Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS One ; 9(12): e115343, 2014.
Article in English | MEDLINE | ID: mdl-25506689

ABSTRACT

West Nile virus (WNV) is a zoonotic virus, which is transmitted by mosquitoes. It is the causative agent of the disease syndrome called West Nile fever. In some human cases, a WNV infection can be associated with severe neurological symptoms. The immune response to WNV is multifactorial and includes both humoral and cellular immunity. T-cell epitope mapping of the WNV envelope (E) protein has been performed in C57BL/6 mice, but not in BALB/c mice. Therefore, we performed in BALB/c mice a T-cell epitope mapping using a series of peptides spanning the WNV envelope (E) protein. To this end, the WNV-E specific T cell repertoire was first expanded by vaccinating BALB/c mice with a DNA vaccine that generates subviral particles that resemble West Nile virus. Furthermore, the WNV structural protein was expressed in Escherichia coli as a series of overlapping 20-mer peptides fused to a carrier-protein. Cytokine-based ELISPOT assays using these purified peptides revealed positive WNV-specific T cell responses to peptides within the different domains of the E-protein.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Viral Envelope Proteins/immunology , West Nile virus/immunology , Animals , Epitope Mapping , Epitopes, T-Lymphocyte/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Envelope Proteins/chemistry
2.
PLoS One ; 9(11): e112568, 2014.
Article in English | MEDLINE | ID: mdl-25392925

ABSTRACT

The mosquito-borne West Nile virus (WNV) causes human and animal disease with outbreaks in several parts of the world including North America, the Mediterranean countries, Central and East Europe, the Middle East, and Africa. Particularly in elderly people and individuals with an impaired immune system, infection with WNV can progress into a serious neuroinvasive disease. Currently, no treatment or vaccine is available to protect humans against infection or disease. The goal of this study was to develop a WNV-vaccine that is safe to use in these high-risk human target populations. We performed a vaccine efficacy study in non-human primates using the contemporary, pathogenic European WNV genotype 1a challenge strain, WNV-Ita09. Two vaccine strategies were evaluated in rhesus macaques (Macaca mulatta) using recombinant soluble WNV envelope (E) ectodomain adjuvanted with Matrix-M, either with or without DNA priming. The DNA priming immunization was performed with WNV-DermaVir nanoparticles. Both vaccination strategies successfully induced humoral and cellular immune responses that completely protected the macaques against the development of viremia. In addition, the vaccine was well tolerated by all animals. Overall, The WNV E protein adjuvanted with Matrix-M is a promising vaccine candidate for a non-infectious WNV vaccine for use in humans, including at-risk populations.


Subject(s)
West Nile Fever/prevention & control , West Nile Virus Vaccines/therapeutic use , West Nile virus/classification , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Culicidae , Europe , Immunity, Cellular , Immunity, Humoral , Injections, Intradermal , Interferon-gamma/immunology , Macaca mulatta , Protein Structure, Tertiary , Viral Envelope Proteins/immunology , Viral Load , Viremia/immunology
3.
PLoS One ; 9(2): e87837, 2014.
Article in English | MEDLINE | ID: mdl-24503579

ABSTRACT

West Nile virus (WNV) is a mosquito-borne flavivirus that is endemic in Africa, the Middle East, Europe and the United States. There is currently no antiviral treatment or human vaccine available to treat or prevent WNV infection. DNA plasmid-based vaccines represent a new approach for controlling infectious diseases. In rodents, DNA vaccines have been shown to induce B cell and cytotoxic T cell responses and protect against a wide range of infections. In this study, we formulated a plasmid DNA vector expressing the ectodomain of the E-protein of WNV into nanoparticles by using linear polyethyleneimine (lPEI) covalently bound to mannose and examined the potential of this vaccine to protect against lethal WNV infection in mice. Mice were immunized twice (prime--boost regime) with the WNV DNA vaccine formulated with lPEI-mannose using different administration routes (intramuscular, intradermal and topical). In parallel a heterologous boost with purified recombinant WNV envelope (E) protein was evaluated. While no significant E-protein specific humoral response was generated after DNA immunization, protein boosting of DNA-primed mice resulted in a marked increase in total neutralizing antibody titer. In addition, E-specific IL-4 T-cell immune responses were detected by ELISPOT after protein boost and CD8(+) specific IFN-γ expression was observed by flow cytometry. Challenge experiments using the heterologous immunization regime revealed protective immunity to homologous and virulent WNV infection.


Subject(s)
Immunity, Cellular , Vaccines, DNA , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody Specificity/immunology , Female , Immunization , Immunization, Secondary , Mice , Nanoparticles/chemistry , Th2 Cells/immunology , Viral Envelope Proteins/genetics , West Nile Fever/mortality , West Nile Virus Vaccines/adverse effects , West Nile virus/genetics
4.
PLoS One ; 8(3): e59198, 2013.
Article in English | MEDLINE | ID: mdl-23555631

ABSTRACT

BACKGROUND: Intranasal delivery of vaccines directed against respiratory pathogens is an attractive alternative to parenteral administration. However, using this delivery route for inactivated vaccines usually requires the use of potent mucosal adjuvants, and no such adjuvant has yet been approved for human use. METHODOLOGY/PRINCIPAL FINDINGS: We have developed a live attenuated Bordetella pertussis vaccine, called BPZE1, and show here that it can be used to present the universal influenza virus epitope M2e to the mouse respiratory tract to prime for protective immunity against viral challenge. Three copies of M2e were genetically fused to the N-terminal domain of filamentous hemagglutinin (FHA) and produced in recombinant BPZE1 derivatives in the presence or absence of endogenous full-length FHA. Only in the absence of FHA intranasal administration of the recombinant BPZE1 derivative induced antibody responses to M2e and effectively primed BALB/c mice for protection against influenza virus-induced mortality and reduced the viral load after challenge. Strong M2e-specific antibody responses and protection were observed after a single nasal administration with the recombinant BPZE1 derivative, followed by a single administration of M2e linked to a virus-like particle without adjuvant, whereas priming alone with the vaccine strain did not protect. CONCLUSIONS/SIGNIFICANCE: Using recombinant FHA-3M2e-producing BPZE1 derivatives for priming and the universal influenza M2e peptide linked to virus-like particles for boosting may constitute a promising approach for needle-free and adjuvant-free nasal vaccination against influenza.


Subject(s)
Adhesins, Bacterial/immunology , Antibodies, Viral/blood , Bordetella pertussis/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/immunology , Virulence Factors, Bordetella/immunology , Adhesins, Bacterial/genetics , Administration, Intranasal , Animals , Bordetella pertussis/genetics , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Respiratory System/drug effects , Respiratory System/immunology , Respiratory System/virology , Survival Analysis , Vaccination , Vaccines, Synthetic , Viral Matrix Proteins/genetics , Virulence Factors, Bordetella/genetics
5.
PLoS One ; 8(3): e59081, 2013.
Article in English | MEDLINE | ID: mdl-23527091

ABSTRACT

The ectodomain of influenza A matrix protein 2 (M2e) is a candidate for a universal influenza A vaccine. We used recombinant Hepatitis B core antigen to produce virus-like particles presenting M2e (M2e-VLPs). We produced the VLPs with and without entrapped nucleic acids and compared their immunogenicity and protective efficacy. Immunization of BALB/c mice with M2e-VLPs containing nucleic acids induced a stronger, Th1-biased antibody response compared to particles lacking nucleic acids. The former also induced a stronger M2e-specific CD4(+) T cell response, as determined by ELISPOT. Mice vaccinated with alum-adjuvanted M2e-VLPs containing the nucleic acid-binding domain were better protected against influenza A virus challenge than mice vaccinated with similar particles lacking this domain, as deduced from the loss in body weight following challenge with X47 (H3N2) or PR/8 virus. Challenge of mice that had been immunized with M2e-VLPs with or without nucleic acids displayed significantly lower mortality, morbidity and lung virus titers than control-immunized groups. We conclude that nucleic acids present in M2e-VLPs correlate with improved immune protection.


Subject(s)
Adaptive Immunity , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , RNA/metabolism , Th1 Cells/immunology , Vaccines, Virus-Like Particle/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Female , Humans , Immunity, Cellular , Influenza Vaccines/metabolism , Influenza, Human/immunology , Influenza, Human/prevention & control , Lung/immunology , Lung/pathology , Lung/virology , Mice , Myeloid Differentiation Factor 88/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Signal Transduction , Vaccination , Vaccines, Virus-Like Particle/metabolism , Viral Load
6.
J Control Release ; 167(2): 157-66, 2013 Apr 28.
Article in English | MEDLINE | ID: mdl-23402727

ABSTRACT

In this report we show that carrier-mediated delivery of mRNA may activate TLR3 signaling in respiratory cells. This activation of the innate immune system was accompanied with a massive production of type 1 interferons and other immunostimulating cytokines. The recognition of mRNA by the innate immune system was also associated with cell death, which proceeded in human respiratory cells via pyroptosis, a form of programmed cell death mediated by substantial overexpression of caspase-1. This indicated that the delivered mRNA is most likely also recognized by NOD-like receptors which regulate caspase-1 production. The viability of murine respiratory cells was less affected by mRNA transfection, which is in line with the lower transfection efficiency, lower innate immune response and the absence of a massive caspase-1 upregulation in these cells. Finally, we also demonstrated that the recognition of the delivered mRNA by the innate immune system had a negative effect on mRNA translation.


Subject(s)
Immunity, Innate , RNA, Messenger/administration & dosage , Toll-Like Receptor 3/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cytokines/metabolism , DNA/administration & dosage , HEK293 Cells , Humans , Liposomes , Luciferases/metabolism , Lung/cytology , Mice , Mice, Inbred BALB C , Transfection
7.
Mol Pharm ; 9(8): 2136-45, 2012 Aug 06.
Article in English | MEDLINE | ID: mdl-22676473

ABSTRACT

Complexes between mRNA and GL67:DOPE:DMPE-PEG5000 (GL67) liposomes were formulated and characterized. Subsequently, the in vitro and in vivo expression characteristics of mRNA/GL67 complexes and pDNA/GL67 complexes, each produced at their optimal ratio, were compared in respiratory cells. Transfection of A549 cells with mRNA/GL67 complexes resulted in a much faster expression than after transfection with pDNA/GL67 complexes. The percentage of GFP-positive cells after mRNA and pDNA transfection peaked after 8 and 24 h, respectively. At these time points the percentage of GFP-positive cells was two times higher after mRNA transfection than after pDNA transfection. Furthermore, the efficacy of mRNA/GL67 complexes was independent of the cell cycle. This was in sharp contrast with pDNA/GL67 complexes that caused only a weak expression in nondividing cells. This confirms that the nuclear barrier is a crucial obstacle for pDNA but not for mRNA. Finally, mRNA/GL67 and pDNA/GL67 complexes encoding luciferase were administered intranasally to the lungs of mice. The mRNA/GL67 complexes did not give rise to a measurable luciferase expression in the murine lungs. In contrast, a detectable bioluminescent signal was present in the lungs of mice that received the pDNA/GL67 complexes. We showed that mRNA/GL67 complexes have a lower stability in biological fluids. Consequently, this may be an explanation for their lower performance in vivo.


Subject(s)
Liposomes/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , Animals , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Transfer Techniques , Humans , Mice , Mice, Inbred BALB C , Transfection/methods
8.
Vet Res ; 43: 16, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22380523

ABSTRACT

West Nile virus (WNV) is a positive-stranded RNA virus belonging to the Flaviviridae family, a large family with 3 main genera (flavivirus, hepacivirus and pestivirus). Among these viruses, there are several globally relevant human pathogens including the mosquito-borne dengue virus (DENV), yellow fever virus (YFV), Japanese encephalitis virus (JEV) and West Nile virus (WNV), as well as tick-borne viruses such as tick-borne encephalitis virus (TBEV). Since the mid-1990s, outbreaks of WN fever and encephalitis have occurred throughout the world and WNV is now endemic in Africa, Asia, Australia, the Middle East, Europe and the Unites States. This review describes the molecular virology, epidemiology, pathogenesis, and highlights recent progress regarding diagnosis and vaccination against WNV infections.


Subject(s)
Birds , Mammals , Vaccination , Viral Vaccines/immunology , West Nile Fever , West Nile virus/immunology , Animals , Humans , Vaccination/veterinary , West Nile Fever/diagnosis , West Nile Fever/epidemiology , West Nile Fever/veterinary , West Nile Fever/virology , West Nile virus/classification , West Nile virus/genetics , West Nile virus/pathogenicity
9.
ACS Nano ; 6(3): 2136-49, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22303914

ABSTRACT

Recombinant antigens hold high potential to develop vaccines against lethal intracellular pathogens and cancer. However, they are poorly immunogenic and fail to induce potent cellular immunity. In this paper, we demonstrate that polymeric multilayer capsules (PMLC) strongly increase antigen delivery toward professional antigen-presenting cells in vivo, including dendritic cells (DCs), macrophages, and B cells, thereby enforcing antigen presentation and stimulating T cell proliferation. A thorough analysis of the T cell response demonstrated their capacity to induce IFN-γ secreting CD4 and CD8 T cells, in addition to follicular T-helper cells, a recently identified CD4 T cell subset supporting antibody responses. On the B cell level, PMLC-mediated antigen delivery promoted the formation of germinal centers, resulting in increased numbers of antibody-secreting plasma cells and elevated antibody titers. The functional relevance of the induced immune responses was validated in murine models of influenza and melanoma. On a mechanistic level, we have demonstrated the capacity of PMLC to activate the NALP3 inflammasome and trigger the release of the potent pro-inflammatory cytokine IL-1ß. Finally, using DC-depleted mice, we have identified DCs as the key mediators of the immunogenic properties of PMLC.


Subject(s)
Drug Carriers/chemistry , Influenza, Human/immunology , Influenza, Human/prevention & control , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Polymers/chemistry , Vaccination/methods , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Biocompatible Materials/chemistry , Capsules , Dextran Sulfate/chemistry , Humans , Immune System/cytology , Immune System/drug effects , Immune System/immunology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Inflammasomes/metabolism , Mice , Peptides/chemistry
10.
Vector Borne Zoonotic Dis ; 11(10): 1371-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21919722

ABSTRACT

Tick-borne encephalitis virus (TBEV) is an important emerging tick-borne viral infection of humans and dogs in Europe. Currently, TBEV surveillance is virtually nonexistent in Belgium, which is considered nonendemic. A commercial enzyme-linked immunosorbent assay (ELISA) was adapted for the detection of TBEV-specific IgG-antibodies in canine sera. Serum samples of Belgian dogs were obtained from three diagnostic laboratories from Northern (n=688) and Southern Belgium (n=192). ELISA-positive and borderline samples were subjected to a TBEV rapid fluorescent focus inhibition confirmation test. One dog was confirmed TBEV seropositive. Several ELISA-positive and borderline sera underwent seroneutralization and hemagglutinin inhibition tests to rule out West Nile and Louping Ill viruses, but tested negative. The clinical history of the seropositive dog could not explain beyond doubt where and when TBEV infection was acquired. Further surveillance is necessary to determine whether this dog remains a single travel-related case or whether it represents an early warning of a possible future emergence of TBEV.


Subject(s)
Antibodies, Viral/blood , Dog Diseases/epidemiology , Encephalitis Viruses, Tick-Borne/isolation & purification , Encephalitis, Tick-Borne/epidemiology , Animals , Belgium/epidemiology , Dog Diseases/immunology , Dog Diseases/virology , Dogs , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/virology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Neutralization Tests , Public Health , Reagent Kits, Diagnostic , Sentinel Surveillance
11.
J Infect Dis ; 203(8): 1063-72, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21450996

ABSTRACT

Influenza A virus infections impose a recurrent and global disease burden. Current antivirals against influenza are not always effective. We assessed the protective potential of monovalent and bivalent Nanobodies (Ablynx) against challenge with this virus. These Nanobodies were derived from llamas and target H5N1 hemagglutinin. Intranasal administration of Nanobodies effectively controlled homologous influenza A virus replication. Administration of Nanobodies before challenge strongly reduced H5N1 virus replication in the lungs and protected mice from morbidity and mortality after a lethal challenge with H5N1 virus. The bivalent Nanobody was at least 60-fold more effective than the monovalent Nanobody in controlling virus replication. In addition, Nanobody therapy after challenge strongly reduced viral replication and significantly delayed time to death. Epitope mapping revealed that the VHH Nanobody binds to antigenic site B in H5 hemagglutinin. Because Nanobodies are small, stable, and simple to produce, they are a promising, novel therapeutic agent against influenza.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibody Specificity , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Nanocapsules , Protein Conformation , Specific Pathogen-Free Organisms , Time Factors
12.
J Gen Virol ; 92(Pt 2): 301-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20965983

ABSTRACT

The recent emergence and rapid spread of the pandemic H1N1 swine influenza virus reminded us once again of the need for a universal influenza vaccine that can elicit heterosubtypic protection. Here, we show the superior immunogenicity and immunoprotective capacity of the full-length matrix protein 2 ectodomain (M2e) peptide coupled to keyhole limpet haemocyanin (KLH) compared with the N-terminal 9 aa residues of M2e (SP1). Immunization with M2e-KLH protected mice against a lethal challenge with influenza A virus and significantly reduced weight loss and lung virus titres. In addition, passive transfer of serum raised in rabbits against M2e-KLH protected mice against a lethal influenza virus challenge, whereas serum from rabbits immunized with SP1-KLH did not. Nevertheless, immunofluorescence staining revealed that rabbit serum raised against SP1-KLH bound specifically to infected Madin-Darby canine kidney cells. We conclude that the peptide SP1 contains an immunogenic epitope that is not sufficient for immunoprotection.


Subject(s)
Immune Sera/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Viral Matrix Proteins/classification , Viral Matrix Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Cell Line , Dogs , Gene Expression Regulation, Viral , Hemocyanins , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza Vaccines/immunology , Mice , Mice, Inbred BALB C , Rabbits , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
13.
J Immunol ; 186(2): 1022-31, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21169548

ABSTRACT

The ectodomain of matrix protein 2 (M2e) of influenza A virus is an attractive target for a universal influenza A vaccine: the M2e sequence is highly conserved across influenza virus subtypes, and induced humoral anti-M2e immunity protects against a lethal influenza virus challenge in animal models. Clinical phase I studies with M2e vaccine candidates have been completed. However, the in vivo mechanism of immune protection induced by M2e-carrier vaccination is unclear. Using passive immunization experiments in wild-type, FcRγ(-/-), FcγRI(-/-), FcγRIII(-/-), and (FcγRI, FcγRIII)(-/-) mice, we report in this study that Fc receptors are essential for anti-M2e IgG-mediated immune protection. M2e-specific IgG1 isotype Abs are shown to require functional FcγRIII for in vivo immune protection but other anti-M2e IgG isotypes can rescue FcγRIII(-/-) mice from a lethal challenge. Using a conditional cell depletion protocol, we also demonstrate that alveolar macrophages (AM) play a crucial role in humoral M2e-specific immune protection. Additionally, we show that adoptive transfer of wild-type AM into (FcγRI, FcγRIII)(-/-) mice restores protection by passively transferred anti-M2e IgG. We conclude that AM and Fc receptor-dependent elimination of influenza A virus-infected cells are essential for protection by anti-M2e IgG.


Subject(s)
Immunoglobulin G/metabolism , Influenza A virus/immunology , Influenza Vaccines/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/prevention & control , Protein Interaction Domains and Motifs/immunology , Receptors, Fc/physiology , Viral Matrix Proteins/immunology , Animals , Cell Death/genetics , Cell Death/immunology , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Immunization, Passive , Immunoglobulin G/toxicity , Influenza A virus/genetics , Influenza Vaccines/genetics , Influenza Vaccines/therapeutic use , Lymphocyte Depletion/methods , Macrophages, Alveolar/pathology , Macrophages, Alveolar/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Protein Interaction Domains and Motifs/genetics , Receptors, Fc/deficiency , Receptors, Fc/therapeutic use , Receptors, IgG/deficiency , Receptors, IgG/metabolism , Receptors, IgG/physiology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/therapeutic use
14.
Vaccine ; 27(45): 6280-3, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19840661

ABSTRACT

Human influenza causes substantial morbidity and mortality. Currently, licensed influenza vaccines offer satisfactory protection if they match the infecting strain, but they come with significant drawbacks. These vaccines are derived from prototype viruses, containing the hemagglutinin of influenza viruses that are likely to cause the next epidemic. Their usefulness against a future pandemic, however, remains problematic. A vaccine based on the ectodomain of influenza matrix protein 2 (M2e) could overcome these drawbacks. M2e is highly conserved in both human and avian influenza A viruses. The low immunogenicity against natural M2e can be overcome by fusing M2e to an appropriate carrier such as Hepatitis B virus-derived virus-like particles. Such chimeric particles can be produced in a simple and safe bacterial expression system, requiring minimal biocontainment, and can be obtained in a pure form. Experiments in animal models have demonstrated that M2e-based vaccines induce protection against a lethal challenge with various influenza A virus subtypes. Furthermore, the production and use of an effective M2e-vaccine could be implemented at any time regardless of seasonality, both in an epidemic as well as in a pandemic preparedness program. In animal models, M2e-vaccines administered parenterally or intranasally protect against disease and mortality following challenge with various influenza A strains. Adjuvants suitable for human use improve protection, which correlates with higher anti-M2e antibody responses of defined subtypes. Recently, Phase I clinical studies with M2e-vaccines have been completed, indicating their safety and immunogenicity. Further clinical development of this universal influenza A vaccine candidate is being pursued in order to validate its protective efficacy in humans.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , Viral Matrix Proteins/immunology , Clinical Trials as Topic , Humans
15.
Expert Rev Vaccines ; 8(4): 499-508, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19348565

ABSTRACT

Influenza vaccines used today are strain specific and need to be adapted every year to try and match the antigenicity of the virus strains that are predicted to cause the next epidemic. The strain specificity of the next pandemic is unpredictable. An attractive alternative approach would be to use a vaccine that matches multiple influenza virus strains, including multiple subtypes. In this review, we focus on the development and clinical potential of a vaccine that is based on the conserved ectodomain of matrix protein 2 (M2) of influenza A virus. Since 1999, a number of studies have demonstrated protection against influenza A virus challenge in animal models using chemical or genetic M2 external domain (M2e) fusion constructs. More recently, Phase I clinical studies have been conducted with M2e vaccine candidates, demonstrating their safety and immunogenicity in humans. Ultimately, and possibly in the near future, efficacy studies in humans should provide proof that this novel vaccine concept can mitigate epidemic and even pandemic influenza A virus infections.


Subject(s)
Influenza Vaccines/immunology , Viral Matrix Proteins/immunology , Animals , Clinical Trials as Topic , Humans , Influenza A virus/genetics , Influenza A virus/immunology , Influenza Vaccines/genetics , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Matrix Proteins/genetics
16.
Vaccine ; 26(51): 6503-7, 2008 Dec 02.
Article in English | MEDLINE | ID: mdl-18835315

ABSTRACT

The extracellular domain of influenza A virus matrix protein 2 (M2e) is strongly conserved. Therefore, vaccines based on M2e can induce broad-spectrum immunity against influenza. We have mainly used recombinant virus-like particles derived from Hepatitis B virus core (HBc) as carrier for efficacious presentation of the M2e antigen. Here, we address whether pre-existing HBc-specific immunity interferes with the protective immune response obtained by M2e-HBc vaccination. Anti-HBc antibodies were induced by immunizing mice with unsubstituted HBc virus-like particles in the presence of two different adjuvants. We demonstrate that pre-existing HBc-specific antibodies affect neither the induction of M2e-specific antibody responses to vaccination with M2e-HBc particles, nor the protective efficacy of the resulting response. These results suggest that vaccination with M2e-HBc can induce protective anti-M2e antibodies even in anti-HBc positive individuals. The implications of these findings are discussed in the context of the clinical development of an M2e-based universal influenza vaccine, which recently successfully completed a Phase I trial.


Subject(s)
Antibodies, Viral/immunology , Hepatitis B Core Antigens/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Female , Influenza A virus/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Recombinant Fusion Proteins/immunology , Viral Load
17.
Cytokine ; 42(1): 62-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18334301

ABSTRACT

Inbred SPRET/Ei mice, derived from Mus spretus, were found to be extremely resistant to infection with a mouse adapted influenza A virus. The resistance was strongly linked to distal chromosome 16, where the interferon-inducible Mx1 gene is located. This gene encodes for the Mx1 protein which stimulates innate immunity to Orthomyxoviruses. The Mx1 gene is defective in most inbred mouse strains, but PCR revealed that SPRET/Ei carries a functional allele. The Mx1 proteins of M. spretus and A2G, the other major resistant strain derived from Mus musculus, share 95.7% identity. We were interested whether the sequence variations between the two Mx1 alleles have functional significance. To address this, we used congenic mouse strains containing the Mx1 gene from M. spretus or A2G in a C57BL/6 background. Using a highly pathogenic influenza virus strain, we found that the B6.spretus-Mx1 congenic mice were better protected against infection than the B6.A2G-Mx1 mice. This effect may be due to different Mx1 induction levels, as was shown by RT-PCR and Western blot. We conclude that SPRET/Ei is a novel Mx1-positive inbred strain useful to study the biology of Mx1.


Subject(s)
GTP-Binding Proteins/metabolism , Influenza A virus/metabolism , Mice, Congenic , Orthomyxoviridae Infections/metabolism , Animals , Base Sequence , Body Weight , Female , GTP-Binding Proteins/genetics , Genetic Linkage , Humans , Immunity, Innate , Lung/cytology , Lung/virology , Male , Mice , Molecular Sequence Data , Myxovirus Resistance Proteins , Sequence Alignment , Survival Rate
18.
J Biol Chem ; 283(17): 11382-7, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18252707

ABSTRACT

Matrix protein 2 (M2) of influenza A is a tetrameric type III membrane protein that functions as a proton-selective channel. The extracellular domain (M2e) has remained nearly invariable since the first human influenza strain was isolated in 1933. By linking a modified form of the leucine zipper of the yeast transcription factor GCN4 to M2e, we obtained a recombinant tetrameric protein, M2e-tGCN4. This protein mimics the quaternary structure of the ectodomain of the natural M2 protein. M2e-tGCN4 was purified, biochemically characterized, and used to immunize BALB/c mice. High M2e-specific serum IgG antibody titers were obtained following either intraperitoneal or intranasal administration. Immunized mice were protected fully against a potentially lethal influenza A virus challenge. Antibodies raised by M2e-tGCN4 immunization specifically bound to the surface of influenza-infected cells and to an M2-expressing cell line. Using a M2e peptide competition enzyme-linked immunosorbent assay with M2-expressing cells as target, we obtained evidence that M2e-tGCN4 induces antibodies that are specific for the native tetrameric M2 ectodomain. Therefore, fusion of an oligomerization domain to the extracellular part of a transmembrane protein allows it to mimic the natural quaternary structure and can promote the induction of oligomer-specific antibodies.


Subject(s)
Influenza Vaccines/chemistry , Viral Matrix Proteins/chemistry , Animals , Chromatography, Gel , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Immunoglobulin G/chemistry , Mass Spectrometry/methods , Mice , Mice, Inbred BALB C , Models, Biological , Molecular Conformation , Peptides/chemistry , Protein Structure, Tertiary
19.
Vaccine ; 24(44-46): 6597-601, 2006 Nov 10.
Article in English | MEDLINE | ID: mdl-16814430

ABSTRACT

M2 is the third integral membrane protein of influenza A. M2e, the extracellular, 23 amino acid residues of M2, has been remarkably conserved in all human influenza A strains. This prompted us to evaluate the use of M2e as a potential broad-spectrum immunogen in a mouse model for influenza infection. Genetic fusion of the M2e and hepatitis B virus core (HBc) coding sequences allowed us to obtain highly immunogenic virus-like particles. This M2e-HBc vaccine induced complete protection in mice against a lethal influenza challenge. Protective immunity was obtained regardless of the position of M2e in the M2e-HBc chimera at the amino-terminus or inserted in the immuno-dominant loop of the HBc protein. Increasing the copy number of M2e inserted at the N-terminus from one to three per monomer (240-720 per particle) significantly enhanced the immune response and reduced the number of vaccinations required for complete protection against a lethal challenge with influenza A virus. A series of M2e-HBc constructs was subsequently combined with CTA1-DD, a recombinant cholera toxin A1 derived mucosal adjuvant, to test its efficacy as an intranasally delivered vaccine. All hybrid VLPs tested with CTA1-DD completely protected mice from a potentially lethal infection and, in addition, significantly reduced morbidity. Overall, increased resistance to influenza challenge in the mice correlated with an enhanced Th1-type M2e-specific antibody response induced by vaccination. These results show that M2e is a valid and versatile vaccine candidate to protect against any strain of human influenza A.


Subject(s)
Administration, Intranasal , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Viral Matrix Proteins/immunology , Adjuvants, Immunologic/pharmacology , Animals , Cholera Toxin/pharmacology , Humans , Immunization , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Influenza, Human/immunology , Mice , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Viral Matrix Proteins/genetics
20.
Vaccine ; 24(5): 544-51, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16169634

ABSTRACT

Mucosal vaccination requires effective and safe adjuvants. We have evaluated the non-toxic adjuvant CTA1-DD for mucosal vaccination against influenza. CTA1-DD contains the enzymatically active CTA1 subunit of cholera toxin (CT) genetically fused to a gene encoding a dimer of the D-fragment from Staphylococcus aureus protein A. CTA1-DD only binds to Ig-receptor carrying cells of the immune system. Nasal administration of the universal influenza vaccine M2e-HBc in combination with CTA1-DD completely protected mice from a potentially lethal infection and significantly reduced morbidity. Sera of mice immunized with M2e-HBc + CTA1-DD revealed IgG subclass profiles consistent with an enhanced Th1-type immunity. When the vaccine was administered intraperitoneally, the adjuvant improved the M2e antibody titer in circulation, but did not significantly reduce the morbidity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cholera Toxin/pharmacology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Recombinant Fusion Proteins/pharmacology , Administration, Intranasal , Animals , Antibodies, Viral/analysis , Antibodies, Viral/biosynthesis , Female , Humans , Immunization , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Influenza, Human/immunology , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...