Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 811
Filtrar
1.
J Immunother Cancer ; 10(2)2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35193929

RESUMEN

BACKGROUND: Oncolytic viruses are a potent form of active immunotherapy, capable of invoking antitumor T-cell responses. Meanwhile, less is known about their effects on immune checkpoints, the main targets for passive immunotherapy of cancer. T-cell immunoglobulin and mucin domain-3 (TIM-3) is a coinhibitory checkpoint driving T-cell exhaustion in cancer. Here we investigated the effects of oncolytic adenovirus on the TIM-3 checkpoint on tumor-infiltrating immune cells and clinical impact in patients with cancer receiving oncolytic immunotherapy. METHODS: Modulation of TIM-3 expression on tumor-infiltrating immune cells was studied preclinically in B16 melanoma following intratumoral treatment with Ad5/3∆24-granulocyte-macrophage colony-stimulating factor oncolytic adenovirus. We conducted a retrospective longitudinal analysis of 15 patients with advanced-stage cancer with tumor-site biopsies before and after oncolytic immunotherapy, treated in the Advanced Therapy Access Program (ISRCTN10141600, April 5, 2011). Following patient stratification with regard to TIM-3 (increase vs decrease in tumors), overall survival and imaging/marker responses were evaluated by log-rank and Fisher's test, while coinhibitory receptors/ligands, transcriptomic changes and tumor-reactive and tumor-infltrating immune cells in biopsies and blood samples were studied by microarray rank-based statistics and immunoassays. RESULTS: Preclinically, TIM-3+ tumor-infiltrating lymphocytes (TILs) in B16 melanoma showed an exhausted phenotype, whereas oncolytic adenovirus treatment significantly reduced the proportion of TIM-3+ TIL subset through recruitment of less-exhausted CD8+ TIL. Decrease of TIM-3 was observed in 60% of patients, which was associated with improved overall survival over TIM-3 increase patients (p=0.004), together with evidence of clinical benefit by imaging and blood analyses. Coinhibitory T-cell receptors and ligands were consistently associated with TIM-3 changes in gene expression data, while core transcriptional exhaustion programs and T-cell dysfunction were enriched in patients with TIM-3 increase, thus identifying patients potentially benefiting from checkpoint blockade. In striking contrast, patients with TIM-3 decrease displayed an acute inflammatory signature, redistribution of tumor-reactive CD8+ lymphocytes and higher influx of CD8+ TIL into tumors, which were associated with the longest overall survival, suggesting benefit from active immunotherapy. CONCLUSIONS: Our results indicate a key role for the TIM-3 immune checkpoint in oncolytic adenoviral immunotherapy. Moreover, our results identify TIM-3 as a potential biomarker for oncolytic adenoviruses and create rationale for combination with passive immunotherapy for a subset of patients.


Asunto(s)
Adenoviridae/patogenicidad , Biomarcadores de Tumor/metabolismo , Linfocitos T CD8-positivos/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Inmunoterapia/métodos , Neoplasias/genética , Virus Oncolíticos/patogenicidad , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Linfocitos T , Microambiente Tumoral
2.
PLoS One ; 16(11): e0258680, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34731182

RESUMEN

OBJECTIVE: To evaluate the pathogenicity of a broad range of 11 possible gastroenteritis viruses, by means of statistical relationships with cases vs. controls, or Ct-values, in order to establish the most appropriate diagnostic panel for our general practitioner (GP) patients in the Netherlands (2010-2012). METHODS: Archived stool samples from 1340 cases and 1100 controls were retested using internally controlled multiplex real-time PCRs for putative pathogenic gastroenteritis viruses: adenovirus, astrovirus, bocavirus, enterovirus, norovirus GI and GII, human parechovirus, rotavirus, salivirus, sapovirus, and torovirus. RESULTS: The prevalence of any virus in symptomatic cases and asymptomatic controls was 16.6% (223/1340) and 10.2% (112/1100), respectively. Prevalence of astrovirus (adjusted odds ratio (aOR) 10.37; 95% confidence interval (CI) 1.34-80.06) and norovirus GII (aOR 3.10; CI 1.62-5.92) was significantly higher in cases versus controls. Rotavirus was encountered only in cases. We did not find torovirus and there was no statistically significant relationship with cases for salivirus (aOR 1,67; (CI) 0.43-6.54)), adenovirus non-group F (aOR 1.20; CI 0.75-1.91), bocavirus (aOR 0.85; CI 0.05-13.64), enterovirus (aOR 0.83; CI 0.50-1.37), human parechovirus (aOR 1.61; CI 0.54-4.77) and sapovirus (aOR 1.15; CI 0.67-1.98). Though adenovirus group F (aOR 6.37; CI 0.80-50.92) and norovirus GI (aOR 2.22, CI: 0.79-6.23) are known enteropathogenic viruses and were more prevalent in cases than in controls, this did not reach significance in this study. The Ct value did not discriminate between carriage and disease in PCR-positive subjects. CONCLUSIONS: In our population, diagnostic gastroenteritis tests should screen for adenovirus group F, astrovirus, noroviruses GI and GII, and rotavirus. Case-control studies as ours are lacking and should also be carried out in populations from other epidemiological backgrounds.


Asunto(s)
Infecciones por Enterovirus/diagnóstico , Heces/virología , Gastroenteritis/diagnóstico , Adenoviridae/genética , Adenoviridae/aislamiento & purificación , Adenoviridae/patogenicidad , Bocavirus/genética , Bocavirus/aislamiento & purificación , Bocavirus/patogenicidad , Preescolar , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Femenino , Gastroenteritis/genética , Gastroenteritis/patología , Gastroenteritis/virología , Médicos Generales , Humanos , Lactante , Masculino , Norovirus/genética , Norovirus/aislamiento & purificación , Norovirus/patogenicidad , Pacientes , Rotavirus/genética , Rotavirus/aislamiento & purificación , Rotavirus/patogenicidad , Sapovirus/genética , Sapovirus/aislamiento & purificación , Sapovirus/patogenicidad
4.
Cancer Biomark ; 32(3): 251-262, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34459386

RESUMEN

BACKGROUND: To explore the suppressive effect of Apoptin-loaded oncolytic adenovirus (Ad-VT) on luciferase-labeled human melanoma cells in vitro and in vivo. METHODS: The stable luciferase-expressing human melanoma cells A375-luc or M14-luc were obtained by transfecting the plasmid pGL4.51 and selection with G418, followed by luciferase activity, genetic stability and bioluminescence intensity assays. In vitro, the inhibitory effects of Ad-VT on A375-luc or M14-luc were evaluated using the MTS cell proliferation, FITC-Annexin V apoptosis detection, transwell migration, Matrigel invasion and scratch assays. The inhibition pathway in Ad-VT-infected A375-luc and M14-luc cells were analyzed by JC-1 staining and Western-blot detection of mitochondrial apoptosis-related proteins. In vivo, the suppressive effects of Ad-VT on A375-luc or M14-luc were assessed by living imaging technology, tumor volume, bioluminescence intensity, survival curves and immunohistochemical analysis of the tumors from the xenograft tumor model BALB/c nude mice. RESULTS: The growth and migration of A375-luc and M14-luc were significantly inhibited by Ad-VT in vitro. The evaluations of A375-luc and M14-luc tumor models in BALB/c nude mice were successfully performed using living imaging technology. Ad-VT had an anti-tumor effect by reducing tumor growth and increasing survival in vivo. Ad-VT significantly changed the mitochondrial membrane potential by triggering the the mitochondrial release of apoptosis-related proteins, AIF (apoptosis inducing factor), ARTS (Apoptosis-Related Proteins), and Cyto-c (cytochrome c) from the mitochondria. CONCLUSION: Ad-VT reduced the mitochondrial membrane potential in A375-luc or M14-luc cells and induced the mitochondrial release of AIF, ARTS and Cyto-C. Ad-VT induced apoptosis in A375-luc or M14-luc cells via the mitochondrial apoptotic pathway.


Asunto(s)
Adenoviridae/patogenicidad , Melanoma/microbiología , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Viruses ; 13(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34372495

RESUMEN

The enteric human adenoviruses of species F (HAdVs-F), which comprise HAdV-F40 and HAdV-F41, are significant pathogens that cause acute gastroenteritis in children worldwide. The early transcription unit 3 (E3) of HAdVs-F is markedly different from that of all other HAdV species. To date, the E3 proteins unique to HAdVs-F have not been characterized and the mechanism by which HAdVs-F evade immune defenses in the gastrointestinal (GI) tract is poorly understood. Here, we show that HAdV-F41 infection of human intestinal HCT116 cells upregulated the expression of MHC class I-related chain A (MIC A) and MIC B relative to uninfected cells. Our results also showed that, for MIC B, this response did not however result in a significant increase of MIC B on the cell surface. Instead, MIC B was largely sequestered intracellularly. Thus, although HAdV-F41 infection of HCT116 cells upregulated MIC B expression, the ligand remained inside infected cells. A similar observation could not be made for MIC A in these cells. Our preliminary findings represent a novel function of HAdVs-F that may enable these viruses to evade immune surveillance by natural killer (NK) cells in the infected gut, thereby paving the way for the future investigation of their unique E3 proteins.


Asunto(s)
Adenoviridae/patogenicidad , Factor 15 de Diferenciación de Crecimiento/clasificación , Factor 15 de Diferenciación de Crecimiento/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Adenoviridae/inmunología , Proteínas Ligadas a GPI/genética , Células HCT116 , Humanos , Filogenia , Análisis de Secuencia de ADN
6.
Viruses ; 13(6)2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200540

RESUMEN

Adenovirus is a common cause of disease in humans and in animals [...].


Asunto(s)
Adenoviridae/patogenicidad , Infecciones por Adenoviridae , Adenovirus Humanos , Animales , Modelos Animales de Enfermedad , Humanos
7.
Int J Mol Sci ; 22(10)2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-34063479

RESUMEN

Adenoviruses are large (~950 Å) and complex non-enveloped, dsDNA icosahedral viruses. They have a pseudo-T = 25 triangulation number with at least 12 different proteins composing the virion. These include the major and minor capsid proteins, core proteins, maturation protease, terminal protein, and packaging machinery. Although adenoviruses have been studied for more than 60 years, deciphering their architecture has presented a challenge for structural biology techniques. An outstanding event was the first near-atomic resolution structure of human adenovirus type 5 (HAdV-C5), solved by cryo-electron microscopy (cryo-EM) in 2010. Discovery of new adenovirus types, together with methodological advances in structural biology techniques, in particular cryo-EM, has lately produced a considerable amount of new, high-resolution data on the organization of adenoviruses belonging to different species. In spite of these advances, the organization of the non-icosahedral core is still a great unknown. Nevertheless, alternative techniques such as atomic force microscopy (AFM) are providing interesting glimpses on the role of the core proteins in genome condensation and virion stability. Here we summarize the current knowledge on adenovirus structure, with an emphasis on high-resolution structures obtained since 2010.


Asunto(s)
Adenoviridae/química , Proteínas Virales/química , Adenoviridae/patogenicidad , Proteínas de la Cápside/química , Microscopía de Fuerza Atómica , Virión/química , Internalización del Virus
8.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33963009

RESUMEN

BACKGROUND: Pediatric high-grade gliomas (pHGGs) are among the most common and incurable malignant neoplasms of childhood. Despite aggressive, multimodal treatment, the outcome of children with high-grade gliomas has not significantly improved over the past decades, prompting the development of innovative approaches. METHODS: To develop an effective treatment, we aimed at improving the suboptimal antitumor efficacy of oncolytic adenoviruses (OAs) by testing the combination with a gene-therapy approach using a bispecific T-cell engager (BiTE) directed towards the erythropoietin-producing human hepatocellular carcinoma A2 receptor (EphA2), conveyed by a replication-incompetent adenoviral vector (EphA2 adenovirus (EAd)). The combinatorial approach was tested in vitro, in vivo and thoroughly characterized at a molecular level. RESULTS: After confirming the relevance of EphA2 as target in pHGGs, documenting a significant correlation with worse clinical outcome of the patients, we showed that the proposed strategy provides significant EphA2-BiTE amplification and enhanced tumor cell apoptosis, on coculture with T cells. Moreover, T-cell activation through an agonistic anti-CD28 antibody further increased the activation/proliferation profiles and functional response against infected tumor cells, inducing eradication of highly resistant, primary pHGG cells. The gene-expression analysis of tumor cells and T cells, after coculture, revealed the importance of both EphA2-BiTE and costimulation in the proposed system. These in vitro observations translated into significant tumor control in vivo, in both subcutaneous and a more challenging orthotopic model. CONCLUSIONS: The combination of OA and EphA2-BiTE gene therapy strongly enhances the antitumor activity of OA, inducing the eradication of highly resistant tumor cells, thus supporting the clinical translation of the approach.


Asunto(s)
Adenoviridae/genética , Anticuerpos Biespecíficos/genética , Neoplasias Encefálicas/terapia , Terapia Genética , Glioma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Receptor EphA2/genética , Adenoviridae/metabolismo , Adenoviridae/patogenicidad , Animales , Anticuerpos Biespecíficos/metabolismo , Apoptosis , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virología , Línea Celular Tumoral , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Femenino , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Glioma/genética , Glioma/metabolismo , Glioma/virología , Humanos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Clasificación del Tumor , Virus Oncolíticos/metabolismo , Virus Oncolíticos/patogenicidad , Receptor EphA2/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Epidemiol Infect ; 149: e96, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33849679

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is pandemic. Prevention and control strategies require an improved understanding of SARS-CoV-2 dynamics. We did a rapid review of the literature on SARS-CoV-2 viral dynamics with a focus on infective dose. We sought comparisons of SARS-CoV-2 with other respiratory viruses including SARS-CoV-1 and Middle East respiratory syndrome coronavirus. We examined laboratory animal and human studies. The literature on infective dose, transmission and routes of exposure was limited specially in humans, and varying endpoints were used for measurement of infection. Despite variability in animal studies, there was some evidence that increased dose at exposure correlated with higher viral load clinically, and severe symptoms. Higher viral load measures did not reflect coronavirus disease 2019 severity. Aerosol transmission seemed to raise the risk of more severe respiratory complications in animals. An accurate quantitative estimate of the infective dose of SARS-CoV-2 in humans is not currently feasible and needs further research. Our review suggests that it is small, perhaps about 100 particles. Further work is also required on the relationship between routes of transmission, infective dose, co-infection and outcomes.


Asunto(s)
COVID-19/transmisión , SARS-CoV-2/patogenicidad , Carga Viral , Adenoviridae/patogenicidad , Animales , COVID-19/epidemiología , COVID-19/prevención & control , COVID-19/virología , Chlorocebus aethiops , Control de Enfermedades Transmisibles , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/virología , Cricetinae , Enterovirus/patogenicidad , Hurones , Humanos , Macaca mulatta , Ratones , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , Orthomyxoviridae/patogenicidad , Virus Sincitiales Respiratorios/patogenicidad , Rhinovirus/patogenicidad , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Síndrome Respiratorio Agudo Grave/epidemiología , Síndrome Respiratorio Agudo Grave/transmisión , Síndrome Respiratorio Agudo Grave/virología , Virosis/epidemiología , Virosis/transmisión , Virosis/virología
11.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33737338

RESUMEN

BACKGROUND: Osteosarcoma is the most common malignant solid tumor that affects bones, however, survival rates of patients with relapsed osteosarcoma have not improved in the last 30 years. Oncolytic virotherapy, which uses viruses designed to selectively replicate in cancer cells, has emerged as a promising treatment for solid tumors. Our group uses mesenchymal stem cells (MSCs) to transport oncolytic adenoviruses (OAds) to the tumor site, a therapeutic strategy called Celyvir. This treatment has been already applied in human patients, canine patients and different mouse models. In parallel, previous results have probed that administration of granulocyte-colony stimulating factor (G-CSF) increased immune infiltration in tumors. We then hypothesized that the mobilization of immune cells by G-CSF may increase the antitumor efficacy of Celyvir treatment by increasing the immune infiltration into the tumors. METHODS: In this study, we use a murine version of Celyvir consisting in murine MSCs carrying the murine OAd dlE102-here called OAd-MSCs-in an immunocompetent model of osteosarcoma. We tested the antitumoral efficacy of the combination of OAd-MSCs plus G-CSF. RESULTS: Our results show that treatment with OAd-MSCs or the union of OAd-MSCs with G-CSF (Combination) significantly reduced tumor growth of osteosarcoma in vivo. Moreover, treated tumors presented higher tumor infiltration of immune cells-especially tumor-infiltrating lymphocytes-and reduced T cell exhaustion, which seems to be enhanced in tumors treated with the Combination. The comparison of our results to those obtained from a cohort of pediatric osteosarcoma patients showed that the virotherapy induces immunological changes similar to those observed in patients with good prognosis. CONCLUSIONS: The results open the possibility of using cellular virotherapy for the treatment of bone cancers. Indeed, its combination with G-CSF may be considered for the improvement of the therapy.


Asunto(s)
Adenoviridae/patogenicidad , Neoplasias Óseas/terapia , Factor Estimulante de Colonias de Granulocitos/farmacología , Agentes Inmunomoduladores/farmacología , Células Madre Mesenquimatosas/virología , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Osteosarcoma/terapia , Adenoviridae/inmunología , Animales , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Neoplasias Óseas/virología , Línea Celular Tumoral , Terapia Combinada , Efecto Citopatogénico Viral , Bases de Datos Genéticas , Humanos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Trasplante de Células Madre Mesenquimatosas , Ratones Endogámicos BALB C , Virus Oncolíticos/inmunología , Osteosarcoma/inmunología , Osteosarcoma/patología , Osteosarcoma/virología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Microambiente Tumoral
12.
Commun Biol ; 4(1): 368, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33742099

RESUMEN

High expression levels of human epidermal growth factor receptor 2 (HER2) have been associated with poor prognosis in patients with pancreatic adenocarcinoma (PDAC). However, HER2-targeting immunotherapies have been unsuccessful to date. Here we increase the breadth, potency, and duration of anti-PDAC HER2-specific CAR T-cell (HER2.CART) activity with an oncolytic adeno-immunotherapy that produces cytokine, immune checkpoint blockade, and a safety switch (CAdTrio). Combination treatment with CAdTrio and HER2.CARTs cured tumors in two PDAC xenograft models and produced durable tumor responses in humanized mice. Modifications to the tumor immune microenvironment contributed to the antitumor activity of our combination immunotherapy, as intratumoral CAdTrio treatment induced chemotaxis to enable HER2.CART migration to the tumor site. Using an advanced PDAC model in humanized mice, we found that local CAdTrio treatment of primary tumor stimulated systemic host immune responses that repolarized distant tumor microenvironments, improving HER2.CART anti-tumor activity. Overall, our data demonstrate that CAdTrio and HER2.CARTs provide complementary activities to eradicate metastatic PDAC and may represent a promising co-operative therapy for PDAC patients.


Asunto(s)
Adenoviridae/patogenicidad , Carcinoma Ductal Pancreático/terapia , Inmunoterapia Adoptiva , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Neoplasias Pancreáticas/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/trasplante , Antígeno B7-H1/inmunología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/virología , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Humanos , Interleucina-12/genética , Masculino , Metástasis de la Neoplasia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/virología , Receptor ErbB-2/genética , Receptores Quiméricos de Antígenos/genética , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Linfocitos T/inmunología , Carga Tumoral , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Sci Rep ; 11(1): 4405, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33623127

RESUMEN

Metagenomic next-generation sequencing (mNGS) holds promise as a diagnostic tool for unbiased pathogen identification and precision medicine. However, its medical utility depends largely on assay simplicity and reproducibility. In the current study, we aimed to develop a streamlined Illumina and Oxford Nanopore-based DNA/RNA library preparation protocol and rapid data analysis pipeline. The Illumina sequencing-based mNGS method was first developed and evaluated using a set of samples with known aetiology. Its sensitivity for RNA viruses (influenza A, H1N1) was < 6.4 × 102 EID50/mL, and a good correlation between viral loads and mapped reads was observed. Then, the rapid turnaround time of Nanopore sequencing was tested by sequencing influenza A virus and adenoviruses. Furthermore, 11 respiratory swabs or sputum samples pre-tested for a panel of pathogens were analysed, and the pathogens identified by Illumina sequencing showed 81.8% concordance with qPCR results. Additional sequencing of cerebrospinal fluid (CSF) samples from HIV-1-positive patients with meningitis/encephalitis detected HIV-1 RNA and Toxoplasma gondii sequences. In conclusion, we have developed a simplified protocol that realizes efficient metagenomic sequencing of a variety of clinical samples and pathogen identification in a clinically meaningful time frame.


Asunto(s)
Adenoviridae/genética , VIH/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Metagenómica/métodos , Técnicas de Diagnóstico Molecular/métodos , Orthomyxoviridae/genética , Adenoviridae/aislamiento & purificación , Adenoviridae/patogenicidad , Líquido Cefalorraquídeo/parasitología , Líquido Cefalorraquídeo/virología , VIH/aislamiento & purificación , VIH/patogenicidad , Humanos , Orthomyxoviridae/aislamiento & purificación , Orthomyxoviridae/patogenicidad , Esputo/virología , Toxoplasma/genética , Toxoplasma/aislamiento & purificación , Toxoplasma/patogenicidad
14.
Infect Dis (Lond) ; 53(2): 142-144, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33073645
15.
Cells ; 9(11)2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182528

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the most lethal cancers in China and existing therapies have been unable to significantly improve prognosis. Oncolytic adenoviruses (OAds) are novel promising anti-tumor drugs and have been evaluated in several cancers including ESCC. However, the antitumour efficacy of the first generation OAds (H101) as single agent is limited. Therefore, more effective OAds are needed. Our previous studies demonstrated that the novel oncolytic adenovirus Ad-TD-nsIL12 (human adenovirus type 5 with E1ACR2, E1B19K, E3gp19K-triple deletions)harboring human non-secretory IL-12 had significant anti-tumor effect, with no toxicity, in a Syrian hamster pancreatic cancer model. In this study, we evaluated the anti-tumor effect of Ad-TD-nsIL12 in human ESCC. The cytotoxicity of Ad-TD-nsIL12, H101 and cisplatin were investigated in two newly established patient-derived tumor cells (PDCs) and a panel of ESCC cell lines in vitro. A novel adenovirus-permissive, immune-deficient Syrian hamster model of PDCs subcutaneous xenograft was established for in vivo analysis of efficacy. The results showed that Ad-TD-nsIL12 was more cytotixic to and replicated more effectively in human ESCC cell lines than H101. Compared with cisplatin and H101, Ad-TD-nsIL12 could significantly inhibit tumor growth and tumor angiogenesis as well as enhance survival rate of animals with no side effects. These findings suggest that Ad-TD-nsIL12 has superior anti-tumor potency against human ESCC with a good safety profile.


Asunto(s)
Adenoviridae/patogenicidad , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Vectores Genéticos/uso terapéutico , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Cricetinae , Modelos Animales de Enfermedad , Vectores Genéticos/farmacología
16.
Immunol Lett ; 228: 64-69, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33031870

RESUMEN

Adoptive transfer of multivirus-specific T cell lines (MVST) is an advanced tool for immunotherapy of virus infections after hematopoietic stem cell transplantation (HSCT). Their preparation includes activation of donor virus-specific T cells by the mixture of oligopeptides derived from immunodominant antigens of several most harmful viruses, i.e. human cytomegalovirus (HCMV), polyomavirus BK (BKV), Epstein-Barr virus (EBV) and adenovirus (ADV). The aim of our study was to find out whether antigenic competition may have an impact on the expansion of virus-specific T cells. MVST from several heathy blood donors were generated using a pulse of overlapping oligopeptides (PepMixes™, derived from the IE1 and pp65 CMV antigens, VP1 and LTAG BKV antigens, BZLF1 and EBNA1 proteins of EBV and hexon protein from ADV) and short time culture in the presence of IL-7 and IL-4. The amount of virus-specific T cells in MVST was measured by ELISPOT and flow cytometry after re-stimulation with individual antigens. To evaluate antigenic competition, MVST were expanded either with a complete set of antigens or with the mixture lacking some of them. MVST expanded with the antigen mixture including CMV antigens contained a lower proportion of the T cells of other antigen specificities. A similar inhibitory effect was not apparent for EBV-derived peptides. The competitive effect of CMV antigens was most pronounced in MVST from CMV-seropositive donors and was mediated by both IE1 and pp65-derived peptides. Antigenic competition did not influence the phenotype of either CMV- or BKV-specific T cells. Both T cell populations had an effector memory phenotype (CD45RO+, CD27-, CCR7-). However, CMV-specific T cells preferentially consist of CD8+ while in BKV-specific T cells, the CD4+ phenotype predominated. Modification of the MVST manufacture protocol to prevent antigenic competition may increase the efficacy of MVST in therapy of BKV infections in HSCT recipients.


Asunto(s)
Adenoviridae/inmunología , Virus BK/inmunología , Citomegalovirus/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Herpesvirus Humano 4/inmunología , Inmunoterapia Adoptiva , Linfocitos T/trasplante , Virosis/terapia , Adenoviridae/patogenicidad , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/terapia , Infecciones por Adenovirus Humanos/virología , Antígenos Virales/inmunología , Virus BK/patogenicidad , Células Cultivadas , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/terapia , Infecciones por Citomegalovirus/virología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/terapia , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Epítopos Inmunodominantes , Activación de Linfocitos , Fenotipo , Infecciones por Polyomavirus/inmunología , Infecciones por Polyomavirus/terapia , Infecciones por Polyomavirus/virología , Linfocitos T/inmunología , Linfocitos T/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/terapia , Infecciones Tumorales por Virus/virología , Virosis/inmunología , Virosis/virología
17.
J Virol ; 94(17)2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32611755

RESUMEN

Recently, the disease of hepatitis-hydropericardium syndrome (HPS) caused by serotype 4 fowl adenovirus (FAdV-4) has spread widely and resulted in huge economic losses to the poultry industry. Although the genome of FAdV-4 has two fiber genes (fiber-1 and fiber-2), the exact role of the genes in the infection of FAdV-4 is barely known. In this study, through superinfection resistance analysis and an interfering assay, we found that fiber-1, but not fiber-2, was the key factor for directly triggering the infection of FAdV-4. The truncation analysis further revealed that both of the shaft and knob domains of fiber-1 were required for the infection. Moreover, the sera against the knob domain were able to block FAdV-4 infection, and the knob-containing fusion protein provided efficient protection against the lethal challenge of FAdV-4 in chickens. All the data demonstrated the significant roles of fiber-1 and its knob domain in directly mediating the infection of FAdV-4, which established a foundation for identifying the receptor of FAdV-4 and developing efficient vaccines against FAdV-4.IMPORTANCE Among 12 serotypes of fowl adenovirus (FAdV), FAdV-1, FAdV-4, and FAdV-10 all carry two fiber genes (i.e., fiber-1 and fiber-2), whereas other serotypes have only one. As important viral surface proteins, the fibers play vital roles in the infection and pathogenesis of FAdV. However, the importance of the fibers to the infection and pathogenesis of FAdV may be different from each other. Recent studies reveal that fiber-2 is identified as a determinant of virulence, but which fiber triggers the infection of FAdV-4 remains unknown. In this study, fiber-1 was identified as a key factor for directly mediating the infection of FAdV-4 through its shaft and knob domains, whereas fiber-2 did not play a role in triggering FAdV-4 infection. The results suggest that fiber-1 and its knob domain may serve as a target for identifying the receptor of FAdV-4 and developing efficient drugs or vaccines against FAdV-4.


Asunto(s)
Infecciones por Adenoviridae/virología , Adenoviridae/genética , Adenoviridae/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Enfermedades de las Aves de Corral/virología , Adenoviridae/patogenicidad , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/patología , Infecciones por Adenoviridae/prevención & control , Animales , Anticuerpos Antivirales , Línea Celular , Pollos/virología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/patología , Enfermedades de las Aves de Corral/prevención & control , Dominios Proteicos , Serogrupo , Vacunas Virales/inmunología
18.
FEBS Lett ; 594(12): 1818-1827, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32538496

RESUMEN

Both well-known and emerging viruses increasingly affect humans and cause disease, sometimes with devastating impact on society. The viruses present in the biosphere are the top predators in the life chain, virtually without enemies, except perhaps the immune system, and harsh environmental physicochemical conditions restricting their dissemination. We know a lot about viruses, but do we know enough? This series of reviews is dedicated to adenoviruses (AdVs), a family of nonenveloped DNA viruses occurring in vertebrates, including humans. AdVs have been the focus of intense research for more than 67 years. Besides causing disease, they have immensely contributed to the advance of life sciences and medicine over the past decades. Recently, AdVs have been widely used as vehicles in gene therapy and vaccination. They continue to provide fundamental insights into virus-host interactions in cells, tissues and organisms, as well as systems and metabolic networks. This special issue of FEBS Letters presents a unique collection of 23 state-of-the-art review articles by leading adenovirologists. In this prelude, I present the chapters, which provide a solid basis for further exploring the rich heritage in adenovirus molecular cell biology, structural biology, genetics, immunology, gene therapy and epidemiology. I conclude with an essential discussion of six blind spots in adenovirology.


Asunto(s)
Infecciones por Adenoviridae/terapia , Adenoviridae/fisiología , Adenoviridae/patogenicidad , Animales , Interacciones Huésped-Patógeno , Humanos , Internalización del Virus
19.
PLoS One ; 15(6): e0232948, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32479490

RESUMEN

BACKGROUND: An outbreak of respiratory disease associated with adenovirus type 7 occurred in a boot camp in China and was characterized by many cases, severe symptoms, and intrapulmonary infection in many patients. METHODS: We implemented a series of comprehensive preventive and control measures. We analyzed the incubation period and generation time by using the maximum likelihood method, assessed the symptom period and hospitalization duration using the Kaplan-Meier method, and estimated the basic reproductive number and dormitory transmission rate by using established methods. RESULTS: The epidemic lasted for 30 days, and 375 individuals were affected. Overall, 109 patients were hospitalized, and 266 individuals were isolated and treated. The median incubation period was 5.2 days (95% confidence interval [CI]: 5.0 to 5.4 days). The median generation time was 7.3 days (95% CI: 7.1 to 7.6 days). The median symptom period was 6 days (95% CI: 6 to 7 days). The median hospitalization duration was 9 days (95% CI: 9 to 11 days). The basic reproductive number was 5.1 (95% CI: 4.6 to 5.6), and the dormitory transmission rate was 0.15 (95% CI: 0.12 to 0.18). CONCLUSION: Patients in the early stage of the epidemic were treated as having a regular cold and were not isolated; therefore, the virus continued to be transmitted to other susceptible individuals.


Asunto(s)
Infecciones por Adenovirus Humanos/epidemiología , Adenoviridae/patogenicidad , Adenovirus Humanos/patogenicidad , Adulto , China/epidemiología , Brotes de Enfermedades/estadística & datos numéricos , Humanos , Infecciones/epidemiología , Masculino , Personal Militar , Adulto Joven
20.
FEBS Lett ; 594(12): 1861-1878, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32472693

RESUMEN

Human adenoviruses (HAdVs) cause widespread acute and persistent infections. Infections are usually mild and controlled by humoral and cell-based immunity. Reactivation of persistently infected immune cells can lead to a life-threatening disease in immunocompromised individuals, especially children and transplant recipients. To date, no effective therapy or vaccine against HAdV disease is available to the public. HAdV-C2 and C5 are the best-studied of more than 100 HAdV types. They persist in infected cells and release their progeny by host cell lysis to neighbouring cells and fluids, a process facilitated by the adenovirus death protein (ADP). ADP consists of about 100 amino acids and harbours a single membrane-spanning domain. It undergoes post-translational processing in endoplasmic reticulum and Golgi compartments, before localizing to the inner nuclear membrane. Here, we discuss the current knowledge on how ADP induces membrane rupture. Membrane rupture is essential for both progression of disease and efficacy of therapeutic viruses in clinical applications, in particular oncolytic therapy.


Asunto(s)
Adenoviridae/patogenicidad , Proteínas E3 de Adenovirus/fisiología , Infecciones por Adenovirus Humanos/patología , Interacciones Huésped-Patógeno/fisiología , Adenoviridae/metabolismo , Infecciones por Adenovirus Humanos/metabolismo , Infecciones por Adenovirus Humanos/virología , Animales , Muerte Celular , Retículo Endoplásmico/metabolismo , Humanos , Proteínas Mad2/metabolismo , Virus Oncolíticos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...