Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Res Sq ; 2023 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-37790486

RESUMO

Lymphoplasmacytic lymphoma (LPL) is an incurable low-grade B-cell lymphoma of the bone marrow. Despite a cumulative risk of progression, there is no approved therapy for patients in the asymptomatic phase. We conducted a first-in-human clinical trial of a novel therapeutic DNA idiotype neoantigen vaccine in nine patients with asymptomatic LPL. Treatment was well tolerated with no dose limiting toxicities. One patient achieved a minor response, and all remaining patients experienced stable disease, with median time to disease progression of 61+ months. Direct interrogation of the tumor microenvironment by single-cell transcriptome analysis revealed an unexpected dichotomous antitumor response, with significantly reduced numbers of clonal tumor mature B-cells, tracked by their unique BCR, and downregulation of genes involved in signaling pathways critical for B-cell survival post-vaccine, but no change in clonal plasma cell subpopulations. Downregulation of HLA class II molecule expression suggested intrinsic resistance by tumor plasma cell subpopulations and cell-cell interaction analyses predicted paradoxical upregulation of IGF signaling post vaccine by plasma cell, but not mature B-cell subpopulations, suggesting a potential mechanism of acquired resistance. Vaccine therapy induced dynamic changes in bone marrow T-cells, including upregulation of signaling pathways involved in T-cell activation, expansion of T-cell clonotypes, increased T-cell clonal diversity, and functional tumor antigen-specific cytokine production, with little change in co-inhibitory pathways or Treg. Vaccine therapy also globally altered cell-cell communication networks across various bone marrow cell types and was associated with reduction of protumoral signaling by myeloid cells, principally non-classical monocytes. These results suggest that this prototype neoantigen vaccine favorably perturbed the tumor immune microenvironment, resulting in reduction of clonal tumor mature B-cell, but not plasma cell subpopulations. Future strategies to improve clinical efficacy may require combinations of neoantigen vaccines with agents which specifically target LPL plasma cell subpopulations, or enable blockade of IGF-1 signaling or myeloid cell checkpoints.

2.
Biomedicines ; 11(3)2023 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-36979775

RESUMO

Peripheral mononuclear blood cells (PBMCs) are the most widely used study materials for immunomonitoring and antigen-specific T-cell identification. However, limited patient PBMCs and low-frequency antigen-specific T cells remain as significant technical challenges. To address these limitations, we established a novel platform comprised of optimized HLA-matched immortalized B cells transfected with mRNA of a prototype viral or tumor antigen conjugated to MHC class-I trafficking domain protein (MITD) to increase the efficiency of epitope expression in antigen-presenting cells (APCs) essential to expanding antigen-specific T cells. When applied to CMV as a model, the IBMAM platform could successfully expand CMV-specific T cells from low-frequency CMV PBMCs from seropositive donors. Additionally, this platform can be applied to the validation of antigen specific TCRs. Together, compared to using APCs with synthesized peptides, this platform is an unlimited, highly efficient, and cost-effective resource in detecting and expanding antigen-specific T cells and validating antigen-specific TCRs.

3.
Vaccines (Basel) ; 11(2)2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36851268

RESUMO

Live attenuated influenza vaccines offer broader and longer-lasting protection in comparison to inactivated influenza vaccines. The neuraminidase (NA) surface glycoprotein of influenza A virus is essential for the release and spread of progeny viral particles from infected cells. In this study, we de novo synthesized the NA gene, in which 62% of codons were synonymously changed based on mammalian codon bias usage. The codon-reprogrammed NA (repNA) gene failed to be packaged into the viral genome, which was achievable with partial restoration of wild-type NA sequence nucleotides at the 3' and 5' termini. Among a series of rescued recombinant viruses, we selected 20/13repNA, which contained 20 and 13 nucleotides of wild-type NA at the 3' and 5' termini of repNA, respectively, and evaluated its potential as a live attenuated influenza vaccine. The 20/13repNA is highly attenuated in mice, and the calculated LD50 was about 10,000-fold higher than that of the wild-type (WT) virus. Intranasal inoculation of the 20/13repNA virus in mice induced viral-specific humoral, cell-mediated, and mucosal immune responses. Mice vaccinated with the 20/13repNA virus were protected from the lethal challenge of both homologous and heterologous viruses. This strategy may provide a new method for the development of live, attenuated influenza vaccines for a better and more rapid response to influenza threats.

4.
Blood Adv ; 7(6): 918-932, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36469551

RESUMO

B-cell activating factor receptor (BAFF-R) is a mature B-cell survival receptor, which is highly expressed in a wide variety of B-cell malignancies but with minimal expression in immature B cells. These properties make BAFF-R an attractive target for therapy of B-cell lymphomas. We generated a novel humanized anti BAFF-R monoclonal antibody (mAb) with high specificity and potent in vitro and in vivo activity against B-cell lymphomas and leukemias. The humanized variants of an original chimeric BAFF-R mAb retained BAFF-R binding affinity and antibody-dependent cellular cytotoxicity (ADCC) against a panel of human cell lines and primary lymphoma samples. Furthermore, 1 humanized BAFF-R mAb clone and its afucosylated version, glycoengineered to optimize the primary mechanism of action, prolonged survival of immunodeficient mice bearing human tumor cell lines or patient-derived lymphoma xenografts in 3 separate models, compared with controls. Finally, the tissue specificity of this humanized mAb was confirmed against a broad panel of normal human tissues. Taken together, we have identified a robust lead-candidate BAFF-R mAb for clinical development.


Assuntos
Linfoma de Células B , Linfoma , Humanos , Camundongos , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos B , Linfoma de Células B/tratamento farmacológico , Anticorpos Monoclonais Humanizados , Linfoma/tratamento farmacológico
5.
Polymers (Basel) ; 14(17)2022 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-36080594

RESUMO

Self-healing materials, especially self-healing polyurea/polyurethane, to replace traditional coating has been of increasing interest in the past decade. The frequency of regular maintenance work can also be reduced as the coating is capable of forming bonds at ruptured sites. This reduces the cost of maintenance and the risk involved in workers engaging in maintenance work. The extremely short curing time of polyurea coating could potentially outweigh the cost due to its short down time. With a high self-healing efficiency, self-healing polyurea could be the ultimate choice of protective coating. This report aims to find the optimum formulation for fabrication of polyurea with a high self-healing efficiency. This is conducted by changing the composition of the components chosen for formulation of polyurea. The choice of isocyanate and amine is varied to explore its impact on chain mobility and microphase separation, which are important factors affecting self-healing efficiency. A series of characterizations, including ATR-FTIR, DSC, optical microscope and mechanical tester, is used to analyze the factors affecting the self-healing efficiency of fabricated polyurea and to eventually determine the best formulation. The ideal formulation of toluene 2,4 diisocyanate-amine (TDI-P1000) polyurea managed to achieve a self-healing of 42%. Further studies could be done to include multiple healing mechanisms after different area of polyurea to boost its self-healing efficiency after repeated healing.

6.
Leukemia ; 36(4): 1015-1024, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35039637

RESUMO

Chimeric antigen receptor (CAR) T cells targeting CD19 mediate potent antitumor effects in B-cell malignancies including acute lymphoblastic leukemia (ALL), but antigen loss remains the major cause of treatment failure. To mitigate antigen escape and potentially improve the durability of remission, we developed a dual-targeting approach using an optimized, bispecific CAR construct that targets both CD19 and BAFF-R. CD19/BAFF-R dual CAR T cells exhibited antigen-specific cytokine release, degranulation, and cytotoxicity against both CD19-/- and BAFF-R-/- variant human ALL cells in vitro. Immunodeficient mice engrafted with mixed CD19-/- and BAFF-R-/- variant ALL cells and treated with a single dose of CD19/BAFF-R dual CAR T cells experienced complete eradication of both CD19-/- and BAFF-R-/- ALL variants, whereas mice treated with monospecific CD19 or BAFF-R CAR T cells succumbed to outgrowths of CD19-/BAFF-R+ or CD19+/BAFF-R- tumors, respectively. Further, CD19/BAFF-R dual CAR T cells showed prolonged in vivo persistence, raising the possibility that these cells may have the potential to promote durable remissions. Together, our data support clinical translation of BAFF-R/CD19 dual CAR T cells to treat ALL.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Antígenos Quiméricos , Animais , Antígenos CD19 , Humanos , Imunoterapia Adotiva , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos Quiméricos/genética , Linfócitos T
7.
Cancer Immunol Immunother ; 69(10): 2139-2145, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32451682

RESUMO

B-cell malignancies can potentially be cured by CD19 chimeric antigen receptor (CAR) T-cell therapy. Although clinical response rates can be up to 93% in acute lymphoblastic leukemia, treatment-related antigen loss and lack of therapeutic persistence contribute to disease relapse. These shortcomings of current CAR T-cell therapy indicate the need for biologically relevant target selection and for improving the efficacy and persistence of the CAR T cells, which we have addressed by developing a novel B-cell activating factor receptor (BAFF-R) CAR T-cell therapy with improved therapeutic persistence. BAFF-R is a B-cell survival receptor and highly expressed in B-cell malignancies. We developed a prototype CAR T cell that efficiently and specifically eliminated BAFF-R expressing human B-cell tumors in several xenogeneic mouse models, including models of CD19 antigen loss. We proceeded with translational development and validation of BAFF-R CAR T cells produced under current good manufacturing practices (cGMP). cGMP-grade BAFF-R CAR T cells underwent in vitro and in vivo validation in established models to confirm that the potency and efficacy of our original research modeling was replicated. Food and Drug Administration required release testing was performed to ensure our BAFF-R CAR T cells meet specifications for new drug products. Completing and exceeding these requirements, the data fully support the initiation of a first-in-human Phase 1 trial for BAFF-R-positive relapsed/refractory (r/r) B-ALL.


Assuntos
Antígenos CD19/imunologia , Receptor do Fator Ativador de Células B/antagonistas & inibidores , Receptor do Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Imunoterapia Adotiva/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Animais , Ensaios Clínicos Fase I como Assunto , Humanos , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia
8.
Sci Transl Med ; 11(511)2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31554741

RESUMO

CAR T cells targeting CD19 provide promising options for treatment of B cell malignancies. However, tumor relapse from antigen loss can limit efficacy. We developed humanized, second-generation CAR T cells against another B cell-specific marker, B cell activating factor receptor (BAFF-R), which demonstrated cytotoxicity against human lymphoma and acute lymphoblastic leukemia (ALL) lines. Adoptively transferred BAFF-R-CAR T cells eradicated 10-day preestablished tumor xenografts after a single treatment and retained efficacy against xenografts deficient in CD19 expression, including CD19-negative variants within a background of CD19-positive lymphoma cells. Four relapsed, primary ALLs with CD19 antigen loss obtained after CD19-directed therapy retained BAFF-R expression and activated BAFF-R-CAR, but not CD19-CAR, T cells. BAFF-R-CAR, but not CD19-CAR, T cells also demonstrated antitumor effects against an additional CD19 antigen loss primary patient-derived xenograft (PDX) in vivo. BAFF-R is amenable to CAR T cell therapy, and its targeting may prevent emergence of CD19 antigen loss variants.


Assuntos
Antígenos CD19/metabolismo , Receptor do Fator Ativador de Células B/metabolismo , Imunoterapia Adotiva , Leucemia de Células B/terapia , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Humanos , Leucemia de Células B/imunologia , Ativação Linfocitária/imunologia , Camundongos , Linfócitos T/imunologia
9.
Cancer Immunol Immunother ; 67(8): 1181-1195, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29855694

RESUMO

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells with an immune suppressive phenotype. They represent a critical component of the immune suppressive niche described in cancer, where they support immune escape and tumor progression through direct effects on both the innate and adaptive immune responses, largely by contributing to maintenance of a high oxidative stress environment. The number of MDSCs positively correlates with protumoral activity, and often diminishes the effectiveness of immunotherapies, which is particularly problematic with the emergence of personalized medicine. Approaches targeting MDSCs showed promising results in preclinical studies and are under active investigation in clinical trials in combination with various immune checkpoint inhibitors. In this review, we discuss MDSC targets and therapeutic approaches targeting MDSC that have the aim of enhancing the existing tumor therapies.


Assuntos
Antineoplásicos/uso terapêutico , Imunoterapia , Células Supressoras Mieloides/imunologia , Neoplasias/tratamento farmacológico , Microambiente Tumoral/imunologia , Animais , Humanos , Neoplasias/imunologia
10.
Mol Ther ; 26(3): 695-707, 2018 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-29433938

RESUMO

Growing evidence links the aggressiveness of non-Hodgkin's lymphoma, especially the activated B cell-like type diffuse large B cell lymphomas (ABC-DLBCLs) to Toll-like receptor 9 (TLR9)/MyD88 and STAT3 transcription factor signaling. Here, we describe a dual-function molecule consisting of a clinically relevant TLR9 agonist (CpG7909) and a STAT3 inhibitor in the form of a high-affinity decoy oligodeoxynucleotide (dODN). The CpG-STAT3dODN blocked STAT3 DNA binding and activity, thus reducing expression of downstream target genes, such as MYC and BCL2L1, in human and mouse lymphoma cells. We further demonstrated that injections (i.v.) of CpG-STAT3dODN inhibited growth of human OCI-Ly3 lymphoma in immunodeficient mice. Moreover, systemic CpG-STAT3dODN administration induced complete regression of the syngeneic A20 lymphoma, resulting in long-term survival of immunocompetent mice. Both TLR9 stimulation and concurrent STAT3 inhibition were critical for immune-mediated therapeutic effects, since neither CpG7909 alone nor CpG7909 co-injected with unconjugated STAT3dODN extended mouse survival. The CpG-STAT3dODN induced expression of genes critical to antigen-processing/presentation and Th1 cell activation while suppressing survival signaling. These effects resulted in the generation of lymphoma cell-specific CD8/CD4-dependent T cell immunity protecting mice from tumor rechallenge. Our results suggest that CpG-STAT3dODN as a systemic/local monotherapy or in combination with PD1 blockade can provide an opportunity for treating patients with B cell NHL.


Assuntos
Antineoplásicos/farmacologia , Linfoma de Células B/imunologia , Linfoma de Células B/metabolismo , Oligonucleotídeos/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Receptor Toll-Like 9/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Imunoterapia , Linfoma de Células B/patologia , Linfoma de Células B/terapia , Camundongos , Terapia de Alvo Molecular , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Transcrição Gênica , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Cancer Res ; 24(5): 1114-1123, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29180606

RESUMO

Purpose: mAbs such as anti-CD20 rituximab are proven therapies in B-cell malignancies, yet many patients develop resistance. Novel therapies against alternative targets are needed to circumvent resistance mechanisms. We sought to generate mAbs against human B-cell-activating factor receptor (BAFF-R/TNFRSF13C), which has not yet been targeted successfully for cancer therapy.Experimental Design: Novel mAbs were generated against BAFF-R, expressed as a natively folded cell surface immunogen on mouse fibroblast cells. Chimeric BAFF-R mAbs were developed and assessed for in vitro and in vivo monotherapy cytotoxicity. The chimeric mAbs were tested against human B-cell tumor lines, primary patient samples, and drug-resistant tumors.Results: Chimeric antibodies bound with high affinity to multiple human malignant B-cell lines and induced potent antibody-dependent cellular cytotoxicity (ADCC) against multiple subtypes of human lymphoma and leukemia, including primary tumors from patients who had relapsed after anti-CD20 therapy. Chimeric antibodies also induced ADCC against ibrutinib-resistant and rituximab-insensitive CD20-deficient variant lymphomas, respectively. Importantly, they demonstrated remarkable in vivo growth inhibition of drug-resistant tumor models in immunodeficient mice.Conclusions: Our method generated novel anti-BAFF-R antibody therapeutics with remarkable single-agent antitumor effects. We propose that these antibodies represent an effective new strategy for targeting and treating drug-resistant B-cell malignancies and warrant further development. Clin Cancer Res; 24(5); 1114-23. ©2017 AACR.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos Imunológicos/farmacologia , Receptor do Fator Ativador de Células B/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Linfoma de Células B/tratamento farmacológico , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos Imunológicos/uso terapêutico , Receptor do Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/imunologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/imunologia , Humanos , Hibridomas , Concentração Inibidora 50 , Linfoma de Células B/sangue , Linfoma de Células B/patologia , Camundongos , Camundongos Endogâmicos BALB C , Dobramento de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Oncoimmunology ; 4(3): e983961, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25949898

RESUMO

Tumor-associated myeloid cells undermine the therapeutic efficacy of cancer immunotherapy by their inhibitory properties on immune effector cells. Development of therapeutic agents to deplete suppressive myeloid cells in tumor microenvironment requires identification of cell-specific targets. A competitive phage display technique on live cells paves the way to discovery of such a target.

13.
Antiviral Res ; 107: 1-5, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24746458

RESUMO

We evaluated the immunogenicity of hemagglutinin (HA) in the context of inactivated H7N9/AH/1/13-PR8 whole-virion. At 4weeks after immunization with 15µg HA, mice produced hemagglutination inhibition (HI) titers of 1:192 and neutralizing antibodies of 1:317. Aluminum hydroxide (alum), or a booster immunization, or both increased HI to 1:768, 1:384, 1:896 and neutralizing antibodies to 1:1868, 1:2302, 1:10,000, respectively. Macaques generated HI of 1:190 or 1:360 and virus neutralizing titers of 1:280 or 1:658 at 3weeks after immunization with HA alone or with alum. Sera from immunized mice and macaques protected mice from infection of A/Anhui/1/2013 (H7N9), suggesting an H7N9 vaccine is immunologically feasible.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Subtipo H7N9 do Vírus da Influenza A/imunologia , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Modelos Animais de Doenças , Feminino , Testes de Inibição da Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunização/métodos , Imunização Passiva , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Macaca , Camundongos Endogâmicos BALB C , Testes de Neutralização , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle
14.
Nat Commun ; 4: 2369, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24022374

RESUMO

Preventing and treating influenza virus infection remain a challenge because of incomplete understanding of the host-pathogen interactions, limited therapeutics and lack of a universal vaccine. So far, methods for monitoring the course of infection with influenza virus in real time in living animals are lacking. Here we report the visualization of influenza viral infection in living mice using an engineered replication-competent influenza A virus carrying luciferase reporter gene. After intranasal inoculation, bioluminescence can be detected in the chest and nasopharyngeal passage of living mice. The intensity of bioluminescence in the chest correlates with the dosage of infection and the viral load in the lung. Bioluminescence in the chest of infected mice diminishes on antiviral treatment. This work provides a novel approach that enables real-time study of influenza virus infection and effects of antiviral therapeutics in living animals.


Assuntos
Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/virologia , Administração Intranasal , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Peso Corporal , Galinhas , Sistemas Computacionais , Cães , Feminino , Genes Reporter , Células HEK293 , Humanos , Imageamento Tridimensional , Vírus da Influenza A/patogenicidade , Luciferases/metabolismo , Medições Luminescentes , Pulmão/patologia , Pulmão/virologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/patologia , Replicação Viral
15.
PLoS One ; 8(6): e66276, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23824680

RESUMO

BACKGROUND: The outbreaks of emerging infectious diseases caused by pathogens such as SARS coronavirus, H5N1, H1N1, and recently H7N9 influenza viruses, have been associated with significant mortality and morbidity in humans. Neutralizing antibodies from individuals who have recovered from an infection confer therapeutic protection to others infected with the same pathogen. However, survivors may not always be available for providing plasma or for the cloning of monoclonal antibodies (mAbs). METHODOLOGY/PRINCIPAL FINDINGS: The genome and the immunoglobulin genes in rhesus macaques and humans are highly homologous; therefore, we investigated whether neutralizing mAbs that are highly homologous to those of humans (human-like) could be generated. Using the H5N1 influenza virus as a model, we first immunized rhesus macaques with recombinant adenoviruses carrying a synthetic gene encoding hemagglutinin (HA). Following screening an antibody phage display library derived from the B cells of immunized monkeys, we cloned selected macaque immunoglobulin heavy chain and light chain variable regions into the human IgG constant region, which generated human-macaque chimeric mAbs exhibiting over 97% homology to human antibodies. Selected mAbs demonstrated potent neutralizing activities against three clades (0, 1, 2) of the H5N1 influenza viruses. The in vivo protection experiments demonstrated that the mAbs effectively protected the mice even when administered up to 3 days after infection with H5N1 influenza virus. In particular, mAb 4E6 demonstrated sub-picomolar binding affinity to HA and superior in vivo protection efficacy without the loss of body weight and obvious lung damage. The analysis of the 4E6 escape mutants demonstrated that the 4E6 antibody bound to a conserved epitope region containing two amino acids on the globular head of HA. CONCLUSIONS/SIGNIFICANCE: Our study demonstrated the generation of neutralizing mAbs for potential application in humans in urgent preparedness against outbreaks of new influenza infections or other virulent infectious diseases.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Neutralizantes/biossíntese , Doenças Transmissíveis Emergentes/terapia , Surtos de Doenças , Influenza Humana/terapia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/imunologia , Humanos , Virus da Influenza A Subtipo H5N1/imunologia , Influenza Humana/epidemiologia , Influenza Humana/imunologia , Macaca mulatta , Camundongos
16.
Hum Vaccin Immunother ; 8(2): 252-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22426370

RESUMO

H5N1 influenza candidate vaccine viruses were developed using the "6+2" approach. The hemagglutinin (HA) and neuraminidase (NA) genes were derived from the popular H5N1 virus and the remaining six internal segments were derived from the A/Puerto Rico/8/34 strain (H1N1, PR8). However, some of these candidate strains have been reported to produce relatively low yields in vaccine manufacture. In this study, we found that the NA vRNA of the A/Vietnam/1194/2004 strain (H5N1, VN1194) was poorly packaged into recombinant viruses with a backbone of PR8 genes, which resulted in the formation of defective virions that did not include the NA vRNA in the genome. Using recombinant DNA techniques, we constructed a chimeric NA gene with the coding region of VN1194 NA flanked by the packaging signal sequence of the PR8 NA gene (41 bp form the 3' end of the vRNA and 67 bp from the 5' end). The packaging of the NA vRNA was restored to normal levels in the recombinant viruses containing the chimeric NA gene. Recombinant viruses containing the chimeric NA replicated much better in chicken embryonated eggs than viruses with the wild-type NA from VN1194. These findings suggest a novel strategy to improve in ovo growth of vaccine strains and to increase the number of vaccine doses available to save people if a pandemic were to occur.


Assuntos
Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Virus da Influenza A Subtipo H5N1/genética , Vacinas contra Influenza/genética , Vacinas contra Influenza/imunologia , Neuraminidase/genética , Proteínas Virais/genética , Animais , Linhagem Celular , Embrião de Galinha , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Virus da Influenza A Subtipo H5N1/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Neuraminidase/imunologia , RNA Viral/genética , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Proteínas Recombinantes de Fusão , Vacinas Sintéticas/imunologia , Proteínas Virais/imunologia , Montagem de Vírus , Cultura de Vírus , Replicação Viral
17.
Virus Res ; 155(1): 156-62, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20883733

RESUMO

Recent outbreaks of highly pathogenic avian influenza (HPAI) H5N1 viruses in poultry and their subsequent transmission to humans have highlighted an urgent need to develop preventive vaccines in the event of a pandemic. In this paper we constructed recombinant adenovirus (rAd)-vectored influenza vaccines expressing different forms of H5 hemagglutinin (HA) from the A/Vietnam/1194/04 (VN/1194/04) virus, a wild-type HA, a sequence codon-optimized HA and a transmembrane (TM) domain-truncated HA. Compared to the rAd vectors expressing the wild-type HA (rAd-04wtHA) and the TM-truncated form of HA (rAd-04optHA-dTM), the rAd vectored vaccine with the sequence codon-optimized HA (rAd-04optHA) showed a tendency to induce much higher hemagglutinin inhibition (HI) antibody titers in mice immunized with a prime-boost vaccine. Furthermore, administration of the rAd-04optHA vaccine to mice could elicit cross-reactive immune responses against the antigenically distinct HK/482/97 virus. Additionally, we constructed another vector containing the codon-optimized HA of the A/Hong Kong/482/97 (HK/482/97) virus. Administration of a bivalent immunization formulation including the rAd-04optHA and rAd-97optHA vaccines to mice induced a stronger immune response against HK/482/97 virus than the monovalent formulation. Taken together, these findings may have some implications for the development of rAd-vectored vaccines in the event of the pandemic spread of HPAI.


Assuntos
Adenoviridae/genética , Portadores de Fármacos , Vetores Genéticos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Animais , Anticorpos Antivirais/sangue , Códon , Reações Cruzadas , Feminino , Testes de Inibição da Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Imunização Secundária/métodos , Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Camundongos , Camundongos Endogâmicos BALB C , Deleção de Sequência
18.
J Virol ; 84(22): 12075-81, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20826692

RESUMO

Replication-competent influenza viruses carrying reporter genes are of great use for basic research, screening of antiviral drugs, and neutralizing of antibodies. In this study, two recombinant influenza A viruses with a neuraminidase (NA) segment harboring enhanced green fluorescent protein (EGFP) in the background of A/PR/8/34 (PR8) were generated. The viral RNA (vRNA)-specific packaging signals for NA were largely retained for efficient packaging. An "autocleave" 2A peptide sequence, which was inserted at the N terminus or the COOH terminus of NA to link with EGFP, enabled NA and EGFP to be expressed monocistronically. Further analysis demonstrated that both viruses, named rPR8-EGFP+NA and rPR8-NA+EGPF, although with some characteristic differences in growth and EGFP expression, could replicate in noncomplementary cells and propagate to large quantities while maintaining genome stability after multiple passages in embryonated eggs. These replication-competent influenza viruses carrying reporter genes are a great addition to the tool set for developing antiviral therapeutics and vaccines and for in vivo studies of viral dissemination and pathogenicity.


Assuntos
Genes Reporter , Engenharia Genética , Vírus da Influenza A/fisiologia , Neuraminidase/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular , Embrião de Galinha , Cães , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Vírus da Influenza A/enzimologia , Vírus da Influenza A/genética , Neuraminidase/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/genética , Montagem de Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...