Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Mol Ther ; 32(7): 2406-2422, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38734899

RESUMO

Vaccinia viruses (VACVs) are versatile therapeutic agents and different features of various VACV strains allow for a broad range of therapeutic applications. Modified VACV Ankara (MVA) is a particularly altered VACV strain that is highly immunogenic, incapable of replicating in mammalian hosts, and broadly used as a safe vector for vaccination. Alternatively, Western Reserve (WR) or Copenhagen (Cop) are VACV strains that efficiently replicate in cancer cells and, therefore, are used to develop oncolytic viruses. However, the immune evasion capacity of WR or Cop hinders their ability to elicit antitumor immune responses, which is crucial for efficacy in the clinic. Here, we describe a new VACV strain named Immune-Oncolytic VACV Ankara (IOVA), which combines efficient replication in cancer cells with induction of immunogenic tumor cell death (ICD). IOVA was engineered from an MVA ancestor and shows superior cytotoxicity in tumor cells. In addition, the IOVA genome incorporates mutations that lead to massive fusogenesis of tumor cells, which contributes to improved antitumor effects. In syngeneic mouse tumor models, the induction of ICD results in robust antitumor immunity directed against tumor neo-epitopes and eradication of large established tumors. These data present IOVA as an improved immunotherapeutic oncolytic vector.


Assuntos
Morte Celular Imunogênica , Terapia Viral Oncolítica , Vírus Oncolíticos , Vaccinia virus , Vaccinia virus/genética , Vaccinia virus/imunologia , Animais , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Camundongos , Humanos , Terapia Viral Oncolítica/métodos , Linhagem Celular Tumoral , Neoplasias/terapia , Neoplasias/imunologia , Replicação Viral , Vetores Genéticos/genética
2.
J. coloproctol. (Rio J., Impr.) ; 43(2): 136-138, Apr.-June 2023. ilus
Artigo em Inglês | LILACS | ID: biblio-1514431

RESUMO

Introduction: Intussusceptions in adults are rare, representing 1% to 5% of intestinal obstructions in this age group. This condition can be caused by benign and malignant lesions acting as lead points, the latter being the most frequent. Furthermore, the diagnosis is challenging due to the non-specific symptoms with variable duration. Case Presentation: A 43-year-old man, with a history of localized clear-cell renal carcinoma (ccRCC) treated 9 years earlier with a right radical nephrectomy, presented with bowel obstruction symptoms. An abdominal computed tomography scan showed an ileocolonic intussusception. Hence, the patient required a right hemicolectomy with ileotransverse anastomosis. The histopathological analysis showed a metastatic ccRC to the terminal ileum causing the intussusception. Discussion: Adult intussusceptions are rare. However, they should be considered in the differential diagnosis of patients with abdominal pain and symptoms of bowel obstruction. Metastases of renal cancer to the small bowel are uncommon and even more so in the form of intussusception. Definitive treatment must be tailored to the patient's condition and underlying cause. (AU)


Assuntos
Humanos , Masculino , Adulto , Carcinoma de Células Renais/patologia , Doenças do Colo , Valva Ileocecal , Intussuscepção/diagnóstico , Neoplasias Renais/patologia , Dor Abdominal
3.
Cancer Gene Ther ; 30(7): 1029-1039, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37016144

RESUMO

cGAMP-specific nucleases (poxins) are a recently described family of proteins dedicated to obstructing cyclic GMP-AMP synthase signaling (cGAS), an important sensor triggered by cytoplasmic viral replication that activates type I interferon (IFN) production. The B2R gene of vaccinia viruses (VACV) codes for one of these nucleases. Here, we evaluated the effects of inactivating the VACV B2 nuclease in the context of an oncolytic VACV. VACV are widely used as anti-cancer vectors due to their capacity to activate immune responses directed against tumor antigens. We aimed to elicit robust antitumor immunity by preventing viral inactivation of the cGAS/STING/IRF3 pathway after infection of cancer cells. Activation of such a pathway is associated with a dominant T helper 1 (Th1) cell differentiation of the response, which benefits antitumor outcomes. Deletion of the B2R gene resulted in enhanced IRF3 phosphorylation and type I IFN expression after infection of tumor cells, while effective VACV replication remained unimpaired, both in vitro and in vivo. In syngeneic mouse tumor models, the absence of the VACV cGAMP-specific nuclease translated into improved antitumor activity, which was associated with antitumor immunity directed against tumor epitopes.


Assuntos
Interferon Tipo I , Poxviridae , Camundongos , Animais , Poxviridae/genética , Nucleotídeos Cíclicos , Vaccinia virus/genética , Vaccinia virus/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Interferon Tipo I/genética , Imunidade , Imunidade Inata/genética
4.
Mol Ther Oncolytics ; 22: 399-409, 2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34553028

RESUMO

Recently, oncolytic vaccinia viruses (VACVs) have shown their potential to provide for clinically effective cancer treatments. The reason for this clinical usefulness is not only the direct destruction of infected cancer cells but also activation of immune responses directed against tumor antigens. For eliciting a robust antitumor immunity, a dominant T helper 1 (Th1) cell differentiation of the response is preferred, and such polarization can be achieved by activating the Toll-like receptor 3 (TLR3)-interferon regulatory factor 3 (IRF3) signaling pathway. However, current VACVs used as oncolytic viruses to date still encode several immune evasion proteins involved in the inhibition of this signaling pathway. By inactivating genes of selected regulatory virus proteins, we aimed for a candidate virus with increased potency to activate cellular antitumor immunity but at the same time with a fully maintained replicative capacity in cancer cells. The removal of up to three key genes (C10L, N2L, and C6L) from VACV did not reduce the strength of viral replication, both in vitro and in vivo, but resulted in the rescue of IRF3 phosphorylation upon infection of cancer cells. In syngeneic mouse tumor models, this activation translated to enhanced cytotoxic T lymphocyte (CTL) responses directed against tumor-associated antigens and neo-epitopes and improved antitumor activity.

5.
Cancer Cell ; 30(1): 108-119, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27374223

RESUMO

Immunotherapies are highly promising cancer treatments, but understanding the factors mediating their resistance remains critical. Successes in randomized clinical testing have supported the growing appreciation that oncolytic virotherapies primarily act as immunotherapies. Here we identified prostaglandin E2 (PGE2) in the tumor as a key mediator of resistance to immunotherapies, including oncolytic vaccinia virotherapy. Elevated levels of PGE2 coupled to suppressive chemokine profiles and high levels of granulocytic myeloid-derived suppressor cells resulted in loss of immunotherapeutic potential. Viral vectors engineered to target PGE2 were capable of overcoming localized immunosuppression leading to profound changes in the tumor's immune status. This allowed the viral vectors to raise robust anti-tumor adaptive immune responses and sensitized established and previously resistant tumors to immunotherapies.


Assuntos
Quimiocinas/metabolismo , Dinoprostona/antagonistas & inibidores , Marcação de Genes/métodos , Hidroxiprostaglandina Desidrogenases/genética , Neoplasias Experimentais/terapia , Terapia Viral Oncolítica/métodos , Animais , Vacinas Anticâncer , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Vetores Genéticos/administração & dosagem , Hidroxiprostaglandina Desidrogenases/farmacologia , Imunoterapia , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Análise de Sobrevida , Resultado do Tratamento , Vaccinia virus/genética
6.
Cell Rep ; 15(2): 264-73, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27050526

RESUMO

The immune response plays a key role in enhancing the therapeutic activity of oncolytic virotherapies. However, to date, investigators have relied on inherent interactions between the virus and the immune system, often coupled to the expression of a single cytokine transgene. Recently, the importance of TLR activation in mediating adaptive immunity has been demonstrated. We therefore sought to influence the type and level of immune response raised after oncolytic vaccinia therapy through manipulation of TLR signaling. Vaccinia naturally activates TLR2, associated with an antibody response, whereas a CTL response is associated with TLR3-TRIF-signaling pathways. We manipulated TLR signaling by vaccinia through deglycosylation of the viral particle to block TLR2 activation and expression of a TRIF transgene. The resulting vector displayed greatly reduced production of anti-viral neutralizing antibody as well as an increased anti-tumor CTL response. Delivery in both naive and pre-treated mice was enhanced and immunotherapeutic activity dramatically improved.


Assuntos
Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica , Transdução de Sinais , Linfócitos T/metabolismo , Receptor 2 Toll-Like/metabolismo , Vaccinia virus/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Glicosilação , Imunoterapia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Biológicos , Necrose , Linfócitos T Citotóxicos/metabolismo , Timidina Quinase/metabolismo
7.
J Clin Invest ; 125(10): 3915-27, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26348895

RESUMO

Optical imaging of whole, living animals has proven to be a powerful tool in multiple areas of preclinical research and has allowed noninvasive monitoring of immune responses, tumor and pathogen growth, and treatment responses in longitudinal studies. However, fluorescence-based studies in animals are challenging because tissue absorbs and autofluoresces strongly in the visible light spectrum. These optical properties drive development and use of fluorescent labels that absorb and emit at longer wavelengths. Here, we present a far-red absorbing fluoromodule-based reporter/probe system and show that this system can be used for imaging in living mice. The probe we developed is a fluorogenic dye called SC1 that is dark in solution but highly fluorescent when bound to its cognate reporter, Mars1. The reporter/probe complex, or fluoromodule, produced peak emission near 730 nm. Mars1 was able to bind a variety of structurally similar probes that differ in color and membrane permeability. We demonstrated that a tool kit of multiple probes can be used to label extracellular and intracellular reporter-tagged receptor pools with 2 colors. Imaging studies may benefit from this far-red excited reporter/probe system, which features tight coupling between probe fluorescence and reporter binding and offers the option of using an expandable family of fluorogenic probes with a single reporter gene.


Assuntos
Compostos de Anilina/análise , Corantes Fluorescentes/análise , Genes Reporter , Microscopia Intravital , Neoplasias Experimentais/ultraestrutura , Imagem Óptica/métodos , Anticorpos de Cadeia Única/análise , Ativação Metabólica , Compostos de Anilina/farmacocinética , Animais , Linhagem Celular , Permeabilidade da Membrana Celular , Cor , Desamino Arginina Vasopressina/farmacologia , Endocitose/efeitos dos fármacos , Fluorescência , Corantes Fluorescentes/farmacocinética , Proteínas de Fluorescência Verde/análise , Células HCT116/transplante , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/química , Neoplasias Peritoneais/química , Neoplasias Peritoneais/ultraestrutura , Receptores de Vasopressinas/análise , Receptores de Vasopressinas/genética , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/metabolismo , Relação Estrutura-Atividade , Frações Subcelulares/metabolismo , Frações Subcelulares/ultraestrutura , Transdução Genética
8.
Clin Cancer Res ; 21(24): 5543-51, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26187615

RESUMO

PURPOSE: Recent data from randomized clinical trials with oncolytic viral therapies and with cancer immunotherapies have finally recapitulated the promise these platforms demonstrated in preclinical models. Perhaps the greatest advance with oncolytic virotherapy has been the appreciation of the importance of activation of the immune response in therapeutic activity. Meanwhile, the understanding that blockade of immune checkpoints (with antibodies that block the binding of PD1 to PDL1 or CTLA4 to B7-2) is critical for an effective antitumor immune response has revitalized the field of immunotherapy. The combination of immune activation using an oncolytic virus and blockade of immune checkpoints is therefore a logical next step. EXPERIMENTAL DESIGN: Here, we explore such combinations and demonstrate their potential to produce enhanced responses in mouse tumor models. Different combinations and regimens were explored in immunocompetent mouse models of renal and colorectal cancer. Bioluminescence imaging and immune assays were used to determine the mechanisms mediating synergistic or antagonistic combinations. RESULTS: Interaction between immune checkpoint inhibitors and oncolytic virotherapy was found to be complex, with correct selection of viral strain, antibody, and timing of the combination being critical for synergistic effects. Indeed, some combinations produced antagonistic effects and loss of therapeutic activity. A period of oncolytic viral replication and directed targeting of the immune response against the tumor were required for the most beneficial effects, with CD8(+) and NK, but not CD4(+) cells mediating the effects. CONCLUSIONS: These considerations will be critical in the design of the inevitable clinical translation of these combination approaches. Clin Cancer Res; 21(24); 5543-51. ©2015 AACR.See related commentary by Slaney and Darcy, p. 5417.


Assuntos
Imunoterapia , Neoplasias/imunologia , Neoplasias/metabolismo , Terapia Viral Oncolítica , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno CTLA-4/antagonistas & inibidores , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Imunidade Celular , Imunomodulação/efeitos dos fármacos , Imunoterapia/métodos , Subunidade alfa de Receptor de Interleucina-2/antagonistas & inibidores , Camundongos , Terapia de Alvo Molecular , Neoplasias/diagnóstico , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Carga Tumoral/efeitos dos fármacos , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/genética , Replicação Viral/efeitos dos fármacos
9.
Clin Cancer Res ; 21(6): 1406-18, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25391696

RESUMO

PURPOSE: Tumor targeting upon intravenous administration and subsequent intratumoral virus dissemination are key features to improve oncolytic adenovirus therapy. VCN-01 is a novel oncolytic adenovirus that combines selective replication conditional to pRB pathway deregulation, replacement of the heparan sulfate glycosaminoglycan putative-binding site KKTK of the fiber shaft with an integrin-binding motif RGDK for tumor targeting, and expression of hyaluronidase to degrade the extracellular matrix. In this study, we evaluate the safety and efficacy profile of this novel oncolytic adenovirus. EXPERIMENTAL DESIGN: VCN-01 replication and potency were assessed in a panel of tumor cell lines. VCN-01 tumor-selective replication was evaluated in human fibroblasts and pancreatic islets. Preclinical toxicity, biodistribution, and efficacy studies were conducted in mice and Syrian hamsters. RESULTS: Toxicity and biodistribution preclinical studies support the selectivity and safety of VCN-01. Antitumor activity after intravenous or intratumoral administration of the virus was observed in all tumor models tested, including melanoma and pancreatic adenocarcinoma, both in immunodeficient mice and immunocompetent hamsters. CONCLUSIONS: Oncolytic adenovirus VCN-01 characterized by the expression of hyaluronidase and the RGD shaft retargeting ligand shows an efficacy-toxicity prolife in mice and hamsters by intravenous and intratumoral administration that warrants clinical testing.


Assuntos
Adenocarcinoma/terapia , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Animais , Sítios de Ligação/genética , Proteínas de Transporte/genética , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Matriz Extracelular/metabolismo , Feminino , Células HEK293 , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Hialuronoglucosaminidase/biossíntese , Hialuronoglucosaminidase/genética , Ilhotas Pancreáticas/virologia , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/genética , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Theranostics ; 2(4): 363-73, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22509200

RESUMO

Biological cancer therapies, such as oncolytic, or replication-selective viruses have advantages over traditional therapeutics as they can employ multiple different mechanisms to target and destroy cancers (including direct cell lysis, immune activation and vascular collapse). This has led to their rapid recent clinical development. However this also makes their pre-clinical and clinical study complex, as many parameters may affect their therapeutic potential and so defining reason for treatment failure or approaches that might enhance their therapeutic activity can be complicated. The ability to non-invasively image viral gene expression in vivo both in pre-clinical models and during clinical testing will considerably enhance the speed of oncolytic virus development as well as increasing the level and type of useful data produced from these studies. Further, subsequent to future clinical approval, imaging of reporter gene expression might be used to evaluate the likelihood of response to oncolytic viral therapy prior to changes in tumor burden. Here different reporter genes used in conjunction with oncolytic viral therapy are described, along with the imaging modalities used to measure their expression, while their applications both in pre-clinical and clinical testing are discussed. Possible future applications for reporter gene expression from oncolytic viruses in the phenotyping of tumors and the personalizing of treatment regimens are also discussed.

11.
Mol Ther ; 18(11): 1960-71, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20808288

RESUMO

Oncolytic adenoviruses are promising anticancer agents due to their ability to self-amplify at the tumor mass. However, tumor stroma imposes barriers difficult to overcome by these agents. Transgene expression is a valuable strategy to counteract these limitations and to enhance antitumor activity. For this purpose, the genetic backbone in which the transgene is inserted should be optimized to render transgene expression compatible with the adenovirus replication cycle and to keep genome size within the encapsidation size limit. In order to design a potent and selective oncolytic adenovirus that keeps intact all the viral functions with minimal increase in genome size, we inserted palindromic E2F-binding sites into the endogenous E1A promoter. The insertion of these sites controlling E1A-Δ24 results in a low systemic toxicity profile in mice. Importantly, the E2F-binding sites also increased the cytotoxicity and the systemic antitumor activity relative to wild-type adenovirus in all cancer models tested. The low toxicity and the increased potency results in improved antitumor efficacy after systemic injection and increased survival of mice carrying tumors. Furthermore, the constrained genome size of this backbone allows an efficient and potent expression of transgenes, indicating that this virus holds promise for overcoming the limitations of oncolytic adenoviral therapy.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Terapia Genética , Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Regiões Promotoras Genéticas/genética , Proteína do Retinoblastoma/fisiologia , Proteínas E1A de Adenovirus/metabolismo , Animais , Western Blotting , Efeito Citopatogênico Viral , Fator de Transcrição E2F1/metabolismo , Vetores Genéticos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/genética , Neoplasias/virologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Transgenes/fisiologia , Células Tumorais Cultivadas , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Ther ; 15(9): 1607-15, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17579575

RESUMO

E2F acts as a transcriptional repressor when bound to unphosphorylated RB during the G(1) or G(0) phase. Upon phosphorylation, E2F is released from the E2F-RB complexes to activate transcription. Tumor cells are characterized by an increase in the level of "free" E2F as a consequence of the absence or hyperphosphorylation of RB. The E2F-1 promoter is a well-characterized E2F-responsive promoter, and it can be used to control adenovirus E1a gene expression as a strategy to achieve tumor-selective expression and replication of an adenovirus. ICOVIR-5 (Ad-DM-E2F-K-Delta24RGD) is an optimized oncolytic adenovirus that combines E1a transcriptional control by an insulated form of the E2F promoter with the Delta24 mutation of E1a to improve the therapeutic index of AdDelta24RGD. ICOVIR-5 also contains the Kozak sequence at the E1a start codon, which is important to restore E1a expression and viral replication to AdwtRGD levels in tumor cells. The unique combination of genetic elements in ICOVIR-5 allows the selectivity for cells with a deregulated E2F-RB pathway to be increased and potent anti-tumoral activity to be maintained. Dose-response toxicological and efficacy studies after a single systemic administration in pre-clinical models in mice are presented to demonstrate that this virus holds promise for treatment of disseminated cancer.


Assuntos
Adenoviridae/genética , Fator de Transcrição E2F1/genética , Terapia Genética/métodos , Neoplasias/terapia , Proteína do Retinoblastoma/genética , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Fator de Transcrição E2F1/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/genética , Neoplasias/patologia , Vírus Oncolíticos/genética , Proteína do Retinoblastoma/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA