Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Expert Rev Vaccines ; 15(5): 607-17, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26588466

RESUMO

T cells are extensively trained on 'self' in the thymus and then move to the periphery, where they seek out and destroy infections and regulate immune response to self-antigens. T cell receptors (TCRs) on T cells' surface recognize T cell epitopes, short linear strings of amino acids presented by antigen-presenting cells. Some of these epitopes activate T effectors, while others activate regulatory T cells. It was recently discovered that T cell epitopes that are highly conserved on their TCR face with human genome sequences are often associated with T cells that regulate immune response. These TCR-cross-conserved or 'redundant epitopes' are more common in proteins found in pathogens that have co-evolved with humans than in other non-commensal pathogens. Epitope redundancy might be the link between pathogens and autoimmune disease. This article reviews recently published data and addresses epitope redundancy, the "elephant in the room" for vaccine developers and T cell immunologists.


Assuntos
Antígenos/genética , Antígenos/metabolismo , Autoimunidade , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Vacinas/imunologia , Animais , Sequência Conservada , Modelos Animais de Doenças , Humanos , Timo/fisiologia
2.
Hum Vaccin Immunother ; 11(9): 2312-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26155959

RESUMO

Computational vaccine design, also known as computational vaccinology, encompasses epitope mapping, antigen selection and immunogen design using computational tools. The iVAX toolkit is an integrated set of tools that has been in development since 1998 by De Groot and Martin. It comprises a suite of immunoinformatics algorithms for triaging candidate antigens, selecting immunogenic and conserved T cell epitopes, eliminating regulatory T cell epitopes, and optimizing antigens for immunogenicity and protection against disease. iVAX has been applied to vaccine development programs for emerging infectious diseases, cancer antigens and biodefense targets. Several iVAX vaccine design projects have had success in pre-clinical studies in animal models and are progressing toward clinical studies. The toolkit now incorporates a range of immunoinformatics tools for infectious disease and cancer immunotherapy vaccine design. This article will provide a guide to the iVAX approach to computational vaccinology.


Assuntos
Antígenos/imunologia , Biologia Computacional/métodos , Descoberta de Drogas/métodos , Epitopos/imunologia , Vacinas Sintéticas/imunologia , Animais , Antígenos/genética , Epitopos/genética , Humanos , Vacinas Sintéticas/genética
3.
Hum Vaccin Immunother ; 11(9): 2241-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26090577

RESUMO

Avian-origin H7N9 influenza is a novel influenza A virus (IAV) that emerged in humans in China in 2013. Using immunoinformatics tools, we identified several H7N9 T cell epitopes with T cell receptor (TCR)-facing residues identical to those of multiple epitopes from human proteins. We hypothesized that host tolerance to these peptides may impair T helper response and contribute to the low titer, weak hemagglutination inhibiting (HI) antibody responses and diminished seroconversion rates that have been observed in human H7N9 infections and vaccine trials. We found that the magnitude of human T effector responses to individual H7N9 peptides was inversely correlated with the peptide's resemblance to self. Furthermore, a promiscuous T cell epitope from the hemagglutinin (HA) protein suppressed responses to other H7N9 peptides when co-administered in vitro. Along with other highly 'human-like' peptides from H7N9, this peptide was also shown to expand FoxP3(+) regulatory T cells (Tregs). Thus, H7N9 may be camouflaged from effective human immune response by T cell epitope sequences that avert or regulate effector T cell responses through host tolerance.


Assuntos
Epitopos de Linfócito T/imunologia , Tolerância Imunológica , Subtipo H7N9 do Vírus da Influenza A/imunologia , Linfócitos T Reguladores/imunologia , Células Cultivadas , Epitopos de Linfócito T/genética , Humanos , Subtipo H7N9 do Vírus da Influenza A/genética , Homologia de Sequência de Aminoácidos
4.
Immunol Cell Biol ; 93(2): 189-97, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385064

RESUMO

Complement fragment C3d covalently attached to antigens enhances immune responses, particularly for antigens lacking T-cell epitopes. Enhancement has been attributed to receptor cross-linking between complement receptor CR2 (CD21) and polysaccharide antigen to surface IgM on naïve B cells. Paradoxically, C3d has still been shown to increase immune responses in CD21 knockout mice, suggesting that an auxiliary activation pathway exists. In prior studies, we demonstrated the CD21-independent C3d adjuvant effect might be due to T-cell recognition of C3d T-helper epitopes processed and presented by major histocompatibility complex class II on the B-cell surface. C3d peptide sequences containing concentrated clusters of putative human C3 T-cell epitopes were identified using the epitope-mapping algorithm, EpiMatrix. These peptide sequences were synthesized and shown in vitro to bind multiple human leukocyte antigen (HLA)-DR alleles with high affinity, and induce interferon-γ responses in healthy donor peripheral blood mononuclear cells. In the present studies, we establish further correlations between HLA binding and HLA-specific lymphocyte reactions with select epitope clusters. In addition, we show that the T-cell phenotype of C3d-specific reactive T cells is CD4(+)CD45RO(+) memory T cells. Finally, mutation of a single T-cell epitope residing within the P28 peptide segment of C3d resulted in significantly diminished adjuvant activity in BALB/c mice. Collectively, these studies support the hypothesis that the paradoxical enhancement of immune responses by C3d in the absence of CD21 is due to internalization and processing of C3d into peptides that activate autoreactive CD4(+) T-helper cells in the context of HLA class II.


Assuntos
Adjuvantes Imunológicos/metabolismo , Complemento C3d/imunologia , Ativação Linfocitária , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Simulação por Computador , Epitopos de Linfócito T/química , Epitopos de Linfócito T/imunologia , Feminino , Marcação de Genes , Antígenos de Histocompatibilidade/imunologia , Humanos , Memória Imunológica/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação , Peptídeos/metabolismo , Ligação Proteica , Doadores de Tecidos
5.
Hum Vaccin Immunother ; 10(12): 3570-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483703

RESUMO

High strain sequence variability, interference with innate immune mechanisms, and epitope deletion are all examples of strategies that pathogens have evolved to subvert host defenses. To this list we would add another strategy: immune camouflage. Pathogens whose epitope sequences are cross-conserved with multiple human proteins at the TCR-facing residues may be exploiting "ignorance and tolerance," which are mechanisms by which mature T cells avoid immune responses to self-antigens. By adopting amino acid configurations that may be recognized by autologous regulatory T cells, pathogens may be actively suppressing protective immunity. Using the new JanusMatrix TCR-homology-mapping tool, we have identified several such 'camouflaged' tolerizing epitopes that are present in the viral genomes of pathogens such as emerging H7N9 influenza. Thus in addition to the overall low number of T helper epitopes that is present in H7 hemaglutinin (as described previously, see http://dx.doi.org/10.4161/hv.24939), the presence of such tolerizing epitopes in H7N9 could explain why, in recent vaccine trials, whole H7N9-HA was poorly immunogenic and associated with low seroconversion rates (see http://dx.doi.org/10.4161/hv.28135). In this commentary, we provide an overview of the immunoinformatics process leading to the discovery of tolerizing epitopes in pathogen genomic sequences, provide a brief summary of laboratory data that validates the discovery, and point the way forward. Removal of viral, bacterial and parasite tolerizing epitopes may permit researchers to develop more effective vaccines and immunotherapeutics in the future.


Assuntos
Evasão da Resposta Imune , Vacinas/imunologia , Reações Cruzadas , Epitopos de Linfócito T , Humanos , Tolerância Imunológica , Subtipo H7N9 do Vírus da Influenza A/imunologia , Receptores de Antígenos de Linfócitos T/fisiologia
6.
Front Microbiol ; 5: 502, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25339942

RESUMO

Despite years of research, vaccines against HIV and HCV are not yet available, due largely to effective viral immunoevasive mechanisms. A novel escape mechanism observed in viruses that cause chronic infection is suppression of viral-specific effector CD4(+) and CD8(+) T cells by stimulating regulatory T cells (Tregs) educated on host sequences during tolerance induction. Viral class II MHC epitopes that share a T cell receptor (TCR)-face with host epitopes may activate Tregs capable of suppressing protective responses. We designed an immunoinformatic algorithm, JanusMatrix, to identify such epitopes and discovered that among human-host viruses, chronic viruses appear more human-like than viruses that cause acute infection. Furthermore, an HCV epitope that activates Tregs in chronically infected patients, but not clearers, shares a TCR-face with numerous human sequences. To boost weak CD4(+) T cell responses associated with persistent infection, vaccines for HIV and HCV must circumvent potential Treg activation that can handicap efficacy. Epitope-driven approaches to vaccine design that involve careful consideration of the T cell subsets primed during immunization will advance HIV and HCV vaccine development.

7.
Vaccine ; 32(26): 3285-92, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24721533

RESUMO

Hepatitis C virus (HCV) is a major cause of liver disease. Spontaneous resolution of infection is associated with broad, MHC class I- (CD8(+)) and class II-restricted (CD4(+)) T cell responses to multiple viral epitopes. Only 20% of patients clear infection spontaneously, however, most develop chronic disease. The response to chemotherapy varies; therapeutic vaccination offers an additional treatment strategy. To date, therapeutic vaccines have demonstrated only limited success in clinical trials. Vector-mediated vaccination with multi-epitope-expressing DNA constructs provides an improved approach. Highly-conserved, HLA-A2-restricted HCV epitopes and HLA-DRB1-restricted immunogenic consensus sequences (ICS, each composed of multiple overlapping and highly conserved epitopes) were predicted using bioinformatics tools and synthesized as peptides. HLA binding activity was determined in competitive binding assays. Immunogenicity and the ability of each peptide to stimulate naïve human T cell recognition and IFN-γ production were assessed in cultures of total PBMCs and in co-cultures composed of peptide-pulsed dendritic cells (DCs) and purified T lymphocytes, cell populations derived from normal blood donors. Essentially all predicted HLA-A2-restricted epitopes and HLA-DRB1-restricted ICS exhibited HLA binding activity and the ability to elicit immune recognition and IFN-γ production by naïve human T cells. The ability of DCs pulsed with these highly-conserved HLA-A2- and -DRB1-restricted peptides to induce naïve human T cell reactivity and IFN-γ production ex vivo demonstrates the potential efficacy of a multi-epitope-based HCV vaccine targeted to dendritic cells.


Assuntos
Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Hepacivirus/imunologia , Linfócitos T/imunologia , Vacinas contra Hepatite Viral/imunologia , Sequência de Aminoácidos , Biologia Computacional , Mapeamento de Epitopos , Antígeno HLA-A2 , Cadeias HLA-DRB1 , Humanos , Interferon gama/imunologia , Peptídeos/imunologia
8.
Hum Vaccin Immunother ; 9(10): 2060-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24045788

RESUMO

The emergence of the pandemic H1N1 strain of influenza in 2009 was associated with a unique w-shaped age-related susceptibility curve, with higher incidence of morbidity and mortality among young persons and lower incidence among older persons, also observed during the 1918 influenza pandemic. Pre-existing H1N1 antibodies were not cross-reactive with the prior seasonal vaccine, forcing influenza experts to scramble to develop a new vaccine specific for the pandemic virus. We hypothesized that response to T-cell epitopes that are cross-conserved between pandemic H1N1 and the 2008 seasonal influenza vaccine strains might have contributed to partial protection from clinical illness among older adults, despite the lack of cross-reactive humoral immunity. Using immunoinformatics tools, we previously identified hemagglutinin and neuraminidase epitopes that were highly conserved between seasonal and pandemic H1N1. Here, we validated predicted CD4(+) T-cell epitopes for their ability to bind HLA and to stimulate interferon-γ production in peripheral blood mononuclear cells from a cohort of donors presenting with influenza-like illness during the 2009 pandemic and a separate cohort immunized with trivalent influenza vaccine in 2011. A limited-epitope heterologous DNA-prime/peptide-boost vaccine composed of these sequences stimulated immune responses and lowered lung viral loads in HLA DR3 transgenic mice challenged with pandemic 2009 H1N1 influenza. Cross-priming with conserved influenza T-cell epitopes such as these may be critically important to T cell-mediated protection against pandemic H1N1 in the absence of cross-protective antibodies.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Antígeno HLA-DR3/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Animais , Vacinas contra Influenza/administração & dosagem , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Transgênicos , Carga Viral
9.
Hum Vaccin Immunother ; 9(7): 1598-607, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23846304

RESUMO

Immune responses to cross-conserved T cell epitopes in novel H1N1 influenza may explain reports of diminished influenza-like illnesses and confirmed infection among older adults, in the absence of cross-reactive humoral immunity, during the 2009 pandemic. These cross-conserved epitopes may prove useful for the development of a universal H1N1 influenza vaccine, therefore, we set out to identify and characterize cross-conserved H1N1 T cell epitopes. An immunoinformatic analysis was conducted using all available pandemic and pre-pandemic HA-H1 and NA-N1 sequences dating back to 1980. Using an approach that balances potential for immunogenicity with conservation, we derived 13 HA and four NA immunogenic consensus sequences (ICS) from a comprehensive analysis of 5,738 HA-H1 and 5,396 NA-N1 sequences. These epitopes were selected because their combined epitope content is representative of greater than 84% of pre-pandemic and pandemic H1N1 influenza strains, their predicted immunogenicity (EpiMatrix) scores were greater than or equal to the 95th percentile of all comparable epitopes, and they were also predicted to be presented by more than four HLA class II archetypal alleles. We confirmed the ability of these peptides to bind in HLA binding assays and to stimulate interferon-γ production in human peripheral blood mononuclear cell cultures. These studies support the selection of the ICS as components of potential group-common H1N1 vaccine candidates and the application of this universal influenza vaccine development approach to other influenza subtypes.


Assuntos
Descoberta de Drogas , Epitopos de Linfócito T/imunologia , Hemaglutininas/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Neuraminidase/imunologia , Linfócitos T CD4-Positivos/imunologia , Reações Cruzadas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Interferon gama/imunologia , Leucócitos Mononucleares/imunologia , Vacinação
10.
R I Med J (2013) ; 96(2): 19-21, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23641421

RESUMO

EpiVax, Inc., is an early-stage informatics and immunology biotechnology company in Providence, Rhode Island. It applies computational tools to harness immunity in three major areas: immunomodulation, biotherapeutic immunogenicity risk assessment and de-risking, and vaccine development. Immunotherapy, bio-better and vaccine candidates under development at EpiVax promise to improve the health outcomes of millions of people affected by devastating immune-related diseases.


Assuntos
Doenças Autoimunes/imunologia , Pesquisa Biomédica/tendências , Biologia Computacional , Descoberta de Drogas/tendências , Indústria Farmacêutica , Epitopos Imunodominantes/imunologia , Imunomodulação , Linfócitos T Reguladores/imunologia , Doenças Autoimunes/tratamento farmacológico , Autoimunidade , Desenho de Fármacos , Humanos , Rhode Island , Vacinas
11.
Endocrinology ; 154(6): 2234-43, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23592747

RESUMO

Development of Graves' disease is related to HLA-DR3. The extracellular domain (ECD) of human TSH receptor (hTSH-R) is a crucial antigen in Graves' disease. hTSH-R peptide 37 (amino acids 78-94) is an important immunogenic peptide in DR3 transgenic mice immunized to hTSH-R. This study examined the epitope recognition in DR3 transgenic mice immunized to hTSH-R protein and evaluated the ability of a mutant hTSH-R peptide to attenuate the immunogenicity of hTSH-R peptide 37. DR3 transgenic mice were immunized to recombinant hTSH-R-ECD protein or peptides. A mutant hTSH-R 37 peptide (ISRIYVSIDATLSQLES: 37 m), in which DR3 binding motif position 5 was mutated V>A, and position 8 Q>S, was synthesized. 37 m should bind to HLA-DR3 but not bind T cell receptors. DR3 transgenic mice were immunized to hTSH-R 37 and 37 m. Mice immunized to hTSH-R-ECD protein developed strong anti-hTSH-R antibody, and antisera reacted strongly with hTSH-R peptides 1-5 (20-94), 21 (258-277), 41 (283-297), 36 (376-389), and 31 (399-418). Strikingly, antisera raised to hTSH-R peptide 37 bound to hTSH-R peptides 1-7 (20-112), 10 (132-50), 33 (137-150), 41, 23 (286-305), 24 (301-320), 36, and 31 as well as to hTSH-R-ECD protein. Both antibody titers to hTSH-R 37 and reaction of splenocytes to hTSH-R 37 were significantly reduced in mice immunized to hTSH-R 37 plus 37 m, compared with mice immunized to hTSH-R 37 alone. The ability of immunization to a single peptide to induce antibodies that bind hTSH-R-ECD protein, and multiple unrelated peptides, is a unique observation. Immunogenic reaction to hTSH-R peptide 37 was partially suppressed by 37 m, and this may contribute to immunotherapy of autoimmune thyroid disease.


Assuntos
Epitopos/imunologia , Antígeno HLA-DR3/imunologia , Fragmentos de Peptídeos/imunologia , Receptores da Tireotropina/imunologia , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Sítios de Ligação/genética , Ensaio de Imunoadsorção Enzimática , Epitopos/metabolismo , Feminino , Doença de Graves/imunologia , Doença de Graves/metabolismo , Doença de Graves/terapia , Antígeno HLA-DR3/genética , Antígeno HLA-DR3/metabolismo , Humanos , Soros Imunes/imunologia , Soros Imunes/metabolismo , Imunização , Masculino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores da Tireotropina/química , Receptores da Tireotropina/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Baço/citologia , Baço/imunologia , Baço/metabolismo
12.
Autoimmun Rev ; 12(3): 436-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22944299

RESUMO

In the course of screening immunoglobulin G (IgG) sequences for T cell epitopes, we identified novel Treg epitope peptides, now called Tregitopes, contained in the highly conserved framework regions of Fab and Fc. Tregitopes may provide one explanation for the expansion and stimulation of Treg cells following intravenous immunoglobulin (IVIg) therapy. Their distinguishing characteristics include in silico signatures that suggest high-affinity binding to multiple human HLA class II DR and conservation across IgG isotypes and mammalian species with only minor amino acid modifications. Tregitopes induce expansion of CD4(+)/CD25(hi)/FoxP3(+) T cells and suppress immune responses to co-incubated antigens in vitro. By comparing the human IgG Tregitopes (hTregitopes 167 and 289, located in the IgG CH1 and CH2 domains) and Fab to murine sequences, we identified class II-restricted murine Tregitope homologs (mTregitopes). In vivo, mTregitopes suppress inflammation and reproducibly induce Tregs to expand. In vitro studies suggest that the Tregitope mechanism of action is to induce Tregs to respond, leading to production of regulatory signals, followed by modulation of dendritic cell phenotype. The identification of Treg epitopes in IgG suggests that additional Tregitopes may also be present in other autologous proteins; methods for identifying and validating such peptides are described here. The discovery of Tregitopes in IgG and other autologous proteins may lead to the development of new insights as to the role of Tregs in autoimmune diseases.


Assuntos
Epitopos de Linfócito T/imunologia , Imunoglobulinas Intravenosas/imunologia , Linfócitos T Reguladores/imunologia , Animais , Mapeamento de Epitopos , Epitopos de Linfócito T/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulinas Intravenosas/metabolismo , Camundongos , Ligação Proteica
13.
Clin Immunol ; 142(3): 320-31, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22222093

RESUMO

T cell-dependent development of anti-Factor VIII (FVIII) antibodies that neutralize FVIII activity is a major obstacle to replacement therapy in hemophilia A. To create a less immunogenic therapeutic protein, recombinant FVIII can be modified to reduce HLA binding of epitopes based on predicted anchoring residues. Here, we used immunoinformatic tools to identify C2 domain HLA DR epitopes and predict site-specific mutations that reduce immunogenicity. Epitope peptides corresponding to original and modified sequences were validated in HLA binding assays and in immunizations of hemophilic E16 mice, DR3 and DR4 mice and DR3×E16 mice. Consistent with immunoinformatic predictions, original epitopes are immunogenic. Immunization with selected modified sequences lowered immunogenicity for particular peptides and revealed residual immunogenicity of incompletely de-immunized modified peptides. The stepwise approach to reduce protein immunogenicity by epitope modification illustrated here is being used to design and produce a functional full-length modified FVIII for clinical use.


Assuntos
Epitopos de Linfócito T/imunologia , Fator VIII/imunologia , Antígenos HLA-DR/imunologia , Alelos , Sequência de Aminoácidos , Animais , Células Cultivadas , Epitopos de Linfócito T/química , Fator VIII/genética , Antígenos HLA-DR/química , Imunização , Camundongos , Camundongos Knockout , Dados de Sequência Molecular
14.
Immunome Res ; 7(2)2011.
Artigo em Inglês | MEDLINE | ID: mdl-25152768

RESUMO

BACKGROUND: One useful application of pattern matching algorithms is identification of major histocompatability complex (MHC) ligands and T-cell epitopes. Peptides that bind to MHC molecules and interact with T cell receptors to stimulate the immune system are critical antigens for protection against infectious pathogens. We describe a genomes-to-vaccine approach to H. pylori vaccine design that takes advantage of immunoinformatics algorithms to rapidly identify T-cell epitope sequences from large genomic datasets. RESULTS: To design a globally relevant vaccine, we used computational methods to identify a core genome comprised of 676 open reading frames (ORFs) from amongst seven genetically and phenotypically diverse H. pylori strains from around the world. Of the 1,241,153 9-mer sequences encoded by these ORFs, 106,791 were identical amongst all seven genomes and 23,654 scored in the top 5% of predicted HLA ligands for at least one of eight archetypal Class II HLA alleles when evaluated by EpiMatrix. To maximize the number of epitopes that can be assessed experimentally, we used a computational algorithm to increase epitope density in 20-25 amino acid stretches by assembling potentially immunogenic 9-mers to be identically positioned as they are in the native protein antigen. 1,805 immunogenic consensus sequences (ICS) were generated. 79% of selected ICS epitopes bound to a panel of 6 HLA Class II haplotypes, representing >90% of the global human population. CONCLUSIONS: The breadth of H. pylori genome datasets was computationally assessed to rapidly and carefully determine a core set of genes. Application of immunoinformatics tools to this gene set accurately predicted epitopes with promising properties for T cell-based vaccine development.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...