1
|
Abo-Zaid MA, Elsapagh RM, Sultan NS, Mawkili W, Hegazy MM, Ismail AH. Allergy Treatment: A Comprehensive Review of Nanoparticle-based Allergen Immunotherapy. FRONT BIOSCI-LANDMRK 2025; 30:26550. [PMID: 40152375 DOI: 10.31083/fbl26550] [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: 09/12/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 03/29/2025]
Abstract
Allergic disorders rising in prevalence globally, affecting a substantial proportion of individuals in industrialized nations. The imbalance in the immune system, characterized by elevated allergen-specific T helper 2 (Th2) cells and immunoglobulin E (IgE) antibodies, is a key factor in allergy development. Allergen-specific immunotherapy (AIT) is the only treatment capable of alleviating allergic symptoms, preventing new sensitizations, and reducing asthma risk in allergic rhinitis patients. Traditional AIT, however, faces challenges such as frequent administration, adverse effects, and inconsistent patient outcomes. Nanoparticle-based approaches have emerged as a promising strategy to enhance AIT. This review explores the utilization of nanoparticles in AIT, highlighting their ability to interact with the immune system and improve therapeutic outcomes. Various types of nanoparticles, including polyesters, polysaccharide polymers, liposomes, protamine-based nanoparticles (NPs), and polyanhydrides, have been employed as adjuvants or carriers to enhance AIT's efficacy and safety. Nanoparticles offer advantages such as allergen protection, improved immune response modulation, targeted cell delivery, and reduced side effects. This review provides an overview of the current landscape of nanoparticle-based allergen immunotherapy, discussing its potential to revolutionize allergy treatment compared to traditional immunotherapy.
Collapse
Affiliation(s)
- Mabrouk A Abo-Zaid
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, 45142 Jazan, Kingdom of Saudi Arabia
| | | | - Nourhan S Sultan
- Biotechnology Department, Faculty of Science, Cairo University, 12613 Giza, Egypt
| | - Wedad Mawkili
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142 Jazan, Kingdom of Saudi Arabia
| | - Maysa M Hegazy
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, 45142 Jazan, Kingdom of Saudi Arabia
| | - Ahmed H Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, 45142 Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
2
|
Zhang M, Li M, Ma J. The role of long-lived plasma cells in viral clearance. JOURNAL OF BIOLOGICAL DYNAMICS 2024; 18:2325523. [PMID: 38445631 DOI: 10.1080/17513758.2024.2325523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 02/21/2024] [Indexed: 03/07/2024]
Abstract
The adaptive immune system has two types of plasma cells (PC), long-lived plasma cells (LLPC) and short-lived plasma cells (SLPC), that differ in their lifespan. In this paper, we propose that LLPC is crucial to the clearance of viral particles in addition to reducing the viral basic reproduction number in secondary infections. We use a sequence of within-host mathematical models to show that, CD8 T cells, SLPC and memory B cells cannot achieve full viral clearance, and the viral load will reach a low positive equilibrium level because of a continuous replenishment of target cells. However, the presence of LLPC is crucial for viral clearance.
Collapse
Affiliation(s)
- Mingran Zhang
- College of Information Science and Technology, Donghua University, Shanghai, People's Republic of China
| | - Meili Li
- College of Science, Donghua University, Shanghai, People's Republic of China
| | - Junling Ma
- Department of Mathematics and Statistics, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
3
|
Adnan N, Haq MA, Akter S, Sajal SMSA, Islam MF, Mou TJ, Jamiruddin MR, Jubyda FT, Islam MS, Tuli JF, Liza SM, Hossain S, Islam Z, Ahmed S, Khandker SS, Hossain R, Ahmed MF, Khondoker MU, Azmuda N, Parvez MAK. Antibody Response after Homologous and Heterologous Prime-Boost COVID-19 Vaccination in a Bangladeshi Residential University Cohort. Vaccines (Basel) 2024; 12:482. [PMID: 38793733 PMCID: PMC11125736 DOI: 10.3390/vaccines12050482] [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: 02/25/2024] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
COVID-19 vaccination strategies, including heterologous prime-boost regimens and additional booster doses, aim to optimize immune responses. However, seroepidemiological studies on immune responses to different COVID-19 vaccine types and schedules remain limited. This study investigated antibody levels following homologous and heterologous prime-and-boost COVID-19 vaccination in Bangladesh. In a cohort of 606 participants who received first/second/booster doses of vaccines (AstraZeneca, Moderna, Pfizer-BioNTech, and Sinopharm), anti-spike IgG and anti-nucleocapsid IgG levels were measured. Antibody titer variations with respect to age, gender, intervals between doses, and prior infection status were analyzed. mRNA vaccines elicited the highest antibody levels after homologous and heterologous boosting. The AstraZeneca booster resulted in a sharp titer decline rate of ~0.04 units per day. Second or booster vaccine doses significantly increased antibody levels, especially in males (p < 0.05). Older age correlated with higher titers, likely reflecting previous infection, which was further confirmed by the elevation of anti-nucleocapsid IgG levels. About 95.5% of non-Sinopharm recipients were anti-nucleocapsid IgG positive, suggesting prior exposure exceeding self-reported infections (12.5%). mRNA and heterologous COVID-19 boosting enhances humoral immunity over homologous prime-boost vector/inactivated vaccination. However, waning immunity merits further investigation across vaccine platforms.
Collapse
Affiliation(s)
- Nihad Adnan
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| | | | - Salma Akter
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| | - S. M. Shafiul Alam Sajal
- Gonoshasthaya-RNA Biotech Limited, Dhaka 1205, Bangladesh; (S.M.S.A.S.); (S.S.K.); (R.H.); (M.U.K.)
| | - Md. Fokhrul Islam
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FF, UK;
- Department of Pharmacy, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Taslin Jahan Mou
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FF, UK;
| | | | - Fatema Tuz Jubyda
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| | - Md. Salequl Islam
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jamsheda Ferdous Tuli
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| | - Syeda Moriam Liza
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| | - Sharif Hossain
- Department of Biotechnology & Genetic Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.H.); (Z.I.)
| | - Zinia Islam
- Department of Biotechnology & Genetic Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.H.); (Z.I.)
| | - Sohel Ahmed
- Department of Biochemistry & Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh;
| | - Shahad Saif Khandker
- Gonoshasthaya-RNA Biotech Limited, Dhaka 1205, Bangladesh; (S.M.S.A.S.); (S.S.K.); (R.H.); (M.U.K.)
| | - Rubel Hossain
- Gonoshasthaya-RNA Biotech Limited, Dhaka 1205, Bangladesh; (S.M.S.A.S.); (S.S.K.); (R.H.); (M.U.K.)
| | - Md. Firoz Ahmed
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| | - Mohib Ullah Khondoker
- Gonoshasthaya-RNA Biotech Limited, Dhaka 1205, Bangladesh; (S.M.S.A.S.); (S.S.K.); (R.H.); (M.U.K.)
- Gonoshasthaya Samaj Vittik Medical College, Savar, Dhaka 1344, Bangladesh
| | - Nafisa Azmuda
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| | - Md. Anowar Khasru Parvez
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh; (S.A.); (T.J.M.); (F.T.J.); (M.S.I.); (J.F.T.); (S.M.L.); (M.F.A.); (N.A.)
| |
Collapse
|
4
|
Roh T, Regan AK, Johnson NM, Hasan NT, Trisha NF, Aggarwal A, Han D. Association of arsenic exposure with measles antibody titers in US children: Influence of sex and serum folate levels. ENVIRONMENT INTERNATIONAL 2024; 183:108329. [PMID: 38071850 DOI: 10.1016/j.envint.2023.108329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/18/2023] [Accepted: 11/14/2023] [Indexed: 01/25/2024]
Abstract
Exposure to arsenic during childhood is associated with various adverse health conditions. However, little is known about the effect of arsenic exposure on vaccine-related humoral immunity in children. We analyzed data from the National Health and Nutrition Examination Survey (2003-2004 and 2009-2010) to study the relationship between urinary arsenic and measles antibody levels in 476 US children aged 6-11. Multivariable linear regression was used to evaluate the association, adjusting for cycle, age, race, body mass index (BMI), serum cotinine, poverty index ratio, and vitamin B12 and selenium intakes. Stratified analyses were conducted by sex and serum folate levels using the median as cutoff (18.7 ng/mL). The measles antibody concentrations in the 3rd and 4th quartiles were found to have significantly decreased by 28.5 % (95 % Confidence Interval (CI) -47.6, -2.28) and 36.8 % (95 % CI -50.2, -19.5), compared to the lowest quartile among boys with serum folate levels lower than 18.7 ng/ml. The serum measles antibody titers significantly decreased by 16.7 % (95 %CI -25.0, -7.61) for each doubling of creatinine-corrected urinary total inorganic arsenic concentrations in the same group. No associations were found in boys with high serum folate levels or in girls. Further prospective studies are needed to validate these findings and develop interventions to protect children from infectious diseases.
Collapse
Affiliation(s)
- Taehyun Roh
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, TX 77843, USA.
| | - Annette K Regan
- School of Nursing and Health Professions, University of San Francisco, San Francisco, CA 94117, USA
| | - Natalie M Johnson
- Department of Environmental and Occupational Health, School of Public Health, Texas A&M University, College Station, TX 77843, USA
| | - Nishat Tasnim Hasan
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, TX 77843, USA
| | - Nusrat Fahmida Trisha
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, TX 77843, USA
| | - Anisha Aggarwal
- Department of Health Behavior, School of Public Health, Texas A&M University, College Station, TX 77843, USA
| | - Daikwon Han
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
5
|
Buscot M, Cremoni M, Graça D, Brglez V, Courjon J, Allouche J, Teisseyre M, Boyer L, Barrière J, Chamorey E, Carles M, Seitz-Polski B. Breakthrough infections due to SARS-CoV-2 Delta variant: relation to humoral and cellular vaccine responses. Front Immunol 2023; 14:1145652. [PMID: 37063916 PMCID: PMC10101330 DOI: 10.3389/fimmu.2023.1145652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
IntroductionCOVID-19 vaccines are expected to provide effective protection. However, emerging strains can cause breakthrough infection in vaccinated individuals. The immune response of vaccinated individuals who have experienced breakthrough infection is still poorly understood.MethodsHere, we studied the humoral and cellular immune responses of fully vaccinated individuals who subsequently experienced breakthrough infection due to the Delta variant of SARS-CoV-2 and correlated them with the severity of the disease.ResultsIn this study, an effective humoral response alone was not sufficient to induce effective immune protection against severe breakthrough infection, which also required effective cell-mediated immunity to SARS-CoV-2. Patients who did not require oxygen had significantly higher specific (p=0.021) and nonspecific (p=0.004) cellular responses to SARS-CoV-2 at the onset of infection than those who progressed to a severe form.DiscussionKnowing both humoral and cellular immune response could allow to adapt preventive strategy, by better selecting patients who would benefit from additional vaccine boosters.Trial registration numbershttps://clinicaltrials.gov, identifier NCT04355351; https://clinicaltrials.gov, identifier NCT04429594.
Collapse
Affiliation(s)
- Matthieu Buscot
- Infectious Diseases Department, Nice University Hospital, Nice, France
| | - Marion Cremoni
- Immunology Laboratory, Archet 1 Hospital, Nice University Hospital, Nice, France
- Clinical Research Unit Côte d’Azur (UR2CA), Côte d’Azur University, Nice, France
| | - Daisy Graça
- Immunology Laboratory, Archet 1 Hospital, Nice University Hospital, Nice, France
| | - Vesna Brglez
- Immunology Laboratory, Archet 1 Hospital, Nice University Hospital, Nice, France
- Clinical Research Unit Côte d’Azur (UR2CA), Côte d’Azur University, Nice, France
| | - Johan Courjon
- Infectious Diseases Department, Nice University Hospital, Nice, France
- Mediterranean Center for Molecular Medicine (C3M), Côte d’Azur University, Nice, France
| | - Jonathan Allouche
- Clinical Research Unit Côte d’Azur (UR2CA), Côte d’Azur University, Nice, France
| | - Maxime Teisseyre
- Clinical Research Unit Côte d’Azur (UR2CA), Côte d’Azur University, Nice, France
| | - Laurent Boyer
- Mediterranean Center for Molecular Medicine (C3M), Côte d’Azur University, Nice, France
| | - Jérôme Barrière
- Department of Oncology, Clinique St Jean, Cagnes sur Mer, France
| | - Emmanuel Chamorey
- Department of Biostatistics, Centre Antoine Lacassagne, Nice, France
| | - Michel Carles
- Infectious Diseases Department, Nice University Hospital, Nice, France
| | - Barbara Seitz-Polski
- Immunology Laboratory, Archet 1 Hospital, Nice University Hospital, Nice, France
- Clinical Research Unit Côte d’Azur (UR2CA), Côte d’Azur University, Nice, France
- *Correspondence: Barbara Seitz-Polski,
| |
Collapse
|
6
|
Peletta A, Lemoine C, Courant T, Collin N, Borchard G. Meeting vaccine formulation challenges in an emergency setting: Towards the development of accessible vaccines. Pharmacol Res 2023; 189:106699. [PMID: 36796463 DOI: 10.1016/j.phrs.2023.106699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Vaccination is considered one of the most successful strategies to prevent infectious diseases. In the event of a pandemic or epidemic, the rapid development and distribution of the vaccine to the population is essential to reduce mortality, morbidity and transmission. As seen during the COVID-19 pandemic, the production and distribution of vaccines has been challenging, in particular for resource-constrained settings, essentially slowing down the process of achieving global coverage. Pricing, storage, transportation and delivery requirements of several vaccines developed in high-income countries resulted in limited access for low-and-middle income countries (LMICs). The capacity to manufacture vaccines locally would greatly improve global vaccine access. In particular, for the development of classical subunit vaccines, the access to vaccine adjuvants is a pre-requisite for more equitable access to vaccines. Vaccine adjuvants are agents required to augment or potentiate, and possibly target the specific immune response to such type of vaccine antigens. Openly accessible or locally produced vaccine adjuvants may allow for faster immunization of the global population. For local research and development of adjuvanted vaccines to expand, knowledge on vaccine formulation is of paramount importance. In this review, we aim to discuss the optimal characteristics of a vaccine developed in an emergency setting by focusing on the importance of vaccine formulation, appropriate use of adjuvants and how this may help overcome barriers for vaccine development and production in LMICs, achieve improved vaccine regimens, delivery and storage requirements.
Collapse
Affiliation(s)
- Allegra Peletta
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| | - Céline Lemoine
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Thomas Courant
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| |
Collapse
|
7
|
Vaccination with an HIV T-cell immunogen induces alterations in the mouse gut microbiota. NPJ Biofilms Microbiomes 2022; 8:104. [PMID: 36585401 PMCID: PMC9801356 DOI: 10.1038/s41522-022-00368-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022] Open
Abstract
The gut microbiota is emerging as a crucial factor modulating vaccine responses; however, few studies have investigated if vaccines, in turn, can alter the microbiota and to what extent such changes may improve vaccine efficacy. To understand the effect of T-cell vaccination on the gut microbiome, we administered an HIV-1 T-cell immunogen (HTI arm) or PBS (control, Mock arm) to C57Bl/6 mice following a heterologous prime-boost scheme. The longitudinal dynamics of the mice gut microbiota was characterized by 16 S ribosomal RNA sequencing in fecal samples collected from cages, as well as from three gut sections (cecum, small and large intestine). Serum and spleen cells were obtained at the last time point of the study to assess immune correlates using IFNγ ELISPOT and cytokine Luminex® assays. Compared with Mock, HTI-vaccinated mice were enriched in Clostridiales genera (Eubacterium xylanophilum group, Roseburia and Ruminococcus) known as primary contributors of anti-inflammatory metabolites, such as short-chain fatty acids. Such shift was observed after the first HTI dose and remained throughout the study follow-up (18 weeks). However, the enriched Clostridiales genera were different between feces and gut sections. The abundance of bacteria enriched in vaccinated animals positively correlated with HTI-specific T-cell responses and a set of pro-inflammatory cytokines, such as IL-6. This longitudinal analysis indicates that, in mice, T-cell vaccination may promote an increase in gut bacteria known to produce anti-inflammatory molecules, which in turn correlate with proinflammatory cytokines, suggesting an adaptation of the gut microbial milieu to T-cell-induced systemic inflammation.
Collapse
|
8
|
Teplensky MH, Distler ME, Kusmierz CD, Evangelopoulos M, Gula H, Elli D, Tomatsidou A, Nicolaescu V, Gelarden I, Yeldandi A, Batlle D, Missiakas D, Mirkin CA. Spherical nucleic acids as an infectious disease vaccine platform. Proc Natl Acad Sci U S A 2022; 119:e2119093119. [PMID: 35312341 PMCID: PMC9168922 DOI: 10.1073/pnas.2119093119] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/05/2022] [Indexed: 12/17/2022] Open
Abstract
SignificanceUsing SARS-CoV-2 as a relevant case study for infectious disease, we investigate the structure-function relationships that dictate antiviral spherical nucleic acid (SNA) vaccine efficacy. We show that the SNA architecture can be rapidly employed to target COVID-19 through incorporation of the receptor-binding domain, and that the resulting vaccine potently activates human cells in vitro and mice in vivo. Furthermore, when challenged with a lethal viral infection, only mice treated with the SNA vaccine survived. Taken together, this work underscores the importance of rational vaccine design for infectious disease to yield vaccines that elicit more potent immune responses to effectively fight disease.
Collapse
Affiliation(s)
- Michelle H. Teplensky
- Department of Chemistry, Northwestern University, Evanston, IL 60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208
| | - Max E. Distler
- Department of Chemistry, Northwestern University, Evanston, IL 60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208
| | - Caroline D. Kusmierz
- Department of Chemistry, Northwestern University, Evanston, IL 60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208
| | | | - Haley Gula
- Howard T. Ricketts Laboratory, Department of Microbiology, University of Chicago, Chicago, IL 60637
| | - Derek Elli
- Howard T. Ricketts Laboratory, Department of Microbiology, University of Chicago, Chicago, IL 60637
| | - Anastasia Tomatsidou
- Howard T. Ricketts Laboratory, Department of Microbiology, University of Chicago, Chicago, IL 60637
| | - Vlad Nicolaescu
- Howard T. Ricketts Laboratory, Department of Microbiology, University of Chicago, Chicago, IL 60637
| | - Ian Gelarden
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Anjana Yeldandi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Daniel Batlle
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Dominique Missiakas
- Howard T. Ricketts Laboratory, Department of Microbiology, University of Chicago, Chicago, IL 60637
| | - Chad A. Mirkin
- Department of Chemistry, Northwestern University, Evanston, IL 60208
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208
| |
Collapse
|
9
|
Ghia CJ, Rambhad G. Pneumococcal Vaccine Recommendations for Old-Age Home Indian Residents: A Literature Review. Gerontol Geriatr Med 2022; 8:23337214221118237. [PMID: 36081416 PMCID: PMC9445461 DOI: 10.1177/23337214221118237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Advancing age is accompanied by decreased immunity, poor health, and physiological changes, which render the elderly population highly susceptible to infectious diseases. We aim to identify the guidelines for pneumococcal vaccines in old-age facilities in India. We performed an extensive review of Indian literature (indexed and non-indexed publications) from 2010 to 2020 using search strings “Pneumococcal vaccine AND Recommendations AND India,” “Pneumococcal vaccine AND Guidelines AND India,” followed by a hand search to identify the most updated versions of recommendations. We reviewed immunization guidelines recommended by nine medical associations and societies in India—Association of Physicians of India (API), Geriatric Society of India (GSI), Indian Society of Nephrology (ISN), Mass Gathering Advisory Board Consensus Recommendation, Indian Medical Association (IMA), Indian Chest Society and National College of Chest Physicians (ICS-NCCP), Research Society for Study of Diabetes in India (RSSDI), Indian Association of Occupational Health Guidelines for Working Adults (IAOH), and API guidelines for immunization during COVID19 pandemic. All bodies recommend pneumococcal vaccines, sequence and preference of which depend on factors such as age, underlying conditions, and immune status. Integration of society recommendations and their implementation into public and private vaccination programs are required to promote adult immunization.
Collapse
|
10
|
Current view on novel vaccine technologies to combat human infectious diseases. Appl Microbiol Biotechnol 2022; 106:25-56. [PMID: 34889981 PMCID: PMC8661323 DOI: 10.1007/s00253-021-11713-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Inactivated and live attenuated vaccines have improved human life and significantly reduced morbidity and mortality of several human infectious diseases. However, these vaccines have faults, such as reactivity or suboptimal efficacy and expensive and time-consuming development and production. Additionally, despite the enormous efforts to develop vaccines against some infectious diseases, the traditional technologies have not been successful in achieving this. At the same time, the concerns about emerging and re-emerging diseases urge the need to develop technologies that can be rapidly applied to combat the new challenges. Within the last two decades, the research of vaccine technologies has taken several directions to achieve safe, efficient, and economic platforms or technologies for novel vaccines. This review will give a brief overview of the current state of the novel vaccine technologies, new vaccine candidates in clinical trial phases 1-3 (listed by European Medicines Agency (EMA) and Food and Drug Administration (FDA)), and vaccines based on the novel technologies which have already been commercially available (approved by EMA and FDA) with the special reference to pandemic COVID-19 vaccines. KEY POINTS: • Vaccines of the new generation follow the minimalist strategy. • Some infectious diseases remain a challenge for the vaccine development. • The number of new vaccine candidates in the late phase clinical trials remains low.
Collapse
|
11
|
Anggraeni R, Ana ID, Wihadmadyatami H. Development of mucosal vaccine delivery: an overview on the mucosal vaccines and their adjuvants. Clin Exp Vaccine Res 2022; 11:235-248. [DOI: 10.7774/cevr.2022.11.3.235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/10/2022] [Indexed: 11/22/2022] Open
Affiliation(s)
- Rahmi Anggraeni
- PT Swayasa Prakarsa, Universitas Gadjah Mada Science Techno Campus, Division of Drugs, Medical Devices, and Functional Food, Yogyakarta, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Hevi Wihadmadyatami
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
12
|
A Blockchain-Based Approach for COVID-19 Vaccine Lifecycle. ENTERP INF SYST-UK 2022. [DOI: 10.1007/978-3-031-08965-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Lynn DJ, Benson SC, Lynn MA, Pulendran B. Modulation of immune responses to vaccination by the microbiota: implications and potential mechanisms. Nat Rev Immunol 2022; 22:33-46. [PMID: 34002068 PMCID: PMC8127454 DOI: 10.1038/s41577-021-00554-7] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 02/05/2023]
Abstract
The need for highly effective vaccines that induce robust and long-lasting immunity has never been more apparent. However, for reasons that are still poorly understood, immune responses to vaccination are highly variable between different individuals and different populations. Furthermore, vaccine immunogenicity is frequently suboptimal in the very populations who are at most risk from infectious disease, including infants, the elderly, and those living in low-income and middle-income countries. Although many factors have the potential to influence vaccine immunogenicity and therefore vaccine effectiveness, increasing evidence from clinical studies and animal models now suggests that the composition and function of the gut microbiota are crucial factors modulating immune responses to vaccination. In this Review, we synthesize this evidence, discuss the immunological mechanisms that potentially mediate these effects and consider the potential of microbiota-targeted interventions to optimize vaccine effectiveness.
Collapse
Affiliation(s)
- David J Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia.
| | - Saoirse C Benson
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
| | - Miriam A Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Bali Pulendran
- Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
14
|
Masurel L, Bianca C, Lemarchand A. Space-velocity thermostatted kinetic theory model of tumor growth. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:5525-5551. [PMID: 34517499 DOI: 10.3934/mbe.2021279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The competition between cancer cells and immune system cells in inhomogeneous conditions is described at cell scale within the framework of the thermostatted kinetic theory. Cell learning is reproduced by increased cell activity during favorable interactions. The cell activity fluctuations are controlled by a thermostat. The direction of cell velocity is changed according to stochastic rules mimicking a dense fluid. We develop a kinetic Monte Carlo algorithm inspired from the direct simulation Monte Carlo (DSMC) method initially used for dilute gases. The simulations generate stochastic trajectories sampling the kinetic equations for the distributions of the different cell types. The evolution of an initially localized tumor is analyzed. Qualitatively different behaviors are observed as the field regulating activity fluctuations decreases. For high field values, i.e. efficient thermalization, cancer is controlled. For small field values, cancer rapidly and monotonously escapes from immunosurveillance. For the critical field value separating these two domains, the 3E's of immunotherapy are reproduced, with an apparent initial elimination of cancer, a long quasi-equilibrium period followed by large fluctuations, and the final escape of cancer, even for a favored production of immune system cells. For field values slightly smaller than the critical value, more regular oscillations of the number of immune system cells are spontaneously observed in agreement with clinical observations. The antagonistic effects that the stimulation of the immune system may have on oncogenesis are reproduced in the model by activity-weighted rate constants for the autocatalytic productions of immune system cells and cancer cells. Local favorable conditions for the launching of the oscillations are met in the fluctuating inhomogeneous system, able to generate a small cluster of immune system cells with larger activities than those of the surrounding cancer cells.
Collapse
Affiliation(s)
- Léon Masurel
- Laboratoire de Physique Théorique de la Matière Condensée, Sorbonne Université, CNRS, 4 place Jussieu, case courrier 121, 75252 Paris Cedex 05, France
| | - Carlo Bianca
- École Supérieure d'Ingénieurs en Génie Électrique, Productique et Management Industriel, Laboratoire Quartz EA 7393, Laboratoire de Recherche en Eco-innovation Industrielle et Energétique, 13 Boulevard de l'Hautil, 95092 Cergy Pontoise Cedex, France
| | - Annie Lemarchand
- Laboratoire de Physique Théorique de la Matière Condensée, Sorbonne Université, CNRS, 4 place Jussieu, case courrier 121, 75252 Paris Cedex 05, France
| |
Collapse
|
15
|
Barrett RS, Wiethoelter A, Halpin K. The Hendra virus vaccine: perceptions regarding the role of antibody titre testing. Aust Vet J 2021; 99:412-418. [PMID: 34151419 DOI: 10.1111/avj.13099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/19/2021] [Accepted: 05/29/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To elucidate veterinarians' and horse owners' perceptions towards the use of Hendra virus (HeV) antibody titre testing and how it influences veterinary advice. METHODS Six semi-structured phone interviews were conducted with veterinarians who have submitted samples for HeV antibody titre testing. Interviews were recorded, transcribed and thematically analysed to identify and report common themes within the data. RESULTS Veterinarians are predominantly using the titre tests as an alternative to vaccination due to clients' fear of vaccine reactions. The high cost of titre testing, the difficulty interpreting titre results and a lack of titre test recognition by authorities were the major barriers reported to using this test. Some veterinarians detailed difficulties communicating titre test procedures and results to their clients. The majority of veterinarians accepted titres of 64 or greater as evidence of protective immunity and would rely on those results for 12 months. However, there was discrepancy of these values and the level of confidence veterinarians had in interpreting the results of HeV antibody titre tests varied. CONCLUSION This study has provided an overview of the attitudes of horse owners and veterinarians towards HeV antibody titre testing. Although evidence for HeV vaccination titres as an indication of protective immunity is still inadequate, it will assist veterinarians in interpreting and communicating titre results.
Collapse
Affiliation(s)
- R S Barrett
- Faculty of Veterinary and Agricultural Science, University of Melbourne, Melbourne, Victoria, 3010, Australia.,Bellarine Veterinary Practice, Newcomb, Victoria, 3219, Australia
| | - A Wiethoelter
- Faculty of Veterinary and Agricultural Science, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - K Halpin
- Australian Centre for Disease Preparedness, Newcomb, Victoria, 3219, Australia
| |
Collapse
|
16
|
Recent advances in nano/microparticle-based oral vaccines. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021; 51:425-438. [PMID: 34150345 PMCID: PMC8196935 DOI: 10.1007/s40005-021-00537-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022]
Abstract
Background Vaccines are often recognized as one of the most cost-effective public health interventions in controlling infectious diseases. Most pathogens infiltrate the body from mucosal sites, primarily from the oral and pulmonary region and reach the systemic circulation where disease manifestation starts. Traditional needle-based vaccines are usually not capable of inducing immunity at the mucosal sites where pathogen infiltrates start, but induces systemic immunity. In contrast to needle-based vaccines, mucosally administered vaccines induce immunity at both the mucosal sites and systemically. The oral route of immunization is the most convenient way to administer the vaccines. However, due to the complicated and hostile gastrointestinal structure and environment, vaccines need to overcome major hurdles while retaining their stability and immunogenicity. Area covered This review will briefly discuss different barriers to oral vaccine development. It gives a brief overview of different types of nano/microparticle-based oral vaccines and discusses how physicochemical characteristics of the particles influence overall immunity after oral immunization. Expert opinion Formulation strategies using novel lipid and polymer-based nano/microparticle platforms retain stability and antigenicity of vaccines against the harsh gastrointestinal condition. The physicochemical properties of particles can be uniquely tailored to prolong the release of antigens, and attached ligands (M-cells and APC-ligands) can precisely target uptake by immune cells. These represent viable strategies for efficient delivery of oral vaccines.
Collapse
|
17
|
Hoes J, Pasmans H, Schurink-van 't Klooster TM, van der Klis FRM, Donken R, Berkhof J, de Melker HE. Review of long-term immunogenicity following HPV vaccination: Gaps in current knowledge. Hum Vaccin Immunother 2021; 18:1908059. [PMID: 34033518 PMCID: PMC8920133 DOI: 10.1080/21645515.2021.1908059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The licensed HPV vaccines are highly efficacious and induce high levels of neutralizing antibody levels, the assumed mediators of protection. However, a correlate of protection against HPV is lacking, and the evidence is still limited as to long-term persistence of antibodies, especially following reduced dosing schedules. The World Health Organization (WHO) urges immunization of young girls as part of the strategy to eliminate cervical cancer, thus long-lasting protection is required. The current review describes long-term follow-up regarding vaccine-induced seropositivity and antibody level development following the different vaccines and dosing schedules. Implications and opportunities of long-term vaccine-induced immune responses are discussed, such as the gaps in monitoring of long-term immunogenicity, the possibilities of reduced dosing schedules, and the importance of evidence for durable immunity.
Collapse
Affiliation(s)
- J Hoes
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.,Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H Pasmans
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - T M Schurink-van 't Klooster
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - F R M van der Klis
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - R Donken
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Berkhof
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H E de Melker
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
18
|
Won JH, Lee H. The Current Status of Drug Repositioning and Vaccine Developments for the COVID-19 Pandemic. Int J Mol Sci 2020; 21:E9775. [PMID: 33371468 PMCID: PMC7767501 DOI: 10.3390/ijms21249775] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022] Open
Abstract
Since the outbreak of coronavirus disease 2019 (COVID-19) was first identified, the world has vehemently worked to develop treatments and vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) at an unprecedented speed. Few of the repositioned drugs for COVID-19 have shown that they were efficacious and safe. In contrast, a couple of vaccines against SARS-CoV-2 will be ready for mass rollout early next year. Despite successful vaccine development for COVID-19, the world will face a whole new set of challenges including scale-up manufacturing, cold-chain logistics, long-term safety, and low vaccine acceptance. We highlighted the importance of knowledge sharing and collaboration to find innovative answers to these challenges and to prepare for newly emerging viruses.
Collapse
Affiliation(s)
- Jung-Hyun Won
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea;
- Center for Convergence Approaches in Drug Development, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea
| | - Howard Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea;
- Center for Convergence Approaches in Drug Development, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Seoul National University, Seoul 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul 03080, Korea
- Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 16229, Korea
| |
Collapse
|
19
|
Han B, Hoang BX. Opinions on the current pandemic of COVID-19: Use functional food to boost our immune functions. J Infect Public Health 2020; 13:1811-1817. [PMID: 32948484 PMCID: PMC7831995 DOI: 10.1016/j.jiph.2020.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/14/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022] Open
Abstract
The pandemic of novel coronavirus caused COVID-19 had resulted in a high number of hospitalizations and deaths and caused a devastating toll on human and society health. The symptoms of the infected patients vary significantly, from life-threatening to mild or even asymptomatic. This clinical observation led to hypothesize on the critical role of host innate immunity in the disease development and progression. As the first defense barrier against microorganisms, the innate immune reaction determines not only the viral infection rate but also immune-mediated response. Therefore, promote healthy behaviors to enhance innate immunity with functional food and nutritional agents may be a rational strategy for minimizing damages caused by viruses to global health.
Collapse
Affiliation(s)
- Bo Han
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Department of Surgery, University of Southern California, Los Angeles, CA 90033, United States
| | - Ba X Hoang
- Nimni-Cordoba Tissue Engineering and Drug Discovery Laboratory, Department of Surgery, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
20
|
Ullah MA, Araf Y, Sarkar B, Moin AT, Reshad RAI, Rahman MDH. Pathogenesis, Diagnosis and Possible Therapeutic Options for COVID-19. JOURNAL OF CLINICAL AND EXPERIMENTAL INVESTIGATIONS 2020. [DOI: 10.29333/jcei/8564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
21
|
Raghupathi V, Ren J, Raghupathi W. Studying Public Perception about Vaccination: A Sentiment Analysis of Tweets. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E3464. [PMID: 32429223 PMCID: PMC7277574 DOI: 10.3390/ijerph17103464] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 01/04/2023]
Abstract
Text analysis has been used by scholars to research attitudes toward vaccination and is particularly timely due to the rise of medical misinformation via social media. This study uses a sample of 9581 vaccine-related tweets in the period January 1, 2019 to April 5, 2019. The time period is of the essence because during this time, a measles outbreak was prevalent throughout the United States and a public debate was raging. Sentiment analysis is applied to the sample, clustering the data into topics using the term frequency-inverse document frequency (TF-IDF) technique. The analyses suggest that most (about 77%) of the tweets focused on the search for new/better vaccines for diseases such as the Ebola virus, human papillomavirus (HPV), and the flu. Of the remainder, about half concerned the recent measles outbreak in the United States, and about half were part of ongoing debates between supporters and opponents of vaccination against measles in particular. While these numbers currently suggest a relatively small role for vaccine misinformation, the concept of herd immunity puts that role in context. Nevertheless, going forward, health experts should consider the potential for the increasing spread of falsehoods that may get firmly entrenched in the public mind.
Collapse
Affiliation(s)
- Viju Raghupathi
- Koppelman School of Business, Brooklyn College of the City University of New York, Brooklyn, NY 11210, USA;
| | - Jie Ren
- Gabelli School of Business, Fordham University, New York, NY 10023, USA;
| | | |
Collapse
|
22
|
Zrzavy T, Kollaritsch H, Rommer PS, Boxberger N, Loebermann M, Wimmer I, Winkelmann A, Zettl UK. Vaccination in Multiple Sclerosis: Friend or Foe? Front Immunol 2019; 10:1883. [PMID: 31440255 PMCID: PMC6693409 DOI: 10.3389/fimmu.2019.01883] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 07/24/2019] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating disease of the central nervous systems (CNS). Disease-modifying treatments (including immunosuppressive treatments) have shown positive effects on the disease course, but are associated with systemic consequences on the immune system and may increase the risk of infections and alter vaccine efficiency. Therefore, vaccination of MS patients is of major interest. Over the last years, vaccine hesitancy has steadily grown especially in Western countries, partly due to fear of sequelae arising from vaccination, especially neurological disorders. The interaction of vaccination and MS has been discussed for decades. In this review, we highlight the immunology of vaccination, provide a review of literature and discuss the clinical consideration of MS, vaccination and immunosuppression. In conclusion, there is consensus that MS cannot be caused by vaccines, neither by inactivated nor by live vaccines. However, particular attention should be paid to two aspects: First, in immunocompromised patients, live vaccines may lead to a stronger immune reaction with signs of the disease against which the patients have been vaccinated, albeit in weakened form. Second, protection provided by vaccination should be controlled in patients who have been vaccinated while receiving immunomodulatory or immunosuppressive treatment. In conclusion, there is evidence that systemic infections can worsen MS, thus vaccination will lower the risk of relapses by reducing the risk of infections. Therefore, vaccination should be in general recommended to MS patients.
Collapse
Affiliation(s)
- Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Herwig Kollaritsch
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Paulus S. Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
| | - Nina Boxberger
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
| | - Micha Loebermann
- Department of Tropical Medicine and Infectious Diseases, University of Rostock, Rostock, Germany
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Uwe K. Zettl
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
- Department of Neurology, University of Rostock, Rostock, Germany
| |
Collapse
|
23
|
Kim MH, Kim HJ, Chang J. Superior immune responses induced by intranasal immunization with recombinant adenovirus-based vaccine expressing full-length Spike protein of Middle East respiratory syndrome coronavirus. PLoS One 2019; 14:e0220196. [PMID: 31329652 PMCID: PMC6645677 DOI: 10.1371/journal.pone.0220196] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) causes an acute and severe lower respiratory illness as well as vomiting, diarrhea, and renal failure. Because no licensed MERS-CoV vaccines are currently available, preventive and therapeutic measures are urgently needed. The surface spike (S) glycoprotein of MERS-CoV, which binds to the cellular receptor dipeptidyl peptidase 4 (DPP4), is considered as a major target for MERS-CoV vaccine development. Here, we designed recombinant replication-deficient adenovirus-based vaccines expressing the N-terminal domain (rAd/NTD) and receptor-binding domain (rAd/RBD) of the MERS-CoV S1 subunit and full-length Spike protein (rAd/Spike). We found that immunization with candidate vaccines via intranasal route induced S1-specific IgG antibodies and neutralizing antibodies against MERS spike pseudotyped virus. Especially, rAd/Spike induced the highest neutralizing antibody titer and the strongest cytokine-induced T cell responses among the three candidate vaccines. To compare the immune responses induced by different administration routes, rAd/Spike was administered via intranasal, sublingual, or intramuscular route. All these administration routes exhibited neutralizing effects in the serum. MERS-CoV-specific neutralizing IgA antibodies in the bronchoalveolar lavage fluid were only induced by intranasal and sublingual administration but not by intramuscular administration. Intranasal administration with rAd/Spike also created resident memory CD8 T cells in the airway and lung parenchyma. Taken together, our results showed that both the humoral and cellular immune responses are highly induced by rAd/Spike administration, suggesting that rAd/Spike may confer protection against MERS-CoV infection.
Collapse
Affiliation(s)
- Myung Hee Kim
- Graduate School of Pharmaceutical Sciences, Ewha Woman’s University, Seoul, Republic of Korea
| | - Hyun Jik Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Woman’s University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
24
|
Kitazawa Y, Ueta H, Sawanobori Y, Katakai T, Yoneyama H, Ueha S, Matsushima K, Tokuda N, Matsuno K. Novel Targeting to XCR1 + Dendritic Cells Using Allogeneic T Cells for Polytopical Antibody Responses in the Lymph Nodes. Front Immunol 2019; 10:1195. [PMID: 31191552 PMCID: PMC6548820 DOI: 10.3389/fimmu.2019.01195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 05/10/2019] [Indexed: 01/23/2023] Open
Abstract
Vaccination strategy that induce efficient antibody responses polytopically in most lymph nodes (LNs) against infections has not been established yet. Because donor-specific blood transfusion induces anti-donor class I MHC antibody production in splenectomized rats, we examined the mechanism and significance of this response. Among the donor blood components, T cells were the most efficient immunogens, inducing recipient T cell and B cell proliferative responses not only in the spleen, but also in the peripheral and gut LNs. Donor T cells soon migrated to the splenic T cell area and the LNs, with a temporary significant increase in recipient NK cells. XCR1+ resident dendritic cells (DCs), but not XCR1− DCs, selectively phagocytosed donor class I MHC+ fragments after 1 day. After 1.5 days, both DC subsets formed clusters with recipient CD4+ T cells, which proliferated within these clusters. Inhibition of donor T cell migration or depletion of NK cells by pretreatment with pertussis toxin or anti-asialoGM1 antibody, respectively, significantly suppressed DC phagocytosis and subsequent immune responses. Three allogeneic strains with different NK activities had the same response but with different intensity. Donor T cell proliferation was not required, indicating that the graft vs. host reaction is dispensable. Intravenous transfer of antigen-labeled and mitotic inhibitor-treated allogeneic, but not syngeneic, T cells induced a polytopical antibody response to labeled antigens in the LNs of splenectomized rats. These results demonstrate a novel mechanism of alloresponses polytopically in the secondary lymphoid organs (SLOs) induced by allogeneic T cells. Donor T cells behave as self-migratory antigen ferries to be delivered to resident XCR1+ DCs with negligible commitment of migratory DCs. Allogeneic T cells may be clinically applicable as vaccine vectors for polytopical prophylactic antibody production even in asplenic or hyposplenic individuals.
Collapse
Affiliation(s)
- Yusuke Kitazawa
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Hisashi Ueta
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Yasushi Sawanobori
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Tomoya Katakai
- Department of Immunology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | | | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Nobuko Tokuda
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Kenjiro Matsuno
- Department of Anatomy (Macro), School of Medicine, Dokkyo Medical University, Tochigi, Japan
| |
Collapse
|
25
|
Abstract
Ebola virus (EBOV) is a single-stranded RNA virus that causes Ebola virus disease (EVD), characterized by excessive inflammation, lymphocyte apoptosis, hemorrhage, and coagulation defects leading to multiorgan failure and shock. Recombinant vesicular stomatitis virus expressing the EBOV glycoprotein (VSV-EBOV), which is highly efficacious against lethal challenge in nonhuman primates, is the only vaccine that successfully completed a phase III clinical trial. Additional studies showed VSV-EBOV provides complete and partial protection to macaques immunized 7 and 3 days before EBOV challenge, respectively. However, the mechanisms by which this live-attenuated vaccine elicits rapid protection are only partially understood. To address this, we carried out a longitudinal transcriptome analysis of host responses in whole-blood samples collected from cynomolgus macaques vaccinated with VSV-EBOV 28, 21, 14, 7, and 3 days before EBOV challenge. Our findings indicate the transcriptional response to the vaccine peaks 7 days following vaccination and contains signatures of both innate antiviral immunity as well as B-cell activation. EBOV challenge 1 week after vaccination resulted in large gene expression changes suggestive of a recall adaptive immune response 14 days postchallenge. Lastly, the timing and magnitude of innate immunity and interferon-stimulated gene expression correlated with viral burden and disease outcome in animals vaccinated 3 days before challenge.IMPORTANCE Ebola virus (EBOV) is the causative agent of Ebola virus disease (EVD), a deadly disease and major public health threat worldwide. A safe and highly efficacious vesicular stomatitis virus-based vaccine against EBOV is the only platform that has successfully completed phase III clinical trials and has been used in recent and ongoing outbreaks. Earlier studies showed that antibodies are the main mode of protection when this vaccine is administered 28 days before EBOV challenge. Recently, we showed this vaccine can provide protection when administered as early as 3 days before challenge and before antibodies are detected. This study seeks to identify the mechanisms of rapid protection, which in turn will pave the way for improved vaccines and therapeutics. Additionally, this study provides insight into host gene expression signatures that could provide early biomarkers to identify infected individuals who are at highest risk of poor outcomes.
Collapse
|
26
|
Reuven EM, Leviatan Ben-Arye S, Yu H, Duchi R, Perota A, Conchon S, Bachar Abramovitch S, Soulillou JP, Galli C, Chen X, Padler-Karavani V. Biomimetic Glyconanoparticle Vaccine for Cancer Immunotherapy. ACS NANO 2019; 13:2936-2947. [PMID: 30840433 PMCID: PMC6756924 DOI: 10.1021/acsnano.8b07241] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Cancer immunotherapy aims to harness the immune system to combat malignant processes. Transformed cells harbor diverse modifications that lead to formation of neoantigens, including aberrantly expressed cell surface carbohydrates. Targeting tumor-associated carbohydrate antigens (TACA) hold great potential for cancer immunotherapy. N-glycolylneuraminic acid (Neu5Gc) is a dietary non-human immunogenic carbohydrate that accumulates on human cancer cells, thereby generating neoantigens. In mice, passive immunotherapy with anti-Neu5Gc antibodies inhibits growth of Neu5Gc-positive tumors. Here, we designed an active cancer vaccine immunotherapy strategy to target Neu5Gc-positive tumors. We generated biomimetic glyconanoparticles using engineered αGal knockout porcine red blood cells to form nanoghosts (NGs) that either express (NGpos) or lack expression (NGneg) of Neu5Gc-glycoconjugates in their natural context. We demonstrated that optimized immunization of "human-like" Neu5Gc-deficient Cmah-/- mice with NGpos glyconanoparticles induce a strong, diverse and persistent anti-Neu5Gc IgG immune response. The resulting anti-Neu5Gc IgG antibodies were also detected within Neu5Gc-positive tumors and inhibited tumor growth in vivo. Using detailed glycan microarray analysis, we further demonstrate that the kinetics and quality of the immune responses influence the efficacy of the vaccine. These findings reinforce the potential of TACA neoantigens and the dietary non-human sialic acid Neu5Gc, in particular, as immunotherapy targets.
Collapse
Affiliation(s)
- Eliran Moshe Reuven
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, 69978, Israel
| | | | - Hai Yu
- Department of Chemistry, University of California-Davis, Davis, CA 95616, USA
| | - Roberto Duchi
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Andrea Perota
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
| | - Sophie Conchon
- Institut de Transplantation–Urologie–Néphrologie, INSERM Unité Mixte de Recherche 1064, Centre Hospitalo Universitaire de Nantes, Nantes 44000, France
| | | | - Jean-Paul Soulillou
- Institut de Transplantation–Urologie–Néphrologie, INSERM Unité Mixte de Recherche 1064, Centre Hospitalo Universitaire de Nantes, Nantes 44000, France
| | - Cesare Galli
- Avantea, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100 Cremona, Italy
- FondazioneAvantea Cremona, Italy
| | - Xi Chen
- Department of Chemistry, University of California-Davis, Davis, CA 95616, USA
| | - Vered Padler-Karavani
- Department of Cell Research and Immunology, Tel Aviv University, Tel Aviv, 69978, Israel
- Corresponding Author: Department of Cell Research & Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978 Israel. Tel: +972-3-640-6737. Fax: +972-3-642-2046.
| |
Collapse
|
27
|
Papp KA, Haraoui B, Kumar D, Marshall JK, Bissonnette R, Bitton A, Bressler B, Gooderham M, Ho V, Jamal S, Pope JE, Steinhart AH, Vinh DC, Wade J. Vaccination Guidelines for Patients With Immune-Mediated Disorders on Immunosuppressive Therapies. J Cutan Med Surg 2018; 23:50-74. [PMID: 30463418 PMCID: PMC6330697 DOI: 10.1177/1203475418811335] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND: Patients with immune-mediated diseases on immunosuppressive therapies have more infectious episodes than healthy individuals, yet vaccination practices by physicians for this patient population remain suboptimal. OBJECTIVES: To evaluate the safety and efficacy of vaccines in individuals exposed to immunosuppressive therapies and provide evidence-based clinical practice recommendations. METHODS: A literature search for vaccination safety and efficacy in patients on immunosuppressive therapies (2009-2017) was conducted. Results were assessed using the Grading of Recommendation, Assessment, Development, and Evaluation system. RESULTS: Several immunosuppressive therapies attenuate vaccine response. Thus, vaccines should be administered before treatment whenever feasible. Inactivated vaccines can be administered without treatment discontinuation. Similarly, evidence suggests that the live zoster vaccine is safe and effective while on select immunosuppressive therapy, although use of the subunit vaccine is preferred. Caution regarding other live vaccines is warranted. Drug pharmacokinetics, duration of vaccine-induced viremia, and immune response kinetics should be considered to determine appropriate timing of vaccination and treatment (re)initiation. Infants exposed to immunosuppressive therapies through breastmilk can usually be immunized according to local guidelines. Intrauterine exposure to immunosuppressive agents is not a contraindication for inactivated vaccines. Live attenuated vaccines scheduled for infants and children ⩾12 months of age, including measles, mumps, rubella, and varicella, can be safely administered as sufficient time has elapsed for drug clearance. CONCLUSIONS: Immunosuppressive agents may attenuate vaccine responses, but protective benefit is generally maintained. While these recommendations are evidence based, they do not replace clinical judgment, and decisions regarding vaccination must carefully assess the risks, benefits, and circumstances of individual patients.
Collapse
Affiliation(s)
- Kim A Papp
- 1 K Papp Clinical Research, Waterloo, ON, Canada.,2 Probity Medical Research, Waterloo, ON, Canada
| | - Boulos Haraoui
- 3 Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Deepali Kumar
- 4 University Health Network, Toronto, ON, Canada.,5 Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - John K Marshall
- 6 Department of Medicine and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | | | - Alain Bitton
- 8 McGill University Health Centre, Montreal, QC, Canada
| | - Brian Bressler
- 9 Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,10 St Paul's Hospital, Vancouver, BC, Canada
| | - Melinda Gooderham
- 2 Probity Medical Research, Waterloo, ON, Canada.,11 Faculty of Medicine, Queen's University, Kingston, ON, Canada
| | - Vincent Ho
- 9 Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Shahin Jamal
- 12 Vancouver Coastal Health, Vancouver, BC, Canada
| | - Janet E Pope
- 13 Faculty of Medicine, University of Western Ontario, London, ON, Canada.,14 St Joseph's Health Care, London, ON, Canada
| | - A Hillary Steinhart
- 5 Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,15 Mount Sinai Hospital, Toronto, ON, Canada
| | - Donald C Vinh
- 8 McGill University Health Centre, Montreal, QC, Canada.,16 Research Institute, McGill University Health Centre, Montreal, QC, Canada
| | - John Wade
- 9 Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,17 Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
28
|
Yan LM, Li OTW, Poh CM, Perera RAPM, Valkenburg SA, Peiris M, Poon LLM. Combined use of live-attenuated and inactivated influenza vaccines to enhance heterosubtypic protection. Virology 2018; 525:73-82. [PMID: 30248524 DOI: 10.1016/j.virol.2018.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 01/04/2023]
Abstract
The limited protection of current commerical vaccines necessitates the investigation of novel vaccine strategies for unpredictable outbreaks. To investigate the feasibility of using vaccines derived from Group 1 influenza A virus to induce broadly cross-reactive immune responses against multiple influenza subtypes, we tested a panel of sequential 4-dose immunization regimens in mice. Mice were treated with inactivated (seasonal H1N1, pandemic H1N1 and H5N1) and vaccinia virus-based H5N1 live-attenuated vaccines in different combinations. Mice were then challenged by viruses of either Group 1 (H1N1) or Group 2 (H3N2, H7N7) influenza virus. All studied sequential 4-dose vaccinations could induce some degrees of heterosubtypic protection in mice. Amongst all these regimens, the combined use of inactivated and live-attenuated vaccines could achieve the best heterologous protection. These results highlight the synergistic effect of combining different vaccine platforms to enhance heterosubtypic protection against influenza viruses.
Collapse
Affiliation(s)
- Li-Meng Yan
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Olive T W Li
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Chek M Poh
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Ranawaka A P M Perera
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie A Valkenburg
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China; HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Malik Peiris
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Leo L M Poon
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
29
|
Stanger KJ, McGregor H, Marenda M, Morton JM, Larsen J. Assessment of the efficacy of an autogenous vaccine against Yersinia pseudotuberculosis in young Merino sheep. N Z Vet J 2018; 67:27-35. [PMID: 30221588 DOI: 10.1080/00480169.2018.1523758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
AIMS To assess the efficacy of an autogenous vaccine against Yersinia pseudotuberculosis III in preventing clinical disease and deaths due to yersiniosis in young Merino sheep, and to determine the effect of vaccination on the prevalence of faecal shedding of pathogenic Yersinia spp., daily liveweight gain, and development of antibodies to Yersinia spp. following vaccination and natural exposure. METHODS In six groups (three groups each from two farms) of young Merino sheep, 148-150 animals were systematically allocated to be vaccinated twice with an autogenous, formalin- killed bacterin vaccine containing Y. pseudotuberculosis serotype III or to remain non-vaccinated. All vaccinated and non-vaccinated sheep were run together in their original groups throughout the trial. Faecal and blood samples were collected, and liveweight measured, at the time of vaccination and subsequently over a 6-month period to determine faecal shedding of Y. enterocolitica and Y. pseudotuberculosis, seroprevalence of antibodies to Yersinia outer membrane proteins (YOP) and changes in liveweight. RESULTS None of the six trial groups experienced an outbreak of clinical yersiniosis during the study period. On Farm A, the prevalence of shedding of either or both Yersinia spp. was <40% on all but one sampling occasions. On Farm B the prevalence of shedding of both Yersinia spp. peaked at 98%, 96 days after vaccination. Mean liveweight and daily liveweight gain at the end of the study were similar in vaccinated and non-vaccinated groups on both farms (p>0.1), as was the prevalence of faecal shedding of Yersinia spp. (p>0.2), and the proportion of animals that became seropositive for antibodies to YOP following vaccination (p>0.1). CONCLUSIONS AND CLINICAL RELEVANCE This vaccine had, at most, limited effects on seroconversion and, under the conditions of this study, had no demonstrable impact on liveweight, mean daily liveweight gain or faecal shedding of Yersinia spp. Further studies are needed to determine the efficacy of this vaccine during outbreaks of yersiniosis or following experimental challenge with pathogenic Yersinia spp..
Collapse
Affiliation(s)
- K J Stanger
- a Faculty of Veterinary and Agricultural Sciences , University of Melbourne , 250 Princes Highway, Werribee , VIC , Australia
| | - H McGregor
- a Faculty of Veterinary and Agricultural Sciences , University of Melbourne , 250 Princes Highway, Werribee , VIC , Australia
| | - M Marenda
- a Faculty of Veterinary and Agricultural Sciences , University of Melbourne , 250 Princes Highway, Werribee , VIC , Australia
| | - J M Morton
- b Jemora Pty Ltd. , PO Box 2277, Geelong , VIC , Australia
| | - Jwa Larsen
- a Faculty of Veterinary and Agricultural Sciences , University of Melbourne , 250 Princes Highway, Werribee , VIC , Australia
| |
Collapse
|
30
|
Quach QH, Ang SK, Chu JHJ, Kah JCY. Size-dependent neutralizing activity of gold nanoparticle-based subunit vaccine against dengue virus. Acta Biomater 2018; 78:224-235. [PMID: 30099200 DOI: 10.1016/j.actbio.2018.08.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 12/17/2022]
Abstract
Dengue results in substantial human morbidity and significant socio-economic impacts, but a specific dengue therapeutic is not available. The currently available dengue vaccine has low efficacy and high rate of adverse effects, necessitating different strategies for the development of a safer and more efficient vaccine against dengue virus. We describe here a hybrid combination of different-sized gold nanoparticles (AuNPs) and domain III of envelope glycoprotein derived from serotype 2 of dengue virus (EDIII) as dengue subunit vaccine. The efficacy of the EDIII-functionalized AuNPs (AuNP-E) to induce neutralizing antibody in BALB/c mice is evaluated. Obtained results show that AuNP-E induced a high level of antibody which mediates serotype-specific neutralization of dengue virus. More importantly, the level of antibody is dependent on both the size of AuNPs and the concentration of AuNP-E, implicating the possibility to modulate it through adjusting these parameters. These results represent an important step towards the development of tetravalent AuNP-based subunit dengue vaccine. STATEMENT OF SIGNIFICANCE This research presents a novel subunit vaccine against dengue virus using a hybrid comprising gold nanoparticles (AuNPs) and domain III of envelop protein (EDIII). We proved the neutralizing activity of anti-EDIII antibody induced in immunized mice on Dengue virus serotype 2 in an AuNP core size and concentration dependent manner. The hybrid concept behind this work could also be adopted for the development of a tetravalent vaccine against four serotypes of Dengue virus.
Collapse
|
31
|
PD-L1, TIM-3, and CTLA-4 Blockade Fails To Promote Resistance to Secondary Infection with Virulent Strains of Toxoplasma gondii. Infect Immun 2018; 86:IAI.00459-18. [PMID: 29967089 DOI: 10.1128/iai.00459-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 01/05/2023] Open
Abstract
T cell exhaustion is a state of hyporesponsiveness that develops during many chronic infections and cancer. Neutralization of inhibitory receptors, or "checkpoint blockade," can reverse T cell exhaustion and lead to beneficial prognoses in experimental and clinical settings. Whether checkpoint blockade can resolve lethal acute infections is less understood but may be beneficial in vaccination protocols that fail to elicit sterilizing immunity. Since a fully protective vaccine for any human parasite has yet to be developed, we explored the efficacy of checkpoint inhibitors in a mouse model of Toxoplasma gondii reinfection. Mice chronically infected with an avirulent type III strain survive reinfection with the type I RH strain but not the MAS, GUY-DOS, and GT1 parasite strains. We report here that mouse susceptibility to secondary infection correlates with the initial parasite burden and that protection against the RH strain is dependent on CD8 but not CD4 T cells in this model. When given a lethal secondary infection, CD8 and CD4 T cells upregulate several coinhibitory receptors, including PD-1, TIM-3, 4-1bb, and CTLA-4. Moreover, the gamma interferon (IFN-γ) response of CD8 but not CD4 T cells is significantly reduced during secondary infection with virulent strains, suggesting that checkpoint blockade may reduce disease severity. However, single and combination therapies targeting TIM-3, CTLA-4, and/or PD-L1 failed to reverse susceptibility to secondary infection. These results suggest that additional host responses, which are refractory to checkpoint blockade, are likely required for immunity to this pathogen.
Collapse
|
32
|
Toh ZQ, Cheow KWB, Russell FM, Hoe E, Reyburn R, Fong J, Tuivaga E, Ratu FT, Nguyen CD, Matanitobua S, Reitsma A, Tabrizi SN, Garland SM, Mulholland EK, Licciardi PV. Cellular Immune Responses 6 Years Following 1, 2, or 3 Doses of Quadrivalent HPV Vaccine in Fijian Girls and Subsequent Responses to a Dose of Bivalent HPV Vaccine. Open Forum Infect Dis 2018; 5:ofy147. [PMID: 30019002 PMCID: PMC6041981 DOI: 10.1093/ofid/ofy147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background This study examined the cellular immunity of 0, 1, 2, and 3 doses of Gardasil vaccine (4vHPV) in girls after 6 years and their responses to a subsequent dose of Cervarix vaccine (2vHPV). Methods A subset of girls (n = 59) who previously received 0, 1, 2, or 3 doses of 4vHPV 6 years earlier were randomly selected from a cohort study of Fijian girls (age 15-19 years). Blood was collected before and 28 days after a dose of 2vHPV. The HPV16- and HPV18-specific cellular immune response was determined by IFNγ-ELISPOT and by measurement of cytokines in peripheral blood mononuclear cell supernatants. Results Six years after 4vHPV vaccination, HPV18-specific responses were significantly lower in the 1- (1D) or 2-dose (2D) recipients compared with 3-dose recipients (2D: IFNγ-ELISPOT: P = .008; cytokines, IFNγ: P = .002; IL-2: P = .022; TNFα: P = .016; IL-10: P = .018; 1D: IL-2: P = .031; IL-10: P = .014). These differences were no longer significant post-2vHPV. No significant differences in HPV16 responses (except IL-2, P < .05) were observed between the 2- or 1-dose recipients and 3-dose recipients. Conclusions These data suggest that cellular immunity following reduced-dose schedules was detectable after 6 years, although the responses were variable between HPV types and dosage groups. The clinical significance of this is unknown. Further studies on the impact of reduced dose schedules are needed, particularly in high-disease burden settings.
Collapse
Affiliation(s)
- Zheng Quan Toh
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | | | - Fiona M Russell
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Edwin Hoe
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Rita Reyburn
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - James Fong
- Ministry of Health and Medical Services, Suva, Fiji
| | | | | | - Cattram D Nguyen
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Andrea Reitsma
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Sepehr N Tabrizi
- Department of Obstetrics and Gynecology, The University of Melbourne, The Royal Women's Hospital and Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Regional HPV Labnet Reference Laboratory, Department of Microbiology and Infectious Disease, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Suzanne M Garland
- Department of Obstetrics and Gynecology, The University of Melbourne, The Royal Women's Hospital and Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Regional HPV Labnet Reference Laboratory, Department of Microbiology and Infectious Disease, The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Edward K Mulholland
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom.,Department of Child Health, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
33
|
Lim H, In HJ, Lee JA, Sik Yoo J, Lee SW, Chung GT, Choi YK, Chung JK, Cho SJ, Lee JW. The immunogenicity and protection effect of an inactivated coxsackievirus A6, A10, and A16 vaccine against hand, foot, and mouth disease. Vaccine 2018; 36:3445-3452. [DOI: 10.1016/j.vaccine.2018.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 04/10/2018] [Accepted: 05/01/2018] [Indexed: 01/17/2023]
|
34
|
Blanco E, Pérez-Andrés M, Arriba-Méndez S, Contreras-Sanfeliciano T, Criado I, Pelak O, Serra-Caetano A, Romero A, Puig N, Remesal A, Torres Canizales J, López-Granados E, Kalina T, Sousa AE, van Zelm M, van der Burg M, van Dongen JJ, Orfao A. Age-associated distribution of normal B-cell and plasma cell subsets in peripheral blood. J Allergy Clin Immunol 2018; 141:2208-2219.e16. [DOI: 10.1016/j.jaci.2018.02.017] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/15/2017] [Accepted: 02/05/2018] [Indexed: 01/12/2023]
|
35
|
Starodubova ES, Kuzmenko YV, Pankova EO, Latanova AA, Preobrazhenskaya OV, Karpov VL. A DNA Construct That Encodes the Rabies Virus Consensus Glycoprotein with a Proteasome Degradation Signal Induces Antibody Production with IgG2A Subtype Predominance. Mol Biol 2018. [DOI: 10.1134/s0026893318030135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Persson G, Pors SE, Thøfner ICN, Bojesen AM. Vaccination with outer membrane vesicles and the fimbrial protein FlfA offers improved protection against lesions following challenge with Gallibacterium anatis. Vet Microbiol 2018; 217:104-111. [PMID: 29615242 DOI: 10.1016/j.vetmic.2018.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 02/16/2018] [Accepted: 03/09/2018] [Indexed: 01/25/2023]
Abstract
Gallibacterium anatis is an opportunistic poultry pathogen belonging to the Pasteurellaceae family. It has been shown to cause oophoritis, salpingitis and peritonitis in hens, as well as being associated with reduced semen quality in cockerels. Widespread multidrug resistance and substantial antigenic variation among strains of Gallibacterium anatis is a major constraint to treatment with antimicrobials and prevention of infection by vaccination. Novel vaccine strategies targeting G. anatis are therefore necessary. Outer membrane vesicles (OMVs) are nanosized vesicles formed from the outer membrane of Gram-negative bacteria. These vesicles have shown promising potential as both adjuvants and as vaccine candidates against numerous bacterial species. A high vesiculating mutant of G. anatis (G. anatis ΔtolR) has previously been made, enabling production of OMVs in large scale. In this study, we elucidated the potential of G. anatis ΔtolR OMVs as adjuvant for the conserved antigens GtxA-N (the N-terminal part of the RTX like toxin Gallibacterium toxin A) and FlfA (F17-like fimbria), as well as evaluated if combinations of OMVs together with antigens could facilitate cross-protective immunity against three different strains of G. anatis. We showed that ΔtolR OMVs function as an adjuvant for GtxA-N by inducing antigen specific antibody production. However, OMVs in combination with GtxA-N failed to induce protection against lesions after challenge infection. In contrast, vaccination with OMVs in combination with FlfA protected against lesions, especially in the salpinx, caused by two diverse strains of G. anatis, thereby indicating a cross-protective potential. No protection against the third G. anatis strain 7990 could be obtained in any of the experimental settings. In conclusion, ΔtolR OMVs and FlfA could serve as potential future vaccine components againt G. anatis.
Collapse
Affiliation(s)
- Gry Persson
- Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Clinical Biochemistry, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Susanne E Pors
- Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Laboratory of Reproductive Biology, Copenhagen University Hospital, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ida C N Thøfner
- Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Anders M Bojesen
- Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
37
|
Role of the Antigen Capture Pathway in the Induction of a Neutralizing Antibody Response to Anthrax Protective Antigen. mBio 2018; 9:mBio.00209-18. [PMID: 29487236 PMCID: PMC5829829 DOI: 10.1128/mbio.00209-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Toxin neutralizing antibodies represent the major mode of protective immunity against a number of toxin-mediated bacterial diseases, including anthrax; however, the cellular mechanisms that lead to optimal neutralizing antibody responses remain ill defined. Here we show that the cellular binding pathway of anthrax protective antigen (PA), the binding component of anthrax toxin, determines the toxin neutralizing antibody response to this antigen. PA, which binds cellular receptors and efficiently enters antigen-presenting cells by receptor-mediated endocytosis, was found to elicit robust anti-PA IgG and toxin neutralizing antibody responses. In contrast, a receptor binding-deficient mutant of PA, which does not bind receptors and only inefficiently enters antigen-presenting cells by macropinocytosis, elicited very poor antibody responses. A chimeric protein consisting of the receptor binding-deficient PA mutant tethered to the binding subunit of cholera toxin, which efficiently enters cells using the cholera toxin receptor rather than the PA receptor, elicited an anti-PA IgG antibody response similar to that elicited by wild-type PA; however, the chimeric protein elicited a poor toxin neutralizing antibody response. Taken together, our results demonstrate that the antigen capture pathway can dictate the magnitudes of the total IgG and toxin neutralizing antibody responses to PA as well as the ratio of the two responses.IMPORTANCE Neutralizing antibodies provide protection against a number of toxin-mediated bacterial diseases by inhibiting toxin action. Therefore, many bacterial vaccines are designed to induce a toxin neutralizing antibody response. We have used protective antigen (PA), the binding component of anthrax toxin, as a model antigen to investigate immune mechanisms important for the induction of robust toxin neutralizing antibody responses. We found that the pathway used by antigen-presenting cells to capture PA dictates the robustness of the neutralizing antibody response to this antigen. These results provide new insights into immune mechanisms that play an important role in the induction of toxin neutralizing antibody responses and may be useful in the design of new vaccines against toxin-mediated bacterial diseases.
Collapse
|
38
|
Bekele Y, Graham RL, Soeria-Atmadja S, Nasi A, Zazzi M, Vicenti I, Naver L, Nilsson A, Chiodi F. Hepatitis B Virus Vaccination in HIV-1-Infected Young Adults: A Tool to Reduce the Size of HIV-1 Reservoirs? Front Immunol 2018; 8:1966. [PMID: 29375579 PMCID: PMC5767726 DOI: 10.3389/fimmu.2017.01966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/19/2017] [Indexed: 01/12/2023] Open
Abstract
During anti-retroviral therapy (ART) HIV-1 persists in cellular reservoirs, mostly represented by CD4+ memory T cells. Several approaches are currently being undertaken to develop a cure for HIV-1 infection through elimination (or reduction) of these reservoirs. Few studies have so far been conducted to assess the possibility of reducing the size of HIV-1 reservoirs through vaccination in virologically controlled HIV-1-infected children. We recently conducted a vaccination study with a combined hepatitis A virus (HAV) and hepatitis B virus (HBV) vaccine in 22 HIV-1-infected children. We assessed the size of the virus reservoir, measured as total HIV-1 DNA copies in blood cells, pre- and postvaccination. In addition, we investigated by immunostaining whether the frequencies of CD4+ and CD8+ T cells and parameters of immune activation and proliferation on these cells were modulated by vaccination. At 1 month from the last vaccination dose, we found that 20 out of 22 children mounted a serological response to HBV; a majority of children had antibodies against HAV at baseline. The number of HIV-1 DNA copies in blood at 1 month postvaccination was reduced in comparison to baseline although this reduction was not statistically significant. A significant reduction of HIV-1 DNA copies in blood following vaccination was found in 12 children. The frequencies of CD4+ (naïve, effector memory) and CD8+ (central memory) T-cell subpopulations changed following vaccinations and a reduction in the activation and proliferation pattern of these cells was also noticed. Multivariate linear regression analysis revealed that the frequency of CD8+ effector memory T cells prior to vaccination was strongly predictive of the reduction of HIV-1 DNA copies in blood following vaccination of the 22 HIV-1-infected children. The results of this study suggest a beneficial effect of vaccination to reduce the size of virus reservoir in HIV-1-infected children receiving ART. A reduced frequency of activated CD4+ cells and an increase in central memory CD8+ T cells were associated with this finding. Further studies should assess whether vaccination is a possible tool to reduce HIV-1 reservoirs.
Collapse
Affiliation(s)
- Yonas Bekele
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rebecka Lantto Graham
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Soeria-Atmadja
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.,Department of Pediatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Aikaterini Nasi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Maurizio Zazzi
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Ilaria Vicenti
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Lars Naver
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.,Department of Pediatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Nilsson
- Department of Women's and Children Health, Karolinska Institutet, Stockholm, Sweden
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
39
|
Schleker T, Jacobsen EM, Mayer B, Strauss G, Debatin KM, Posovszky C. Preserved in vitro immunoreactivity in children receiving long-term immunosuppressive therapy due to inflammatory bowel disease or autoimmune hepatitis. Mol Cell Pediatr 2018; 5:1. [PMID: 29352427 PMCID: PMC5775189 DOI: 10.1186/s40348-018-0079-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 01/04/2018] [Indexed: 01/04/2023] Open
Abstract
Background Children with inflammatory bowel disease (IBD) or autoimmune hepatitis (AIH) are at risk for severe infections. This is partially a result of their chronic disease condition but, moreover, a side effect of their immunosuppressive therapy. Currently, vaccinations with live vaccines are regarded as contraindicated under immunosuppressive therapy, mainly because of concerns about side effects and a lack of data showing an adequate immune reaction. As there is no systematic study on the individual immunoreactivity under immunosuppressive therapy in this patient group, we analyzed the lymphocyte subgroups and immunoreactivity of lymphocytes in children with IBD or AIH with and without immunosuppressive therapy in vitro. Methods We collected whole blood samples from 17 children with IBD or AIH on high-level immunosuppression (IS) (group 1) and 8 on low-level IS (group 2) in comparison with 6 patients without systemic IS (group 3). After Ficoll separation of peripheral mononuclear cells, the samples were analyzed by flow cytometry to determine the lymphocyte subgroups. Furthermore, we stimulated the isolated lymphocytes with phytohemagglutinin (PHA), tetanus antigen, and adenovirus antigen and measured their proliferation by incorporation of H3-thymidine detected in a beta counter. The statistical evaluation was performed by Kruskal-Wallis test and Mann-Whitney U test using a bilateral level of significance of α = 5%. Results Patients with low- or high-level IS showed no significant difference in the number of lymphocytes or T cells. Interestingly, IS did not influence the lymphocyte proliferation assay significantly regarding median reaction to PHA, tetanus antigen, or adenovirus antigen between the three groups. However, comparing all immunosuppressed patients to the patients without IS, there was a significant difference towards stimulation with tetanus antigen. Conclusions Contrary to expectations of a strong influence of IS therapy on the immunoreactivity, this study showed only minor differences between the groups with high-level, low-level, and no IS. Particularly, the in vitro reactivity to adenovirus antigen was nearly the same in all three groups. We assume that—provided a normal distribution and count of lymphocyte subgroups—patients with moderate immunosuppression might be capable of raising an effective immune response to inactivated and live vaccines. Electronic supplementary material The online version of this article (10.1186/s40348-018-0079-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Teresa Schleker
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Eva-Maria Jacobsen
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Carsten Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany. .,Pediatric Gastroenterology and Nutrition, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Eythstr. 24, 89075, Ulm, Germany.
| |
Collapse
|
40
|
Rosengard HC, Wheat CM, Tilson MP, Cuda JD. Lichen planus following tetanus-diphtheria-acellular pertussis vaccination: A case report and review of the literature. SAGE Open Med Case Rep 2018; 6:2050313X17750335. [PMID: 29326823 PMCID: PMC5758953 DOI: 10.1177/2050313x17750335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 11/21/2017] [Indexed: 11/15/2022] Open
Abstract
Lichen planus is an inflammatory dermatosis with a prevalence of approximately 1%. Recent meta-analyses show that patients with hepatitis C virus have a 2.5- to 4.5-fold increased risk of developing lichen planus. Lichen planus has also followed vaccinations and has specifically been attributed to the hepatitis B vaccine, the influenza vaccine, and the tetanus–diphtheria–acellular pertussis vaccine. We describe a case of lichen planus in a hepatitis C virus–infected African American male occurring in temporal association with the administration of the tetanus–diphtheria–acellular pertussis vaccine. The patient’s presentation was clinically consistent with lichen planus and confirmed by biopsy. It is likely that many cases of vaccine-induced lichen planus have gone unpublished or unrecognized. In areas with high prevalence of hepatitis C virus infection, we may expect to see more cases of vaccine-induced lichen planus especially in light of the updated Centers for Disease Control and Prevention tetanus–diphtheria–acellular pertussis vaccination recommendations. This case serves to educate healthcare providers about vaccine-induced lichen planus and, in particular, the need to counsel hepatitis C virus–infected patients about a potential risk of developing lichen planus following vaccination. We also reflect on current theories suggesting the T-cell–mediated pathogenesis of lichen planus and the role that hepatitis C virus and toxoid or protein vaccines may play in initiating the disease.
Collapse
Affiliation(s)
- Heather C Rosengard
- Department of Dermatology, School of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Chikoti M Wheat
- Department of Dermatology, School of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Matthew P Tilson
- Department of Dermatology, School of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan D Cuda
- Department of Dermatology, School of Medicine, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
41
|
A brief review on the immunological scenario and recent developmental status of vaccines against enteric fever. Vaccine 2017; 35:6359-6366. [DOI: 10.1016/j.vaccine.2017.09.066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/11/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023]
|
42
|
Voysey M, Fanshawe TR, Kelly DF, O’Brien KL, Kandasamy R, Shrestha S, Thorson S, Hinds J, Pollard AJ. Serotype-Specific Correlates of Protection for Pneumococcal Carriage: An Analysis of Immunity in 19 Countries. Clin Infect Dis 2017; 66:913-920. [DOI: 10.1093/cid/cix895] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/19/2017] [Indexed: 01/27/2023] Open
Affiliation(s)
- Merryn Voysey
- Nuffield Department of Primary Care Health Sciences, University of Oxford, United Kingdom
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and National Institute for Health Research Oxford Biomedical Research Centre, United Kingdom
| | - Thomas R Fanshawe
- Nuffield Department of Primary Care Health Sciences, University of Oxford, United Kingdom
| | - Dominic F Kelly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and National Institute for Health Research Oxford Biomedical Research Centre, United Kingdom
| | - Katherine L O’Brien
- International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Rama Kandasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and National Institute for Health Research Oxford Biomedical Research Centre, United Kingdom
| | - Shrijana Shrestha
- Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Stephen Thorson
- Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Jason Hinds
- Institute for Infection and Immunity, St George’s University of London, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and National Institute for Health Research Oxford Biomedical Research Centre, United Kingdom
| |
Collapse
|
43
|
Obanewa O, Newell ML. Maternal nutritional status during pregnancy and infant immune response to routine childhood vaccinations. Future Virol 2017; 12:525-536. [PMID: 29225661 PMCID: PMC5716389 DOI: 10.2217/fvl-2017-0021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/08/2017] [Indexed: 01/08/2023]
Abstract
To systematically review the association between maternal nutritional status in pregnancy and infant immune response to childhood vaccines. We reviewed literature on maternal nutrition during pregnancy, fetal immune system and vaccines and possible relationships. Thereafter, we undertook a systematic review of the literature of maternal nutritional status and infant vaccine response, extracted relevant information, assessed quality of the nine papers identified and present findings in a narrative format. From limited evidence of average quality, intrauterine nutrition deficiency could lead to functional deficit in the infant's immune function; child vaccine response may thus be negatively affected by maternal malnutrition. Response to childhood vaccination may be associated with fetal and early life environment; evaluation of programs should take this into account.
Collapse
Affiliation(s)
- Olayinka Obanewa
- Human Development & Health, Global Health Research Institute, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Marie-Louise Newell
- Human Development & Health, Global Health Research Institute, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
44
|
Tsvetkov YE, Gening ML, Kurbatova EA, Akhmatova NK, Nifantiev NE. Oligosaccharide ligand tuning in design of third generation carbohydrate pneumococcal vaccines. PURE APPL CHEM 2017. [DOI: 10.1515/pac-2016-1123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractStreptococcus pneumoniae can cause many types of dangerous infectious diseases such as otitis media, pneumonia, meningitis and others that are more common in the very young and very old age. Available to date commercial vaccines based on capsular polysaccharides of S. pneumoniae of clinically important strains (first generation carbohydrate vaccines) and conjugated vaccines based on these polysaccharides (second generation carbohydrate vaccines) have certain limitations in protective efficiency. However, the efficiency of vaccines can be increased by the use of third generation vaccines based on synthetic oligosaccharide ligands representing in their structures the protective epitopes of capsular polysaccharides. The proper choice of an optimal oligosaccharide ligand is the most important step in the design of third generation carbohydrate vaccines. Herein we overview our works on the synthesis of three oligosaccharides corresponding to one, “one and a half” and two repeating units of S. pneumoniae type 14 capsular polysaccharide, immunogenic conjugates thereof and comparative immunological study of their conjugates with bovine serum albumin, which was used as a model protein carrier. The ability of obtained products to raise antibodies specific to capsular polysaccharide and homologous oligosaccharides, the induction of phagocytosis by immune antisera and active protection of immunized animals from S. pneumoniae type 14 infection were evaluated. On the basis of the results obtained tetrasaccharide comprising the repeating unit of S. pneumoniae type 14 capsular polysaccharide is an optimal carbohydrate ligand to be used as a part of the third generation carbohydrate pneumococcal vaccine.
Collapse
Affiliation(s)
- Yury E. Tsvetkov
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
| | - Marina L. Gening
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
| | - Ekaterina A. Kurbatova
- Laboratory of Therapeutic Vaccines, Mechnikov Research Institute for Vaccines and Sera, Malyi Kazennyi Pereulok 5a, 105064 Moscow, Russia
| | - Nelly K. Akhmatova
- Laboratory of Therapeutic Vaccines, Mechnikov Research Institute for Vaccines and Sera, Malyi Kazennyi Pereulok 5a, 105064 Moscow, Russia
| | - Nikolay E. Nifantiev
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia, e-mail:
| |
Collapse
|
45
|
Effects of prior influenza virus vaccination on maternal antibody responses: Implications for achieving protection in the newborns. Vaccine 2017; 35:5283-5290. [PMID: 28778612 DOI: 10.1016/j.vaccine.2017.05.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/28/2017] [Accepted: 05/16/2017] [Indexed: 01/29/2023]
Abstract
BACKGROUND In the US, influenza vaccination is recommended annually to everyone ≥6months. Prior receipt of influenza vaccine can dampen antibody responses to subsequent vaccination. This may have implications for pregnant women and their newborns, groups at high risk for complications from influenza infection. OBJECTIVE This study examined effects of prior vaccination on maternal and cord blood antibody levels in a cohort of pregnant women in the US. STUDY DESIGN Influenza antibody titers were measured in 141 pregnant women via the hemagglutination inhibition (HAI) assay prior to receipt of quadrivalent influenza vaccine, 30days post-vaccination, and at delivery (maternal and cord blood). Logistic regression analyses adjusting for age, BMI, parity, gestational age at vaccination, and year of vaccination compared HAI titers, seroprotection, and seroconversion in women with versus without vaccination in the prior year. RESULTS Compared to those without vaccination in the previous year (n=50), women with prior vaccination (n=91) exhibited higher baseline antibody titers and/or seroprotection rates against all four strains after controlling for covariates. Prior vaccination also predicted lower antibody responses and seroconversion rates at one month post-vaccination. However, at delivery, there were no significant differences in antibody titers or seroprotection rates in women or newborns, and no meaningful differences in the efficiency of antibody transfer, as indicated by the ratio of cord blood to maternal antibody titers at the time of delivery. CONCLUSION In this cohort of pregnant women, receipt of influenza vaccine the previous year predicted higher baseline antibody titers and decreased antibody responses at one month post-vaccination against all influenza strains. However, prior maternal vaccination did not significantly affect either maternal antibody levels at delivery or antibody levels transferred to the neonate. This study is registered with the NIH as a clinical trial (NCT02148874).
Collapse
|
46
|
van der Heiden M, Boots AMH, Bonacic Marinovic AA, de Rond LGH, van Maurik M, Tcherniaeva I, Berbers GAM, Buisman AM. Novel Intervention in the Aging Population: A Primary Meningococcal Vaccine Inducing Protective IgM Responses in Middle-Aged Adults. Front Immunol 2017; 8:817. [PMID: 28769927 PMCID: PMC5515833 DOI: 10.3389/fimmu.2017.00817] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/27/2017] [Indexed: 12/14/2022] Open
Abstract
Introduction Vaccine responses are often reduced in the elderly, leaving part of the elderly population vulnerable to infectious diseases. Timely vaccination may offer a solution for strengthening memory immunity before reaching old age, which classifies middle-aged persons as a target age group for vaccine interventions. However, knowledge regarding the immunogenicity of primary immunizations in middle-aged adults is lacking. We determined the immunogenicity of a primary meningococcal vaccine towards which no or (very) low pre-vaccination immunity exists in middle-aged adults (NTR4636). Methods A vaccine containing multiple meningococcal groups (tetravalent) conjugated to tetanus toxoid (MenACWY-TT) was administered to middle-aged adults (50–65 years of age, N = 204) in a phase IV single-center and open-label study. Blood samples were taken pre-, 7 days, 28 days, and 1 year post-vaccination. Functional antibody titers were measured with the serum bactericidal assay (SBA). Meningococcal- and tetanus-specific antibody responses were determined with a fluorescent bead-based multiplex immunoassay. A bi-exponential decay model was used to estimate long-term protection. Results In the majority of the participants, the meningococcal vaccine clearly induced naïve responses to meningococci W (MenW) and meningococci Y (MenY) as compared to a booster response to meningococci C (MenC). After 28 days, 94, 99, and 97% of the participants possessed a protective SBA titer for MenC, MenW, and MenY, respectively, which was maintained in 76, 94, and 86% 1 year post-vaccination. At this 1-year time point, significantly lower SBA titers were found in participants without a pre-vaccination SBA titer. Overall, protective antibody titers were predicted to persist after 10 years in 40–60% of the participants. The SBA titers correlated well with the meningococcal-specific IgM responses, especially for MenW and MenY. Interestingly, these IgM responses were negatively correlated with age. Conclusion Primary immunization with a tetravalent meningococcal vaccine was highly immunogenic in middle-aged adults, inducing protective antibody titers in the vast majority of the participants lasting for at least 1 year. The age-related decrease in highly functional IgM responses argues in favor of vaccination against de novo antigens before reaching old age and, hence, middle-aged persons are an age group of interest for future vaccine interventions to protect the aging population.
Collapse
Affiliation(s)
- Marieke van der Heiden
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands.,Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Annemieke M H Boots
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Axel A Bonacic Marinovic
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lia G H de Rond
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Marjan van Maurik
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Irina Tcherniaeva
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Guy A M Berbers
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Anne-Marie Buisman
- Centre for Infectious Disease Control (Cib), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
47
|
Kurbatova EA, Akhmatova NK, Akhmatova EA, Egorova NB, Yastrebova NE, Sukhova EV, Yashunsky DV, Tsvetkov YE, Gening ML, Nifantiev NE. Neoglycoconjugate of Tetrasaccharide Representing One Repeating Unit of the Streptococcus pneumoniae Type 14 Capsular Polysaccharide Induces the Production of Opsonizing IgG1 Antibodies and Possesses the Highest Protective Activity As Compared to Hexa- and Octasaccharide Conjugates. Front Immunol 2017; 8:659. [PMID: 28626461 PMCID: PMC5454037 DOI: 10.3389/fimmu.2017.00659] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 05/18/2017] [Indexed: 01/19/2023] Open
Abstract
Identifying protective synthetic oligosaccharide (OS) epitopes of Streptococcus pneumoniae capsular polysaccharides (CPs) is an indispensable step in the development of third-generation carbohydrate pneumococcal vaccines. Synthetic tetra-, hexa-, and octasaccharide structurally related to CP of S. pneumoniae type 14 were coupled to bovine serum albumin (BSA), adjuvanted with aluminum hydroxide, and tested for their immunogenicity in mice upon intraperitoneal prime-boost immunizations. Injections of the conjugates induced production of opsonizing anti-OS IgG1 antibodies (Abs). Immunization with the tetra- and octasaccharide conjugates stimulated the highest titers of the specific Abs. Further, the tetrasaccharide ligand demonstrated the highest ability to bind OS and CP Abs. Murine immune sera developed against tetra- and octasaccharide conjugates promoted pathogen opsonization to a higher degree than antisera against conjugated hexasaccharide. For the first time, the protective activities of these glycoconjugates were demonstrated in mouse model of generalized pneumococcal infections. The tetrasaccharide conjugate possessed the highest protective activities. Conversely, the octasaccharide conjugate had lower protective activities and the lowest one showed the hexasaccharide conjugate. Sera against all of the glycoconjugates passively protected naive mice from pneumococcal infections. Given that the BSA-tetrasaccharide induced the most abundant yield of specific Abs and the best protective activity, this OS may be regarded as the most promising candidate for the development of conjugated vaccines against S. pneumoniae type 14 infections.
Collapse
Affiliation(s)
- Ekaterina A. Kurbatova
- Laboratory of Therapeutic Vaccines, Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - Nelli K. Akhmatova
- Laboratory of Therapeutic Vaccines, Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - Elina A. Akhmatova
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda B. Egorova
- Laboratory of Therapeutic Vaccines, Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - Natalya E. Yastrebova
- Laboratory of Therapeutic Vaccines, Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - Elena V. Sukhova
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Dmitriy V. Yashunsky
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yury E. Tsvetkov
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Marina L. Gening
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay E. Nifantiev
- Laboratory of Glycoconjugate Chemistry, N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
48
|
Carithers DS. Examining the role of macrolides and host immunity in combatting filarial parasites. Parasit Vectors 2017; 10:182. [PMID: 28410595 PMCID: PMC5391593 DOI: 10.1186/s13071-017-2116-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 03/28/2017] [Indexed: 11/10/2022] Open
Abstract
Macrocyclic lactones (MLs), specifically the avermectins and milbemycins, are known for their effectiveness against a broad spectrum of disease-causing nematodes and arthropods in humans and animals. In most nematodes, drugs in this class induce paralysis, resulting in starvation, impaired ability to remain associated with their anatomical environment, and death of all life stages. Initially, this was also thought to be the ML mode of action against filarial nematodes, but researchers have not been able to validate these characteristic effects of immobilization/starvation of MLs in vitro, even at higher doses than are possible in vivo. Relatively recently, ML receptor sites exclusively located proximate to the excretory-secretory (ES) apparatus were identified in Brugia malayi microfilaria and an ML-induced suppression of secretory protein release by B. malayi microfilariae was demonstrated in vitro. It is hypothesized here that suppression of these ES proteins prevents the filarial worm from interfering with the host's complement cascade, reducing the ability of the parasite to evade the immune system. Live microfilariae and/or larvae, thus exposed, are attacked and presented to the host's innate immune mechanisms and are ultimately killed by the immune response, not the ML drug. These live, exposed filarial worms stimulate development of innate, cellular and humoral immune responses that when properly stimulated, are capable of clearing all larvae or microfilariae present in the host, regardless of their individual sensitivity to MLs. Additional research in this area can be expected to improve our understanding of the relationships among filarial worms, MLs, and the host immune system, which likely would have implications in filarial disease management in humans and animals.
Collapse
Affiliation(s)
- Doug S Carithers
- Boehringer Ingelheim, 3239 Satellite Boulevard, Duluth, GA, 30096, USA.
| |
Collapse
|
49
|
Curran D, de Ridder M, Van Effelterre T. The impact of assumptions regarding vaccine-induced immunity on the public health and cost-effectiveness of hepatitis A vaccination: Is one dose sufficient? Hum Vaccin Immunother 2016; 12:2765-2771. [PMID: 27428611 PMCID: PMC5137541 DOI: 10.1080/21645515.2016.1203495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/01/2016] [Accepted: 06/15/2016] [Indexed: 11/26/2022] Open
Abstract
Hepatitis A vaccination stimulates memory cells to produce an anamnestic response. In this study, we used a mathematical model to examine how long-term immune memory might convey additional protection against clinical/icteric infections. Dynamic and decision models were used to estimate the expected number of cases, and the costs and quality-adjusted life-years (QALYs), respectively. Several scenarios were explored by assuming: (1) varying duration of vaccine-induced immune memory, (2) and/or varying levels of vaccine-induced immune memory protection (IMP), (3) and/or varying levels of infectiousness in vaccinated individuals with IMP. The base case analysis assumed a time horizon of 25 y (2012 - 2036), with additional analyses over 50 and 75 y. The analyses were conducted in the Mexican public health system perspective. In the base case that assumed no vaccine-induced IMP, the 2-dose hepatitis A vaccination strategy was cost-effective compared with the 1-dose strategy over the 3 time horizons. However, it was not cost-effective if we assumed additional IMP durations of at least 10 y in the 25-y horizon. In the 50- and 75-y horizons, the 2-dose strategy was always cost-effective, except when 100% reduction in the probability of icteric Infections, 75% reduction in infectiousness, and mean durations of IMP of at least 50 y were assumed. This analysis indicates that routine vaccination of toddlers against hepatitis A virus would be cost-effective in Mexico using a single-dose vaccination strategy. However, the cost-effectiveness of a second dose depends on the assumptions of additional protection by IMP and the time horizon over which the analysis is performed.
Collapse
|
50
|
Ciabattini A, Pettini E, Fiorino F, Pastore G, Andersen P, Pozzi G, Medaglini D. Modulation of Primary Immune Response by Different Vaccine Adjuvants. Front Immunol 2016; 7:427. [PMID: 27781036 PMCID: PMC5066114 DOI: 10.3389/fimmu.2016.00427] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/28/2016] [Indexed: 01/09/2023] Open
Abstract
Adjuvants contribute to enhancing and shaping the vaccine immune response through different modes of action. Here early biomarkers of adjuvanticity after primary immunization were investigated using four different adjuvants combined with the chimeric tuberculosis vaccine antigen H56. C57BL/6 mice were immunized by the subcutaneous route with different vaccine formulations, and the modulation of primary CD4+ T cell and B cell responses was assessed within draining lymph nodes, blood, and spleen, 7 and 12 days after priming. Vaccine formulations containing the liposome system CAF01 or a squalene-based oil-in-water emulsion (o/w squalene), but not aluminum hydroxide (alum) or CpG ODN 1826, elicited a significant primary antigen-specific CD4+ T cell response compared to antigen alone, 7 days after immunization. The effector function of activated CD4+ T cells was skewed toward a Th1/Th17 response by CAF01, while a Th1/Th2 response was elicited by o/w squalene. Differentiation of B cells in short-lived plasma cells, and subsequent early H56-specific IgG secretion, was observed in mice immunized with o/w squalene or CpG adjuvants. Tested adjuvants promoted the germinal center reaction with different magnitude. These results show that the immunological activity of different adjuvants can be characterized by profiling early immunization biomarkers after primary immunization. These data and this approach could give an important contribution to the rational development of heterologous prime–boost vaccine immunization protocols.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut , Copenhagen , Denmark
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena , Siena , Italy
| |
Collapse
|