours (LOS) and antimicrobial therapy for ≥ 5 days or death <5 days while receiving therapy. Regression models were used to assess risk of death or LOS by 120 days and LOS by 120 days among survivors to discharge or 120 days, adjusting for gestational age and other covariates. RESULTS Of 34,396 infants studied, 504 (1.5%) had EOS. After adjustment, risk of death or LOS by 120 days did not differ overall for infants with EOS compared with those without EOS [risk ratio (RR): 0.99 (0.89-1.09)] but was reduced in infants born at <25 weeks gestation [RR: 0.87 (0.76-0.99), P = .048]. Among survivors, no difference in LOS risk was found overall for infants with versus without EOS [RR: 0.88 (0.75-1.02)], but LOS risk was reduced in infants with birth weight 401-750 g who had EOS [RR: 0.80 (0.64-0.99), P = .047]. CONCLUSIONS Risk of LOS after EOS was not increased in VLBW infants. Surprisingly, risk of LOS following EOS appeared to be reduced in the smallest, most premature infants, underscoring the need for age-specific analyses of immune function.
Collapse
Affiliation(s)
- James L Wynn
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Flanagan KL, van Crevel R, Curtis N, Shann F, Levy O. Heterologous ("nonspecific") and sex-differential effects of vaccines: epidemiology, clinical trials, and emerging immunologic mechanisms. Clin Infect Dis 2013; 57:283-9. [PMID: 23572484 DOI: 10.1093/cid/cit209] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence from epidemiologic, clinical, and immunologic studies indicates that vaccines can influence morbidity and mortality independent of vaccine-specific B-cell or T-cell immunity. For example, the live attenuated measles vaccine and BCG vaccine may reduce mortality from infections other than measles or tuberculosis, respectively. Immunologists call these heterologous effects and epidemiologists have called them nonspecific effects, indicating that they manifest against a broad range of pathogens/disease. These effects differ by sex, can be beneficial or detrimental, and appear to be mediated by mechanisms including innate immune memory (also known as "trained immunity") and cross-reacting lymphocytes. Herein we review recent studies in this emerging field based on a meeting of experts, the recent Optimmunize meeting, held in Copenhagen, Denmark, in August 2012. Further characterization of these effects is likely to expand the way vaccines are evaluated and alter the manner and sequence in which they are given.
Collapse
|
50
|
Dowling DJ, Tan Z, Prokopowicz ZM, Palmer CD, Matthews MAH, Dietsch GN, Hershberg RM, Levy O. The ultra-potent and selective TLR8 agonist VTX-294 activates human newborn and adult leukocytes. PLoS One 2013; 8:e58164. [PMID: 23483986 PMCID: PMC3587566 DOI: 10.1371/journal.pone.0058164] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 02/01/2013] [Indexed: 11/20/2022] Open
Abstract
Background Newborns display distinct immune responses that contribute to susceptibility to infection and reduced vaccine responses. Toll-like receptor (TLR) agonists may serve as vaccine adjuvants, when given individually or in combination, but responses of neonatal leukocytes to many TLR agonists are diminished. TLR8 agonists are more effective than other TLR agonists in activating human neonatal leukocytes in vitro, but little is known about whether different TLR8 agonists may distinctly activate neonatal leukocytes. We characterized the in vitro immuno-stimulatory activities of a novel benzazepine TLR8 agonist, VTX-294, in comparison to imidazoquinolines that activate TLR8 (R-848; (TLR7/8) CL075; (TLR8/7)), with respect to activation of human newborn and adult leukocytes. Effects of VTX-294 and R-848 in combination with monophosphoryl lipid A (MPLA; TLR4) were also assessed. Methods TLR agonist specificity was assessed using TLR-transfected HEK293 cells expressing a NF-κB reporter gene. TLR agonist-induced cytokine production was measured in human newborn cord and adult peripheral blood using ELISA and multiplex assays. Newborn and adult monocytes were differentiated into monocyte-derived dendritic cells (MoDCs) and TLR agonist-induced activation assessed by cytokine production (ELISA) and co-stimulatory molecule expression (flow cytometry). Results VTX-294 was ∼100x more active on TLR8- than TLR7-transfected HEK cells (EC50, ∼50 nM vs. ∼5700 nM). VTX-294-induced TNF and IL-1β production were comparable in newborn cord and adult peripheral blood, while VTX-294 was ∼ 1 log more potent in inducing TNF and IL-1β production than MPLA, R848 or CL075. Combination of VTX-294 and MPLA induced greater blood TNF and IL-1β responses than combination of R-848 and MPLA. VTX-294 also potently induced expression of cytokines and co-stimulatory molecules HLA-DR and CD86 in human newborn MoDCs. Conclusions VTX-294 is a novel ultra-potent TLR8 agonist that activates newborn and adult leukocytes and is a candidate vaccine adjuvant in both early life and adulthood.
Collapse
Affiliation(s)
- David J. Dowling
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Zhen Tan
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Pediatrics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zofia M. Prokopowicz
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christine D. Palmer
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Gregory N. Dietsch
- VentiRx Pharmaceuticals, Inc., Seattle, Washington, United States of America
| | - Robert M. Hershberg
- VentiRx Pharmaceuticals, Inc., Seattle, Washington, United States of America
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|