Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 23(8): 1183-1192, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35902637

RESUMO

Anti-programmed death-1 (anti-PD-1) immunotherapy reinvigorates CD8 T cell responses in patients with cancer but PD-1 is also expressed by other immune cells, including follicular helper CD4 T cells (Tfh) which are involved in germinal centre responses. Little is known, however, about the effects of anti-PD-1 immunotherapy on noncancer immune responses in humans. To investigate this question, we examined the impact of anti-PD-1 immunotherapy on the Tfh-B cell axis responding to unrelated viral antigens. Following influenza vaccination, a subset of adults receiving anti-PD-1 had more robust circulating Tfh responses than adults not receiving immunotherapy. PD-1 pathway blockade resulted in transcriptional signatures of increased cellular proliferation in circulating Tfh and responding B cells compared with controls. These latter observations suggest an underlying change in the Tfh-B cell and germinal centre axis in a subset of immunotherapy patients. Together, these results demonstrate dynamic effects of anti-PD-1 therapy on influenza vaccine responses and highlight analytical vaccination as an approach that may reveal underlying immune predisposition to adverse events.


Assuntos
Vacinas contra Influenza , Adulto , Humanos , Imunidade Humoral , Estações do Ano , Linfócitos T Auxiliares-Indutores , Vacinação
2.
Antimicrob Agents Chemother ; 65(12): e0077221, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34543092

RESUMO

Antivirals are urgently needed to combat the global SARS-CoV-2/COVID-19 pandemic, supplement existing vaccine efforts, and target emerging SARS-CoV-2 variants of concern. Small molecules that interfere with binding of the viral spike receptor binding domain (RBD) to the host angiotensin-converting enzyme II (ACE2) receptor may be effective inhibitors of SARS-CoV-2 cell entry. Here, we screened 512 pure compounds derived from natural products using a high-throughput RBD/ACE2 binding assay and identified (-)-hopeaphenol, a resveratrol tetramer, in addition to vatalbinoside A and vaticanol B, as potent and selective inhibitors of RBD/ACE2 binding and viral entry. For example, (-)-hopeaphenol disrupted RBD/ACE2 binding with a 50% inhibitory concentration (IC50) of 0.11 µM, in contrast to an IC50 of 28.3 µM against the unrelated host ligand/receptor binding pair PD-1/PD-L1 (selectivity index, 257.3). When assessed against the USA-WA1/2020 variant, (-)-hopeaphenol also inhibited entry of a VSVΔG-GFP reporter pseudovirus expressing SARS-CoV-2 spike into ACE2-expressing Vero-E6 cells and in vitro replication of infectious virus in cytopathic effect and yield reduction assays (50% effective concentrations [EC50s], 10.2 to 23.4 µM) without cytotoxicity and approaching the activities of the control antiviral remdesivir (EC50s, 1.0 to 7.3 µM). Notably, (-)-hopeaphenol also inhibited two emerging variants of concern, B.1.1.7/Alpha and B.1.351/Beta in both viral and spike-containing pseudovirus assays with similar or improved activities over the USA-WA1/2020 variant. These results identify (-)-hopeaphenol and related stilbenoid analogues as potent and selective inhibitors of viral entry across multiple SARS-CoV-2 variants of concern.


Assuntos
COVID-19 , Estilbenos , Humanos , Pandemias , Fenóis , Ligação Proteica , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo
3.
Virol J ; 18(1): 242, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34876153

RESUMO

BACKGROUND: Chronic hepatitis B virus (HBV) infection (CHB) is a significant public health problem that could benefit from treatment with immunomodulators. Here we describe a set of therapeutic HBV vaccines that target the internal viral proteins. METHODS: Vaccines are delivered by chimpanzee adenovirus vectors (AdC) of serotype 6 (AdC6) and 7 (AdC7) used in prime only or prime-boost regimens. The HBV antigens are fused into an early T cell checkpoint inhibitor, herpes simplex virus (HSV) glycoprotein D (gD), which enhances and broadens vaccine-induced cluster of differentiation (CD8)+ T cell responses. RESULTS: Our results show that the vaccines are immunogenic in mice. They induce potent CD8+ T cell responses that recognize multiple epitopes. CD8+ T cell responses increase after a boost, although the breadth remains similar. In mice, which carry high sustained loads of HBV particles due to a hepatic infection with an adeno-associated virus (AAV)8 vector expressing the 1.3HBV genome, CD8+ T cell responses to the vaccines are attenuated with a marked shift in the CD8+ T cells' epitope recognition profile. CONCLUSIONS: Our data show that in different stains of mice including those that carry a human major histocompatibility complex (MHC) class I antigen HBV vaccines adjuvanted with a checkpoint inhibitor induce potent and broad HBV-specific CD8+ T cell responses and lower but still detectable CD4+ T cell responses. CD8+ T cell responses are reduced and their epitope specificity changes in mice that are chronically exposed to HBV antigens. Implications for the design of therapeutic HBV vaccines are discussed.


Assuntos
Hepatite B Crônica , Hepatite B , Animais , Linfócitos T CD8-Positivos , Epitopos de Linfócito T/genética , Vacinas contra Hepatite B , Vírus da Hepatite B/genética , Camundongos , Infecção Persistente
4.
Mol Ther ; 28(3): 771-783, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-31839483

RESUMO

Transfer of genes by adeno-associated virus (AAV) vectors is benefiting patients with particular genetic defects. Challenges remain by rejection of AAV-transduced cells, which may be caused by CD8+ T lymphocytes directed to AAV capsid antigens. Reducing the number of CpG motifs from the genome of AAV vectors reduces expansion of naive T cells directed against an epitope within the capsid. In contrast, AAV capsid-specific memory CD8+ T cells respond more vigorously to AAV vectors lacking CpG motifs than to those with CpG motifs presumably reflecting dampening of T cell expansion by cytokines from the innate immune system. Depending on the purification method, AAV vector preparations can contain substantial amounts of empty AAV particles that failed to package the genome. Others have used empty particles as decoys to AAV-neutralizing antibodies. We tested if empty AAV vectors given alone or mixed with genome-containing AAV vectors induce proliferation of naive or memory CD8+ T cells directed to an antigen within an AAV capsid. Naive CD8+ T cells failed to respond to empty AAV vectors, which in contrast induced expansion of AAV-specific memory CD8+ T cells.


Assuntos
Composição de Bases , Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Dependovirus/genética , Dependovirus/imunologia , Vetores Genéticos/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Proteínas do Capsídeo/química , Técnicas de Transferência de Genes , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Memória Imunológica , Ativação Linfocitária/imunologia , Camundongos , Motivos de Nucleotídeos , Transdução Genética
6.
Cell Immunol ; 342: 103722, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-29195742

RESUMO

Although gene transfer using adeno-associated virus (AAV) vectors has made tremendous progress in recent years, challenges remain due to vector-specific adaptive immune responses. Specifically, AAV-neutralizing antibodies reduce AAV-transduction rates, while CD8+ T cells directed to AAV capsid antigens cause rejection of AAV-transduced cells. This has been addressed clinically by excluding humans with pre-existing AAV-neutralizing antibodies from gene transfer trials or by using immunosuppression or reduced doses of vectors expressing improved transgene products to blunt or circumvent destructive T cell responses. Although these approaches have met with success for treatment of some diseases, most notably hemophilia B, they may not be suitable for others. Pre-clinical models are thus needed to test alternative options to sidestep pre-existing AAV-neutralizing antibodies, to prevent their induction following gene transfer and to block the detrimental effects of CD8+ T cells directed to AAV capsid antigens. This chapter describes some of the available, although not yet perfect, models that can assess immune responses to AAV gene transfer.


Assuntos
Dependovirus/imunologia , Vetores Genéticos/imunologia , Imunidade Adaptativa , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/imunologia , Quimera , Dependovirus/genética , Humanos , Modelos Animais
7.
Clin Infect Dis ; 67(3): 327-333, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-29471464

RESUMO

Background: Influenza vaccination aims to prevent infection by influenza virus and reduce associated morbidity and mortality; however, vaccine effectiveness (VE) can be modest, especially for subtype A(H3N2). Low VE has been attributed to mismatches between the vaccine and circulating influenza strains and to the vaccine's elicitation of protective immunity in only a subset of the population. The low H3N2 VE in the 2012-2013 season was attributed to egg-adaptive mutations that created antigenic mismatch between the actual vaccine strain (IVR-165) and both the intended vaccine strain (A/Victoria/361/2011) and the predominant circulating strains (clades 3C.2 and 3C.3). Methods: We investigated the basis of low VE in 2012-2013 by determining whether vaccinated and unvaccinated individuals were infected by different viral strains and by assessing the serologic responses to IVR-165, A/Victoria/361/2011, and 3C.2 and 3C.3 strains in an adult cohort before and after vaccination. Results: We found no significant genetic differences between the strains that infected vaccinated and unvaccinated individuals. Vaccination increased titers to A/Victoria/361/2011 and 3C.2 and 3C.3 representative strains as much as to IVR-165. These results are consistent with the hypothesis that vaccination boosted cross-reactive immune responses instead of specific responses against unique vaccine epitopes. Only approximately one-third of the cohort achieved a ≥4-fold increase in titer. Conclusions: In contrast to analyses based on ferret studies, low H3N2 VE in 2012-2013 in adults does not appear to be due to egg adaptation of the vaccine strain. Instead, low VE might have been caused by low vaccine immunogenicity in a subset of the population.


Assuntos
Imunogenicidade da Vacina , Vírus da Influenza A Subtipo H3N2/genética , Vacinas contra Influenza/imunologia , Influenza Humana/epidemiologia , Adaptação Fisiológica , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos Virais/imunologia , Estudos de Coortes , Reações Cruzadas , Ovos/virologia , Furões , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Vírus da Influenza A Subtipo H3N2/isolamento & purificação , Vacinas contra Influenza/uso terapêutico , Influenza Humana/prevenção & controle , Mutação , Filogenia , Estações do Ano
8.
Cancer Immunol Immunother ; 67(10): 1533-1544, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30051333

RESUMO

Human immunotherapy with checkpoint blockades has achieved significant breakthroughs in recent years. In this study, a checkpoint blockade vaccine for canine melanoma was tested for safety and immunogenicity. Five healthy adult dogs received a mixture of three replication-defective chimpanzee-derived adenoviral vectors, one expressing mouse fibroblast-associated protein (mFAP) and the others expressing canine melanoma-associated antigens Trp-1 or Trp-2 fused into Herpes Simplex-1 glycoprotein D, a checkpoint inhibitor of herpes virus entry mediator (HVEM) pathways. The vaccine mixture was shown to be well tolerated and increased frequencies of canineTrp-1-specific activated CD8+ and CD4+ T cells secreting interferon-(IFN)-γ, tumor necrosis factor (TNF)-α, or interleukin (IL)-2 alone or in combinations in four and five out of five dogs, respectively. To avoid excessive bleeds, responses to cTrp-2 were not analyzed. All dogs responded with increased frequencies of mFAP-specific activated CD8+ and CD4+ T cells. The results of this safety/immunogenicity trial invite further testing of this checkpoint blockade vaccine combination in dogs with melanoma.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Imunoterapia , Melanoma/terapia , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Cães , Endopeptidases , Gelatinases/genética , Interferon gama/metabolismo , Interleucina-2/metabolismo , Masculino , Melanoma/genética , Melanoma/imunologia , Proteínas de Membrana/genética , Camundongos , Oxirredutases/genética , Serina Endopeptidases/genética
9.
Immun Ageing ; 15: 19, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30186359

RESUMO

BACKGROUND: The elderly patient population is the most susceptible to influenza virus infection and its associated complications. Polypharmacy is common in the aged, who often have multiple co-morbidities. Previous studies have demonstrated that commonly used prescription drugs can have extensive impact on immune defenses and responses to vaccination. In this study, we examined how the dynamics of immune responses to the two influenza A virus strains of the trivalent inactivated influenza vaccine (TIV) can be affected by patient's history of using the prescription drugs Metformin, NSAIDs or Statins. RESULTS: We provide evidence for differential antibody (Ab) production, B-cell phenotypic changes, alteration in immune cell proportions and transcriptome-wide perturbation in individuals with a history of long-term medication use, compared with non-users. We noted a diminished response to TIV in the elderly on Metformin, whereas those on NSAIDs or Statins had higher baseline responses but reduced relative increases in virus-neutralizing Abs (VNAs) or Abs detected by an enzyme-linked immunosorbent assay (ELISA) following vaccination. CONCLUSION: Collectively, our findings suggest novel pathways that might underlie how long-term medication use impacts immune response to influenza vaccination in the elderly. They provide a strong rationale for targeting the medication-immunity interaction in the aged population to improve vaccination responses.

10.
Proc Natl Acad Sci U S A ; 112(2): 518-23, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25550504

RESUMO

An effective T-cell-based AIDS vaccine should induce strong HIV-specific CD8(+) T cells in mucosal tissues without increasing the availability of target cells for the virus. Here, we evaluated five immunization strategies that include Human adenovirus-5 (AdHu5), Chimpanzee adenovirus-6 (AdC6) or -7 (AdC7), Vaccinia virus (VV), and DNA given by electroporation (DNA/EP), all expressing Simian immunodeficiency virus group specific antigen/transactivator of transcription (SIV(mac239Gag/Tat)). Five groups of six rhesus macaques (RMs) each were vaccinated with DNA/EP-AdC6-AdC7, VV-AdC6-AdC7, DNA/-EP-VV-AdC6, DNA/EP-VV-AdC7, or AdHu5-AdHu5-AdHu5 and were challenged repeatedly with low-dose intrarectal SIVmac239. Upon challenge, there were no significant differences among study groups in terms of virus acquisition or viral load after infection. When taken together, the immunization regimens did not protect against SIV acquisition compared with controls but did result in an ∼ 1.6-log decline in set-point viremia. Although all immunized RMs had detectable SIV-specific CD8(+) T cells in blood and rectal mucosa, we found no correlation between the number or function of these SIV-specific CD8(+) T cells and protection against SIV acquisition. Interestingly, RMs experiencing breakthrough infection showed significantly higher prechallenge levels of CD4(+)C-C chemokine receptor type 5 (CCR5)(+)HLA-DR(+) T cells in the rectal biopsies (RB) than animals that remained uninfected. In addition, among the infected RMs, the percentage of CD4(+)CCR5(+)Ki-67(+) T cells in RBs prechallenge correlated with higher early viremia. Overall, these data suggest that the levels of activated CD4(+)CCR5(+) target T cells in the rectal mucosa may predict the risk of SIV acquisition in RMs vaccinated with vectors that express SIVGag/Tat.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Vacinas contra a SAIDS/imunologia , Vírus da Imunodeficiência Símia/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Produtos do Gene gag/imunologia , Produtos do Gene tat/imunologia , Humanos , Imunidade Celular , Imunidade nas Mucosas , Ativação Linfocitária , Macaca mulatta/imunologia , Macaca mulatta/virologia , Receptores CCR5/metabolismo , Reto/imunologia , Reto/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/patogenicidade , Vacinação/métodos , Viremia/imunologia , Viremia/prevenção & controle
11.
J Virol ; 89(15): 7841-51, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25995265

RESUMO

UNLABELLED: Influenza infection causes severe disease and death in humans. In traditional vaccine research and development, a single high-dose virus challenge of animals is used to evaluate vaccine efficacy. This type of challenge model may have limitations. In the present study, we developed a novel challenge model by infecting mice repeatedly in short intervals with low doses of influenza A virus. Our results show that compared to a single high-dose infection, mice that received repeated low-dose challenges showed earlier morbidity and mortality and more severe disease. They developed higher vial loads, more severe lung pathology, and greater inflammatory responses and generated only limited influenza A virus-specific B and T cell responses. A commercial trivalent influenza vaccine protected mice against a single high and lethal dose of influenza A virus but was ineffective against repeated low-dose virus challenges. Overall, our data show that the repeated low-dose influenza A virus infection mouse model is more stringent and may thus be more suitable to select for highly efficacious influenza vaccines. IMPORTANCE: Influenza epidemics and pandemics pose serious threats to public health. Animal models are crucial for evaluating the efficacy of influenza vaccines. Traditional models based on a single high-dose virus challenge may have limitations. Here, we describe a new mouse model based on repeated low-dose influenza A virus challenges given within a short period. Repeated low-dose challenges caused more severe disease in mice, associated with higher viral loads and increased lung inflammation and reduced influenza A virus-specific B and T cell responses. A commercial influenza vaccine that was shown to protect mice from high-dose challenge was ineffective against repeated low-dose challenges. Overall, our results show that the low-dose repeated-challenge model is more stringent and may therefore be better suited for preclinical vaccine efficacy studies.


Assuntos
Modelos Animais de Doenças , Vírus da Influenza A Subtipo H1N1/fisiologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Camundongos , Animais , Anticorpos Antivirais/imunologia , Embrião de Galinha , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Influenza Humana/imunologia , Influenza Humana/patologia , Influenza Humana/virologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos Endogâmicos C57BL
12.
J Immunol ; 193(4): 1836-46, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25024391

RESUMO

Two vaccines expressing CD4(+) and CD8(+) T cell epitopes of melanoma-associated Ags (MAAs) by a chimpanzee-derived replication-defective AdC68 vector were compared in a mouse model of melanoma. In one vaccine, termed AdC68-gDMelapoly, the epitopes were expressed as a fusion protein within HSV-1 glycoprotein D (gD), which blocks immunoinhibitory signaling through the herpes virus entry mediator pathway. The other vaccine, termed AdC68-Melapoly, expressed only the MAA epitopes. AdC68-gDMelapoly induced more potent MAA-specific CD8(+) T cell responses especially to the subdominant MAA epitopes. Upon prophylactic vaccination, mice that developed CD8(+) T cell responses to the two vaccines that were comparable in magnitude showed equal protection against tumor challenge. When mice were first challenged with tumor cells and then vaccinated results differed. In animals with comparable CD8(+) T cell responses, the AdC68-gDMelapoly vaccine was more efficacious compared with the AdC68-Melapoly vaccine in delaying tumor growth. This effect was linked to reduced expression of 2B4, LAG-3, and programmed death-1 on tumor-infiltrating MAA-specific CD8(+) T cells elicited by the gD-adjuvanted vaccine, suggesting that CD8(+) T cells induced in presence of gD are less susceptible to tumor-driven exhaustion.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Melanoma/terapia , Proteínas do Envelope Viral/imunologia , Adjuvantes Imunológicos , Animais , Antígenos CD/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Quimioterapia Adjuvante , Epitopos de Linfócito T/imunologia , Feminino , Hemaglutininas Virais/genética , Hemaglutininas Virais/imunologia , Herpesvirus Humano 1/imunologia , Ativação Linfocitária/imunologia , Melanoma/imunologia , Melanoma/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/biossíntese , Receptores Imunológicos/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Família de Moléculas de Sinalização da Ativação Linfocitária , Proteínas do Envelope Viral/genética , Proteína do Gene 3 de Ativação de Linfócitos
13.
Blood ; 121(12): 2224-33, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23325831

RESUMO

Recent clinical trials have shown that evasion of CD8(+) T-cell responses against viral capsid is critical for successful liver-directed gene therapy with adeno-associated viral (AAV) vectors for hemophilia. Preclinical models to test whether use of alternate serotypes or capsid variants could avoid this deleterious response have been lacking. Here, the ability of CD8(+) T cells ("cap-CD8," specific for a capsid epitope presented by human B*0702 or murine H2-L(d) molecules) to target AAV-infected hepatocytes was investigated. In a murine model based on adoptive transfer of ex vivo expanded cap-CD8, AAV2-transduced livers showed CD8(+) T-cell infiltrates, transaminitis, significant reduction in factor IX transgene expression, and loss of transduced hepatocytes. AAV8 gene transfer resulted in prolonged susceptibility to cap-CD8, consistent with recent clinical findings. In contrast, using an AAV2(Y-F) mutant capsid, which is known to be less degraded by proteasomes, preserved transgene expression and largely avoided hepatotoxicity. In vitro assays confirmed reduced major histocompatibility complex class I presentation of this capsid and killing of human or murine hepatocytes compared with AAV2. In conclusion, AAV capsids can be engineered to substantially reduce the risk of destruction by cytotoxic T lymphocytes, whereas use of alternative serotypes per se does not circumvent this obstacle.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/imunologia , Dependovirus/fisiologia , Terapia Genética/métodos , Vetores Genéticos/fisiologia , Hepatócitos/imunologia , Transferência Adotiva/métodos , Animais , Linfócitos T CD8-Positivos/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Células Cultivadas , Dependovirus/genética , Dependovirus/imunologia , Dependovirus/metabolismo , Engenharia Genética , Vetores Genéticos/genética , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Transdução Genética
14.
Mol Ther ; 22(1): 42-51, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24077034

RESUMO

Self-complementary adeno-associated viral (AAV) vectors expressing human factor IX (hF.IX) have achieved transient or sustained correction of hemophilia B in human volunteers. High doses of AAV2 or AAV8 vectors delivered to the liver caused in several patients an increase in transaminases accompanied by a rise in AAV capsid-specific T cells and a decrease in circulating hF.IX levels suggesting immune-mediated destruction of vector-transduced cells. Kinetics of these adverse events differed in patients receiving AAV2 or AAV8 vectors causing rise in transaminases at 3 versus 8 weeks after vector injection, respectively. To test if CD8+ T cells to AAV8 vectors, which are similar to AAV2 vectors are fully-gutted vectors and thereby fail to encode structural viral proteins, could cause damage at this late time point, we tested in a series of mouse studies how long major histocompatibility (MHC) class I epitopes within AAV8 capsid can be presented to CD8+ T cells. Our results clearly show that depending on the vectors' genome, CD8+ T cells can detect such epitopes on AAV8's capsid for up to 6 months indicating that the capsid of AAV8 degrades slowly in mice.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Capsídeo/imunologia , Dependovirus/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Genoma , Animais , Proteínas do Capsídeo/imunologia , Dependovirus/genética , Epitopos de Linfócito T , Vetores Genéticos/normas , Humanos , Memória Imunológica , Ativação Linfocitária/imunologia , Masculino , Camundongos , Controle de Qualidade , Especificidade do Receptor de Antígeno de Linfócitos T , Transdução Genética
15.
Infect Immun ; 82(2): 793-807, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478093

RESUMO

Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antiprotozoários/sangue , Feminino , Imunoglobulina G/sangue , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Malária Vivax/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium vivax/genética , Poli I-C/administração & dosagem , Proteínas de Protozoários/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
16.
Retrovirology ; 11: 8, 2014 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-24444350

RESUMO

BACKGROUND: A key goal for HIV-1 envelope immunogen design is the induction of cross-reactive neutralizing antibodies (nAbs). As AIDS vaccine recipients will not be exposed to strains exactly matching any immunogens due to multiple HIV-1 quasispecies circulating in the human population worldwide, heterologous SHIV challenges are essential for realistic vaccine efficacy testing in primates. We assessed whether polyclonal IgG, isolated from rhesus monkeys (RMs) with high-titer nAbs (termed SHIVIG), could protect RMs against the R5-tropic tier-2 SHIV-2873Nip, which was heterologous to the viruses or HIV-1 envelopes that had elicited SHIVIG. RESULTS: SHIVIG demonstrated binding to HIV Gag, Tat, and Env of different clades and competed with the broadly neutralizing antibodies b12, VRC01, 4E10, and 17b. SHIVIG neutralized tier 1 and tier 2 viruses, including SHIV-2873Nip. NK-cell depletion decreased the neutralizing activity of SHIVIG 20-fold in PBMC assays. Although SHIVIG neutralized SHIV-2873Nip in vitro, this polyclonal IgG preparation failed to prevent acquisition after repeated intrarectal low-dose virus challenges, but at a dose of 400 mg/kg, it significantly lowered peak viremia (P = 0.001). Unexpectedly, single-genome analysis revealed a higher number of transmitted variants at the low dose of 25 mg/kg, implying increased acquisition at low SHIVIG levels. In vitro, SHIVIG demonstrated complement-mediated Ab-dependent enhancement of infection (C'-ADE) at concentrations similar to those observed in plasmas of RMs treated with 25 mg/kg of SHIVIG. CONCLUSION: Our primate model data suggest a dual role for polyclonal anti-HIV-1 Abs depending on plasma levels upon virus encounter.


Assuntos
Síndrome da Imunodeficiência Adquirida/prevenção & controle , Anticorpos Neutralizantes/administração & dosagem , Proteção Cruzada , Anticorpos Anti-HIV/administração & dosagem , HIV-1/imunologia , Imunização Passiva/métodos , Imunoglobulina G/administração & dosagem , Síndrome da Imunodeficiência Adquirida/virologia , Animais , Modelos Animais de Doenças , Macaca mulatta , Vírus da Imunodeficiência Símia/imunologia , Resultado do Tratamento
17.
J Virol ; 87(24): 13904-10, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24089556

RESUMO

Epstein-Barr virus (EBV) is a vaccine/immunotherapy target due to its association with several human malignancies. EBNA-1 is an EBV protein consistently expressed in all EBV-associated cancers. Herein, EBNA-1-specific T cell epitopes were evaluated after AdC-rhEBNA-1 immunizations in chronically lymphocryptovirus-infected rhesus macaques, an EBV infection model. Preexisting rhEBNA-1-specific responses were augmented in 4/12 animals, and new epitopes were recognized in 5/12 animals after vaccinations. This study demonstrated that EBNA-1-specific T cells can be expanded by vaccination.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/imunologia , Infecções por Herpesviridae/veterinária , Lymphocryptovirus/imunologia , Macaca mulatta , Doenças dos Primatas/imunologia , Linfócitos T/imunologia , Animais , Mapeamento de Epitopos , Epitopos de Linfócito T/química , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Antígenos Nucleares do Vírus Epstein-Barr/administração & dosagem , Antígenos Nucleares do Vírus Epstein-Barr/química , Antígenos Nucleares do Vírus Epstein-Barr/genética , Feminino , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Lymphocryptovirus/genética , Macaca mulatta/genética , Macaca mulatta/imunologia , Macaca mulatta/virologia , Masculino , Doenças dos Primatas/tratamento farmacológico , Doenças dos Primatas/virologia , Linfócitos T/virologia , Vacinação , Vacinas Virais/administração & dosagem , Vacinas Virais/química , Vacinas Virais/genética , Vacinas Virais/imunologia
18.
J Virol ; 87(17): 9420-30, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23804645

RESUMO

Human adenovirus (AdHu)-based candidate AIDS vaccine can provide protection from simian immunodeficiency virus (SIV) transmission and disease progression. However, their potential use may be limited by widespread preexisting immunity to the vector. In contrast, preexisting immunity to chimpanzee adenoviruses (AdC) is relatively rare. In this study, we utilized two regimens of prime-boost immunizations with AdC serotype SAd-V23 (also called AdC6) and SAd-V24 (also called AdC7) expressing SIV Gag/Tat to test their immunogenicity and ability to protect rhesus macaques (RMs) from a repeated low-dose SIVmac239 challenge. Both AdC6 followed by AdC7 (AdC6/7) and AdC7 followed by AdC6 (AdC7/6) induced robust SIV Gag/Tat-specific T cell responses as measured by tetramer staining and functional assays. However, no significant protection from SIV transmission was observed in either AdC7/6- or AdC7/6-vaccinated RMs. Interestingly, in the RMs showing breakthrough infections, AdC7/6-SIV immunization was associated with a transient but significant (P = 0.035 at day 90 and P = 0.033 at day 120 postinfection) reduction in the setpoint viral load compared to unvaccinated controls. None of the measured immunological markers (i.e., number or functionality of SIV-specific CD8(+) and CD4(+) T cell responses and level of activated and/or CCR5(+) CD4(+) target cells) at the time of challenge correlated with protection from SIV transmission in the AdC-SIV-vaccinated RMs. The robust immunogenicity observed in all AdC-immunized RMs and the transient signal of protection from SIV replication exhibited by AdC7/6-vaccinated RMs even in the absence of any envelope immunogen suggest that AdC-based vectors may represent a promising platform for candidate AIDS vaccines.


Assuntos
Adenovirus dos Símios/imunologia , Produtos do Gene gag/imunologia , Produtos do Gene tat/imunologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas contra a AIDS/genética , Vacinas contra a AIDS/imunologia , Adenovirus dos Símios/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Produtos do Gene gag/genética , Produtos do Gene tat/genética , Vetores Genéticos , Humanos , Imunização Secundária , Pan troglodytes/virologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Doenças Retais , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/genética , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral
19.
J Transl Med ; 12: 25, 2014 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-24460861

RESUMO

BACKGROUND: Self-complementary adeno-associated virus (scAAV) vectors have become a desirable vector for therapeutic gene transfer due to their ability to produce greater levels of transgene than single-stranded AAV (ssAAV). However, recent reports have suggested that scAAV vectors are more immunogenic than ssAAV. In this study, we investigated the effects of a self-complementary genome during gene therapy with a therapeutic protein, human factor IX (hF.IX). METHODS: Hemophilia B mice were injected intramuscularly with ss or scAAV1 vectors expressing hF.IX. The outcome of gene transfer was assessed, including transgene expression as well as antibody and CD8⁺ T cell responses to hF.IX. RESULTS: Self-complementary AAV1 vectors induced similar antibody responses (which eliminated systemic hF.IX expression) but stronger CD8⁺ T cell responses to hF.IX relative to ssAAV1 in mice with F9 gene deletion. As a result, hF.IX-expressing muscle fibers were effectively eliminated in scAAV-treated mice. In contrast, mice with F9 nonsense mutation (late stop codon) lacked antibody or T cell responses, thus showing long-term expression regardless of the vector genome. CONCLUSIONS: The nature of the AAV genome can impact the CD8⁺ T cell response to the therapeutic transgene product. In mice with endogenous hF.IX expression, however, this enhanced immunogenicity did not break tolerance to hF.IX, suggesting that the underlying mutation is a more important risk factor for transgene-specific immunity than the molecular form of the AAV genome.


Assuntos
Dependovirus/genética , Fator IX/genética , Fator IX/uso terapêutico , Terapia Genética , Vetores Genéticos/genética , Hemofilia B/terapia , Imunidade/genética , Animais , Anticorpos Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Capsídeo/imunologia , Códon sem Sentido/genética , Técnicas de Transferência de Genes , Genoma/genética , Hemofilia B/genética , Hemofilia B/imunologia , Humanos , Camundongos
20.
J Immunol ; 188(4): 1933-41, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22246631

RESUMO

Aging is associated with suboptimal CD8 T cell responses to viral infections. It is not clear whether these poor responses are due to environmental influences or quantitative and qualitative changes in the pool of responding CD8 T cells. Our studies demonstrated several deleterious age-related changes in the pool of Ag-specific CD8 T cells that respond to infection. The majority of CD8 T cells from uninfected aged mice was CD44(Hi) and had increased expression of inhibitory receptors including PD1, LAG3, 2B4, and CD160. These aged CD44(Hi) CD8 T cells were transcriptionally similar to exhausted CD8 T cells found during chronic infections. In addition, the number of virus-specific precursors in aged mice prior to infection was decreased up to 10-fold, and many of these Ag-specific precursors had high expression of CD44 and PD1. Finally, TCR transgenic studies demonstrated that the CD44(Hi) Ag-specific CD8 T cells from unimmunized aged and young mice were qualitatively inferior compared with CD44(Lo) CD8 T cells from aged or young donors. Thus, a decrease in precursor frequency as well as qualitative changes of CD8 T cells during aging are directly related to impaired immunity.


Assuntos
Envelhecimento/imunologia , Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Antígenos CD/biossíntese , Linfócitos T CD8-Positivos/metabolismo , Feminino , Proteínas Ligadas por GPI/biossíntese , Receptores de Hialuronatos/análise , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Receptor de Morte Celular Programada 1/biossíntese , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Imunológicos/biossíntese , Família de Moléculas de Sinalização da Ativação Linfocitária , Proteína do Gene 3 de Ativação de Linfócitos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa