Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Infect Dis ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836471

ABSTRACT

BACKGROUND: We determined the relationships between cytokine expression in sputum and clinical data to characterise and understand Chronic Obstructive Pulmonary Disease (COPD) exacerbations in COPD patients. METHODS: We measured 30 cytokines in 936 sputum samples, collected at stable state (ST) and exacerbation (EX) visits from 99 participants in the Acute Exacerbation and Respiratory InfectionS in COPD (AERIS) study (NCT01360398, www.clinicaltrials.gov). We determined their longitudinal expression and examined differential expression based on disease status or exacerbation type. RESULTS: Of the cytokines, 17 were suitable for analysis. As for disease states, in EX sputum samples, IL-17A, TNF-α, IL-1ß, and IL-10 were significantly increased compared to ST sputum samples, but a logistic mixed model could not predict disease state. As for exacerbation types, bacteria-associated exacerbations showed higher expression of IL-17A, TNF-α, IL-1ß, and IL-1α. IL-1α, IL-1ß, and TNF-α were identified as suitable biomarkers for bacteria-associated exacerbation. Bacteria-associated exacerbations also formed a cluster separate from other exacerbation types in principal component analysis. CONCLUSIONS: Measurement of cytokines in sputum from COPD patients could help identify bacteria-associated exacerbations based on increased concentrations of IL-1α, IL-1ß, or TNF-α. This finding may provide a point-of-care assessment to distinguish a bacterial exacerbation of COPD from other exacerbation types.

2.
Vaccine ; 41(3): 724-734, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36564274

ABSTRACT

The candidate Adjuvant System AS37 contains a synthetic toll-like receptor agonist (TLR7a) adsorbed to alum. In a phase I study (NCT02639351), healthy adults were randomised to receive one dose of licensed alum-adjuvanted meningococcal serogroup C (MenC-CRM197) conjugate vaccine (control) or MenC-CRM197 conjugate vaccine adjuvanted with AS37 (TLR7a dose 12.5, 25, 50 or 100 µg). A subset of 66 participants consented to characterisation of peripheral whole blood transcriptomic responses, systemic cytokine/chemokine responses and multiple myeloid and lymphoid cell responses as exploratory study endpoints. Blood samples were collected pre-vaccination, 6 and 24 h post-vaccination, and 3, 7, 28 and 180 days post-vaccination. The gene expression profile in whole blood showed an early, AS37-specific transcriptome response that peaked at 24 h, increased with TLR7a dose up to 50 µg and generally resolved within one week. Five clusters of differentially expressed genes were identified, including those involved in the interferon-mediated antiviral response. Evaluation of 30 cytokines/chemokines by multiplex assay showed an increased level of interferon-induced chemokine CXCL10 (IP-10) at 24 h and 3 days post-vaccination in the AS37-adjuvanted vaccine groups. Increases in activated plasmacytoid dendritic cells (pDC) and intermediate monocytes were detected 3 days post-vaccination in the AS37-adjuvanted vaccine groups. T follicular helper (Tfh) cells increased 7 days post-vaccination and were maintained at 28 days post-vaccination, particularly in the AS37-adjuvanted vaccine groups. Moreover, most of the subjects that received vaccine containing 25, 50 and 100 µg TLR7a showed an increased MenC-specific memory B cell responses versus baseline. These data show that the adsorption of TLR7a to alum promotes an immune signature consistent with TLR7 engagement, with up-regulation of interferon-inducible genes, cytokines and frequency of activated pDC, intermediate monocytes, MenC-specific memory B cells and Tfh cells. TLR7a 25-50 µg can be considered the optimal dose for AS37, particularly for the adjuvanted MenC-CRM197 conjugate vaccine.


Subject(s)
Aluminum Hydroxide , Meningococcal Vaccines , Adult , Humans , Interferons , Toll-Like Receptor 7 , Antiviral Agents , Vaccines, Conjugate , Adjuvants, Immunologic , Cytokines , Systems Analysis
3.
Sci Rep ; 9(1): 11520, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31395915

ABSTRACT

Self-emulsification is routinely used for oral delivery of lipophilic drugs in vivo, with the emulsion forming in vivo. We modified this technique to prepare novel oil-in-water emulsions of varying droplet size and composition on bench to enable adjuvanted vaccine delivery. We used these formulations to show that smaller droplets (20 nm) were much less effective as adjuvants for an influenza vaccine in mice than the emulsion droplet size of commercial influenza vaccine adjuvants (~160 nm). This was unexpected, given the many claims in the literature of the advantages of smaller particulates. We also undertook cell-recruitment mechanistic studies at site of injection and draining lymph nodes to directly address the question of why the smaller droplets were less effective. We discovered that emulsion droplet size and composition have a considerable impact on the ability to recruit immune cells to the injection site. We believe that further work is warranted to more extensively explore the question of whether, the smaller is not 'better', is a more common observation for particulate adjuvants.


Subject(s)
Adjuvants, Pharmaceutic/administration & dosage , Emulsions/chemistry , Influenza Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Drug Compounding , Female , Immunity, Cellular , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Particle Size
4.
ACS Infect Dis ; 5(9): 1546-1558, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31290323

ABSTRACT

Mannosylation of Lipid Nanoparticles (LNP) can potentially enhance uptake by Antigen Presenting Cells, which are highly abundant in dermal tissues, to improve the potency of Self Amplifying mRNA (SAM) vaccines in comparison to the established unmodified LNP delivery system. In the current studies, we evaluated mannosylated LNP (MLNP), which were obtained by incorporation of a stable Mannose-cholesterol amine conjugate, for the delivery of an influenza (hemagglutinin) encoded SAM vaccine in mice, by both intramuscular and intradermal routes of administration. SAM MLNP exhibited in vitro enhanced uptake in comparison to unglycosylated LNP from bone marrow-derived dendritic cells, and in vivo more rapid onset of the antibody response, independent of the route. The increased binding antibody levels also translated into higher functional hemagglutinin inhibition titers, particularly following intradermal administration. T cell assay on splenocytes from immunized mice also showed an increase in antigen specific CD8+ T responses, following intradermal administration of MLNP SAM vaccines. Induction of enhanced antigen specific CD4+ T cells, correlating with higher IgG2a antibody responses, was also observed. Hence, the present work illustrates the benefit of mannosylation of LNPs to achieve a faster immune response with SAM vaccines and these observations could contribute to the development of novel skin delivery systems for SAM vaccines.


Subject(s)
Cholesterol/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/administration & dosage , Mannose/chemistry , Orthomyxoviridae Infections/prevention & control , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/virology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/virology , Female , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immunoglobulin G/metabolism , Influenza Vaccines/chemical synthesis , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Injections, Intradermal , Mice , Nanoparticles , Orthomyxoviridae Infections/immunology , Particle Size , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/immunology
5.
Vaccine ; 37(30): 4204-4213, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31227353

ABSTRACT

Self-amplifying mRNAs (SAM)-based vaccines have been shown to induce a robust immune response in various animal species against both viral and bacterial pathogens. Due to their synthetic nature and to the versatility of the manufacturing process, SAM technology may represent an attractive solution for rapidly producing novel vaccines, which is particularly critical in case of pandemic infections or diseases mediated by newly emerging pathogens. Recent published data support the hypothesis that Antigen Presenting Cells (APCs) are responsible for CD8+ T-cell priming after SAM vaccination, suggesting cross-priming as the key mechanism for antigen presentation by SAM vaccines. In our study we investigated the possibility to enhance the immune response induced in mice by a single immunization with SAM by increasing the recruitment of APCs at the site of injection. To enhance SAM immunogenicity, we selected murine granulocyte-macrophage colony-stimulating factor (GM-CSF) as a model chemoattractant for APCs, and developed a SAM-GM-CSF vector. We evaluated whether the use of SAM-GM-CSF in combination with a SAM construct encoding the Influenza A virus nucleoprotein (NP) would lead to an increase of APC recruitment and NP-specific immune response. We indeed observed that the administration of SAM-GM-CSF enhances the recruitment of APCs at the injection site. Consistently with our hypothesis, co-administration of SAM-GM-CSF with SAM-NP significantly improved the magnitude of NP-specific CD8+ T-cell response both in terms of frequency of cytotoxic antigen-specific CD8+ T-cells and their functional activity in vivo. Furthermore, co-immunization with SAM-GM-CSF and SAM-NP provided an increase in protection against a lethal challenge with influenza virus. In conclusion, we demonstrated that increased recruitment of APCs at the site of injection is associated with an enhanced effectiveness of SAM vaccination and might be a powerful tool to potentiate the efficacy of RNA vaccination.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , RNA, Messenger/metabolism , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , CD8-Positive T-Lymphocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Influenza, Human/immunology , Influenza, Human/prevention & control , Mice , Mice, Inbred C57BL
6.
J Infect Dis ; 213(12): 1876-85, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26908732

ABSTRACT

BACKGROUND: Most preclinical studies assess vaccine effectiveness in single-pathogen infection models. This is unrealistic given that humans are continuously exposed to different commensals and pathogens in sequential and mixed infections. Accordingly, complications from secondary bacterial infection are a leading cause of influenza-associated morbidity and mortality. New vaccination strategies are needed to control infections on simultaneous fronts. METHODS: We compared different anti-influenza vaccines for their protective potential in a model of viral infection with bacterial superinfection. Mice were immunized with H1N1/A/California/7/2009 subunit vaccines, formulated with different adjuvants inducing either T-helper type 1 (Th1) (MF59 plus CpG)-, Th1/2 (MF59)-, or Th17 (LTK63)-prone immune responses and were sequentially challenged with mouse-adapted influenza virus H1N1/A/Puerto Rico/8/1934 and Staphylococcus aureus USA300, a clonotype emerging as a leading contributor in postinfluenza pneumonia in humans. RESULTS: Unadjuvanted vaccine controlled single viral infection, yet mice had considerable morbidity from viral disease and bacterial superinfection. In contrast, all adjuvanted vaccines efficiently protected mice in both conditions. Interestingly, the Th1-inducing formulation was superior to Th1/2 or Th17 inducers. CONCLUSIONS: Our studies should help us better understand how differential immunity to influenza skews immune responses toward coinfecting bacteria and discover novel modes to prevent bacterial superinfections in the lungs of persons with influenza.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Superinfection/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Bacterial Toxins/administration & dosage , Enterotoxins/administration & dosage , Escherichia coli Proteins/administration & dosage , Female , Humans , Immunization , Influenza Vaccines/administration & dosage , Influenza, Human/complications , Influenza, Human/microbiology , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/administration & dosage , Polysorbates/administration & dosage , Specific Pathogen-Free Organisms , Squalene/administration & dosage , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology , Superinfection/microbiology
7.
Vaccine ; 32(20): 2382-8, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24434044

ABSTRACT

Influenza is a vaccine-preventable disease that remains a major health problem world-wide. Needle and syringe are still the primary delivery devices, and injection of liquid vaccine into the muscle is still the primary route of immunization. Vaccines could be more convenient and effective if they were delivered by the mucosal route. Elicitation of systemic and mucosal innate and adaptive immune responses, such as pathogen neutralizing antibodies (including mucosal IgA at the site of pathogen entry) and CD4(+) T-helper cells (especially the Th17 subset), have a critical role in vaccine-mediated protection. In the current study, a sublingual subunit influenza vaccine formulated with or without mucosal adjuvant was evaluated for systemic and mucosal immunogenicity and compared to intranasal and intramuscular vaccination. Sublingual administration of adjuvanted influenza vaccine elicited comparable antibody titers to those elicited by intramuscular immunization with conventional influenza vaccine. Furthermore, influenza-specific Th17 cells or neutralizing mucosal IgA were detected exclusively after mucosal immunization.


Subject(s)
Administration, Sublingual , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Th17 Cells/immunology , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Hemagglutination Inhibition Tests , Humans , Immunity, Mucosal , Immunoglobulin A/immunology , Influenza A Virus, H1N1 Subtype , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Vaccines, Subunit/administration & dosage
8.
Proc Natl Acad Sci U S A ; 110(52): 21095-100, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324152

ABSTRACT

Vaccines are the most effective agents to control infections. In addition to the pathogen antigens, vaccines contain adjuvants that are used to enhance protective immune responses. However, the molecular mechanism of action of most adjuvants is ill-known, and a better understanding of adjuvanticity is needed to develop improved adjuvants based on molecular targets that further enhance vaccine efficacy. This is particularly important for tuberculosis, malaria, AIDS, and other diseases for which protective vaccines do not exist. Release of endogenous danger signals has been linked to adjuvanticity; however, the role of extracellular ATP during vaccination has never been explored. Here, we tested whether ATP release is involved in the immune boosting effect of four common adjuvants: aluminum hydroxide, calcium phosphate, incomplete Freund's adjuvant, and the oil-in-water emulsion MF59. We found that intramuscular injection is always associated with a weak transient release of ATP, which was greatly enhanced by the presence of MF59 but not by all other adjuvants tested. Local injection of apyrase, an ATP-hydrolyzing enzyme, inhibited cell recruitment in the muscle induced by MF59 but not by alum or incomplete Freund's adjuvant. In addition, apyrase strongly inhibited influenza-specific T-cell responses and hemagglutination inhibition titers in response to an MF59-adjuvanted trivalent influenza vaccine. These data demonstrate that a transient ATP release is required for innate and adaptive immune responses induced by MF59 and link extracellular ATP with an enhanced response to vaccination.


Subject(s)
Adenosine Triphosphate/metabolism , Adjuvants, Immunologic/pharmacology , CD4-Positive T-Lymphocytes/immunology , Muscle, Skeletal/metabolism , Polysorbates/pharmacology , Squalene/pharmacology , Vaccination/methods , Aluminum Hydroxide/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , Calcium Phosphates/immunology , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Freund's Adjuvant/immunology , Lipids/immunology , Luminescent Measurements , Mice , Mice, Inbred BALB C , Specific Pathogen-Free Organisms , Squalene/immunology
9.
Vaccine ; 31(33): 3363-9, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23684834

ABSTRACT

MF59 is a safe and effective vaccine adjuvant that has been used in a licensed seasonal influenza vaccine for 15 years. The purpose of the present studies was to directly address a question that has been asked of us on many occasions: "which is the adjuvant active component of MF59?". Since we have recently gained a number of insights on how MF59 works as an adjuvant, we were able to use these approaches to evaluate if the individual components of MF59 (squalene oil, the surfactants Span 85 and Tween 80 or the citrate buffer) showed any direct immunostimulatory activity. We assessed the ability of the individual components to stimulate the innate and adaptive immune responses that we have shown to be indicative of MF59-mediated adjuvanticity. No immune stimulatory capacities could be attributed to squalene, Tween 80 or the citrate buffer alone. Instead, we found that the lipophilic surfactant Span 85 contributes to activation of the muscle transcriptome. However, despite this local activation, Span 85 alone - like the other single components of MF59 - is not sufficient to induce an adjuvant effect. Only the fully formulated MF59 emulsion induces all the established hallmarks of innate and adaptive immune activation, which includes activation of genes indicative of transendothelial cell migration, strong influx of immune cells into the injection site and their enhanced antigen uptake and transport to the lymph nodes. These observations may have important implications in the design of optimal emulsion-based vaccine adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Emulsions/pharmacology , Polysorbates/pharmacology , Squalene/pharmacology , Adaptive Immunity , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Viral/blood , Antigen Presentation/drug effects , CD4-Positive T-Lymphocytes/immunology , Emulsions/chemistry , Female , Hemagglutination Inhibition Tests , Hexoses/pharmacology , Humans , Immunity, Innate , Influenza Vaccines/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation , Macrophages/immunology , Mice , Mice, Inbred BALB C , Monocytes/immunology , Polysorbates/chemistry , Squalene/chemistry , Transcriptome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...