Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Mol Ther Nucleic Acids ; 32: 794-806, 2023 Jun 13.
Article En | MEDLINE | ID: mdl-37346973

The use of modified nucleosides is an important approach to mitigate the intrinsic immunostimulatory activity of exogenous mRNA and to increase its translation for mRNA therapeutic applications. However, for vaccine applications, the intrinsic immunostimulatory nature of unmodified mRNA could help induce productive immunity. Additionally, the ionizable lipid nanoparticles (LNPs) used to deliver mRNA vaccines can possess immunostimulatory properties that may influence the impact of nucleoside modification. Here we show that uridine replacement with N1-methylpseudouridine in an mRNA vaccine encoding influenza hemagglutinin had a significant impact on the induction of innate chemokines/cytokines and a positive impact on the induction of functional antibody titers in mice and macaques when MC3 or KC2 LNPs were used as delivery systems, while it impacted only minimally the titers obtained with L319 LNPs, indicating that the impact of nucleoside modification on mRNA vaccine efficacy varies with LNP composition. In line with previous observations, we noticed an inverse correlation between the induction of high innate IFN-α titers in the macaques and antigen-specific immune responses. Furthermore, and consistent with the species specificity of pathogen recognition receptors, we found that the effect of uridine replacement did not strictly translate from mice to non-human primates.

2.
Front Immunol ; 14: 1100161, 2023.
Article En | MEDLINE | ID: mdl-36845117

Introduction: Pulmonary-resident memory T cells (TRM) and B cells (BRM) orchestrate protective immunity to reinfection with respiratory pathogens. Developing methods for the in situ detection of these populations would benefit both research and clinical settings. Methods: To address this need, we developed a novel in situ immunolabelling approach combined with clinic-ready fibre-based optical endomicroscopy (OEM) to detect canonical markers of lymphocyte tissue residency in situ in human lungs undergoing ex vivo lung ventilation (EVLV). Results: Initially, cells from human lung digests (confirmed to contain TRM/BRM populations using flow cytometry) were stained with CD69 and CD103/CD20 fluorescent antibodies and imaged in vitro using KronoScan, demonstrating it's ability to detect antibody labelled cells. We next instilled these pre-labelled cells into human lungs undergoing EVLV and confirmed they could still be visualised using both fluorescence intensity and lifetime imaging against background lung architecture. Finally, we instilled fluorescent CD69 and CD103/CD20 antibodies directly into the lung and were able to detect TRM/BRM following in situ labelling within seconds of direct intra-alveolar delivery of microdoses of fluorescently labelled antibodies. Discussion: In situ, no wash, immunolabelling with intra-alveolar OEM imaging is a novel methodology with the potential to expand the experimental utility of EVLV and pre-clinical models.


Immunologic Memory , Lung , Humans , Lung/diagnostic imaging , CD8-Positive T-Lymphocytes , Lymphocytes
3.
Sci Rep ; 12(1): 6570, 2022 04 21.
Article En | MEDLINE | ID: mdl-35449223

The World Health Organization protocol for rabies post-exposure prophylaxis (PEP) recommends extensive wound washing, immediate vaccination, and administration of rabies immunoglobulin (RIG) in severe category III exposures. Some studies have shown that RIG can interfere with rabies vaccine immunogenicity to some extent. We investigated the interference of RIG on a next generation highly purified Vero cell rabies vaccine candidate (PVRV-NG) versus standard-of-care vaccines in a previously described hamster model. The interference of either human (h) or equine (e) RIG on the immune response elicited by PVRV-NG, Verorab® (purified Vero cell rabies vaccine, PVRV), and Imovax® Rabies (human diploid cell rabies vaccine; HDCV) was evaluated using the 4-dose Essen PEP regimen. The anti-rabies seroneutralizing titers and specific serum IgM titers were measured by fluorescent antibody virus neutralization test and enzyme-linked immunosorbent assay, respectively, for the vaccines administered with or without RIG. The RIG interference on PVRV-NG, observed transiently at Day 7, was similar to that on PVRV and tended to be lower than that on HDCV using both read-outs. In summary, the results generated in the hamster model showed that RIG induced similar or less interference on PVRV-NG than the standard-of-care vaccines.


Blood Group Antigens , Rabies Vaccines , Rabies virus , Rabies , Animals , Antibodies, Viral , Chlorocebus aethiops , Cricetinae , Horses , Humans , Immunoglobulins , Immunologic Factors , Post-Exposure Prophylaxis , Rabies/prevention & control , Rabies Vaccines/immunology , Rabies virus/immunology , Vero Cells
4.
Hum Vaccin Immunother ; 17(7): 2336-2348, 2021 07 03.
Article En | MEDLINE | ID: mdl-33427044

Adjuvants are central to the efficacy of subunit vaccines. Although several new adjuvants have been approved in human vaccines over the last decade, the panel of adjuvants in licensed human vaccines remains small. There is still a need for novel adjuvants that can be safely used in humans, easy to source and to formulate with a wide range of antigens and would be broadly applicable to a wide range of vaccines. In this article, using the Respiratory Syncytial Virus (RSV) nanoparticulate prefusion F model antigen developed by Sanofi, we demonstrate in the macaque model that the polyacrylate (PAA)-based adjuvant SPA09 is well tolerated and increases vaccine antigen-specific humoral immunity (sustained neutralizing antibodies, memory B cells and mucosal immunity) and elicits strong TH1-type responses (based on IFNγ and IL-2 ELISpots) in a dose-dependent manner. These data warrant further development of the SPA09 adjuvant for evaluation in clinical trials.


Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing , Antibodies, Viral , Humans , Immunity, Cellular , Immunity, Humoral , Macaca fascicularis
5.
NPJ Vaccines ; 5(1): 19, 2020.
Article En | MEDLINE | ID: mdl-32194996

The recent spread of Zika virus (ZIKV) through the Americas and Caribbean and its devastating consequences for pregnant women and their babies have driven the search for a safe and efficacious ZIKV vaccine. Among the vaccine candidates, a first-generation ZIKV purified inactivated vaccine (ZPIV), adjuvanted with aluminum hydroxide, developed by the Walter Reed Army Institute of Research (WRAIR), has elicited high seroconversion rates in participants in three phase-I clinical trials. In collaboration with the WRAIR, Sanofi Pasteur (SP) optimized the production scale, culture and purification conditions, and increased the regulatory compliance, both of which are critical for clinical development and licensure of this vaccine. Using a clinical batch of the first-generation ZPIV as a benchmark, we report that different doses of the optimized vaccine (ZPIV-SP) elicited sustained neutralizing antibodies, specific T- and memory B-cells, and provided complete protection against a ZIKV challenge in cynomolgus macaques. These data provide evidence that the ZPIV-SP vaccine performs at least as well as the ZPIV vaccine, and provide support for continued development in the event of future ZIKV outbreaks.

6.
Vaccine ; 37(20): 2679-2686, 2019 05 06.
Article En | MEDLINE | ID: mdl-30967310

After decades of inconsequential infections, and sporadic outbreaks in the Asia-Pacific region between 2007 and 2013, Zika virus caused a widespread epidemic in South America in 2015 that was complicated by severe congenital infections. After the WHO declared a Public Health Emergency of International Concern in February 2016, vaccine development efforts based on different platforms were initiated. Several candidates have since been evaluated in clinical phase I studies. Of these, a Zika purified inactivated vaccine (ZPIV), adjuvanted with aluminum hydroxide, developed by the Walter Reed Army Institute of Research (WRAIR), yielded high seroconversion rates. Sanofi Pasteur further optimized the vaccine in terms of production scale, purification conditions and regulatory compliance, using its experience in flavivirus vaccine development. Here we report that the resulting optimized vaccine (ZPIV-SP) elicited robust seroneutralizing antibody responses and provided complete protection from homologous Zika virus strain challenge in immunocompetent BALB/c mice. ZPIV-SP also showed improved immunogenicity compared with the first-generation vaccine, and improved efficacy in the more permissive interferon receptor-deficient A129 mice. Finally, analysis of the IgG response directed towards nonstructural protein 1 (NS1) suggests that viral NS1 was efficiently removed during the optimized purification process of ZPIV-SP. Together, these results suggest that the optimized vaccine is well suited for further evaluation in larger animal models and late-stage clinical studies.


Immunogenicity, Vaccine , Vaccine Potency , Vaccines, Inactivated/immunology , Viral Vaccines/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Knockout , Viral Envelope Proteins/immunology , Zika Virus/genetics , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
7.
J Virol ; 92(12)2018 06 15.
Article En | MEDLINE | ID: mdl-29593041

Recent data obtained with the live-attenuated tetravalent dengue CYD-TDV vaccine showed higher protective efficacy against dengue virus type 4 (DENV-4) than against DENV-2. In contrast, results from previous studies in nonhuman primates predicted comparable high levels of protection against each serotype. Maximum viral loads achieved in macaques by subcutaneous inoculation of DENV are generally much lower than those observed in naturally dengue virus-infected humans. This may contribute to an overestimation of vaccine efficacy. Using more-stringent DENV infection conditions consisting of the intravenous inoculation of 107 50% cell culture infectious doses (CCID50) in CYD-TDV-vaccinated macaques, complete protection (i.e., undetectable viral RNA) was achieved in all 6 monkeys challenged with DENV-4 and in 6/18 of those challenged with DENV-2, including transiently positive animals. All other infected macaques (12/18) developed sustained DENV-2 RNAemia (defined as detection of viral RNA in serum samples) although 1 to 3 log10 units below the levels achieved in control animals. Similar results were obtained with macaques immunized with either CYD-TDV or monovalent (MV) CYD-2. This suggests that partial protection against DENV-2 was mediated mainly by CYD-2 and not by the other CYDs. Postchallenge induction of strong anamnestic responses, suggesting efficient vaccine priming, likely contributed to the reduction of DENV-2 RNAemia. Finally, an inverse correlation between DENV RNA titers postchallenge and vaccine-induced homotypic neutralizing antibody titers prechallenge was found, emphasizing the key role of these antibodies in controlling DENV infection. Collectively, these data show better agreement with reported data on CYD-TDV clinical vaccine efficacy against DENV-2 and DENV-4. Despite inherent limitations of the nonhuman primate model, these results reinforce its value in assessing the efficacy of dengue vaccines.IMPORTANCE The nonhuman primate (NHP) model is the most widely recognized tool for assessing the protective activity of dengue vaccine candidates, based on the prevention of postinfection DENV viremia. However, its use has been questioned after the recent CYD vaccine phase III trials, in which moderate protective efficacy against DENV-2 was reported, despite full protection against DENV-2 viremia previously being demonstrated in CYD-vaccinated monkeys. Using a reverse translational approach, we show here that the NHP model can be improved to achieve DENV-2 protection levels that show better agreement with clinical efficacy. With this new model, we demonstrate that the injection of the CYD-2 component of the vaccine, in either a monovalent or a tetravalent formulation, is able to reduce DENV-2 viremia in all immunized animals, and we provide clear statistical evidence that DENV-2-neutralizing antibodies are able to reduce viremia in a dose-dependent manner.


Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Vaccines/immunology , Dengue Virus/immunology , Dengue/immunology , Viral Load/veterinary , Animals , Dengue/prevention & control , Dengue/virology , Dengue Virus/genetics , Disease Models, Animal , Macaca fascicularis , Male , RNA, Viral/blood , Treatment Outcome , Vaccination , Viral Load/immunology , Viremia/prevention & control
8.
PLoS Negl Trop Dis ; 10(7): e0004830, 2016 07.
Article En | MEDLINE | ID: mdl-27459266

BACKGROUND: Two large-scale efficacy studies with the recombinant yellow fever-17D-dengue virus, live-attenuated, tetravalent dengue vaccine (CYD-TDV) candidate undertaken in Asia (NCT01373281) and Latin America (NCT01374516) demonstrated significant protection against dengue disease during two years' active surveillance (active phase). Long-term follow up of participants for breakthrough disease leading to hospitalization is currently ongoing (hospital phase). METHODOLOGY/PRINCIPAL FINDINGS: We assessed the cytokine profile in acute sera from selected participants hospitalized (including during the active phase) up to the beginning of the second year of long-term follow up for both studies. The serum concentrations of 38 cytokines were measured in duplicate using the Milliplex Human Cytokine MAGNETIC BEAD Premixed 38 Plex commercial kit (Millipore, Billerica, MA, USA). Partial least squares discriminant analyses did not reveal any difference in the overall cytokine profile of CYD-TDV and placebo recipients hospitalized for breakthrough dengue regardless of stratification used. In addition, there was no difference in the cytokine profile for breakthrough dengue among those aged <9 years versus those aged ≥ 9 years. CONCLUSIONS/SIGNIFICANCE: These exploratory findings show that CYD-TDV does not induce a particular immune profile versus placebo, corroborating the clinical profile observed.


Cytokines/blood , Dengue Vaccines/administration & dosage , Dengue Virus/isolation & purification , Dengue/therapy , Vaccines, Attenuated/administration & dosage , Adolescent , Antibodies, Viral/blood , Child , Child, Preschool , Dengue/blood , Dengue/immunology , Dengue/virology , Dengue Vaccines/immunology , Dengue Virus/classification , Dengue Virus/genetics , Female , Hospitalization/statistics & numerical data , Humans , Infant , Male , Vaccines, Attenuated/immunology
9.
Vaccine ; 34(7): 914-22, 2016 Feb 10.
Article En | MEDLINE | ID: mdl-26768129

Intradermal (ID) vaccination constitutes a promising approach to induce anti-infectious immunity. This route of immunization has mostly been studied with influenza split-virion vaccines. However, the efficacy of ID vaccination for sub-unit vaccines in relation to underlying skin innate immunity remains to be explored for wider application in humans. Relevant animal models that more closely mimic human skin immunity than the widely used mouse models are therefore necessary. Here, we show in domestic swine, which shares striking anatomic and functional properties with human skin, that a single ID delivery of pseudorabies virus (PRV) glycoproteins without added adjuvant is sufficient to trigger adaptive cellular and humoral immune responses, and to confer protection from a lethal respiratory infection with PRV. Analysis of early events at the skin injection site revealed up-regulation of pro-inflammatory cytokine and chemokine genes, recruitment of neutrophils and monocytes and accumulation of inflammatory DC. We further show that the sustained induction of pro-inflammatory cytokine genes results from the combined effects of skin puncture, liquid injection in the dermis and viral antigens. These data highlight that immune protection against respiratory infection can be induced by ID vaccination with a subunit vaccine and reveal that adjuvant requirements are circumvented by the mechanical and antigenic stress caused by ID injection, which triggers innate immunity and mobilization of inflammatory DC at the immunization site. ID vaccination with sub-unit vaccines may thus represent a safe and efficient solution for protection against respiratory infections in swine and possibly also in humans, given the similarity of skin structure and function in both species.


Immunity, Innate , Pseudorabies/prevention & control , Skin/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytokines , Dendritic Cells/immunology , Female , Immunity, Cellular , Immunity, Humoral , Injections, Intradermal , Swine , Vaccination/methods , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Vaccines/immunology
10.
Hum Vaccin Immunother ; 11(6): 1449-55, 2015.
Article En | MEDLINE | ID: mdl-25997147

A tuberculosis (TB) vaccine consisting of a recombinant fusion protein (H4) and a novel TLR9 adjuvant (IC31) is in clinical development. To better understand the H4-IC31 ratio, we measured the binding capacity of IC31 for H4 protein and immunized mice with formulations that contained limiting to excess ratios of IC31 to H4. An immunomodulated H4-specific IFNγ response was only observed when IC31 was present in excess of H4. Since TLR expression is species-specific and the vaccine is intended to boost BCG-primed immunity, we questioned whether data in mice would translate to humans. To address this question, we used the fresh human Whole Blood (hWB) recovered from BCG-vaccinated subjects to screen H4-IC31 formulations. We found IC31 modulation in hWB to be quite distinct from the TLR4-Adjuvant. Unlike TLR4-Adjuvant, IC31 formulations did not induce the pro-inflammatory cytokine TNFα, but modulated a robust H4-specific IFNγ response after 12 d of culture. We then re-stimulated the fresh hWB of 5 BCG-primed subjects with formulations that had excess or limiting IC31 binding for H4 protein and again found that an immunomodulated H4-specific IFNγ response needed an excess of IC31. Finally, we monitored the zeta (ζ) potential of H4-IC31 formulations and found that the overall charge of H4-IC31 particles changes from negative to positive once IC31 is in greater than 9-fold excess. Using two diverse yet mutually supportive approaches, we confirm the need for an excess of IC31 adjuvant in H4 TB vaccine formulations and suggest surface potential may be an important factor.


Antigens, Bacterial/immunology , Immunomodulation , Oligodeoxyribonucleotides/pharmacology , Oligopeptides/pharmacology , Tuberculosis Vaccines/immunology , Animals , Cells, Cultured , Chemistry, Pharmaceutical , Drug Combinations , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Mice , Vaccination/methods
11.
Hum Vaccin Immunother ; 10(9): 2713-20, 2014.
Article En | MEDLINE | ID: mdl-25483667

Intanza® 9 µg (Sanofi Pasteur), a trivalent split-virion vaccine administered by intradermal (ID) injection, was approved in Europe in 2009 for the prevention of seasonal influenza in adults 18 to 59 years. Here, we examined the immune responses induced in adults by the ID 9 µg vaccine and the standard trivalent intramuscular (IM) vaccine (Vaxigrip® 15 µg, Sanofi Pasteur). This trial was a randomized, controlled, single-center, open-label study in healthy adults 18 to 40 years of age during the 2007/8 influenza season. Subjects received a single vaccination with the ID 9 µg (n=38) or IM 15 µg (n=42) vaccine. Serum, saliva, and peripheral blood mononuclear cells were collected up to 180 days post-vaccination. Geometric mean hemagglutination inhibition titers, seroprotection rates, seroconversion rates, and pre-vaccination-to-post-vaccination ratios of geometric mean hemagglutination inhibition titers did not differ between the two vaccines. Compared with pre-vaccination, the vaccines induced similar increases in vaccine-specific circulating B cells at day 7 but did not induce significant increases in vaccine-specific memory B cells at day 180. Cell-mediated immunity to all three vaccine strains, measured in peripheral blood mononuclear cells, was high at baseline and not increased by either vaccine. Neither vaccine induced a mucosal immune response. These results show that the humoral and cellular immune responses to the ID 9 µg vaccine are similar to those to the standard IM 15 µg vaccine.


Immunity, Cellular , Immunity, Humoral , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Adolescent , Adult , Antibodies, Viral/analysis , Antibodies, Viral/blood , B-Lymphocytes/immunology , Europe , Female , Hemagglutination Inhibition Tests , Humans , Injections, Intradermal , Injections, Intramuscular , Leukocytes, Mononuclear/immunology , Male , Saliva/immunology , Serum/immunology , Young Adult
12.
Vaccine ; 28(18): 3076-9, 2010 Apr 19.
Article En | MEDLINE | ID: mdl-20193791

Pandemic influenza vaccines have been manufactured using the A/California/07/2009 (H1N1) strain as recommended by the World Health Organization. We evaluated in mice the immunogenicity of pandemic (H1N1) 2009 vaccine and the impact of prior vaccination against seasonal trivalent influenza vaccines (TIV) on antibody responses against pandemic (H1N1) 2009. In naïve mice, a single dose of unadjuvanted H1N1 vaccine (3 microg of HA) was shown to elicit hemagglutination inhibition (HI) antibody titers >40, a titer associated with protection in humans against seasonal influenza. A second vaccine dose of pandemic (H1N1) 2009 vaccine strongly increased these titers, which were consistently higher in mice previously primed with TIV than in naïve mice. At a low immunization dose (0.3 microg of HA), the AF03-adjuvanted vaccine elicited higher HI antibody titers than the corresponding unadjuvanted vaccines in both naïve and TIV-primed animals, suggesting a potential for antigen dose-sparing. These results are in accordance with the use in humans of a split-virion inactivated pandemic (H1N1) 2009 vaccine formulated with or without AF03 adjuvant to protect children and young adults against influenza A (H1N1) 2009 infection.


Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Vaccination/methods , Animals , Female , Hemagglutination Inhibition Tests , Immunization, Secondary/methods , Mice , Mice, Inbred BALB C
...