Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
ACS Infect Dis ; 10(5): 1552-1560, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38623820

ABSTRACT

Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.


Subject(s)
Antibodies, Viral , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , Tyrosine , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Tyrosine/chemistry , Tyrosine/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Mice, Inbred BALB C , Influenza A Virus, H3N2 Subtype/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Lung/virology , Lung/immunology , Administration, Intranasal , Injections, Intramuscular , Cytokines , Cross Protection , Viroporin Proteins
2.
J Immunother ; 40(5): 155-163, 2017 06.
Article in English | MEDLINE | ID: mdl-28368960

ABSTRACT

We have previously shown that inhibition of the proteasome causes defective ribosomal products to be shunted into autophagosomes and subsequently released from tumor cells as defective ribosomal products in Blebs (DRibbles). These DRibbles serve as an excellent source of antigens for cross-priming of tumor-specific T cells. Here, we examine the role of ubiquitinated proteins (Ub-proteins) in this pathway. Using purified Ub-proteins from tumor cells that express endogenous tumor-associated antigen or exogenous viral antigen, we tested the ability of these proteins to stimulate antigen-specific T-cell responses, by activation of monocyte-derived dendritic cells generated from human peripheral blood mononuclear cells. Compared with total cell lysates, we found that purified Ub-proteins from both a gp100-specific melanoma cell line and from a lung cancer cell line expressing cytomegalovirus pp65 antigen produced a significantly higher level of IFN-γ in gp100- or pp65-specific T cells, respectively. In addition, Ub-proteins from an allogeneic tumor cell line could be used to stimulate tumor-infiltrating lymphocytes isolated and expanded from non-small cell lung cancer patients. These results establish that Ub-proteins provide a relevant source of antigens for cross-priming of antitumor immune responses in a variety of settings, including endogenous melanoma and exogenous viral antigen presentation, as well as antigen-specific tumor-infiltrating lymphocytes. Thus, ubiquitin can be used as an affinity tag to enrich for unknown tumor-specific antigens from tumor cell lysates to stimulate tumor-specific T cells ex vivo or to be used as vaccines to target short-lived proteins.


Subject(s)
Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic , Aluminum Oxide/immunology , Antigens, Neoplasm/immunology , Autophagy , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cross-Priming , Humans , Interferon-gamma/metabolism , Lung Neoplasms/therapy , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma/therapy , Phosphoproteins/immunology , Ribosomes/immunology , T-Lymphocytes/transplantation , Ubiquitinated Proteins/immunology , Viral Matrix Proteins/immunology , gp100 Melanoma Antigen/immunology
3.
Expert Rev Vaccines ; 16(2): 123-136, 2017 02.
Article in English | MEDLINE | ID: mdl-27653543

ABSTRACT

INTRODUCTION: Current influenza vaccines can prevent disease caused by influenza viruses but require annual administration and almost yearly reformulation. An attractive alternative approach would be to use a vaccine that provides broad and, ideally, lifelong protection against all influenza A and B virus strains. The extracellular domain of matrix protein 2 (M2e) of influenza A viruses is conserved and thus fits well in such a broadly protective vaccine. Areas covered: Recent advances in M2e vaccine design, the mode of action of M2e-based immunity and clinical progress of M2-based influenza vaccines. Expert commentary: Many M2e vaccine have been successfully tested for efficacy against a panel of divergent influenza viruses in animal models. More recently, clinical studies have been conducted with M2e vaccine candidates, which demonstrated their safety and immunogenicity in humans. Efficacy studies in humans are still needed to provide evidence that an M2e-based vaccine can protect against human influenza.


Subject(s)
Influenza Vaccines/immunology , Viral Matrix Proteins/immunology , Animals , Clinical Trials as Topic , Disease Models, Animal , Drug Discovery/trends , Drug Evaluation, Preclinical , Humans , Influenza Vaccines/adverse effects , Influenza Vaccines/genetics , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Matrix Proteins/genetics
4.
PLoS One ; 10(3): e0117203, 2015.
Article in English | MEDLINE | ID: mdl-25756283

ABSTRACT

Nanoparticles are increasingly used to adjuvant vaccine formulations due to their biocompatibility, ease of manufacture and the opportunity to tailor their size, shape, and physicochemical properties. The efficacy of similarly-sized silica (Si-OH), poly (D,L-lactic-co-glycolic acid) (PLGA) and poly caprolactone (PCL) nanoparticles (nps) to adjuvant recombinant capsomere presenting antigenic M2e modular peptide from Influenza A virus (CapM2e) was investigated in vivo. Formulation of CapM2e with Si-OH or PLGA nps significantly boosted the immunogenicity of modular capsomeres, even though CapM2e was not actively attached to the nanoparticles prior to injection (i.e., formulation was by simple mixing). In contrast, PCL nps showed no significant adjuvant effect using this simple-mixing approach. The immune response induced by CapM2e alone or formulated with nps was antibody-biased with very high antigen-specific antibody titer and less than 20 cells per million splenocytes secreting interferon gamma. Modification of silica nanoparticle surface properties through amine functionalization and pegylation did not lead to significant changes in immune response. This study confirms that simple mixing-based formulation can lead to effective adjuvanting of antigenic protein, though with antibody titer dependent on nanoparticle physicochemical properties.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Pharmaceutic/administration & dosage , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Nanoparticles/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Pharmaceutic/chemistry , Animals , Female , Hepatitis A Antibodies/metabolism , Influenza Vaccines/chemistry , Lactic Acid/chemistry , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Polyesters/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Silicon Dioxide/chemistry , Surface Properties , Viral Matrix Proteins/immunology
5.
Antiviral Res ; 111: 60-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25218949

ABSTRACT

Influenza virus is a global health concern due to its unpredictable pandemic potential. Frequent mutations of surface molecules, hemagglutinin (HA) and neuraminidase (NA), contribute to low efficacy of the annual flu vaccine and therapeutic resistance to standard antiviral agents. The populations at high risk of influenza virus infection, such as the elderly and infants, generally mount low immune responses to vaccines, and develop severe disease after infection. Novel therapeutics with high effectiveness and mutation resistance are needed. Previously, we described the generation of a fully human influenza virus matrix protein 2 (M2) specific monoclonal antibody (mAb), Z3G1, which recognized the majority of M2 variants from natural viral isolates, including highly pathogenic avian strains. Passive immunotherapy with Z3G1 significantly protected mice from the infection when administered either prophylactically or 1-2days post infection. In the present study, we showed that Z3G1 significantly protected mice from lethal infection when treatment was initiated 3days post infection. In addition, therapeutic administration of Z3G1 reduced lung viral titers in mice infected with different viral strains, including amantadine and oseltamivir-resistant strains. Furthermore, prophylactic and therapeutic administration of Z3G1 sustained O2 saturation and reduced lung pathology in monkeys infected with a pandemic H1N1 strain. Finally, de-fucosylated Z3G1 with an IgG1/IgG3 chimeric Fc region was generated (AccretaMab® Z3G1), and showed increased ADCC and CDC in vitro. Our data suggest that the anti-M2 mAb Z3G1 has great potential as a novel anti-flu therapeutic agent.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/administration & dosage , Immunization, Passive , Influenza A virus/drug effects , Influenza, Human/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Drug Evaluation, Preclinical , Female , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/physiology , Influenza A virus/physiology , Influenza, Human/immunology , Influenza, Human/virology , Macaca , Male , Mice , Mice, Inbred C57BL , Viral Matrix Proteins/immunology
6.
Vaccine ; 32(29): 3664-9, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24793947

ABSTRACT

Nanotechnology promises a revolution in medicine including through new vaccine approaches. The use of nanoparticles in vaccination has, to date, focused on attaching antigen directly to or within nanoparticle structures to enhance antigen uptake by immune cells. Here we question whether antigen incorporation with the nanoparticle is actually necessary to boost vaccine effectiveness. We show that the immunogenicity of a sub-unit protein antigen was significantly boosted by formulation with silica nanoparticles even without specific conjugation of antigen to the nanoparticle. We further show that this effect was observed only for virus-sized nanoparticles (50 nm) but not for larger (1,000 nm) particles, demonstrating a pronounced effect of nanoparticle size. This non-attachment approach has potential to radically simplify the development and application of nanoparticle-based formulations, leading to safer and simpler nanoparticle applications in vaccine development.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens/immunology , Influenza Vaccines/immunology , Nanoparticles/administration & dosage , Aluminum Hydroxide/administration & dosage , Animals , Antibodies, Viral/blood , Escherichia coli/metabolism , Genetic Vectors , Immunoglobulin G/blood , Mice, Inbred BALB C , Particle Size , Silicon Dioxide/administration & dosage , Viral Matrix Proteins/immunology
7.
Vet Microbiol ; 162(2-4): 623-630, 2013 Mar 23.
Article in English | MEDLINE | ID: mdl-23265240

ABSTRACT

Since 2003, the highly pathogenic avian influenza (HPAI) H5N1 has become a serious problem in animals and an increasing threat to public health. To develop effective vaccines for H5 HPAI in chickens, virus-like particles (VLP) were produced using a baculovirus expression system. The particles comprised hemagglutinin (HA) alone (HA-VLP) or HA in combination with a matrix protein (M1; HAM-VLP) derived from a recent clade 2.3.2.1 H5N1 HPAI virus. To compare the immunogenicity and protective efficacy of these VLPs, 10 µg HAM-VLP, the equivalent amounts of HA incorporated HA-VLP or whole inactivated virus (WIV), were emulsified with mineral oil and used to immunize chickens. The serum hemagglutination inhibition antibody levels induced by HA-VLP and HAM-VLP were comparable to WIV. Antibodies to nucleoprotein were detected only in the WIV group. Immunized chickens in each group survived and were protected against a lethal homologous virus challenge, showing no clinical signs of infection. The challenge virus was detected intermittently in some oropharyngeal swabs, but not in cloacal swabs or various organs, which means that VLPs and WIV provide protection against systemic but not local virus replication in chickens. After the challenge, the HA-VLP group showed significantly increased serum antibody levels compared to the HAM-VLP and WIV groups, and some chickens in the HA-VLP group seroconverted with respect to nucleoprotein. Taken together, these results suggest that VLPs may be an effective method for controlling HPAI in chickens. They could be applied to a differentiating infected from vaccinated animals (DIVA) strategy. In addition, it is likely that HAM-VLP is more efficacious than HA-VLP in chickens.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza in Birds/prevention & control , Vaccines, Virus-Like Particle/administration & dosage , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Baculoviridae/genetics , Baculoviridae/immunology , Chickens , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza in Birds/immunology , Influenza in Birds/virology , Sf9 Cells , Specific Pathogen-Free Organisms , Vaccination/veterinary , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology , Viral Matrix Proteins/genetics , Virion/genetics , Virion/immunology
8.
Vaccine ; 30(46): 6527-33, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22959982

ABSTRACT

Vaccination is the most effective means for preventing influenza-associated morbidity and mortality. Since the influenza virus mutates frequently, the virus strains for new vaccine production should be changed according to predicted epidemic strains. The extracellular domain of matrix protein 2 (M2e) is 24 amino acids long, which is highly conserved and therefore a good target for the development of a universal vaccine which may protect against a much wider range of influenza A virus strains. However its low antigenicity and immunogenicity, which are related to its small size, poses a big challenge for vaccine development. Multiple antigen peptide system (MAP) is based on an inert core molecule of radially branching lysine dendrites onto which a number of peptide antigens are anchored. Tuftsin is an immuno-stimulant molecule peptide. Here we developed a novel peptide vaccine by connecting a tuftsin to a branched, four-copy M2e. Not only did this increase the molecular mass, but also potentiate the immunogenicity. Two branched peptides, (M2e)4-tuftsin and (M2e)4-G4(tuftsin was replaced with four glycines), and a M2e monomer were synthesized using standard solid-phase methods. In vitro and in vivo studies were performed to compare their antigenicity and immunogenicity. Experiments in BALB/c mice demonstrated that the branched M2e could induce stronger humoral and cellular immune responses than the M2e monomer, and (M2e)4-tuftsin induced stronger humoral and cellular immune response than (M2e)4-G4. After lethal challenge with influenza virus PR8 strain, up to 80% of the animals in the (M2e)4-tuftsin vaccinated group still survived, in contrast to 44% in the (M2e)4-G4 group and 30% in the M2e monomer group. The combination of branched polypeptides and tuftsin in vaccine design is presented here for the first time, and the results show that the new construct is a promising candidate for a universal vaccine against the influenza A virus.


Subject(s)
Antigens, Viral/immunology , Influenza Vaccines/immunology , Tuftsin/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/chemistry , Disease Models, Animal , Female , Influenza Vaccines/administration & dosage , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Survival Analysis , Tuftsin/chemistry , Viral Matrix Proteins/chemistry
9.
Bing Du Xue Bao ; 26(3): 189-94, 2010 May.
Article in Chinese | MEDLINE | ID: mdl-20572339

ABSTRACT

M2 protein of type A influenza virus is a good candidate for universal influenza vaccine, exotoxin A of Pseudomonas aeruginosa may facilitate the immunogenicity of M2 protein. We constructed and expressed a prokaryotic expression plasmid containing a chimeric gene of M2 extracellular coding region and a partial PEA gene, and observed the immunoprotection in BALB/c mice vaccinated with the fusion protein. The fusion protein (ntPE-M2e) was generated by inserting the coding sequence of the M2e in place of Ib loop in PEA. This fusion protein was used to immunize BALB/c mice by subcutaneously injection with incomplete Freund's adjuvant and boost at weeks 3 and 7. The immunized mice were challenged with influenza virus strain A/PR/34/8. The fusion protein (ntPE-M2e) immunization protected mice against lethal viral challenge. ELISA and ELISPOT results demonstrated that the fusion protein could induce a strong systemic immune response against synthetic M2e peptide, and virus replication in the lungs of mice was inhibited in comparison with the control. This study provides foundation for developing broad-spectrum vaccines against type A influenza viruses.


Subject(s)
ADP Ribose Transferases/genetics , Bacterial Toxins/genetics , Exotoxins/genetics , Influenza A virus , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Virulence Factors/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Female , Gene Expression , Immunization , Influenza A virus/immunology , Influenza A virus/physiology , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Viral Matrix Proteins/biosynthesis , Viral Matrix Proteins/isolation & purification , Pseudomonas aeruginosa Exotoxin A
10.
Biol Chem ; 389(5): 551-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18953722

ABSTRACT

Human cytomegalovirus (HCMV) is contained by T-lymphocyte responses focused towards the major tegument protein pp65. To systematically identify T-cell epitopes, we applied the following strategy: 441 overlapping 15mer peptides spanning the entire HCMV pp65 antigen in 1-aa steps were screened in enzyme-linked immunospot (ELispot) assays for interferon gamma (IFN-gamma) secretion by peripheral blood mononuclear cells (PBMCs) from nine healthy HCMV-seropositive subjects expressing human leukocyte antigen (HLA)-A2. This analysis confirmed a number of previously known epitopes and revealed several new ones. A total of 26 epitopes were identified, including 14 HLA-A2, four HLA-B7, -B35, -812 and -B44 restricted class I epitopes, six class II epitopes, and two epitopes of unknown restriction. Three novel HLA-A2 epitopes were confirmed using T2-cells, and one peptide for which only binding data had been published so far was verified. Two novel class II epitopes were confirmed by intracellular cytokine staining. Responses were usually oligoclonal against up to seven HLA-A2 epitopes, albeit with a few dominating epitopes. Clusters of overlapping epitopes (hot-spots) were identified. These and the newly identified T-cell epitopes may be of great value for epitope-based immunotherapeutic approaches, including peptide vaccines.


Subject(s)
Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Phosphoproteins/immunology , T-Lymphocytes/immunology , Viral Matrix Proteins/immunology , Amino Acid Sequence , Animals , Cytomegalovirus Vaccines/chemical synthesis , Drug Evaluation, Preclinical , Flow Cytometry , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Molecular Sequence Data , Monocytes/immunology , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology
12.
J Immunol Methods ; 334(1-2): 11-20, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18329663

ABSTRACT

Current flu vaccines are based on killed or attenuated virus vaccines that must be altered each year to include the hemagglutinin and neuraminidase genes from a strain of virus predicted to predominate in the coming year. A vaccine that could protect against multiple strains of influenza A and B would be a major asset in the fight against flu-related mortality and morbidity. To support development of such a vaccine, we have developed a Flu Multiplex Assay based on a Luminex platform to assess serum antibody levels to two conserved peptides derived from influenza A (M2 protein) and influenza B (hemagglutinin protein). The peptides were synthesized with a biotin label and subsequently coupled to two different LumAvidin microspheres. We then tested various sera against both types of peptide in the multiplex assay format. The data show that sera from Rhesus macaques immunized with a single peptide react only with the homologous peptide while Rhesus macaques immunized with both peptides respond well to both peptides. Additionally, we were able to specifically compete reactivity to both peptides. We have tested serial bleeds from 100 pediatric patients at ages ranging from 16 to 56 weeks as well as single bleeds from over 100 healthy adults. No overall trend in titer relative to pediatric age was detected. Both demographics exhibited a minimal response to either the A/M2 or B/HA0 peptides. However, the average titer for the pediatric serum samples was significantly lower than that found in the adult population. The adult population exhibited a higher prevalence of low reactive samples. Assay reagents and parameters have been optimized and the assay is shown to be repeatable and robust. The assay will be used to support clinical vaccine trials of a bivalent peptide vaccine.


Subject(s)
Antibodies, Viral/blood , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunoassay/methods , Influenza A virus/immunology , Influenza B virus/immunology , Influenza Vaccines/immunology , Viral Matrix Proteins/immunology , Adolescent , Adult , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Female , Humans , Infant , Macaca mulatta , Male , Peptides/immunology , Reference Standards , Sensitivity and Specificity
13.
Emerg Infect Dis ; 13(3): 426-35, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17552096

ABSTRACT

Changes in influenza viruses require regular reformulation of strain-specific influenza vaccines. Vaccines based on conserved antigens provide broader protection. Influenza matrix protein 2 (M2) is highly conserved across influenza A subtypes. To evaluate its efficacy as a vaccine candidate, we vaccinated mice with M2 peptide of a widely shared consensus sequence. This vaccination induced antibodies that cross-reacted with divergent M2 peptide from an H5N1 subtype. A DNA vaccine expressing full-length consensus-sequence M2 (M2-DNA) induced M2-specific antibody responses and protected against challenge with lethal influenza. Mice primed with M2-DNA and then boosted with recombinant adenovirus expressing M2 (M2-Ad) had enhanced antibody responses that crossreacted with human and avian M2 sequences and produced T-cell responses. This M2 prime-boost vaccination conferred broad protection against challenge with lethal influenza A, including an H5N1 strain. Vaccination with M2, with key sequences represented, may provide broad protection against influenza A.


Subject(s)
Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Ion Channels/immunology , Orthomyxoviridae Infections/prevention & control , Vaccination , Viral Matrix Proteins/immunology , Adenoviridae/metabolism , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cross Reactions/immunology , Drug Evaluation, Preclinical , Female , Genes, Viral , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Immunization Schedule , Influenza Vaccines/immunology , Injections, Intramuscular , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Orthomyxoviridae Infections/blood , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Sequence Alignment , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
15.
Hum Immunol ; 65(5): 565-70, 2004 May.
Article in English | MEDLINE | ID: mdl-15172458

ABSTRACT

Cytomegalovirus (CMV) is responsible for significant morbidity and mortality in immunocompromised patients undergoing allogeneic hematopoietic stem cell transplantation. The limitations of antiviral drugs and a better understanding of the cellular immune response to CMV has lead to the development of alternative therapies that restore host cellular immunity to CMV. Infusion of donor T lymphocytes results in variable protection against CMV but a high incidence of graft-versus-host disease in the allogeneic setting. To prevent this complication and further improve anti-CMV immune response, several groups have developed new approaches, such as the introduction of a suicide gene to control alloreactivity against the host or the selective activation of CMV-specific T cells by antigen-presenting cells expressing CMV antigens introduced by gene transfer. Depending on the target cells and the strategy chosen, adenovirus, retrovirus or poxviruses derived vectors are used for gene transfer. The protocols as well as the preclinical and clinical results obtained in the field of anti-CMV immunotherapy using gene transfer are reported and discussed.


Subject(s)
Cytomegalovirus Infections/therapy , Genetic Therapy/methods , Immunotherapy/methods , T-Lymphocytes/immunology , Antigen-Presenting Cells/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/prevention & control , Dendritic Cells/immunology , Genes, Transgenic, Suicide/immunology , Genetic Vectors/genetics , Graft vs Host Disease/prevention & control , Graft vs Host Disease/therapy , Herpesvirus 4, Human/genetics , Humans , Immunocompromised Host/immunology , Leukocytes, Mononuclear/immunology , Phosphoproteins/genetics , Phosphoproteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Thymidine Kinase/genetics , Transfection , Vaccination , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
16.
J Mol Biol ; 335(1): 193-207, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14659750

ABSTRACT

The expression of intracellular antibodies (intrabodies) in eukaryotic cells has provided a powerful tool to manipulate microbial and cellular signaling pathways in a highly precise manner. However, there have been several technical issues that have restricted their more widespread use. In particular, single-chain antibodies (sFv) have been reported to fold poorly in the reducing environment of the cytoplasm and as such there has been a reluctance to use sFv-phage libraries as a source of intrabodies unless a pre-selection step to identify these rare sFvs from natural libraries or libraries of engineering sFvs that could fold properly in the absence of disulfide bonds were used. Here, we investigated whether target specific sFvs that are isolated from a 15 billion member non-immune human sFv-phage display library could be directly screened in pools as intrabodies without prior knowledge of their individual identity or purity within pools of antigen-specific sFvs. As the target, we used a synthetic transformation effector site 1 (TES1) polypeptide comprising the membrane-most proximal 34 amino acid residues of the carboxy-terminal cytoplasmic tail of the oncogenic latent membrane protein 1 (LMP1) of Epstein Barr virus, which serves as a docking site for adapter proteins of the tumor necrosis factor (TNF) receptor (TNFR)-associated factor (TRAF) family. Anti-TES1 sFvs, initially identified by phage ELISA screens, were grouped into pools according to the absorbance reading of the antigen-specific phage ELISA assays and then transferred as pools into eukaryotic expression vectors and expressed as cytoplasmic intrabodies. Using the pooling strategy, there was no loss of individual anti-TES1 sFvs in the transfer from prokaryotic to eukaryotic expression vectors. In addition, the initial assignments into sFv pools based on phage ELISA readings allowed the segregation of individual anti-TES1 sFvs into discrete or minimally overlapping intrabody pools. Further assessment of the biological activity of the anti-TES1 intrabody pools demonstrated that they were all able to selectively block F-LMP1-induced NFkappaB activity that was mediated through the TES1-site and to bind LMP1 protein with high efficiency. This direct phage to intrabody screening (DPIS) strategy should allow investigators to bypass much of the in vitro sFv characterization that is often not predictive of in vivo intrabody function and provide a more efficient use of large native and synthetic sFv phage libraries already in existence to identify intrabodies that are active in vivo.


Subject(s)
Drug Evaluation, Preclinical/methods , Homeodomain Proteins , Immunoglobulin Fragments/immunology , NF-kappa B/antagonists & inhibitors , Peptide Library , Viral Matrix Proteins/immunology , Antibodies/immunology , Binding Sites/immunology , Cell Line , Cytoskeletal Proteins , Cytosol/immunology , DNA-Binding Proteins , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fragments/isolation & purification , RNA-Binding Proteins , Transcription Factors
17.
Blood ; 100(10): 3681-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12393676

ABSTRACT

Epitope vaccines have shown promise for inducing cellular immune responses in animal models of infectious disease. In cases where cellular immunity was augmented, peptide vaccines composed of covalently linked minimal cytotoxic T-lymphocyte (CTL) and T-helper (T(H)) epitopes generally showed the most efficacy. To address a clinical vaccine strategy for cytomegalovirus (CMV) in the context of HCT (hematopoietic cell transplantation), we observed that linking the synthetically derived pan-DR epitope peptide (PADRE) or one of several tetanus T(H) epitopes to the immunodominant human leukocyte antigen (HLA) A*0201-restricted CTL epitope from CMV-pp65 to create a fusion peptide caused robust cytotoxic cellular immune responses in HLA A*0201/K(b) transgenic mice. Significantly, the fusion peptides are immunogenic when administered in saline solution by either subcutaneous or intranasal routes. CpG-containing single-stranded DNA (ss-oligodeoxynucleotide [ODN]) added to the fusion peptides dramatically up-regulated immune recognition by either route. Notably, target cells that either expressed full-length pp65 protein from vaccinia viruses or were sensitized with the CTL epitope encoded in the vaccine were recognized by splenic effectors from immunized animals. Visualization of murine peptide-specific CTL by flow cytometry was accomplished using an HLA A*0201 tetramer complexed with the pp65(495-503) CTL epitope. T(H)-CTL epitope fusion peptides in combination with CpG ss-ODN represent a new strategy for parenteral or mucosal delivery of vaccines in a safe and effective manner that has applicability for control or prophylaxis of infectious disease, especially in situations such as vaccination of donors or recipients of HCT, where highly inflammatory adjuvants are not desired.


Subject(s)
Antigens, Viral/immunology , Cytomegalovirus Vaccines/administration & dosage , HLA Antigens/immunology , Peptides/immunology , Phosphoproteins/immunology , Viral Matrix Proteins/immunology , Animals , Antigens, Viral/metabolism , Cytomegalovirus/immunology , Cytomegalovirus Vaccines/chemistry , Cytomegalovirus Vaccines/immunology , Drug Carriers/administration & dosage , Drug Carriers/pharmacology , Drug Evaluation, Preclinical , Epitopes/administration & dosage , Epitopes/immunology , Epitopes, T-Lymphocyte/immunology , HLA Antigens/genetics , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Immunization , Malaria Vaccines/chemical synthesis , Malaria Vaccines/immunology , Mice , Mice, Transgenic , Oligodeoxyribonucleotides/immunology , Peptides/administration & dosage , Peptides/chemical synthesis , Phosphoproteins/chemical synthesis , Tetanus/immunology , Viral Matrix Proteins/chemical synthesis
18.
Cytokine ; 9(2): 73-82, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9071557

ABSTRACT

The capacity of adjuvants to activate Ag-presenting cells during the induction of the primary immune response is of critical importance for the development of protective immunity to a number of pathogens. In this context, interleukin 12 (IL-12) has a key role by controlling the differentiation of T helper cells and favouring the expansion of Th1 cells. The capacity of iscoms with influenza virus Ag (flu-iscoms) and iscom matrix with EBV gp340 Ag to induce IL-12 was analysed in mice. The flu-iscom drives the immune response towards a Th1 type subsequent to IL-12 induction as measured in the serum of H2b, H2d and H2k mice. The iscom presenting the Ag and adjuvant in the same particle was considerably more efficient than the formulation of matrix and Ag in separate particles. Inhibition experiments with mAb neutralizing IL-12, interferon gamma (IFN-gamma) or IL-4, the latter two cytokines representing the Th1 and Th2 type of responses, showed that iscoms induce a broader immune response than that involving IL-12. This was shown by the additional effect that IL-4 neutralization had on the immune response to iscoms. Anti-IL 12 reduced the specific total Ab as well as IgG1, IgG2a and IgG2b while anti-IL 4 influenced the response to iscom by decreasing IgG2a and increasing IgG1. Further, the neutralization experiments indicate that IL-12 has a broader effect than IFN-gamma on the Ab response by influencing the production of IgG1, IgG2a and IgG2b.


Subject(s)
Adjuvants, Immunologic , Antibody Formation , Interleukin-12/immunology , Plants, Medicinal/immunology , Animals , Antibodies, Monoclonal , Haplotypes , Mice , Mice, Inbred BALB C , Micelles , Neutralization Tests , T-Lymphocytes, Helper-Inducer/immunology , Viral Matrix Proteins/immunology
19.
Vaccine ; 5(4): 307-14, 1987 Dec.
Article in English | MEDLINE | ID: mdl-3124365

ABSTRACT

The influence of detergents on the immunogenic activity of the major outer membrane protein of Neisseria gonorrhoeae was investigated. Most detergents tested were found to enhance the immune response. This effect was synergistic with the adjuvant activity of AlPO4. The combination of detergent and AlPO4 showed a stronger adjuvant activity than Freund's complete adjuvant. The adjuvant effect was only observed with protein preparations with very low lipopolysaccharide content. The immunostimulating effect of detergents was also observed with meningococcal group C polysaccharide conjugated to a Haemophilus influenzae type b outer membrane protein and with the fusion protein of measles virus. The influence of some detergent parameters (critical micelle concentration, hydrophile-lipophile balance, charge) was investigated.


Subject(s)
Aluminum Compounds , Aluminum/pharmacology , Antibody Formation/drug effects , Bacterial Outer Membrane Proteins/immunology , Detergents/pharmacology , Phosphates/pharmacology , Surface-Active Agents/pharmacology , Viral Matrix Proteins/immunology , Animals , Freund's Adjuvant , Lipopolysaccharides , Mice , Neisseria gonorrhoeae , Polysaccharides
SELECTION OF CITATIONS
SEARCH DETAIL