1
|
Brown L, Bingham J, Pulliam J, Mthombothi Z, Sereo T, Kamupira M, Botha S, Molema K, Maseti E, Schönfeldt M, Mabhena N, Prabdial-Sing N, von Gottberg A, McCarthy K, van Schalkwyk C. Estimation of the poliovirus type 2 immunity gap in South Africa. Vaccine 2024; 42:126062. [PMID: 38969540 PMCID: PMC11413476 DOI: 10.1016/j.vaccine.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 07/07/2024]
Abstract
In the context of polio eradication efforts, accurate assessment of vaccination programme effectiveness is essential to public health planning and decision making. Such assessments are often based on zero-dose children, estimated using the number of children who did not receive the first dose of the Diphtheria-Tetanus-Pertussis containing vaccine as a proxy. Our study introduces a novel approach to directly estimate the number of children susceptible to poliovirus type 2 (PV2) and uses this approach to provide district-level estimates for South Africa of susceptible children born between 2017 and 2022. We used district-level data on annual doses of inactivated poliovirus vaccine (IPV) administered, live births, and population sizes, from 2017 through 2022. We imputed missing vaccination data, implemented flexible assumptions regarding dose distribution in the eligible population, and used estimated efficacy values for one, two, three, and four doses of IPV, to compute the number of susceptible and immune children by birth year. We validated our approach by comparing an intermediary output with zero-dose children (ZDC) estimated using data reported by WHO/UNICEF Estimates of National Immunization Coverage (WUENIC). Our results indicate high heterogeneity in susceptibility to PV2 across South Africa's 52 districts as of the end of 2022. In children under 5 years, PV2 susceptibility ranged from approximately 30 % in districts including Xhariep (31.9 %), Ekurhuleni (30.1 %), and Central Karoo (29.8 %), to less than 4 % in Sarah Baartman (1.9 %), Buffalo City (2.1 %), and eThekwini (3.2 %). Our susceptibility estimates were consistently higher than ZDC over the timeframe. We estimated that ZDC decreased nationally from 155,168 (152,737-158,523) in 2017 to 108,593 in 2021, and increased to 127,102 in 2022, a trend consistent with ZDC derived from data reported by WUENIC. While our approach provides a more comprehensive profile of PV2 susceptibility, our susceptibility and ZDC estimates generally agree in the ranking of districts according to risk.
Collapse
Affiliation(s)
- Lauren Brown
- South African Centre for Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | - Jeremy Bingham
- South African Centre for Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa.
| | - Juliet Pulliam
- South African Centre for Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | - Zinhle Mthombothi
- South African Centre for Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | - Tumelo Sereo
- South African Centre for Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| | | | - Sonia Botha
- Western Cape Department of Health, Expanded Programme on Immunisation, City of Cape Town, South Africa
| | - Koko Molema
- National Department of Health, Expanded Programme on Immunisation, Pretoria, South Africa
| | - Elizabeth Maseti
- National Department of Health, Expanded Programme on Immunisation, Pretoria, South Africa
| | - Marione Schönfeldt
- National Department of Health, Expanded Programme on Immunisation, Pretoria, South Africa
| | | | - Nishi Prabdial-Sing
- National Institute for Communicable Diseases, Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Anne von Gottberg
- National Institute for Communicable Diseases, Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Kerrigan McCarthy
- National Institute for Communicable Diseases, Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - Cari van Schalkwyk
- South African Centre for Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
2
|
Verhasselt V, Marchant A, Kollmann TR. Per Os to Protection - Targeting the Oral Route to Enhance Immune-mediated Protection from Disease of the Human Newborn. J Mol Biol 2024; 436:168718. [PMID: 39094783 DOI: 10.1016/j.jmb.2024.168718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Affiliation(s)
- Valerie Verhasselt
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, School of Medicine, University of Western Australia, Perth, WA, Australia; Immunology and Breastfeeding Team, Telethon Kids Institute, Perth, WA, Australia
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Tobias R Kollmann
- Dalhousie University, Department of Microbiology & Immunology, Pediatric Infectious Diseases, Canada.
| |
Collapse
|
3
|
Riller Q, Schmutz M, Fourgeaud J, Fischer A, Neven B. Protective role of antibodies in enteric virus infections: Lessons from primary and secondary immune deficiencies. Immunol Rev 2024. [PMID: 39340232 DOI: 10.1111/imr.13402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Enteric viruses are the main cause of acute gastroenteritis worldwide with a significant morbidity and mortality, especially among children and aged adults. Some enteric viruses also cause disseminated infections and severe neurological manifestations such as poliomyelitis. Protective immunity against these viruses is not well understood in humans, with most knowledge coming from animal models, although the development of poliovirus and rotavirus vaccines has extended our knowledge. In a classical view, innate immunity involves the recognition of foreign DNA or RNA by pathogen recognition receptors leading to the production of interferons and other inflammatory cytokines. Antigen uptake and presentation to T cells and B cells then activate adaptive immunity and, in the case of the mucosal immunity, induce the secretion of dimeric IgA, the more potent immunoglobulins in viral neutralization. The study of Inborn errors of immunity (IEIs) offers a natural opportunity to study nonredundant immunity toward pathogens. In the case of enteric viruses, patients with a defective production of antibodies are at risk of developing neurological complications. Moreover, a recent description of patients with low or absent antibody production with protracted enteric viral infections associated with hepatitis reinforces the prominent role of B cells and immunoglobulins in the control of enteric virus.
Collapse
Affiliation(s)
- Quentin Riller
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France
- IHU-Imagine, Paris, France
| | - Muriel Schmutz
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France
- IHU-Imagine, Paris, France
| | - Jacques Fourgeaud
- Université Paris Cité, FETUS, Paris, France
- Microbiology Department, AP-HP, Hôpital Necker, Paris, France
| | - Alain Fischer
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM UMRS 1163, Institut Imagine, Paris, France
- Collège de France, Paris, France
| | - Bénédicte Neven
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France
- IHU-Imagine, Paris, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
4
|
Saleem AF, Kazi ZU, Zehra SM, Parkar S, Macklin G, Sifontes G, Mainou BA, Alam M, Lopez Cavestany R, Mach O. Mucosal Immunity to Poliovirus in Children 0-15 Years of Age: A Community-Based Study in Karachi, Pakistan in 2019. J Infect Dis 2024; 230:736-740. [PMID: 38195177 DOI: 10.1093/infdis/jiae006] [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: 07/13/2023] [Revised: 12/30/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024] Open
Abstract
This study assesses poliovirus type 1 (PV1) immunity in children to inform the contribution of mucosal immunity in and prevention of poliovirus circulation. A community-based study was conducted in periurban Karachi, Pakistan. Randomly selected children (0-15 years of age) received oral poliovirus vaccine (OPV) challenge dose. Blood and stool samples were collected at several time points and evaluated for polio-neutralizing antibodies and serotype-specific poliovirus, respectively. Eighty-one of 589 (14%) children excreted PV1 7 days post-OPV challenge; 70 of 81 (86%) were seropositive at baseline. Twelve of 610 (2%) were asymptomatic wild poliovirus type 1 (WPV1) excretors. Most poliovirus excretors had humoral immunity, suggesting mucosal immunity in these children likely waned or never developed. Without mucosal immunity, they are susceptible to poliovirus infection, shedding, and transmission. Asymptomatic WPV1 excretion suggests undetected poliovirus circulation within the community.
Collapse
Affiliation(s)
- Ali Faisal Saleem
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zaubina Umar Kazi
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Sadia Parkar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Grace Macklin
- Polio Eradication, World Health Organization, Geneva, Switzerland
| | - Giovanna Sifontes
- Polio and Picornavirus Laboratory Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Bernardo A Mainou
- Polio and Picornavirus Laboratory Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Masroor Alam
- Department of Virology, National Institute of Health, Islamabad, Pakistan
| | | | - Ondrej Mach
- Polio Eradication, World Health Organization, Geneva, Switzerland
| |
Collapse
|
5
|
Biswas M, Nurunnabi M, Khatun Z. Understanding Mucosal Physiology and Rationale of Formulation Design for Improved Mucosal Immunity. ACS APPLIED BIO MATERIALS 2024; 7:5037-5056. [PMID: 38787767 DOI: 10.1021/acsabm.4c00395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
The oral and nasal cavities serve as critical gateways for infectious pathogens, with microorganisms primarily gaining entry through these routes. Our first line of defense against these invaders is the mucosal membrane, a protective barrier that shields the body's internal systems from infection while also contributing to vital functions like air and nutrient intake. One of the key features of this mucosal barrier is its ability to protect the physiological system from pathogens. Additionally, mucosal tolerance plays a crucial role in maintaining homeostasis by regulating the pH and water balance within the body. Recognizing the importance of the mucosal barrier, researchers have developed various mucosal formulations to enhance the immune response. Mucosal vaccines, for example, deliver antigens directly to mucosal tissues, triggering local immune stimulation and ultimately inducing systemic immunity. Studies have shown that lipid-based formulations such as liposomes and virosomes can effectively elicit both local and systemic immune responses. Furthermore, mucoadhesive polymeric particles, with their prolonged delivery to target sites, have demonstrated an enhanced immune response. This Review delves into the critical role of material selection and delivery approaches in optimizing mucosal immunity.
Collapse
Affiliation(s)
- Mila Biswas
- Department of Electrical and Computer Engineering, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Department of Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Zehedina Khatun
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| |
Collapse
|
6
|
He X, Jiang N, Li Y. Saccharomyces cerevisiae cells that display norovirus P induce both systemic and mucosal neutralizing antibodies. Virology 2024; 594:110034. [PMID: 38460411 DOI: 10.1016/j.virol.2024.110034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/11/2024]
Abstract
The human norovirus (HuNov) is the leading cause of acute gastroenteritis (AGE) worldwide. Mucosal secretory IgA (sIgA) in the gastrointestinal tract interrupts the interaction between host cells and HuNov, thus inhibiting viral infection. In this study, we constructed a recombinant Saccharomyces cerevisiae (S. cerevisiae) expressing the HuNov P protein (GII. 4) and evaluated its immunogenicity in mice after oral delivery. First, the recombinant S. cerevisiae (EBY100/pYD1-P) efficiently expressed P, as evidenced by western blotting and indirect fluorescent assay. Second, after oral administration, EBY100/pYD1-P, especially the high-dose group (5 × 109 clone formation units), elicited systemic and mucosal immune responses characterized by significant sera IgG, IgA, and mucosal sIgA. More importantly, these antibodies showed a substantial neutralization effect against P. Lastly, EBY100/pYD1-P induced significant P-specific IFN-γ-secreting T cells and IL4-secreting T cells. Collectively, the recombinant S. cerevisiae expressing HuNov P is a promising mucosal vaccine candidate against HuNov.
Collapse
Affiliation(s)
- Xin He
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary, Huazhong Agricultural University, Wuhan, China
| | - Nianzhu Jiang
- School of Biological Engineering and Food, Hubei University of Technology, Wuhan, China
| | - Yaoming Li
- Department of Biology of Mucosal Pathogen, College of Life Science and Technology, Wuhan University of Bioengineering, Wuhan, China; Applied Biotechnology Research Center, Wuhan University of Bioengineering, Wuhan, China.
| |
Collapse
|
7
|
Thompson KM, Badizadegan K. Review of Poliovirus Transmission and Economic Modeling to Support Global Polio Eradication: 2020-2024. Pathogens 2024; 13:435. [PMID: 38921733 PMCID: PMC11206708 DOI: 10.3390/pathogens13060435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 06/27/2024] Open
Abstract
Continued investment in the development and application of mathematical models of poliovirus transmission, economics, and risks leads to their use in support of polio endgame strategy development and risk management policies. This study complements an earlier review covering the period 2000-2019 and discusses the evolution of studies published since 2020 by modeling groups supported by the Global Polio Eradication Initiative (GPEI) partners and others. We systematically review modeling papers published in English in peer-reviewed journals from 2020-2024.25 that focus on poliovirus transmission and health economic analyses. In spite of the long-anticipated end of poliovirus transmission and the GPEI sunset, which would lead to the end of its support for modeling, we find that the number of modeling groups supported by GPEI partners doubled and the rate of their publications increased. Modeling continued to play a role in supporting GPEI and national/regional policies, but changes in polio eradication governance, decentralized management and decision-making, and increased heterogeneity in modeling approaches and findings decreased the overall impact of modeling results. Meanwhile, the failure of the 2016 globally coordinated cessation of type 2 oral poliovirus vaccine use for preventive immunization and the introduction of new poliovirus vaccines and formulation, increased the complexity and uncertainty of poliovirus transmission and economic models and policy recommendations during this time.
Collapse
|
8
|
Ma B, Tao M, Li Z, Zheng Q, Wu H, Chen P. Mucosal vaccines for viral diseases: Status and prospects. Virology 2024; 593:110026. [PMID: 38373360 DOI: 10.1016/j.virol.2024.110026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Virus-associated infectious diseases are highly detrimental to human health and animal husbandry. Among all countermeasures against infectious diseases, prophylactic vaccines, which developed through traditional or novel approaches, offer potential benefits. More recently, mucosal vaccines attract attention for their extraordinary characteristics compared to conventional parenteral vaccines, particularly for mucosal-related pathogens. Representatively, coronavirus disease 2019 (COVID-19), a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), further accelerated the research and development efforts for mucosal vaccines by thoroughly investigating existing strategies or involving novel techniques. While several vaccine candidates achieved positive progresses, thus far, part of the current COVID-19 mucosal vaccines have shown poor performance, which underline the need for next-generation mucosal vaccines and corresponding platforms. In this review, we summarized the typical mucosal vaccines approved for humans or animals and sought to elucidate the underlying mechanisms of these successful cases. In addition, mucosal vaccines against COVID-19 that are in human clinical trials were reviewed in detail since this public health event mobilized all advanced technologies for possible solutions. Finally, the gaps in developing mucosal vaccines, potential solutions and prospects were discussed. Overall, rational application of mucosal vaccines would facilitate the establishing of mucosal immunity and block the transmission of viral diseases.
Collapse
Affiliation(s)
- Bingjie Ma
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Mengxiao Tao
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Zhili Li
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Quanfang Zheng
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Haigang Wu
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Peirong Chen
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China.
| |
Collapse
|
9
|
Marchant A, Van Damme P, Plotkin S, Neels P, Cassetti MC, Cramer J, Gruber MF, Goldblatt D, King D, Hartig-Merkel W, Vandeputte J. Enabling the evaluation of COVID-19 vaccines with correlates of protection. Biologicals 2024; 85:101723. [PMID: 37976940 DOI: 10.1016/j.biologicals.2023.101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
In February 2023, a meeting about correlates of protection (CoPs) against COVID-19 was organized by the International Alliance for Biological Standardization, the European Plotkin Institute for Vaccinology, and Vaccinopolis. The meeting aimed at reviewing the evidence, drawing conclusions, and identifying knowledge gaps. Collection of evidence is not straightforward. Neutralizing antibodies correlate with protection and are used for immunobridging studies within and between vaccine platforms for approval of new COVID-19 vaccines. In preparation for the next pandemic, it is vital that rapidly authorized initial vaccines are available to perform immunobridging studies very early. Additional components of the immune response likely contribute to protection against symptomatic infection. Current evidence is strongest for T lymphocytes and binding antibodies. Further studies are needed to consolidate this evidence and define their potential role in the evaluation of vaccines. For evaluation of mucosal vaccines, identifying CoPs against infection and transmission is key; further research is needed to identify and standardize methods suitable for clinical studies. CoPs for broadly protective beta-coronavirus vaccines remain a critical area of research. The knowledge, expertise, and capacity exist to conduct clinical studies using different designs in different populations to discover and validate CoPs, facilitating and accelerating evaluation of novel vaccines/vaccination platforms.
Collapse
Affiliation(s)
- Arnaud Marchant
- Institute for Medical Immunology (IMI), European Plotkin Institute for Vaccinology (EPIV), Université libre de Bruxelles, 808 Route de Lennik, 1070, Brussels, Belgium.
| | - Pierre Van Damme
- Centre for the Evaluation of Vaccination, European Plotkin Institute for Vaccinology (EPIV), Vaccine & Infectious Disease Institute, University of Antwerp, Faculty of Medicine & Health Sciences, Vaccinopolis/University of Antwerp, Drie Eikenstraat 663, 2650, Edegem, Belgium.
| | - Stanley Plotkin
- University of Pennsylvania, 18902, Doylestown, Pennsylvania, United States.
| | - Pieter Neels
- International Alliance for Biological Standardization (IABS), Rue Mina-Audemars 3, 1204, Genève, Switzerland.
| | - Maria Cristina Cassetti
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), 20852, United States.
| | - Jakob Cramer
- Coalition for Epidemic Preparedness Innovations (CEPI), Gibbs Building, 215 Euston Rd, Bloomsbury, London, NW1 2BE, United Kingdom.
| | - Marion F Gruber
- International AIDS Vaccine Initiative (IAVI), 125 Broad Street, New York, NY, 10004, United States.
| | - David Goldblatt
- Infection, Immunity, and Inflammation Department, Great Ormond Street Institute of Child Health, University College London and Great Ormond Street Children's Hospital, NHS Foundation Trust, 30 Guilford Street, London, WC1N 1EH, United Kingdom.
| | - Deborah King
- Wellcome Trust, Gibbs Building, 215 Euston Road, Bloomsbury, London, NW1 2BE, United Kingdom.
| | - Wendy Hartig-Merkel
- P95 Epidemiology & Pharmacovigilance, Koning Leopold III Laan 1, 3001, Leuven, Belgium.
| | - Joris Vandeputte
- International Alliance for Biological Standardization (IABS), Rue Mina-Audemars 3, 1204, Genève, Switzerland.
| |
Collapse
|
10
|
Devaux CA, Pontarotti P, Levasseur A, Colson P, Raoult D. Is it time to switch to a formulation other than the live attenuated poliovirus vaccine to prevent poliomyelitis? Front Public Health 2024; 11:1284337. [PMID: 38259741 PMCID: PMC10801389 DOI: 10.3389/fpubh.2023.1284337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
The polioviruses (PVs) are mainly transmitted by direct contact with an infected person through the fecal-oral route and respiratory secretions (or more rarely via contaminated water or food) and have a primary tropism for the gut. After their replication in the gut, in rare cases (far less than 1% of the infected individuals), PVs can spread to the central nervous system leading to flaccid paralysis, which can result in respiratory paralysis and death. By the middle of the 20th century, every year the wild polioviruses (WPVs) are supposed to have killed or paralyzed over half a million people. The introduction of the oral poliovirus vaccines (OPVs) through mass vaccination campaigns (combined with better application of hygiene measures), was a success story which enabled the World Health Organization (WHO) to set the global eradication of poliomyelitis as an objective. However this strategy of viral eradication has its limits as the majority of poliomyelitis cases today arise in individuals infected with circulating vaccine-derived polioviruses (cVDPVs) which regain pathogenicity following reversion or recombination. In recent years (between January 2018 and May 2023), the WHO recorded 8.8 times more cases of polio which were linked to the attenuated OPV vaccines (3,442 polio cases after reversion or recombination events) than cases linked to a WPV (390 cases). Recent knowledge of the evolution of RNA viruses and the exchange of genetic material among biological entities of the intestinal microbiota, call for a reassessment of the polio eradication vaccine strategies.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique (CNRS-SNC5039), Marseille, France
| | - Pierre Pontarotti
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique (CNRS-SNC5039), Marseille, France
| | - Anthony Levasseur
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Philippe Colson
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| |
Collapse
|
11
|
de Swart RL, Belov GA. Advantages and challenges of Newcastle disease virus as a vector for respiratory mucosal vaccines. Curr Opin Virol 2023; 62:101348. [PMID: 37591130 DOI: 10.1016/j.coviro.2023.101348] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 07/18/2023] [Indexed: 08/19/2023]
Abstract
Newcastle disease virus (NDV) is an avian pathogen with an unsegmented negative-strand RNA genome. Properties such as the ease of genome modification, respiratory tract tropism, and self-limiting replication in mammals make NDV an attractive vector for vaccine development. Experimental NDV-based vaccines against multiple human and animal pathogens elicited both systemic and mucosal immune responses and were protective in preclinical animal studies, but their real-life efficacy remains to be demonstrated. Only recently, the first results of clinical trials of NDV-based vaccines against SARS-CoV-2 became available, highlighting the challenges that need to be overcome to fully realize the potential of NDV as a platform for the rapid development of economically affordable and effective mucosal vaccines.
Collapse
Affiliation(s)
- Rik L de Swart
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, the Netherlands.
| | - George A Belov
- Department of Veterinary Medicine and Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD, USA.
| |
Collapse
|
12
|
Snider CJ, Zaman K, Wilkinson AL, Binte Aziz A, Yunus M, Haque W, Jones KAV, Wei L, Estivariz CF, Konopka-Anstadt JL, Mainou BA, Patel JC, Lickness JS, Pallansch MA, Wassilak SGF, Steven Oberste M, Anand A. Poliovirus type 1 systemic humoral and intestinal mucosal immunity induced by monovalent oral poliovirus vaccine, fractional inactivated poliovirus vaccine, and bivalent oral poliovirus vaccine: A randomized controlled trial. Vaccine 2023; 41:6083-6092. [PMID: 37652822 PMCID: PMC10895964 DOI: 10.1016/j.vaccine.2023.08.055] [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: 06/20/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND To inform response strategies, we examined type 1 humoral and intestinal immunity induced by 1) one fractional inactivated poliovirus vaccine (fIPV) dose given with monovalent oral poliovirus vaccine (mOPV1), and 2) mOPV1 versus bivalent OPV (bOPV). METHODS We conducted a randomized, controlled, open-label trial in Dhaka, Bangladesh. Healthy infants aged 5 weeks were block randomized to one of four arms: mOPV1 at age 6-10-14 weeks/fIPV at 6 weeks (A); mOPV1 at 6-10-14 weeks/fIPV at 10 weeks (B); mOPV1 at 6-10-14 weeks (C); and bOPV at 6-10-14 weeks (D). Immune response at 10 weeks and cumulative response at 14 weeks was assessed among the modified intention-to-treat population, defined as seroconversion from seronegative (<1:8 titers) to seropositive (≥1:8) or a four-fold titer rise among seropositive participants sustained to age 18 weeks. We examined virus shedding after two doses of mOPV1 with and without fIPV, and after the first mOPV1 or bOPV dose. The trial is registered at ClinicalTrials.gov (NCT03722004). FINDINGS During 18 December 2018 - 23 November 2019, 1,192 infants were enrolled (arms A:301; B:295; C:298; D:298). Immune responses at 14 weeks did not differ after two mOPV1 doses alone (94% [95% CI: 91-97%]) versus two mOPV1 doses with fIPV at 6 weeks (96% [93-98%]) or 10 weeks (96% [93-98%]). Participants who received mOPV1 and fIPV at 10 weeks had significantly lower shedding (p < 0·001) one- and two-weeks later compared with mOPV1 alone. Response to one mOPV1 dose was significantly higher than one bOPV dose (79% versus 67%; p < 0·001) and shedding two-weeks later was significantly higher after mOPV1 (76% versus 56%; p < 0·001) indicating improved vaccine replication. Ninety-nine adverse events were reported, 29 serious including two deaths; none were attributed to study vaccines. INTERPRETATION Given with the second mOPV1 dose, fIPV improved intestinal immunity but not humoral immunity. One mOPV1 dose induced higher humoral and intestinal immunity than bOPV. FUNDING U.S. Centers for Disease Control and Prevention.
Collapse
Affiliation(s)
- Cynthia J Snider
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Khalequ Zaman
- icddr,b, 68 Shahid Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh
| | - Amanda L Wilkinson
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA.
| | - Asma Binte Aziz
- icddr,b, 68 Shahid Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh
| | - Mohammad Yunus
- icddr,b, 68 Shahid Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh
| | - Warda Haque
- icddr,b, 68 Shahid Tajuddin Ahmed Sarani, Dhaka 1212, Bangladesh
| | - Kathryn A V Jones
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Ling Wei
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Concepcion F Estivariz
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | | | - Bernardo A Mainou
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Jaymin C Patel
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Jacquelyn S Lickness
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Mark A Pallansch
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Steven G F Wassilak
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - M Steven Oberste
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| | - Abhijeet Anand
- U.S. Centers for Disease Control and Prevention, 1600 Clifton Road Northeast, Atlanta, GA 30329, USA
| |
Collapse
|
13
|
Cohn IS, Hunter CA. Mining for crypto protection: a search for Cryptosporidium antibodies reveals antigens associated with immunity. J Clin Invest 2023; 133:e171966. [PMID: 37581310 PMCID: PMC10425208 DOI: 10.1172/jci171966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
Infectious diarrhea is a major cause of morbidity and mortality, particularly for children in low- and middle-income countries. Cryptosporidium is a diarrheal pathogen for which there is no vaccine and current therapies are only partially effective. In this issue of the JCI, Gilchrist, Campo, and colleagues surveyed a large cohort of Bangladeshi children to profile antibody responses against an array of Cryptosporidium proteins. They discovered 233 proteins to which children developed antibodies, identified seven as being associated with protection from reinfection, and provided insights regarding the longevity of Cryptosporidium antibodies and the development of antibody breadth. In this commentary, we discuss the burden of disease caused by Cryptosporidium and how these studies highlight the strategies to better manage this parasite.
Collapse
|
14
|
Kalkowska DA, Badizadegan K, Thompson KM. Outbreak management strategies for cocirculation of multiple poliovirus types. Vaccine 2023:S0264-410X(23)00429-2. [PMID: 37121801 DOI: 10.1016/j.vaccine.2023.04.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/11/2023] [Indexed: 05/02/2023]
Abstract
Prior modeling studies showed that current outbreak management strategies are unlikely to stop outbreaks caused by type 1 wild polioviruses (WPV1) or circulating vaccine-derived polioviruses (cVDPVs) in many areas, and suggested increased risks of outbreaks with cocirculation of more than one type of poliovirus. The surge of type 2 poliovirus transmission that began in 2019 and continues to date, in conjunction with decreases in preventive supplemental immunization activities (SIAs) for poliovirus types 1 and 3, has led to the emergence of several countries with cocirculation of more than one type of poliovirus. Response to these emerging cocirculation events is theoretically straightforward, but the different formulations, types, and inventories of oral poliovirus vaccines (OPVs) available for outbreak response present challenging practical questions. In order to demonstrate the implications of using different vaccine options and outbreak campaign strategies, we applied a transmission model to a hypothetical population with conditions similar to populations currently experiencing outbreaks of cVDPVs of both types 1 and 2. Our results suggest prevention of the largest number of paralytic cases occurs when using (1) trivalent OPV (tOPV) (or coadministering OPV formulations for all three types) until one poliovirus outbreak type dies out, followed by (2) using a type-specific OPV until the remaining poliovirus outbreak type also dies out. Using tOPV first offers a lower overall expected cost, but this option may be limited by the willingness to expose populations to type 2 Sabin OPV strains. For strategies that use type 2 novel OPV (nOPV2) concurrently administered with bivalent OPV (bOPV, containing types 1 and 3 OPV) emerges as a leading option, but questions remain about feasibility, logistics, type-specific take rates, and coadministration costs.
Collapse
|
15
|
Kok TW, Izzo AA, Costabile M. Intracellular immunoglobulin A (icIgA) in protective immunity and vaccines. Scand J Immunol 2023; 97:e13253. [PMID: 36597220 DOI: 10.1111/sji.13253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/20/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023]
Abstract
Virus neutralization at respiratory mucosal surfaces is important in the prevention of infection. Mucosal immunity is mediated mainly by extracellular secretory immunoglobulin A (sIgA) and its role has been well studied. However, the protective role of intracellular specific IgA (icIgA) is less well defined. Initially, in vitro studies using epithelial cell lines with surface expressed polymeric immunoglobulin receptor (pIgR) in transwell culture chambers have shown that icIgA can neutralize influenza, parainfluenza, HIV, rotavirus and measles viruses. This effect appears to involve an interaction between polymeric immunoglobulin A (pIgA) and viral particles within an intracellular compartment, since IgA is transported across the polarized cell. Co-localization of specific icIgA with influenza virus in patients' (virus culture positive) respiratory epithelial cells using well-characterized antisera was initially reported in 2018. This review provides a summary of in vitro studies with icIgA on colocalization and neutralization of the above five viruses. Two other highly significant respiratory infectious agents with severe global impacts viz. SARS-2 virus (CoViD pandemic) and the intracellular bacterium-Mycobacterium tuberculosis-are discussed. Further studies will provide more detailed understanding of the mechanisms and kinetics of icIgA neutralization in relation to viral entry and early replication steps with a specific focus on mucosal infections. This will inform the design of more effective vaccines against infectious agents transmitted via the mucosal route.
Collapse
Affiliation(s)
- Tuck-Weng Kok
- University of Adelaide, Faculty of Health & Medical Sciences and School of Biological Sciences, Adelaide, South Australia, Australia
| | - Angelo A Izzo
- University of Sydney, Tuberculosis Research Program, Centenary Institute, Camperdown, New South Wales, Australia
| | - Maurizio Costabile
- University of South Australia, Clinical and Health Sciences and Centre for Cancer Biology, Adelaide, South Australia, Australia
| |
Collapse
|
16
|
Erdem R, De Coster I, Withanage K, Mercer LD, Marchant A, Taton M, Cools N, Lion E, Cassels F, Higgins D, Ivinson K, Locke E, Mahmood K, Wright PF, Gast C, White JA, Ackerman ME, Konopka-Anstadt JL, Mainou BA, Van Damme P. Safety, tolerability, and immunogenicity of inactivated poliovirus vaccine with or without E.coli double mutant heat-labile toxin (dmLT) adjuvant in healthy adults; a phase 1 randomized study. Vaccine 2023; 41:1657-1667. [PMID: 36746739 PMCID: PMC9996288 DOI: 10.1016/j.vaccine.2023.01.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/01/2023] [Accepted: 01/21/2023] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inactivated trivalent poliovirus vaccine (IPV) induces humoral immunity, which protects against paralytic poliomyelitis but does not induce sufficient mucosal immunity to block intestinal infection. We assessed the intestinal immunity in healthy adults in Belgium conferred by a co-formulation of IPV with the mucosal adjuvant double mutant Labile Toxin (dmLT) derived from Escherichia coli. METHODS Healthy fully IPV-vaccinated 18-45-year-olds were randomly allocated to three groups: on Day 1 two groups received one full dose of IPV (n = 30) or IPV + dmLT (n = 30) in a blinded manner, and the third received an open-label dose of bivalent live oral polio vaccine (bOPV types 1 and 3, n = 20). All groups received a challenge dose of bOPV on Day 29. Participants reported solicited and unsolicited adverse events (AE) using study diaries. Mucosal immune responses were measured by fecal neutralization and IgA on Days 29 and 43, with fecal shedding of challenge viruses measured for 28 days. Humoral responses were measured by serum neutralizing antibody (NAb). RESULTS Solicited and unsolicited AEs were mainly mild-to-moderate and transient in all groups, with no meaningful differences in rates between groups. Fecal shedding of challenge viruses in both IPV groups exceeded that of the bOPV group but was not different between IPV and IPV + dmLT groups. High serum NAb responses were observed in both IPV groups, alongside modest levels of fecal neutralization and IgA. CONCLUSIONS Addition of dmLT to IPV administered intramuscularly neither affected humoral nor intestinal immunity nor decreased fecal virus shedding following bOPV challenge. The tolerability of the dose of dmLT used in this study may allow higher doses to be investigated for impact on mucosal immunity. Registered on ClinicalTrials.gov - NCT04232943.
Collapse
Affiliation(s)
- Rahsan Erdem
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Ilse De Coster
- Vaccine & Infectious Disease Institute, Centre for the Evaluation of Vaccination, University of Antwerp, Edegem, Belgium.
| | - Kanchanamala Withanage
- Vaccine & Infectious Disease Institute, Centre for the Evaluation of Vaccination, University of Antwerp, Edegem, Belgium
| | - Laina D Mercer
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Brussels, Belgium
| | - Martin Taton
- Institute for Medical Immunology, Université libre de Bruxelles, Brussels, Belgium
| | - Nathalie Cools
- Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
| | - Eva Lion
- Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
| | - Fred Cassels
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Deborah Higgins
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Karen Ivinson
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Emily Locke
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Kutub Mahmood
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | | | - Chris Gast
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Jessica A White
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | | | | | - Bernardo A Mainou
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Pierre Van Damme
- Vaccine & Infectious Disease Institute, Centre for the Evaluation of Vaccination, University of Antwerp, Edegem, Belgium
| |
Collapse
|
17
|
Ma B, Zhou Y, Hu Y, Duan H, Sun Z, Wang P, Li W, Han W, Qi H. Mapping Resident Immune Cells in the Murine Ocular Surface and Lacrimal Gland by Flow Cytometry. Ocul Immunol Inflamm 2023; 31:748-759. [PMID: 36867079 DOI: 10.1080/09273948.2023.2182327] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
BACKGROUND The ocular surface and lacrimal gland have a frontline position in mucosal immunology. However, there have been few updates to the immune cell atlas of these tissues in recent years. PURPOSE To map the immune cells in murine ocular surface tissues and lacrimal gland. METHODS Central and peripheral corneas, conjunctiva, and lacrimal gland were dissociated into single cell suspensions, followed by flow cytometry. Discrepancy of immune cells between the central and peripheral corneas was compared. In the conjunctiva and lacrimal gland, myeloid cells were clustered by tSNE and FlowSOM based on the expression of F4/80, Ly6C, Ly6G, and MHC II. ILCs, type 1 immune cells, and type 3 immune cells were analyzed. RESULTS The number of immune cells in peripheral corneas was about 16 folds of that in central corneas. B cells accounted for 8.74% of immune cells in murine peripheral corneas. In the conjunctiva and lacrimal gland, most myeloid cells tended out to be monocytes, macrophages, and classical dendritic cells (cDCs). ILC3 were 6.28% and 3.63% of ILCs in the conjunctiva and lacrimal gland, respectively. Th1, Tc1, and NK cells were predominant type 1 immune cells. γδ T17 cells and ILC3 outnumbered Th17 cells among type 3 T cells. CONCLUSION B cells resident in murine corneas were reported for the first time. Additionally, we proposed a strategy of clustering myeloid cells to better understand their heterogeneity in the conjunctiva and lacrimal gland based on tSNE and FlowSOM. Furthermore, we identified the ILC3 in the conjunctiva and lacrimal gland for the first time. Compositions of type 1 and type 3 immune cells were summarized. Our study provides a fundamental reference and novel insights for ocular surface immune homeostasis and diseases.
Collapse
Affiliation(s)
- Baikai Ma
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Beijing, China
| | - Yifan Zhou
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Beijing, China
| | - Yuzhe Hu
- Department of Immunology, Peking University Health Science Center, Beijing, China.,NHC Key Laboratory of Medical Immunology, Beijing, China.,Peking University Center for Human Disease Genomics, Beijing, China
| | - Hongyu Duan
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Beijing, China
| | - Zhengze Sun
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, Peking University Health Science Center, Beijing, China.,NHC Key Laboratory of Medical Immunology, Beijing, China.,Peking University Center for Human Disease Genomics, Beijing, China
| | - Wei Li
- Eye Institute of Xiamen University, Xiamen, China.,Xiang'an Hospital of Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Wenling Han
- Department of Immunology, Peking University Health Science Center, Beijing, China.,NHC Key Laboratory of Medical Immunology, Beijing, China.,Peking University Center for Human Disease Genomics, Beijing, China
| | - Hong Qi
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Beijing, China
| |
Collapse
|
18
|
Kumar P, Bird C, Holland D, Joshi SB, Volkin DB. Current and next-generation formulation strategies for inactivated polio vaccines to lower costs, increase coverage, and facilitate polio eradication. Hum Vaccin Immunother 2022; 18:2154100. [PMID: 36576132 PMCID: PMC9891683 DOI: 10.1080/21645515.2022.2154100] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/29/2022] Open
Abstract
Implementation of inactivated polio vaccines (IPV) containing Sabin strains (sIPV) will further enable global polio eradication efforts by improving vaccine safety during use and containment during manufacturing. Moreover, sIPV-containing vaccines will lower costs and expand production capacity to facilitate more widespread use in low- and middle-income countries (LMICs). This review focuses on the role of vaccine formulation in these efforts including traditional Salk IPV vaccines and new sIPV-containing dosage forms. The physicochemical properties and stability profiles of poliovirus antigens are described. Formulation approaches to lower costs include developing multidose and combination vaccine formats as well as improving storage stability. Formulation strategies for dose-sparing and enhanced mucosal immunity include employing adjuvants (e.g. aluminum-salt and newer adjuvants) and/or novel delivery systems (e.g. ID administration with microneedle patches). The potential for applying these low-cost formulation development strategies to other vaccines to further improve vaccine access and coverage in LMICs is also discussed.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Christopher Bird
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - David Holland
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
19
|
Medeiros MM, Ingham AC, Nanque LM, Correia C, Stegger M, Andersen PS, Fisker AB, Benn CS, Lanaspa M, Silveira H, Abrantes P. Oral polio revaccination is associated with changes in gut and upper respiratory microbiomes of infants. Front Microbiol 2022; 13:1016220. [PMID: 36386704 PMCID: PMC9649904 DOI: 10.3389/fmicb.2022.1016220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
After the eradication of polio infection, the plan is to phase-out the live-attenuated oral polio vaccine (OPV). Considering the protective non-specific effects (NSE) of OPV on unrelated pathogens, the withdrawal may impact child health negatively. Within a cluster-randomized trial, we carried out 16S rRNA deep sequencing analysis of fecal and nasopharyngeal microbial content of Bissau–Guinean infants aged 4–8 months, before and after 2 months of OPV revaccination (revaccinated infants = 47) vs. no OPV revaccination (control infants = 47). The aim was to address changes in the gut and upper respiratory bacterial microbiotas due to revaccination. Alpha-diversity for both microbiotas increased similarly over time in OPV-revaccinated infants and controls, whereas greater changes over time in the bacterial composition of gut (padjusted < 0.001) and upper respiratory microbiotas (padjusted = 0.018) were observed in the former. Taxonomic analysis of gut bacterial microbiota revealed a decrease over time in the median proportion of Bifidobacterium longum for all infants (25–14.3%, p = 0.0006 in OPV-revaccinated infants and 25.3–11.6%, p = 0.01 in controls), compatible with the reported weaning. Also, it showed a restricted increase in the median proportion of Prevotella_9 genus in controls (1.4–7.1%, p = 0.02), whereas in OPV revaccinated infants an increase over time in Prevotellaceae family (7.2–17.4%, p = 0.005) together with a reduction in median proportion of potentially pathogenic/opportunistic genera such as Escherichia/Shigella (5.8–3.4%, p = 0.01) were observed. Taxonomic analysis of upper respiratory bacterial microbiota revealed an increase over time in median proportions of potentially pathogenic/opportunistic genera in controls, such as Streptococcus (2.9–11.8%, p = 0.001 and Hemophilus (11.3–20.5%, p = 0.03), not observed in OPV revaccinated infants. In conclusion, OPV revaccination was associated with a healthier microbiome composition 2 months after revaccination, based on a more abundant and diversified bacterial community of Prevotellaceae and fewer pathogenic/opportunistic organisms. Further information on species-level differentiation and functional analysis of microbiome content are warranted to elucidate the impact of OPV-associated changes in bacterial microbiota on child health.
Collapse
Affiliation(s)
- Márcia Melo Medeiros
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Lisboa, Portugal
- *Correspondence: Márcia Melo Medeiros,
| | - Anna Cäcilia Ingham
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Line Møller Nanque
- Bandim Health Project, Bissau, Guinea-Bissau
- Bandim Health Project, Odense Patient Data Explorative Network, Institute of Clinical Research, Odense University Hospital/University of Southern Denmark, Odense, Denmark
| | | | - Marc Stegger
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Paal Skyt Andersen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Ane Baerent Fisker
- Bandim Health Project, Bissau, Guinea-Bissau
- Bandim Health Project, Odense Patient Data Explorative Network, Institute of Clinical Research, Odense University Hospital/University of Southern Denmark, Odense, Denmark
| | - Christine Stabell Benn
- Bandim Health Project, Bissau, Guinea-Bissau
- Bandim Health Project, Odense Patient Data Explorative Network, Institute of Clinical Research, Odense University Hospital/University of Southern Denmark, Odense, Denmark
- Danish Institute for Advanced Study, University of Southern Denmark, Odense, Denmark
| | - Miguel Lanaspa
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Lisboa, Portugal
| | - Henrique Silveira
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Lisboa, Portugal
| | - Patrícia Abrantes
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Lisboa, Portugal
| |
Collapse
|
20
|
Quarleri J. Poliomyelitis is a current challenge: long-term sequelae and circulating vaccine-derived poliovirus. GeroScience 2022; 45:707-717. [PMID: 36260265 PMCID: PMC9886775 DOI: 10.1007/s11357-022-00672-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/12/2022] [Indexed: 02/03/2023] Open
Abstract
For more than 20 years, the World Health Organization Western Pacific Region (WPR) has been polio-free. However, two current challenges are still polio-related. First, around half of poliomyelitis elderly survivors suffer late poliomyelitis sequelae with a substantial impact on daily activities and quality of life, experiencing varying degrees of residual weakness as they age. The post-polio syndrome as well as accelerated aging may be involved. Second, after the worldwide Sabin oral poliovirus (OPV) vaccination, the recent reappearance of strains of vaccine-derived poliovirus (VDPV) circulating in the environment is worrisome and able to persistent person-to-person transmission. Such VDPV strains exhibit atypical genetic characteristics and reversed neurovirulence that can cause paralysis similarly to wild poliovirus, posing a significant obstacle to the elimination of polio. Immunization is essential for preventing paralysis in those who are exposed to the poliovirus. Stress the necessity of maintaining high vaccination rates because declining immunity increases the likelihood of reemergence. If mankind wants to eradicate polio in the near future, measures to raise immunization rates and living conditions in poorer nations are needed, along with strict observation. New oral polio vaccine candidates offer a promissory tool for this goal.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina. .,Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
21
|
Deere D, Ryan U. Current assumptions for quantitative microbial risk assessment (QMRA) of Norovirus contamination of drinking water catchments due to recreational activities: an update. JOURNAL OF WATER AND HEALTH 2022; 20:1543-1557. [PMID: 36308498 DOI: 10.2166/wh.2022.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Contamination of drinking water from Norovirus (NoV) and other waterborne viruses is a major public health concern globally. Increasingly, quantitative microbial risk assessment (QMRA) is being used to assess the various risks from waterborne pathogens and evaluate control strategies. As urban populations grow and expand, there is increasing demand for recreational activities in drinking water catchments. QMRA relies on context-specific data to map out the pathways by which viruses can enter water and be transferred to drinking water consumers and identify risk factors and appropriate controls. This review examines the current evidence base and assumptions for QMRA analysis of NoV and other waterborne viral pathogens and recommends numerical values based on the most recent evidence to better understand the health risks associated with recreators in Australian drinking water sources; these are broadly applicable to all drinking water sources where recreational access is allowed. Key issues include the lack of an agreed upon data and dose-response models for human infectious NoV genotypes, faecal shedding by bathers, the extent of NoV infectivity and aggregation, resistance (secretor status) to NoV and the extent of secondary transmission.
Collapse
Affiliation(s)
- Dan Deere
- Water Futures and Water Research Australia, Sydney, Australia
| | - Una Ryan
- Harry Butler Institute, Murdoch University, Perth, Australia E-mail:
| |
Collapse
|
22
|
Hill M, Bandyopadhyay AS, Pollard AJ. Emergence of vaccine-derived poliovirus in high-income settings in the absence of oral polio vaccine use. Lancet 2022; 400:713-715. [PMID: 35988575 DOI: 10.1016/s0140-6736(22)01582-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 01/20/2023]
Affiliation(s)
- Matilda Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, UK.
| | | | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 7LE, UK
| |
Collapse
|
23
|
Chen W, Wang J, Du L, Chen J, Zheng Q, Li P, Du B, Fang X, Liao Z. Kefir microbiota and metabolites stimulate intestinal mucosal immunity and its early development. Crit Rev Food Sci Nutr 2022; 64:1371-1384. [PMID: 36039934 DOI: 10.1080/10408398.2022.2115975] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Kefir consists of a large number of probiotics, which can regulate or shape the balance of intestinal microbiota, and enhance the host's immune response. Kefir microbiota can shape the mucosal immunity of the body through SCFAs, EPS, polypeptides, lactic acid, and other metabolites and microbial antigens themselves, and this shaping may have time windows and specific pathways. Kefir can regulate antibody SIgA and IL-10 levels to maintain intestinal homeostasis, and its secreted SIgA can shape the stable microbiota system by wrapping and binding different classes of microorganisms. The incidence of intestinal inflammation is closely linked to the development and maturation of intestinal mucosal immunity. Based on summarizing the existing research results on Kefir, its metabolites, and immune system development, this paper proposes to use Kefir, traditional fermented food with natural immune-enhancing components and stable functional microbiota, as an intervention method. Early intervention in the immune system may seize the critical window period of mucosal immunity and stimulate the development and maturation of intestinal mucosal immunity in time.
Collapse
Affiliation(s)
- Weizhe Chen
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jie Wang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Liyu Du
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Junjie Chen
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Qikai Zheng
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Pan Li
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Bing Du
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Xiang Fang
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Zhenlin Liao
- College of Food Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
24
|
HAMURO K, SAITO H, SAITO T, KOHDA N. Identification of antigens recognized by salivary IgA using microbial protein microarrays. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2022; 41:177-184. [PMID: 36258770 PMCID: PMC9533030 DOI: 10.12938/bmfh.2022-014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/15/2022] [Indexed: 11/05/2022]
Abstract
Secretory IgA plays an important role in the mucosal immune system for protection against pathogens. However, the antigens recognized by these antibodies have only been partially studied. We comprehensively investigated the antigens bound by salivary IgA in healthy adults using microbial protein microarrays. This confirmed that saliva contained IgA antibodies that bind to a variety of pathogenic microorganisms, including spike proteins of severe acute respiratory syndrome coronavirus 2, severe acute respiratory syndrome coronavirus, Middle East respiratory syndrome coronavirus, and other human coronavirus species. Also, many subtypes and strains of influenza virus were bound, regardless of the seasonal or vaccine strains. Salivary IgA also bound many serogroups and serovars of Escherichia coli and Salmonella. Taken together, these findings suggest that salivary IgA, which exhibits broad reactivity, is likely an essential element of the mucosal immune system at the forefront of defense against infection.
Collapse
Affiliation(s)
- Koji HAMURO
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical
Co., Ltd., 3-31-13 Saigawa, Otsu, Shiga 520-0002, Japan
| | - Hiroshi SAITO
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical
Co., Ltd., 3-31-13 Saigawa, Otsu, Shiga 520-0002, Japan
| | - Takao SAITO
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical
Co., Ltd., 3-31-13 Saigawa, Otsu, Shiga 520-0002, Japan
| | - Noriyuki KOHDA
- Otsu Nutraceuticals Research Institute, Otsuka Pharmaceutical
Co., Ltd., 3-31-13 Saigawa, Otsu, Shiga 520-0002, Japan
| |
Collapse
|
25
|
Lockhart A, Mucida D, Parsa R. Immunity to enteric viruses. Immunity 2022; 55:800-818. [PMID: 35545029 PMCID: PMC9257994 DOI: 10.1016/j.immuni.2022.04.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022]
Abstract
Pathogenic enteric viruses are a major cause of morbidity and mortality, particularly among children in developing countries. The host response to enteric viruses occurs primarily within the mucosa, where the intestinal immune system must balance protection against pathogens with tissue protection and tolerance to harmless commensal bacteria and food. Here, we summarize current knowledge in natural immunity to enteric viruses, highlighting specialized features of the intestinal immune system. We further discuss how knowledge of intestinal anti-viral mechanisms can be translated into vaccine development with particular focus on immunization in the oral route. Research reveals that the intestine is a complex interface between enteric viruses and the host where environmental factors influence susceptibility and immunity to infection, while viral infections can have lasting implications for host health. A deeper mechanistic understanding of enteric anti-viral immunity with this broader context can ultimately lead to better vaccines for existing and emerging viruses.
Collapse
Affiliation(s)
- Ainsley Lockhart
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| | - Roham Parsa
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
26
|
Crothers JW, Ross Colgate E, Cowan KJ, Dickson DM, Walsh M, Carmolli M, Wright PF, Norton EB, Kirkpatrick BD. Intradermal fractional-dose inactivated polio vaccine (fIPV) adjuvanted with double mutant Enterotoxigenic Escherichia coli heat labile toxin (dmLT) is well-tolerated and augments a systemic immune response to all three poliovirus serotypes in a randomized placebo-controlled trial. Vaccine 2022; 40:2705-2713. [PMID: 35367069 PMCID: PMC9024222 DOI: 10.1016/j.vaccine.2022.03.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 11/19/2022]
Abstract
Eradication of poliomyelitis globally is constrained by fecal shedding of live polioviruses, both wild-type and vaccine-derived strains, into the environment. Although inactivated polio vaccines (IPV) effectively protect the recipient from clinical poliomyelitis, fecal shedding of live virus still occurs following infection with either wildtype or vaccine-derived strains of poliovirus. In the drive to eliminate the last cases of polio globally, improvements in both oral polio vaccines (OPV) (to prevent reversion to virulence) and injectable polio vaccines (to improve mucosal immunity and prevent viral shedding) are underway. The E. coli labile toxin with two or "double" attenuating mutations (dmLT) may boost immunologic responses to IPV, including at mucosal sites. We performed a double-blinded phase I controlled clinical trial to evaluate safety, tolerability, as well as systemic and mucosal immunogenicity of IPV adjuvanted with dmLT, given as a fractional (1/5th) dose intradermally (fIPV-dmLT). Twenty-nine volunteers with no past exposure to OPV were randomized to a single dose of fIPV-dmLT or fIPV alone. fIPV-dmLT was well tolerated, although three subjects had mild but persistent induration and hyperpigmentation at the injection site. A ≥ 4-fold rise in serotype-specific neutralizing antibody (SNA) titers to all three serotypes was seen in 84% of subjects receiving fIPV-dmLT vs. 50% of volunteers receiving IPV alone. SNA titers were higher in the dmLT-adjuvanted group, but only differences in serotype 1 were significant. Mucosal immune responses, as measured by polio serotype specific fecal IgA were minimal in both groups and differences were not seen. fIPV-dmLT may offer a benefit over IPV alone. Beyond NAB responses protecting the individual, studies demonstrating the ability of fIPV-dmLT to prevent viral shedding are necessary. Studies employing controlled human infection models, using monovalent OPV post-vaccine are ongoing. Studies specifically in children may also be necessary and additional biomarkers of mucosal immune responses in this population are needed. Clinicaltrials.gov Identifer: NCT03922061.
Collapse
Affiliation(s)
- Jessica W Crothers
- Department of Pathology and Laboratory Medicine, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA.
| | - Elizabeth Ross Colgate
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Kelly J Cowan
- Department of Pediatrics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Dorothy M Dickson
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - MaryClaire Walsh
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Marya Carmolli
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Elizabeth B Norton
- Department of Immunology and Microbiology, Tulane University, New Orleans, LA, USA
| | - Beth D Kirkpatrick
- Microbiology and Molecular Genetics, Vaccine Testing Center, University of Vermont Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
27
|
Huang J, Ding Y, Yao J, Zhang M, Zhang Y, Xie Z, Zuo J. Nasal Nanovaccines for SARS-CoV-2 to Address COVID-19. Vaccines (Basel) 2022; 10:vaccines10030405. [PMID: 35335037 PMCID: PMC8952855 DOI: 10.3390/vaccines10030405] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19 is still prevalent around the globe. Although some SARS-CoV-2 vaccines have been distributed to the population, the shortcomings of vaccines and the continuous emergence of SARS-CoV-2 mutant virus strains are a cause for concern. Thus, it is vital to continue to improve vaccines and vaccine delivery methods. One option is nasal vaccination, which is more convenient than injections and does not require a syringe. Additionally, stronger mucosal immunity is produced under nasal vaccination. The easy accessibility of the intranasal route is more advantageous than injection in the context of the COVID-19 pandemic. Nanoparticles have been proven to be suitable delivery vehicles and adjuvants, and different NPs have different advantages. The shortcomings of the SARS-CoV-2 vaccine may be compensated by selecting or modifying different nanoparticles. It travels along the digestive tract to the intestine, where it is presented by GALT, tissue-resident immune cells, and gastrointestinal lymph nodes. Nasal nanovaccines are easy to use, safe, multifunctional, and can be distributed quickly, demonstrating strong prospects as a vaccination method for SARS-CoV-2, SARS-CoV-2 variants, or SARS-CoV-n.
Collapse
Affiliation(s)
- Jialu Huang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Yubo Ding
- Nanhua Hospital Affiliated to University of South China, Hengyang Medical School, University of South China, Hengyang 421002, China; (Y.D.); (J.Y.)
| | - Jingwei Yao
- Nanhua Hospital Affiliated to University of South China, Hengyang Medical School, University of South China, Hengyang 421002, China; (Y.D.); (J.Y.)
| | - Minghui Zhang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Yu Zhang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Zhuoyi Xie
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
| | - Jianhong Zuo
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; (J.H.); (M.Z.); (Y.Z.); (Z.X.)
- Nanhua Hospital Affiliated to University of South China, Hengyang Medical School, University of South China, Hengyang 421002, China; (Y.D.); (J.Y.)
- The Third Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang 421900, China
- Correspondence: ; Tel.: +86-7345-675219
| |
Collapse
|
28
|
Affiliation(s)
- Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany.
| | - Ana Izcue
- Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
29
|
Wright PF, Prevost-Reilly AC, Natarajan H, Brickley EB, Connor RI, Wieland-Alter WF, Miele AS, Weiner JA, Nerenz RD, Ackerman ME. OUP accepted manuscript. J Infect Dis 2022; 226:1204-1214. [PMID: 35188974 PMCID: PMC8903457 DOI: 10.1093/infdis/jiac065] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/18/2022] [Indexed: 01/08/2023] Open
Abstract
Background A longitudinal study was performed to determine the breadth, kinetics, and correlations of systemic and mucosal antibody responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Methods Twenty-six unvaccinated adults with confirmed coronavirus disease 2019 (COVID-19) were followed for 6 months with 3 collections of blood, nasal secretions, and stool. Control samples were obtained from 16 unvaccinated uninfected individuals. SARS-CoV-2 neutralizing and binding antibody responses were respectively evaluated by pseudovirus assays and multiplex bead arrays. Results Neutralizing antibody responses to SARS-CoV-2 were detected in serum and respiratory samples for 96% (25/26) and 54% (14/26), respectively, of infected participants. Robust binding antibody responses against SARS-CoV-2 spike protein and S1, S2, and receptor binding (RBD) domains occurred in serum and respiratory nasal secretions, but not in stool samples. Serum neutralization correlated with RBD-specific immunoglobulin (Ig)G, IgM, and IgA in serum (Spearman ρ = 0.74, 0.66, and 0.57, respectively), RBD-specific IgG in respiratory secretions (ρ = 0.52), disease severity (ρ = 0.59), and age (ρ = 0.40). Respiratory mucosal neutralization correlated with RBD-specific IgM (ρ = 0.42) and IgA (ρ = 0.63). Conclusions Sustained antibody responses occurred after SARS-CoV-2 infection. Notably, there was independent induction of IgM and IgA binding antibody and neutralizing responses in systemic and respiratory compartments. These observations have implications for current vaccine strategies and understanding SARS-CoV-2 reinfection and transmission.
Collapse
Affiliation(s)
- Peter F Wright
- Correspondence: Peter F. Wright, MD, Division of Infectious Disease and International Health, 330 Borwell, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756 ()
| | | | | | | | - Ruth I Connor
- Division of Infectious Disease and International Health, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Wendy F Wieland-Alter
- Division of Infectious Disease and International Health, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | | | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Robert D Nerenz
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|