1
|
Weinberger B. Adjuvant strategies to improve vaccination of the elderly population. Curr Opin Pharmacol 2018; 41:34-41. [PMID: 29677646 DOI: 10.1016/j.coph.2018.03.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/30/2018] [Indexed: 10/17/2022]
Abstract
Immunosenescence contributes to increased incidence and severity of many infections in old age and is responsible for impaired immunogenicity and efficacy of vaccines. Adjuvants are one strategy to enhance immunogenicity of vaccines. The oil-in-water emulsions MF59TM and AS03, as well as a virosomal vaccine have been licensed in seasonal or pandemic influenza vaccines and are/were used successfully in the elderly. AS01, a liposome-based adjuvant comprising two immunostimulants has recently been approved in a recombinant protein vaccine for older adults, which showed very high efficacy against herpes zoster in clinical trials. Several adjuvants for use in the older population are in clinical and preclinical development and will hopefully improve vaccines for this age group in the future.
Collapse
Affiliation(s)
- Birgit Weinberger
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria.
| |
Collapse
|
2
|
Vono M, Eberhardt CS, Mohr E, Auderset F, Christensen D, Schmolke M, Coler R, Meinke A, Andersen P, Lambert PH, Mastelic-Gavillet B, Siegrist CA. Overcoming the Neonatal Limitations of Inducing Germinal Centers through Liposome-Based Adjuvants Including C-Type Lectin Agonists Trehalose Dibehenate or Curdlan. Front Immunol 2018. [PMID: 29541075 PMCID: PMC5835515 DOI: 10.3389/fimmu.2018.00381] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Neonates and infants are more vulnerable to infections and show reduced responses to vaccination. Consequently, repeated immunizations are required to induce protection and early life vaccines against major pathogens such as influenza are yet unavailable. Formulating antigens with potent adjuvants, including immunostimulators and delivery systems, is a demonstrated approach to enhance vaccine efficacy. Yet, adjuvants effective in adults may not meet the specific requirements for activating the early life immune system. Here, we assessed the neonatal adjuvanticity of three novel adjuvants including TLR4 (glucopyranosyl lipid adjuvant-squalene emulsion), TLR9 (IC31®), and Mincle (CAF01) agonists, which all induce germinal centers (GCs) and potent antibody responses to influenza hemagglutinin (HA) in adult mice. In neonates, a single dose of HA formulated into each adjuvant induced T follicular helper (TFH) cells. However, only HA/CAF01 elicited significantly higher and sustained antibody responses, engaging neonatal B cells to differentiate into GCs already after a single dose. Although antibody titers remained lower than in adults, HA-specific responses induced by a single neonatal dose of HA/CAF01 were sufficient to confer protection against influenza viral challenge. Postulating that the neonatal adjuvanticity of CAF01 may result from the functionality of the C-type lectin receptor (CLR) Mincle in early life we asked whether other C-type lectin agonists would show a similar neonatal adjuvanticity. Replacing the Mincle agonist trehalose 6,6′-dibehenate by Curdlan, which binds to Dectin-1, enhanced antibody responses through the induction of similar levels of TFH, GCs and bone marrow high-affinity plasma cells. Thus, specific requirements of early life B cells may already be met after a single vaccine dose using CLR-activating agonists, identified here as promising B cell immunostimulators for early life vaccines when included into cationic liposomes.
Collapse
Affiliation(s)
- Maria Vono
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Christiane Sigrid Eberhardt
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland.,WHO Collaborative Center for Vaccine Immunology, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Elodie Mohr
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Floriane Auderset
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Dennis Christensen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Mirco Schmolke
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Rhea Coler
- Infectious Disease Research Institute, Seattle, WA, United States
| | | | - Peter Andersen
- Vaccine Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Paul-Henri Lambert
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Beatris Mastelic-Gavillet
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- WHO Collaborative Center for Vaccine Immunology, Department of Pathology-Immunology, University of Geneva, Geneva, Switzerland.,WHO Collaborative Center for Vaccine Immunology, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
Abstract
In spite of current influenza vaccines being immunogenic, evolution of the influenza virus can reduce efficacy and so influenza remains a major threat to public health. One approach to improve influenza vaccines is to include adjuvants; substances that boost the immune response. Adjuvants are particularly beneficial for influenza vaccines administered during a pandemic when a rapid response is required or for use in patients with impaired immune responses, such as infants and the elderly. This review outlines the current use of adjuvants in human influenza vaccines, including what they are, why they are used and what is known of their mechanism of action. To date, six adjuvants have been used in licensed human vaccines: Alum, MF59, AS03, AF03, virosomes and heat labile enterotoxin (LT). In general these adjuvants are safe and well tolerated, but there have been some rare adverse events when adjuvanted vaccines are used at a population level that may discourage the inclusion of adjuvants in influenza vaccines, for example the association of LT with Bell's Palsy. Improved understanding about the mechanisms of the immune response to vaccination and infection has led to advances in adjuvant technology and we describe the experimental adjuvants that have been tested in clinical trials for influenza but have not yet progressed to licensure. Adjuvants alone are not sufficient to improve influenza vaccine efficacy because they do not address the underlying problem of mismatches between circulating virus and the vaccine. However, they may contribute to improved efficacy of next-generation influenza vaccines and will most likely play a role in the development of effective universal influenza vaccines, though what that role will be remains to be seen.
Collapse
Affiliation(s)
- John S Tregoning
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| | - Ryan F Russell
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| | - Ekaterina Kinnear
- a Mucosal Infection and Immunity group, Section of Virology, Department of Medicine , St Mary's Campus, Imperial College London , UK
| |
Collapse
|
4
|
He L, Su J, Ming M, Bernardo L, Chen T, Gisonni-Lex L, Gajewska B. Flow cytometry: An efficient method for antigenicity measurement and particle characterization on an adjuvanted vaccine candidate H4-IC31 for tuberculosis. J Immunol Methods 2017; 452:39-45. [PMID: 29056527 DOI: 10.1016/j.jim.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/16/2017] [Accepted: 10/18/2017] [Indexed: 11/17/2022]
Abstract
We have developed an accurate, precise and stability-indicating flow cytometry (FC) based assay to directly measure antigenicity of H4 protein (also known as HyVac4) in a vaccine formulation of H4-IC31, without desorbing the H4 protein from the IC31 adjuvant. This method involves immuno-staining of H4-IC31 complex with anti-H4 monoclonal antibodies (mAbs) followed by FC analysis. The assay is not only able to consistently measure H4 antigenicity levels in H4-IC31 stored under normal condition at 2-8°C, but also able to detect changes in H4 antigenicity after H4-IC31 undergoes heat stress or freeze-thawing. In addition, the FC method is able to characterize particle morphology while measuring antigenicity. The biological relevance of the changes in H4 antigenicity detected by the FC assay was supported by an in vitro cell based functional assay using human PBMCs to measure IFN-gamma (IFN-γ) secretion upon re-stimulation with H4-IC31. Our results show that the FC based antigenicity assay can efficiently monitor the biological and physicochemical properties of H4-IC31 and is an indicator for adjuvanted vaccine product stability.
Collapse
Affiliation(s)
- Liwei He
- Sanofi Pasteur, Analytical Research and Development, 1755 Steeles Avenue West, Toronto, Canada.
| | - Jin Su
- Sanofi Pasteur, Analytical Research and Development, 1755 Steeles Avenue West, Toronto, Canada
| | - Marin Ming
- Sanofi Pasteur, Analytical Research and Development, 1755 Steeles Avenue West, Toronto, Canada
| | - Lidice Bernardo
- Sanofi Pasteur, Analytical Research and Development, 1755 Steeles Avenue West, Toronto, Canada
| | - Tricia Chen
- Sanofi Pasteur, Analytical Research and Development, 1755 Steeles Avenue West, Toronto, Canada
| | - Lucy Gisonni-Lex
- Sanofi Pasteur, Analytical Research and Development, 1755 Steeles Avenue West, Toronto, Canada
| | - Beata Gajewska
- Sanofi Pasteur, Analytical Research and Development, 1755 Steeles Avenue West, Toronto, Canada
| |
Collapse
|
5
|
Update on Chlamydia trachomatis Vaccinology. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00543-16. [PMID: 28228394 DOI: 10.1128/cvi.00543-16] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Attempts to produce a vaccine to protect against Chlamydia trachomatis-induced trachoma were initiated more than 100 years ago and continued for several decades. Using whole organisms, protective responses were obtained. However, upon exposure to C. trachomatis, disease exacerbation developed in some immunized individuals, precluding the implementation of the vaccine. Evidence of the role of C. trachomatis as a sexually transmitted pathogen started to emerge in the 1960s, and it soon became evident that it can cause acute infections and long-term sequelae in women, men, and newborns. The main focus of this minireview is to summarize recent findings and discuss formulations, including antigens, adjuvants, routes, and delivery systems for immunization, primarily explored in the female mouse model, with the goal of implementing a vaccine against C. trachomatis genital infections.
Collapse
|
6
|
Falkeborn T, Hinkula J, Olliver M, Lindberg A, Maltais AK. The intranasal adjuvant Endocine™ enhances both systemic and mucosal immune responses in aged mice immunized with influenza antigen. Virol J 2017; 14:44. [PMID: 28253901 PMCID: PMC5335733 DOI: 10.1186/s12985-017-0698-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/02/2017] [Indexed: 01/17/2023] Open
Abstract
Despite availability of annual influenza vaccines, influenza causes significant morbidity and mortality in the elderly. This is at least in part a result of immunosenescence; the age-dependent decrease in immunological competence that results in greater susceptibility to infections and reduced responses to vaccination. To improve protective immune responses in this age group, new vaccines strategies, such as the use of adjuvants, are needed. Here, we evaluated the mucosal vaccine adjuvant Endocine™, formulated with split influenza antigen and administered intranasally in aged (20-month old) mice. Humoral immune responses were assessed and compared to unadjuvanted intranasal and subcutaneous vaccines. We show that formulation with Endocine™ significantly enhances hemagglutination inhibition (HI) titers, as well as serum IgG and mucosal IgA antibody titers, compared to both types of unadjuvanted vaccines. Thus, our results indicate that intranasal vaccination with Endocine™ is a possible approach for the development of mucosal influenza vaccines for the elderly.
Collapse
Affiliation(s)
- Tina Falkeborn
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jorma Hinkula
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Marie Olliver
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | - Alf Lindberg
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | | |
Collapse
|
7
|
Soleimani S, Shahsavandi S, Maddadgar O. Improvement influenza HA2 DNA vaccine cellular and humoral immune responses with Mx bio adjuvant. Biologicals 2016; 46:6-10. [PMID: 28027847 DOI: 10.1016/j.biologicals.2016.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 11/19/2022] Open
Abstract
Immunization with DNA vaccines as a novel alternative to conventional vaccination strategy requires adjuvant for improving vaccine efficacy. The conserved immunogenic HA2 subunit, which harbors neutralizing epitopes is a promising vaccine candidate against influenza viruses. In this study, for the first time we explore the idea of using host interferon inducible Mx protein to increase the immunogenicity of HA2 H9N2 influenza DNA vaccine. The potency and safety of the Mx adjuvanted-HA2 vaccine was evaluated in BALB/c mice by different prime-boost strategies. To assess the effect of the vaccination on the virus clearance rate, mice were challenged with homologous influenza virus. Administration of the adjuvanted vaccine and boosting with the same regimen could effectively enhance both humoral and cellular immune responses in treated mice. These data demonstrated that Mx as host defense peptide can be potentiated for improving influenza vaccine efficacy.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Chemotherapy, Adjuvant/methods
- Enzyme-Linked Immunosorbent Assay
- Female
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- Immunization, Secondary/methods
- Influenza A Virus, H9N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Mice, Inbred BALB C
- Myxovirus Resistance Proteins/administration & dosage
- Myxovirus Resistance Proteins/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Treatment Outcome
- Vaccination/methods
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Sina Soleimani
- Faculty of Veterinary Medicine, Tehran University, P.O. Box 14155-6453, Tehran, Iran; Razi Vaccine & Serum Research Institute, Agricultural Research Education and Extension Organization, P.O. Box 31975-148, Karaj, Iran
| | - Shahla Shahsavandi
- Razi Vaccine & Serum Research Institute, Agricultural Research Education and Extension Organization, P.O. Box 31975-148, Karaj, Iran.
| | - Omid Maddadgar
- Faculty of Veterinary Medicine, Tehran University, P.O. Box 14155-6453, Tehran, Iran
| |
Collapse
|
8
|
Chikh G, Luu R, Patel S, Davis HL, Weeratna RD. Effects of KLK Peptide on Adjuvanticity of Different ODN Sequences. Vaccines (Basel) 2016; 4:vaccines4020014. [PMID: 27153098 PMCID: PMC4931631 DOI: 10.3390/vaccines4020014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/20/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023] Open
Abstract
Endosomal Toll-like receptors (TLR) such as TLR3, 7, 8 and 9 recognize pathogen associated nucleic acids. While DNA sequence does influence degree of binding to and activation of TLR9, it also appears to influence the ability of the ligand to reach the intracellular endosomal compartment. The KLK (KLKL5KLK) antimicrobial peptide, which is immunostimulatory itself, can translocate into cells without cell membrane permeabilization and thus can be used for endosomal delivery of TLR agonists, as has been shown with the IC31 formulation that contains an oligodeoxynucleotide (ODN) TLR9 agonist. We evaluated the adjuvant activity of KLK combined with CpG or non-CpG (GpC) ODN synthesized with nuclease resistant phosphorothioate (S) or native phosphodiester (O) backbones with ovalbumin (OVA) antigen in mice. As single adjuvants, CpG(S) gave the strongest enhancement of OVA-specific immunity and the addition of KLK provided no benefit and was actually detrimental for some readouts. In contrast, KLK enhanced the adjuvant effects of CpG(O) and to a lesser extent of GpC (S), which on their own had little or no activity. Indeed while CD8 T cells, IFN-γ secretion and humoral response to vaccine antigen were enhanced when CpG(O) was combined with KLK, only IFN-γ secretion was enhanced when GpC (S) was combined to KLK. The synergistic adjuvant effects with KLK/ODN combinations were TLR9-mediated since they did not occur in TLR9 knock-out mice. We hypothesize that a nuclease resistant ODN with CpG motifs has its own mechanism for entering cells to reach the endosome. For ODN without CpG motifs, KLK appears to provide an alternate mechanism for accessing the endosome, where it can activate TLR9, albeit with lower potency than a CpG ODN. For nuclease sensitive (O) backbone ODN, KLK may also provide protection from nucleases in the tissues.
Collapse
Affiliation(s)
- Ghania Chikh
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, Ottawa, ON K2K 3A2, Canada.
| | - Rachel Luu
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, Ottawa, ON K2K 3A2, Canada.
| | - Shobhna Patel
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, Ottawa, ON K2K 3A2, Canada.
| | - Heather L Davis
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, Ottawa, ON K2K 3A2, Canada.
| | - Risini D Weeratna
- Pfizer Vaccine Immunotherapeutics, Ottawa Laboratories, Ottawa, ON K2K 3A2, Canada.
| |
Collapse
|
9
|
Soleimani S, Madadgar O, Shahsavandi S, Mahravani H, Lotfi M. In Silico Analysis of HA2/Mx Chimera Peptide for Developing an Adjuvanted Vaccine to Induce Immune Responses Against Influenza Viruses. Adv Pharm Bull 2016; 5:629-36. [PMID: 26793608 DOI: 10.15171/apb.2015.085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 05/11/2015] [Accepted: 05/14/2015] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The direct transmission of avian influenza viruses to human and increasing drug resisted strains posing new threats for public health. Therefore, development of efficient vaccines is needed to generate protective and persistent immunity to the viruses. METHODS Three motifs of Mx protein sequence in human, mouse and poultry located in interferon induced (GTP ase) domain were candidate as biologic adjuvant for enhancing the immune responses against influenza virus. Chimera proteins composed with the conserved HA2 subunit of influenza virus and the Mx motifs named HA2/Mx were modeled and evaluated by in silico analysis includes bioinformatics algorithms in order to explore biological characteristics of these peptides. RESULTS Amongst the predicted models, HA2/Mx1 peptide showed the better results following protein structures prediction, antigenic epitopes determination and model quality evaluation. Comparative homology modeling was performed with Swiss Model and the model was validated using ProSA. Epitope predictions revealed the construct could induce both B and T cell epitopes that expect a high immune response. CONCLUSION Taken together, these data indicate that the HA2/Mx1 chimera peptide can be potentiated for developing an adjuvant-fused influenza vaccine capable of stimulating effective immune response.
Collapse
Affiliation(s)
- Sina Soleimani
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran. ; Razi Vaccine & Serum Research Institute, Karaj, Iran
| | - Omid Madadgar
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | | | - Mohsen Lotfi
- Razi Vaccine & Serum Research Institute, Karaj, Iran
| |
Collapse
|
10
|
Knudsen NPH, Olsen A, Buonsanti C, Follmann F, Zhang Y, Coler RN, Fox CB, Meinke A, D'Oro U, Casini D, Bonci A, Billeskov R, De Gregorio E, Rappuoli R, Harandi AM, Andersen P, Agger EM. Different human vaccine adjuvants promote distinct antigen-independent immunological signatures tailored to different pathogens. Sci Rep 2016; 6:19570. [PMID: 26791076 PMCID: PMC4726129 DOI: 10.1038/srep19570] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/15/2015] [Indexed: 01/20/2023] Open
Abstract
The majority of vaccine candidates in clinical development are highly purified proteins and peptides relying on adjuvants to enhance and/or direct immune responses. Despite the acknowledged need for novel adjuvants, there are still very few adjuvants in licensed human vaccines. A vast number of adjuvants have been tested pre-clinically using different experimental conditions, rendering it impossible to directly compare their activity. We performed a head-to-head comparison of five different adjuvants Alum, MF59®, GLA-SE, IC31® and CAF01 in mice and combined these with antigens from M. tuberculosis, influenza, and chlamydia to test immune-profiles and efficacy in infection models using standardized protocols. Regardless of antigen, each adjuvant had a unique immunological signature suggesting that the adjuvants have potential for different disease targets. Alum increased antibody titers; MF59® induced strong antibody and IL-5 responses; GLA-SE induced antibodies and Th1; CAF01 showed a mixed Th1/Th17 profile and IC31® induced strong Th1 responses. MF59® and GLA-SE were strong inducers of influenza HI titers while CAF01, GLA-SE and IC31® enhanced protection to TB and chlamydia. Importantly, this is the first extensive attempt to categorize clinical-grade adjuvants based on their immune profiles and protective efficacy to inform a rational development of next generation vaccines for human use.
Collapse
Affiliation(s)
- Niels Peter H Knudsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Cecilia Buonsanti
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Yuan Zhang
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA, USA
| | | | | | - Ugo D'Oro
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Daniele Casini
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Alessandra Bonci
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Rolf Billeskov
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Ennio De Gregorio
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Rino Rappuoli
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Ali M Harandi
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
11
|
Guimarães LE, Baker B, Perricone C, Shoenfeld Y. Vaccines, adjuvants and autoimmunity. Pharmacol Res 2015; 100:190-209. [PMID: 26275795 PMCID: PMC7129276 DOI: 10.1016/j.phrs.2015.08.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 12/15/2022]
Abstract
Vaccines and autoimmunity are linked fields. Vaccine efficacy is based on whether host immune response against an antigen can elicit a memory T-cell response over time. Although the described side effects thus far have been mostly transient and acute, vaccines are able to elicit the immune system towards an autoimmune reaction. The diagnosis of a definite autoimmune disease and the occurrence of fatal outcome post-vaccination have been less frequently reported. Since vaccines are given to previously healthy hosts, who may have never developed the disease had they not been immunized, adverse events should be carefully accessed and evaluated even if they represent a limited number of occurrences. In this review of the literature, there is evidence of vaccine-induced autoimmunity and adjuvant-induced autoimmunity in both experimental models as well as human patients. Adjuvants and infectious agents may exert their immune-enhancing effects through various functional activities, encompassed by the adjuvant effect. These mechanisms are shared by different conditions triggered by adjuvants leading to the autoimmune/inflammatory syndrome induced by adjuvants (ASIA syndrome). In conclusion, there are several case reports of autoimmune diseases following vaccines, however, due to the limited number of cases, the different classifications of symptoms and the long latency period of the diseases, every attempt for an epidemiological study has so far failed to deliver a connection. Despite this, efforts to unveil the connection between the triggering of the immune system by adjuvants and the development of autoimmune conditions should be undertaken. Vaccinomics is a field that may bring to light novel customized, personalized treatment approaches in the future.
Collapse
Affiliation(s)
- Luísa Eça Guimarães
- The Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Britain Baker
- The Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Carlo Perricone
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Italy
| | - Yehuda Shoenfeld
- The Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer, Israel; Incumbent of the Laura Schwarz-kipp chair for research of autoimmune diseases, Sackler Faculty of Medicine, Tel-Aviv University, Israel.
| |
Collapse
|
12
|
Soema PC, Kompier R, Amorij JP, Kersten GFA. Current and next generation influenza vaccines: Formulation and production strategies. Eur J Pharm Biopharm 2015; 94:251-63. [PMID: 26047796 DOI: 10.1016/j.ejpb.2015.05.023] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
Abstract
Vaccination is the most effective method to prevent influenza infection. However, current influenza vaccines have several limitations. Relatively long production times, limited vaccine capacity, moderate efficacy in certain populations and lack of cross-reactivity are important issues that need to be addressed. We give an overview of the current status and novel developments in the landscape of influenza vaccines from an interdisciplinary point of view. The feasibility of novel vaccine concepts not only depends on immunological or clinical outcomes, but also depends on biotechnological aspects, such as formulation and production methods, which are frequently overlooked. Furthermore, the next generation of influenza vaccines is addressed, which hopefully will bring cross-reactive influenza vaccines. These developments indicate that an exciting future lies ahead in the influenza vaccine field.
Collapse
Affiliation(s)
- Peter C Soema
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands; Division of Drug Delivery and Technology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| | - Ronald Kompier
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands; FluConsult, Noordwijk, The Netherlands
| | - Jean-Pierre Amorij
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands.
| | - Gideon F A Kersten
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands; Division of Drug Delivery and Technology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| |
Collapse
|
13
|
Wang C, Li X, Wu T, Li D, Niu M, Wang Y, Zhang C, Cheng X, Chen P. Bursin-like peptide (BLP) enhances H9N2 influenza vaccine induced humoral and cell mediated immune responses. Cell Immunol 2014; 292:57-64. [DOI: 10.1016/j.cellimm.2014.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/16/2014] [Accepted: 09/20/2014] [Indexed: 11/16/2022]
|
14
|
Activation of the RIG-I pathway during influenza vaccination enhances the germinal center reaction, promotes T follicular helper cell induction, and provides a dose-sparing effect and protective immunity. J Virol 2014; 88:13990-4001. [PMID: 25253340 DOI: 10.1128/jvi.02273-14] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Pattern recognition receptors (PRR) sense certain molecular patterns uniquely expressed by pathogens. Retinoic-acid-inducible gene I (RIG-I) is a cytosolic PRR that senses viral nucleic acids and induces innate immune activation and secretion of type I interferons (IFNs). Here, using influenza vaccine antigens, we investigated the consequences of activating the RIG-I pathway for antigen-specific adaptive immune responses. We found that mice immunized with influenza vaccine antigens coadministered with 5'ppp-double-stranded RNA (dsRNA), a RIG-I ligand, developed robust levels of hemagglutination-inhibiting antibodies, enhanced germinal center reaction, and T follicular helper cell responses. In addition, RIG-I activation enhanced antibody affinity maturation and plasma cell responses in the draining lymph nodes, spleen, and bone marrow and conferred protective immunity against virus challenge. Importantly, activation of the RIG-I pathway was able to reduce the antigen requirement by 10- to 100-fold in inducing optimal influenza-specific cellular and humoral responses, including protective immunity. The effects induced by 5'ppp-dsRNA were significantly dependent on type I IFN and IPS-1 (an adapter protein downstream of the RIG-I pathway) signaling but were independent of the MyD88- and TLR3-mediated pathways. Our results show that activation of the RIG-I-like receptor pathway programs the innate immunity to achieve qualitatively and quantitatively enhanced protective cellular adaptive immune responses even at low antigen doses, and this indicates the potential utility of RIG-I ligands as molecular adjuvants for viral vaccines. IMPORTANCE The recently discovered RNA helicase family of RIG-I-like receptors (RLRs) is a critical component of host defense mechanisms responsible for detecting viruses and triggering innate antiviral cytokines that help control viral replication and dissemination. In this study, we show that the RLR pathway can be effectively exploited to enhance adaptive immunity and protective immune memory against viral infection. Our results show that activation of the RIG-I pathway along with influenza vaccination programs the innate immunity to induce qualitatively and quantitatively superior protective adaptive immunity against pandemic influenza viruses. More importantly, RIG-I activation at the time of vaccination allows induction of robust adaptive responses even at low vaccine antigen doses. These results highlight the potential utility of exploiting the RIG-I pathway to enhance viral-vaccine-specific immunity and have broader implications for designing better vaccines in general.
Collapse
|
15
|
Levast B, Awate S, Babiuk L, Mutwiri G, Gerdts V, van Drunen Littel-van den Hurk S. Vaccine Potentiation by Combination Adjuvants. Vaccines (Basel) 2014; 2:297-322. [PMID: 26344621 PMCID: PMC4494260 DOI: 10.3390/vaccines2020297] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/22/2014] [Accepted: 03/28/2014] [Indexed: 01/02/2023] Open
Abstract
Adjuvants are crucial components of vaccines. They significantly improve vaccine efficacy by modulating, enhancing, or extending the immune response and at the same time reducing the amount of antigen needed. In contrast to previously licensed adjuvants, current successful adjuvant formulations often consist of several molecules, that when combined, act synergistically by activating a variety of immune mechanisms. These "combination adjuvants" are already registered with several vaccines, both in humans and animals, and novel combination adjuvants are in the pipeline. With improved knowledge of the type of immune responses needed to successfully induce disease protection by vaccination, combination adjuvants are particularly suited to not only enhance, but also direct the immune responses desired to be either Th1-, Th2- or Th17-biased. Indeed, in view of the variety of disease and population targets for vaccine development, a panel of adjuvants will be needed to address different disease targets and populations. Here, we will review well-known and new combination adjuvants already licensed or currently in development-including ISCOMs, liposomes, Adjuvant Systems Montanides, and triple adjuvant combinations-and summarize their performance in preclinical and clinical trials. Several of these combination adjuvants are promising having promoted improved and balanced immune responses.
Collapse
Affiliation(s)
- Benoît Levast
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
| | - Sunita Awate
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
| | - Lorne Babiuk
- University Hall, University of Alberta, Edmonton, AB T6G 2J9, Canada.
| | - George Mutwiri
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
- School of Public Health, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| | - Volker Gerdts
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
- Veterinary Microbiology, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada.
| | - Sylvia van Drunen Littel-van den Hurk
- VIDO-Intervac, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK S7N 5E3, Canada.
- Microbiology and Immunology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
16
|
Even-Or O, Samira S, Ellis R, Kedar E, Barenholz Y. Adjuvanted influenza vaccines. Expert Rev Vaccines 2014; 12:1095-108. [PMID: 24053401 DOI: 10.1586/14760584.2013.825445] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Influenza is one of the most common causes of human morbidity and mortality that is preventable by vaccination. Immunization with available vaccines provides incomplete protection against illness caused by influenza virus, especially in high-risk groups such as the elderly and young children. Thus, more efficacious vaccines are needed for the entire population, and all the more so for high-risk groups. One way to improve immune responses and protection is to formulate the vaccine with antigen carriers and/or adjuvants, which can play an important role in improving immune responses and delivery to antigen-presenting cells, especially for a vaccine like influenza that is based on protein antigens usually administered without a carrier or adjuvant. In this review, the authors present an overview of available vaccines, focusing on research and development of new adjuvants used in influenza vaccines, as well as adjuvanted influenza vaccines aimed to improve immune responses, protection and breadth of coverage for influenza.
Collapse
Affiliation(s)
- Orli Even-Or
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, P.O. Box 12272, Jerusalem 91120, Israel
| | | | | | | | | |
Collapse
|
17
|
Mutwiri G, Gerdts V, van Drunen Littel-van den Hurk S, Auray G, Eng N, Garlapati S, Babiuk LA, Potter A. Combination adjuvants: the next generation of adjuvants? Expert Rev Vaccines 2014; 10:95-107. [DOI: 10.1586/erv.10.154] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
18
|
Del Giudice G, Rappuoli R. Inactivated and adjuvanted influenza vaccines. Curr Top Microbiol Immunol 2014; 386:151-80. [PMID: 25038938 DOI: 10.1007/82_2014_406] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inactivated influenza vaccines are produced every year to fight against the seasonal epidemics of influenza. Despite the nonoptimal coverage, even in subjects at risk like the elderly, pregnant women, etc., these vaccines significantly reduce the burden of mortality and morbidity linked to the influenza infection. Importantly, these vaccines have also contributed to reduce the impact of the last pandemics. Nevertheless, the performance of these vaccines can be improved mainly in those age groups, like children and the elderly, in which their efficacy is suboptimal. The use of adjuvants has proven effective to this scope. Oil-in-water adjuvants like MF59 and AS03 have been licensed and widely used, and shown efficacious in preventing influenza infection in the last pandemic. MF59-adjuvanted inactivated vaccine was more efficacious than non-adjuvanted vaccine in preventing influenza infection in young children and in reducing hospitalization due to the influenza infection in the elderly. Other adjuvants are now at different stages of development and some are being tested in clinical trials. The perspective remains to improve the way inactivated vaccines are prepared and to accelerate their availability, mainly in the case of influenza pandemics, and to enhance their efficacy/effectiveness for a more successful impact at the public health level.
Collapse
Affiliation(s)
- Giuseppe Del Giudice
- Research and Development, Novartis Vaccines, Via Fiorentina 1, 53100, Siena, Italy,
| | | |
Collapse
|
19
|
Nasal and skin delivery of IC31(®)-adjuvanted recombinant HSV-2 gD protein confers protection against genital herpes. Vaccine 2012; 30:4361-8. [PMID: 22682292 DOI: 10.1016/j.vaccine.2012.02.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 12/28/2011] [Accepted: 02/06/2012] [Indexed: 11/21/2022]
Abstract
Genital herpes caused by herpes simplex virus type 2 (HSV-2) remains the leading cause of genital ulcers worldwide. Given the disappointing results of the recent genital herpes vaccine trials in humans, development of novel vaccine strategies capable of eliciting protective mucosal and systemic immune responses to HSV-2 is urgently required. Here we tested the ability of the adjuvant IC31(®) in combination with HSV-2 glycoprotein D (gD) used through intranasal (i.n.), intradermal (i.d.), or subcutaneous (s.c.) immunization routes for induction of protective immunity against genital herpes infection in C57BL/6 mice. Immunization with gD plus IC31(®) through all three routes of immunization developed elevated gD-specific serum antibody responses with HSV-2 neutralizing activity. Whereas the skin routes promoted the induction of a mixed IgG2c/IgG1 isotype profile, the i.n. route only elicited IgG1 antibodies. All immunization routes were able to induce gD-specific IgG antibody responses in the vaginas of mice immunized with IC31(®)-adjuvanted gD. Although specific lymphoproliferative responses were observed in splenocytes from mice of most groups vaccinated with IC31(®)-adjuvanted gD, only i.d. immunization resulted in a significant splenic IFN-γ response. Further, immunization with gD plus IC31(®) conferred 80-100% protection against an otherwise lethal vaginal HSV-2 challenge with amelioration of viral replication and disease severity in the vagina. These results warrant further exploration of IC31(®) for induction of protective immunity against genital herpes and other sexually transmitted infections.
Collapse
|
20
|
Pattacini L, Mize GJ, Graham JB, Fluharty TR, Graham TM, Lingnau K, Wizel B, Perdiguero B, Esteban M, Pantaleo G, Shen M, Spies GA, McElrath MJ, Lund JM. A novel HIV vaccine adjuvanted by IC31 induces robust and persistent humoral and cellular immunity. PLoS One 2012; 7:e42163. [PMID: 22848738 PMCID: PMC3405041 DOI: 10.1371/journal.pone.0042163] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 07/02/2012] [Indexed: 11/18/2022] Open
Abstract
The HIV vaccine strategy that, to date, generated immune protection consisted of a prime-boost regimen using a canarypox vector and an HIV envelope protein with alum, as shown in the RV144 trial. Since the efficacy was weak, and previous HIV vaccine trials designed to generate antibody responses failed, we hypothesized that generation of T cell responses would result in improved protection. Thus, we tested the immunogenicity of a similar envelope-based vaccine using a mouse model, with two modifications: a clade C CN54gp140 HIV envelope protein was adjuvanted by the TLR9 agonist IC31®, and the viral vector was the vaccinia strain NYVAC-CN54 expressing HIV envelope gp120. The use of IC31® facilitated immunoglobulin isotype switching, leading to the production of Env-specific IgG2a, as compared to protein with alum alone. Boosting with NYVAC-CN54 resulted in the generation of more robust Th1 T cell responses. Moreover, gp140 prime with IC31® and alum followed by NYVAC-CN54 boost resulted in the formation and persistence of central and effector memory populations in the spleen and an effector memory population in the gut. Our data suggest that this regimen is promising and could improve the protection rate by eliciting strong and long-lasting humoral and cellular immune responses.
Collapse
Affiliation(s)
- Laura Pattacini
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Gregory J. Mize
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jessica B. Graham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Tayler R. Fluharty
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Tisha M. Graham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | | | | | - Beatriz Perdiguero
- Centro Nacional de Biotecnologia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Mariano Esteban
- Centro Nacional de Biotecnologia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Department of Medicine and Swiss Vaccine Research Institute, Lausanne University Hospital, Lausanne, Switzerland
| | - Mingchao Shen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Gregory A. Spies
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Jennifer M. Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
21
|
NS1-truncated live attenuated virus vaccine provides robust protection to aged mice from viral challenge. J Virol 2012; 86:10293-301. [PMID: 22787224 DOI: 10.1128/jvi.01131-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunological changes associated with age contribute to the high rates of influenza virus morbidity and mortality in the elderly. Compounding this problem, aged individuals do not respond to vaccination as well as younger, healthy adults. Efforts to increase protection to this demographic group are of utmost importance, as the proportion of the population above the age of 65 is projected to increase in the coming decade. Using a live influenza virus with a truncated nonstructural protein 1 (NS1), we are able to stimulate cellular and humoral immune responses of aged mice comparable to levels seen in young mice. Impressively, a single vaccination provided protection following stringent lethal challenge in aged mice.
Collapse
|
22
|
Biswas S, Chang H, Sarkis PTN, Fikrig E, Zhu Q, Marasco WA. Humoral immune responses in humanized BLT mice immunized with West Nile virus and HIV-1 envelope proteins are largely mediated via human CD5+ B cells. Immunology 2012; 134:419-33. [PMID: 22044090 DOI: 10.1111/j.1365-2567.2011.03501.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BLT mice, constructed by surgical implantation of human fetal thymus-liver tissues and intravenous delivery of autologous CD34+ haematopoietic stem cells into adult non-obese diabetic/severe combined immunodeficiency mice, were evaluated for vaccine-induced humoral immune responses. Following engraftment, these mice developed a human lymphoid system; however, the majority of the peripheral human B lymphocytes displayed an immature phenotype as evidenced by surface CD10 expression. Over 50% of the human B cells in the periphery but not in the bone marrow also expressed the CD5 antigen, which is found only infrequently on mature follicular B cells in humans. A single intramuscular immunization with recombinant viral envelope antigens, e.g., HIVgp140 and West Nile Virus envelope proteins, together with the immune stimulatory KLK/ODN1a composition) [corrected] adjuvant resulted in seroconversion characterized by antigen-specific human antibodies predominantly of the IgM isotype. However, repeated booster immunizations did not induce secondary immune responses as evidenced by the lack of class switching and specific IgM levels remaining relatively unchanged. Interestingly, the peripheral CD19+ CD5+ but not the CD19+ CD5- human B lymphocytes displayed a late developing CD27+ IgM+ memory phenotype, suggesting that the CD5+ B-cell subset, previously implicated in 'natural antibody' production, may play a role in the vaccine-induced antibody response. Furthermore, human T lymphocytes from these mice demonstrated suboptimal proliferative responses and loss of co-stimulatory surface proteins ex vivo that could be partially reversed with human interleukin-2 and interleukin-7. Therefore, vaccine-induced immune responses in BLT mice resemble a T-cell-independent pathway that can potentially be modulated in vivo by the exogenous delivery of human cytokines/growth factors.
Collapse
Affiliation(s)
- Subhabrata Biswas
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | |
Collapse
|
23
|
Novel protein-based pneumococcal vaccines administered with the Th1-promoting adjuvant IC31 induce protective immunity against pneumococcal disease in neonatal mice. Infect Immun 2011; 80:461-8. [PMID: 22025519 DOI: 10.1128/iai.05801-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Streptococcus pneumoniae is responsible for many vaccine-preventable deaths, annually causing around 1 million deaths in children younger than 5 years of age. A new generation of pneumococcal vaccines based on conserved proteins is being developed. We evaluated the immunogenicities and protective efficacies of four pneumococcal protein vaccine candidates, PcsB, StkP, PsaA, and PspA, in a neonatal mouse model. Mice were immunized three times and challenged intranasally with virulent pneumococci. All four proteins were immunogenic in neonatal mice, and antibody (Ab) responses were significantly enhanced by the novel adjuvant IC31, which consists of an antibacterial peptide (KLKL5KLK) and a synthetic oligodeoxynucleotide, ODN1a, that signals through Toll-like receptor 9 (TLR9). Two single proteins, StkP and PspA, combined with IC31 significantly reduced pneumococcal bacteremia but had no effects on lung infection. Three proteins, PcsB, StkP, and PsaA, were evaluated with alum or IC31. IC31 enhanced Ab responses and avidity to all three proteins, whereas alum enhanced Ab responses and avidity to StkP and PsaA only. Mice receiving the trivalent protein formulation with IC31 had significantly reduced bacteremia and lung infection compared to unvaccinated mice, but the level of protection was dependent on the dose of IC31. When PspA was added to the trivalent protein formulation, the dose of IC31 needed to obtain protective immunity could be reduced. These results demonstrate that a novel pneumococcal protein-based vaccine is immunogenic at an early age of mice and emphasize the benefits of using a combination of conserved proteins and an effective adjuvant to elicit potent protective immunity against invasive pneumococcal disease.
Collapse
|
24
|
Rosenkrands I, Vingsbo-Lundberg C, Bundgaard TJ, Lindenstrøm T, Enouf V, van der Werf S, Andersen P, Agger EM. Enhanced humoral and cell-mediated immune responses after immunization with trivalent influenza vaccine adjuvanted with cationic liposomes. Vaccine 2011; 29:6283-91. [PMID: 21722683 DOI: 10.1016/j.vaccine.2011.06.040] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 06/09/2011] [Accepted: 06/10/2011] [Indexed: 11/18/2022]
Abstract
The recent pandemic caused by new influenza A (H1N1) has emphasized the need for improved influenza vaccines with enhanced immune responses that ideally include longlived humoral and CMI responses and mediate a broad protection. This study demonstrates that administration of trivalent influenza vaccine (TIV) with the cationic liposome adjuvant system CAF01 enhances the humoral immune response as measured by hemagglutinin inhibition titers and influenza-specific serum antibody titers, and promote a strong Th1 response with augmented levels of IL-1β, IL-2, IL-12, IFN-γ and TNF-α. Furthermore, high levels of IL-17 are detected in agreement with CAF01's ability to promote TH17 responses. Importantly, the Th1/Th17 cytokine profile is still maintained 20 weeks after the last vaccination. The CAF01 adjuvanted influenza vaccine reduces weight loss and temperature decrease and results in complete survival of mice challenged with the drifted H1N1 influenza strain A/PR/8/34. Overall, the results suggest that CAF01 is a potent adjuvant system for future, improved influenza vaccines.
Collapse
Affiliation(s)
- Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, 5 Orestads Boulevard, DK-2300 Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Bernardo L, Pavón A, Hermida L, Gil L, Valdés I, Cabezas S, Linares R, Alvarez M, Silva R, Guillén G, Nagy E, Schlick P, Guzmán MG. The two component adjuvant IC31® potentiates the protective immunity induced by a dengue 2 recombinant fusion protein in mice. Vaccine 2011; 29:4256-63. [PMID: 21447316 DOI: 10.1016/j.vaccine.2011.03.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 02/27/2011] [Accepted: 03/12/2011] [Indexed: 02/03/2023]
Abstract
Here we evaluated the suitability of the synthetic adjuvant IC31® to potentiate the protective capacity of PD5 protein (domain III of the envelope protein of dengue 2 virus fused to the carrier protein P64k). Unlike Alum, PD5 mixed with IC31® induced complete protection against virus challenge in mice and increased IFN-γ secretion after in vitro re-stimulation. The induced antibody response was highly specific to the homologous serotype and showed both IgG1 and IgG2a subtypes. IC31® is a promising adjuvant for PD5 recombinant protein based vaccination against dengue. Future work should address the suitability of PD5/IC31® formulations in non-human primate models.
Collapse
Affiliation(s)
- Lidice Bernardo
- Department of Virology, PAHO/WHO Collaborating Center for the Study of Dengue and its Vector, Pedro Kourí Tropical Medicine Institute, Autopista Novia del Mediodía, km 6 ½ P.O. Box 601 Marianao 13, Havana, Cuba
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Henriksen-Lacey M, Korsholm KS, Andersen P, Perrie Y, Christensen D. Liposomal vaccine delivery systems. Expert Opin Drug Deliv 2011; 8:505-19. [DOI: 10.1517/17425247.2011.558081] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
|
28
|
Influenza Vaccines: What Do We Want and How Can We Get It? CROSSROADS BETWEEN INNATE AND ADAPTIVE IMMUNITY III 2011; 780:161-74. [DOI: 10.1007/978-1-4419-5632-3_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
29
|
Aichinger MC, Ginzler M, Weghuber J, Zimmermann L, Riedl K, Schütz G, Nagy E, von Gabain A, Schweyen R, Henics T. Adjuvating the adjuvant: facilitated delivery of an immunomodulatory oligonucleotide to TLR9 by a cationic antimicrobial peptide in dendritic cells. Vaccine 2010; 29:426-36. [PMID: 21093498 DOI: 10.1016/j.vaccine.2010.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 09/16/2010] [Accepted: 11/02/2010] [Indexed: 01/19/2023]
Abstract
IC31(®) is a novel bi-component vaccine adjuvant consisting of the peptide KLKL(5)KLK (KLK) and the TLR9 agonist oligonucleotide d(IC)(13) (ODN1a). While membrane-interacting properties of KLK and immuno-modulating capabilities of ODN1a have been characterized in detail, little is known of how these two molecules function together and synergize in interacting with their primary target cells, dendritic cells (DCs). We have found that KLK-triggered aggregates entrapped ODN1a and these complexes readily associated with the DC cell surface. KLK stimulated the uptake and internalization of ODN1a via endocytosis, while the bulk of the peptide remained associated with the cell periphery. ODN1a co-localized with early and late endosomes as well as endoplasmic reticular structures. ODN1a co-localized with TLR9 positive compartments following KLK mediated uptake. These features did not depend on the expression of TLR-9. Our results reveal novel mechanisms that allow KLK to enhance the effects of the TLR-9 ligand ODN1a in immunomodulation.
Collapse
Affiliation(s)
- Michael C Aichinger
- Department of Genetics, Max F. Perutz Laboratories, Dr. Bohrgasse 9, 1030 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Morgan EL, Thoman ML, Sanderson SD, Phillips JA. A novel adjuvant for vaccine development in the aged. Vaccine 2010; 28:8275-9. [PMID: 20965299 DOI: 10.1016/j.vaccine.2010.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 09/28/2010] [Accepted: 10/05/2010] [Indexed: 12/31/2022]
Abstract
A conformationally-biased, response-selective agonist of human C5a(65-74) (EP67) activated antigen presenting cells (APC) from aged C57Bl/6 mice in vitro and the generation of antigen (Ag)-specific antibody (Ab) responses in aged mice in vivo. EP67, induced the release of the pro-inflammatory cytokines IL-6, TNFα, and INFγ from splenic APCs obtained from both aged and young mice. Both aged and young mice produced high Ag-specific IgG Ab titers when immunized with EP67-containing vaccines to ovalbumin (OVA-EP67) and to a protein (rPrp1) from the cell wall of Coccidioides (rPrp1-EP67). Immunization with EP67-containing vaccines resulted in higher IgG titers in both young and aged mice compared to mice immunized with OVA adsorbed to alum (OVA/alum) and Prp1 admixed with CpG (rPrp1 +CpG). Aged and young mice immunized with the EP67-containing vaccines generated higher titers of IgG1 and IgG2b relative to their aged-matched counterparts immunized with OVA/alum or Prp1 +CpG. These results indicate that EP67 induces humoral immunity in aged mice not obtainable with alum and CpG. These results support the use of EP67 as a potential vaccine adjuvant suited to the elderly.
Collapse
Affiliation(s)
- Edward L Morgan
- San Diego State University, Biosciences Center, 5500 Campanile Drive, San Diego, CA 92182, USA.
| | | | | | | |
Collapse
|
31
|
|
32
|
An adjuvant for the induction of potent, protective humoral responses to an H5N1 influenza virus vaccine with antigen-sparing effect in mice. J Virol 2010; 84:8639-49. [PMID: 20538850 DOI: 10.1128/jvi.00596-10] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intramuscular administration of inactivated influenza virus vaccine is the main vaccine platform used for the prevention of seasonal influenza virus infection. In clinical trials, inactivated H5N1 vaccines have been shown to be safe and capable of eliciting immune correlates of protection. However, the H5N1 vaccines are poorly immunogenic compared to seasonal influenza virus vaccines. Needle-free vaccination would be more efficient and economical in a pandemic, and the development of an effective and safe mucosal adjuvant will be an important milestone. A stabilized chemical analog of double-stranded RNA, PIKA, was previously reported to be a potent mucosal adjuvant in a murine model. While PIKA stimulates dendritic cells in vitro, little was known about its receptor and adjuvanting mechanism in vivo. In this study, we demonstrated that the immunostimulatory effect of PIKA resulted in an increased number of mature antigen-presenting cells, with the induction of proinflammatory cytokines at the inoculation site. In addition, coadministration of PIKA with a poorly immunogenic H5N1 subunit vaccine led to antigen sparing and quantitative and qualitative improvements of the immune responses over those achieved with an unadjuvanted vaccine in mice. The adjuvanted vaccine provided protection against lethal challenge with homologous and heterologous H5N1 wild-type viruses. Mice lacking functional TLR3 showed diminished cytokine production with PIKA stimulation, diminished antibody responses, and reduced protective efficacy against wild-type virus challenge following vaccination. These data suggest that TLR3 is important for the optimal performance of PIKA as an adjuvant. With its good safety profile and antigen-sparing effect, PIKA could be an attractive adjuvant for use in future pandemics.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW To summarize recent progress in the development of adjuvants with a special focus on adjuvants that enhance B-cell responses to protein-based vaccines. Both established and new experimental approaches are described and also briefly we discuss how adjuvants and virus-based vaccines interact with the immune system. RECENT FINDINGS Two new adjuvants were recently approved for human applications and many others are in preclinical or clinical testing. Significant advances were made to describe the mechanism of action of adjuvants. For example, aluminum hydroxide salts were shown to engage Nalp3, a member of the cytosolic NOD-like receptors and activation of B cells via invariant natural killer cell presentation of alpha-galactosylceramide was described. The effects of Toll-like receptor ligands on B-cell differentiation were further characterized and a peptide derived from IPS-1, a cytosolic signaling molecule, was shown to provide adjuvant effect. Stimulation of protective antibodies against HIV-1 may require extensive antibody affinity maturation, thus long-term exposure or repeated administration of antigen may be needed to induce effective B-cell responses. SUMMARY Advances in our understanding of how specific signaling pathways link innate and adaptive immunity provides a basis for the design of improved adjuvants to promote broad and potent B-cell responses.
Collapse
|
34
|
Hagenaars N, Verheul RJ, Mooren I, de Jong PH, Mastrobattista E, Glansbeek HL, Heldens JG, van den Bosch H, Hennink WE, Jiskoot W. Relationship between structure and adjuvanticity of N,N,N-trimethyl chitosan (TMC) structural variants in a nasal influenza vaccine. J Control Release 2009; 140:126-33. [DOI: 10.1016/j.jconrel.2009.08.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 08/13/2009] [Accepted: 08/17/2009] [Indexed: 11/29/2022]
|
35
|
Abstract
Some adjuvants may exert adverse effects upon injection or, on the other hand, may not trigger a full immunological reaction. The mechanisms underlying adjuvant adverse effects are under renewed scrutiny because of the enormous implications for vaccine development. In the search for new and safer adjuvants, several new adjuvants were developed by pharmaceutical companies utilizing new immunological and chemical innovations. The ability of the immune system to recognize molecules that are broadly shared by pathogens is, in part, due to the presence of special immune receptors called toll-like receptors (TLRs) that are expressed on leukocyte membranes. The very fact that TLR activation leads to adaptive immune responses to foreign entities explains why so many adjuvants used today in vaccinations are developed to mimic TLR ligands. Alongside their supportive role, adjuvants were found to inflict by themselves an illness of autoimmune nature, defined as ‘the adjuvant diseases’. The debatable question of silicone as an adjuvant and connective tissue diseases, as well as the Gulf War syndrome and macrophagic myofaciitis which followed multiple injections of aluminium-based vaccines, are presented here. Owing to the adverse effects exerted by adjuvants, there is no doubt that safer adjuvants need to be developed and incorporated into future vaccines. Other needs in light of new vaccine technologies are adjuvants suitable for use with mucosally delivered vaccines, DNA vaccines, cancer and autoimmunity vaccines. In particular, there is demand for safe and non-toxic adjuvants able to stimulate cellular (Th1) immunity. More adjuvants were approved to date besides alum for human vaccines, including MF59 in some viral vaccines, MPL, AS04, AS01B and AS02A against viral and parasitic infections, virosomes for HBV, HPV and HAV, and cholera toxin for cholera. Perhaps future adjuvants occupying other putative receptors will be employed to bypass the TLR signaling pathway completely in order to circumvent common side effects of adjuvant-activated TLRs such as local inflammation and the general malaise felt because of the costly whole-body immune response to antigen. Lupus (2009) 18, 1217—1225.
Collapse
Affiliation(s)
- E. Israeli
- Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - N. Agmon-Levin
- Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel, Department of Medicine 'B', Sheba Medical Center, Tel-Hashomer, Israel
| | - M. Blank
- Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Y. Shoenfeld
- Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel, Sackler Faculty of Medicine, Incumbent of the Laura Schwarz-Kip Chair for Research of Autoimmune Diseases, Tel-Aviv University, Israel,
| |
Collapse
|
36
|
Hartikka J, Bozoukova V, Yang CK, Ye M, Rusalov D, Shlapobersky M, Vilalta A, Wei Q, Rolland A, Smith LR. Vaxfectin®, a cationic lipid-based adjuvant for protein-based influenza vaccines. Vaccine 2009; 27:6399-403. [DOI: 10.1016/j.vaccine.2009.06.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
37
|
A liposome-based mycobacterial vaccine induces potent adult and neonatal multifunctional T cells through the exquisite targeting of dendritic cells. PLoS One 2009; 4:e5771. [PMID: 19492047 PMCID: PMC2685976 DOI: 10.1371/journal.pone.0005771] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 05/07/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In the search for more potent and safer tuberculosis vaccines, CAF01 was identified as a remarkable formulation. Based on cationic liposomes and including a synthetic mycobacterial glycolipid as TLR-independent immunomodulator, it induces strong and protective T helper-1 and T helper-17 adult murine responses to Ag85B-ESAT-6, a major mycobacterial fusion protein. Here, we assessed whether these properties extend to early life and how CAF01 mediates its adjuvant properties in vivo. METHODS/FINDINGS Following adult or neonatal murine immunization, Ag85B-ESAT-6/CAF01 similarly reduced the post-challenge bacterial growth of M. bovis BCG, whereas no protection was observed using Alum as control. This protection was mediated by the induction of similarly strong Th1 and Th17 responses in both age groups. Multifunctional Th1 cells were already elicited after a single vaccine dose and persisted at high levels for at least 6 months even after neonatal priming. Unexpectedly, this potent adjuvanticity was not mediated by a massive targeting/activation of dendritic cells: in contrast, very few DCs in the draining lymph nodes were bearing the labeled antigen/adjuvant. The increased expression of the CD40 and CD86 activation markers was restricted to the minute portion of adjuvant-bearing DCs. However, vaccine-associated activated DCs were recovered several days after immunization. CONCLUSION The potent adult and neonatal adjuvanticity of CAF01 is associated in vivo with an exquisite but prolonged DC uptake and activation, fulfilling the preclinical requirements for novel tuberculosis vaccines to be used in early life.
Collapse
|
38
|
Olafsdottir TA, Lingnau K, Nagy E, Jonsdottir I. IC31, a two-component novel adjuvant mixed with a conjugate vaccine enhances protective immunity against pneumococcal disease in neonatal mice. Scand J Immunol 2009; 69:194-202. [PMID: 19281531 DOI: 10.1111/j.1365-3083.2008.02225.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
IC31 is a novel adjuvant which combines the immunostimulatory effects of an 11-mer antibacterial peptide (KLKL(5)KLK) and a synthetic oligodeoxynucleotide (ODN1a) which is a Toll-like receptor 9 agonist without containing cytosine phosphate guanine (CpG) motifs. The effects of IC31 on neonatal immune response to vaccination have not been reported. Neonatal mice were immunized once or twice with a Streptococcus pneumoniae serotype 1 polysaccharide conjugate containing Tetanus Toxoid (Pnc1-TT) carrier protein, with or without IC31 or CpG-ODN. IC31 significantly enhanced IgG1, IgG2a and IgG2b antibodies (Ab) to the serotype 1 polysaccharide. One dose of Pnc1-TT and low dose IC31 elicited high Ab levels that protected the neonatal mice completely from bacteraemia and significantly reduced lung infection following i.n. challenge with serotype 1 pneumococcal strain. One-sixth of an adult murine dose of IC31 was sufficient and optimal for induction of protective immunity in neonatal mice. Two doses of Pnc1-TT with or without adjuvants protected the neonatal mice completely, but more rapid Ab response was observed when IC31 was given with the Pnc1-TT. IC31 is a promising new adjuvant for neonatal vaccinations, rapidly enhancing protective humoral responses when combined with Pnc1-TT.
Collapse
Affiliation(s)
- T A Olafsdottir
- Department of Immunology, Landspitali University Hospital, Reykjavik, Iceland
| | | | | | | |
Collapse
|
39
|
Kamath AT, Rochat AF, Valenti MP, Agger EM, Lingnau K, Andersen P, Lambert PH, Siegrist CA. Adult-like anti-mycobacterial T cell and in vivo dendritic cell responses following neonatal immunization with Ag85B-ESAT-6 in the IC31 adjuvant. PLoS One 2008; 3:e3683. [PMID: 18997860 PMCID: PMC2577009 DOI: 10.1371/journal.pone.0003683] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2008] [Accepted: 10/23/2008] [Indexed: 11/17/2022] Open
Abstract
Background With the exception of some live vaccines, e.g. BCG, subunit vaccines formulated with “classical” adjuvants do not induce similar responses in neonates as in adults. The usual neonatal profile is characterized by lower levels of TH1-associated biomarkers. This has hampered the development of new neonatal vaccines for diseases that require early protection. Tuberculosis is one of the major targets for neonatal immunization. In this study, we assessed the immunogenicity of a novel candidate vaccine comprising a mycobacterial fusion protein, Ag85B-ESAT-6, in a neonatal murine immunization model. Methods/Findings The Ag85B-ESAT-6 fusion protein was formulated either with a classical alum based adjuvant or with the novel IC31® adjuvant. Following neonatal or adult immunization, 3 parameters were studied in vivo: (1) CD4+ T cell responses, (2) vaccine targeting/activation of dendritic cells (DC) and (3) protection in a surrogate mycobacterial challenge model. Conversely to Alum, IC31® induced in both age groups strong Th1 and Th17 responses, characterized by multifunctional T cells expressing IL-2 and TNF-α with or without IFN-γ. In the draining lymph nodes, a similarly small number of DC contained the adjuvant and/or the antigen following neonatal or adult immunization. Expression of CD40, CD80, CD86 and IL-12p40 production was focused on the minute adjuvant-bearing DC population. Again, DC targeting/activation was similar in adults and neonates. These DC/T cell responses resulted in an equivalent reduction of bacterial growth following infection with M. bovis BCG, whereas no protection was observed when Alum was used as adjuvant. Conclusion Neonatal immunization with the IC31®- adjuvanted Ag85B-ESAT-6 subunit vaccine elicited adult-like multifunctional protective anti-mycobacterial T cell responses through the induction of an adult pattern of in vivo DC activation.
Collapse
Affiliation(s)
- Arun T Kamath
- World Health Organization Collaborating Center for Vaccinology and Neonatal Immunology, Department of Pathology-Immunology and Pediatrics, University of Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|