Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Br J Cancer ; 117(1): 51-55, 2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-28557974

RESUMO

BACKGROUND: Despite recent therapeutic and diagnostic advances, prostate cancer remains the second leading cause of cancer-related deaths among men in the Western world. Oncolytic viruses that replicate selectively in tumour cells represent a novel treatment candidate for these malignancies. METHODS: We analysed infectivity of avirulent Semliki Firest virus SFV-VA7 in human prostate cancer cell lines VCaP, LNCaP and 22Rv1 and in nonmalignant prostate epithelial cell line RWPE-1. Therapeutic potency of SFV-VA7 was evaluated in subcutaneous and orthotopic mouse LNCaP xenograft models. RESULTS: SFV-VA7 infected and killed the tested human prostate cancer cell lines irrespective of their hormone response status, while the nonmalignant prostate epithelial cell line RWPE-1 proved highly virus resistant. Notably, a single peritoneal dose of SFV-VA7 was sufficient to eradicate all subcutaneous and orthotopic LNCaP tumours. CONCLUSIONS: Our results indicate that SFV-VA7 is a novel, promising therapeutic virus against prostate cancer warranting further testing in early clinical trials.


Assuntos
Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/terapia , Vírus da Floresta de Semliki , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Transplante de Neoplasias , Vírus Oncolíticos , Próstata , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Virol ; 89(20): 10637-47, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26269187

RESUMO

UNLABELLED: Glioblastoma is a terminal disease with no effective treatment currently available. Among the new therapy candidates are oncolytic viruses capable of selectively replicating in cancer cells, causing tumor lysis and inducing adaptive immune responses against the tumor. However, tumor antiviral responses, primarily mediated by type I interferon (IFN-I), remain a key problem that severely restricts viral replication and oncolysis. We show here that the Semliki Forest virus (SFV) strain SFV4, which causes lethal encephalitis in mice, is able to infect and replicate independent of the IFN-I defense in mouse glioblastoma cells and cell lines originating from primary human glioblastoma patient samples. The ability to tolerate IFN-I was retained in SFV4-miRT124 cells, a derivative cell line of strain SFV4 with a restricted capacity to replicate in neurons due to insertion of target sites for neuronal microRNA 124. The IFN-I tolerance was associated with the viral nsp3-nsp4 gene region and distinct from the genetic loci responsible for SFV neurovirulence. In contrast to the naturally attenuated strain SFV A7(74) and its derivatives, SFV4-miRT124 displayed increased oncolytic potency in CT-2A murine astrocytoma cells and in the human glioblastoma cell lines pretreated with IFN-I. Following a single intraperitoneal injection of SFV4-miRT124 into C57BL/6 mice bearing CT-2A orthotopic gliomas, the virus homed to the brain and was amplified in the tumor, resulting in significant tumor growth inhibition and improved survival. IMPORTANCE: Although progress has been made in development of replicative oncolytic viruses, information regarding their overall therapeutic potency in a clinical setting is still lacking. This could be at least partially dependent on the IFN-I sensitivity of the viruses used. Here, we show that the conditionally replicating SFV4-miRT124 virus shares the IFN-I tolerance of the pathogenic wild-type SFV, thereby allowing efficient targeting of a glioma that is refractory to naturally attenuated therapy vector strains sensitive to IFN-I. This is the first evidence of orthotopic syngeneic mouse glioma eradication following peripheral alphavirus administration. Our findings indicate a clear benefit in harnessing the wild-type virus replicative potency in development of next-generation oncolytic alphaviruses.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Interferon Tipo I/imunologia , MicroRNAs/imunologia , Vírus Oncolíticos/fisiologia , Vírus da Floresta de Semliki/fisiologia , Idoso , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/virologia , Linhagem Celular Tumoral , Células Clonais , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação da Expressão Gênica , Glioblastoma/imunologia , Glioblastoma/mortalidade , Glioblastoma/virologia , Humanos , Interferon Tipo I/genética , Masculino , Camundongos , MicroRNAs/genética , Neurônios/imunologia , Neurônios/patologia , Neurônios/virologia , Terapia Viral Oncolítica/métodos , Transdução de Sinais , Análise de Sobrevida , Carga Tumoral , Replicação Viral
3.
J Pediatr Gastroenterol Nutr ; 60(5): 626-31, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25522308

RESUMO

OBJECTIVES: Determination of antibodies to synthetic deamidated gliadin peptides (anti-DGPs) may work as an alternative or complement the commonly used test for tissue transglutaminase antibodies (TGA) in the diagnosis of celiac disease (CD). We analyzed the performance of a time-resolved immunofluorometric anti-DGP assay (TR-IFMA) in the diagnosis of CD in children and also retrospectively analyzed the appearance of anti-DGP antibodies before TGA seroconversion. METHODS: The study included 92 children with biopsy-confirmed CD. Serum samples were taken at the time or just before the clinical diagnosis. The control group comprised 82 TGA-negative children who were positive for human leucocyte antigen-DQ2 or -DQ8. RESULTS: Based on receiver operating characteristic curves, the optimal cutoff value for immunoglobulin (Ig) A anti-DGP positivity was 153 arbitrary units (AUs) with a sensitivity of 92.4% and specificity of 97.6% and that for IgG anti-DGP 119 AU, with a sensitivity of 97.8% and specificity of 97.6%. All 92 children with CD were either IgA or IgG anti-DGP positive at the time of diagnosis. Sera from 48 children with CD were also analyzed retrospectively before the diagnosis. Anti-DGP antibodies preceded TGA positivity in 35 of the 48 children with CD and appeared a median of 1 year earlier. CONCLUSIONS: The TR-IFMA assay for detecting anti-DGP antibodies shows high sensitivity and specificity for the diagnosis of CD in children. In a majority of our study population, anti-DGP seropositivity preceded TGA positivity, indicating that earlier detection of CD may be possible by monitoring anti-DGP antibodies frequently in genetically susceptible children.


Assuntos
Doença Celíaca/sangue , Doença Celíaca/diagnóstico , Glutens/imunologia , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Adolescente , Doença Celíaca/genética , Criança , Pré-Escolar , Diagnóstico Precoce , Feminino , Fluorimunoensaio , Proteínas de Ligação ao GTP/imunologia , Cadeias alfa de HLA-DQ/genética , Cadeias beta de HLA-DQ/genética , Humanos , Lactente , Masculino , Proteína 2 Glutamina gama-Glutamiltransferase , Curva ROC , Estudos Retrospectivos , Testes Sorológicos , Fatores de Tempo , Transglutaminases/imunologia
4.
BMC Vet Res ; 11: 170, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26215394

RESUMO

BACKGROUND: Dogs suffer from spontaneous tumors which may be amenable to therapies developed for human cancer patients, and dogs may serve as large-animal cancer models. A non-pathogenic Semliki Forest virus vector VA7-EGFP previously showed promise in targeting human tumor xenografts in mice, but the oncolytic capacity of the virus in canine cancer cells and the safety of the virus in higher mammals such as dogs, are not known. We therefore assessed the oncolytic potency of VA7-EGFP against canine cancer cells by infectivity and viability assays in two dog solid tumor cell lines. Furthermore we performed a 3-week safety study in two adult Beagles which received a single intravenous injection of ~2 × 10(5) plaque forming units of parental A7(74) strain. RESULTS: VA7-EGFP was able to replicate in and kill both canine cancer cell lines tested. No adverse events were observed in either of the two virus-injected adult Beagles and no infective virus could be recovered from any of the biological samples collected over the course of the study. Neutralizing antibodies to Semliki Forest virus became detectable in the dogs at 5 days post infection and remained elevated until study termination. CONCLUSIONS: Based on these results, testing of the oncolytic potential of attenuated Semliki Forest virus in canine cancer patients appears feasible.


Assuntos
Vacinas Anticâncer/efeitos adversos , Doenças do Cão/induzido quimicamente , Animais , Anticorpos Neutralizantes/sangue , Linhagem Celular Tumoral , Cães , Feminino , Vírus da Floresta de Semliki , Replicação Viral/fisiologia
5.
J Virol ; 87(1): 335-44, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23077310

RESUMO

Artificial target sequences for tissue-specific miRNAs have recently been introduced as a new means for altering the tissue tropism of viral replication. This approach can be used to improve the safety of oncolytic viruses for cancer virotherapy by restricting their replication in unwanted tissues, such as the liver. Semliki Forest virus (SFV) is a positive-strand RNA virus and, similar to the related alphaviruses, like Sindbis virus, has potential as a gene therapy vector and an oncolytic virotherapy agent, but this potential is limited by the neurovirulence of these alphaviruses. Here, we have generated a replicative SFV4 carrying six tandem targets for the neuron-specific miR124 between the viral nonstructural protein 3 and 4 (nsp3 and nsp4) genes. When administered intraperitoneally into adult BALB/c mice, SFV4-miRT124 displayed an attenuated spread into the central nervous system (CNS) and greatly increased survival. Peripheral replication was not affected, indicating neuron-specific attenuation. Moreover, a strong protective SFV immunity was elicited in these animals. Intracranial infection of adult mice with SFV4-miRT124 showed greatly reduced infection of neurons in the brain but led to the infection of oligodendrocytes in the corpus callosum. Taken together, our data show that miR124-mediated attenuation of neurovirulence is a feasible and promising strategy for generating safer oncolytic alphavirus virotherapy agents.


Assuntos
MicroRNAs/metabolismo , RNA Viral/metabolismo , Vírus da Floresta de Semliki/patogenicidade , Tropismo Viral , Replicação Viral , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Encéfalo/virologia , Linhagem Celular , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , RNA Viral/genética , Vírus da Floresta de Semliki/fisiologia , Análise de Sobrevida
6.
J Virol ; 87(4): 2363-6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23221568

RESUMO

Attenuated Semliki Forest virus (SFV) may be suitable for targeting malignant glioma due to its natural neurotropism, but its replication in brain tumor cells may be restricted by innate antiviral defenses. We attempted to facilitate SFV replication in glioma cells by combining it with vaccinia virus, which is capable of antagonizing such defenses. Surprisingly, we found parenchymal mouse brain tumors to be refractory to both viruses. Also, vaccinia virus appears to be sensitive to SFV-induced antiviral interference.


Assuntos
Glioma/terapia , Vírus Oncolíticos/crescimento & desenvolvimento , Vírus Oncolíticos/imunologia , Vírus da Floresta de Semliki/crescimento & desenvolvimento , Vírus da Floresta de Semliki/imunologia , Vaccinia virus/crescimento & desenvolvimento , Vaccinia virus/imunologia , Animais , Modelos Animais de Doenças , Camundongos
7.
Bioconjug Chem ; 25(1): 4-10, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24341748

RESUMO

In this paper, novel firefly luciferase-specific inhibitor compounds (FLICs) are evaluated as potential tools for cellular trafficking of transporter conjugates. As a proof-of-concept, we designed FLICs that were suitable for solid phase peptide synthesis and could be covalently conjugated to peptides via an amide bond. The spacer between inhibitor and peptide was optimized to gain efficient inhibition of recombinant firefly luciferase (FLuc) without compromising the activity of the model peptides. The hypothesis of using FLICs as tools for cellular trafficking studies was ensured with U87Fluc glioblastoma cells expressing firefly luciferase. Results show that cell penetrating peptide (penetratin) FLIC conjugate 9 inhibited FLuc penetrated cells efficiently (IC50 = 1.6 µM) and inhibited bioluminescence, without affecting the viability of the cells. Based on these results, peptide-FLIC conjugates can be used for the analysis of cellular uptake of biomolecules in a new way that can at the same time overcome some downsides seen with other methods. Thus, FLICs can be considered as versatile tools that broaden the plethora of methods that take advantage of the bioluminescence phenomena.


Assuntos
Proteínas de Transporte/química , Vaga-Lumes/enzimologia , Isoxazóis/química , Isoxazóis/farmacologia , Luminescência , Animais , Proteínas de Transporte/metabolismo , Peptídeos Penetradores de Células , Relação Dose-Resposta a Droga , Humanos , Isoxazóis/farmacocinética , Cinética , Luciferases de Vaga-Lume/antagonistas & inibidores , Luciferases de Vaga-Lume/metabolismo , Medições Luminescentes , Estrutura Molecular , Relação Estrutura-Atividade , Fatores de Tempo , Distribuição Tecidual
8.
Biomacromolecules ; 15(4): 1534-42, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24628489

RESUMO

We present two facile approaches for introducing multivalent displays of tyrosine sulfate mimetic ligands on the surface of cellulose nanocrystals (CNCs) for application as viral inhibitors. We tested the efficacy of cellulose nanocrystals, prepared either from cotton fibers or Whatman filter paper, to inhibit alphavirus infectivity in Vero (B) cells. Cellulose nanocrystals were produced by sulfuric acid hydrolysis leading to nanocrystal surfaces decorated with anionic sulfate groups. When the fluorescent marker expressing Semliki Forest virus vector, VA7-EGFP, was incubated with CNCs, strong inhibition of virus infectivity was achieved, up to 100 and 88% for cotton and Whatman CNCs, respectively. When surface sulfate groups of CNCs were exchanged for tyrosine sulfate mimetic groups (i.e. phenyl sulfonates), improved viral inhibition was attained. Our observations suggest that the conjugation of target-specific functionalities to CNC surfaces provides a means to control their antiviral activity. Multivalent CNCs did not cause observable in vitro cytotoxicity to Vero (B) cells or human corneal epithelial (HCE-T) cells, even within the 100% virus-inhibitory concentrations. Based on the similar chemistry of known polyanionic inhibitors, our results suggest the potential application of CNCs as inhibitors of other viruses, such as human immunodeficiency virus (HIV) and herpes simplex viruses.


Assuntos
Infecções por Alphavirus/tratamento farmacológico , Antivirais/química , Antivirais/farmacologia , Celulose/síntese química , Nanopartículas/química , Nanotecnologia/métodos , Vírus da Floresta de Semliki/efeitos dos fármacos , Tirosina/análogos & derivados , Animais , Biomimética , Celulose/química , Chlorocebus aethiops , Epitélio Corneano/citologia , Epitélio Corneano/efeitos dos fármacos , Genes erbB-1 , Ligantes , Nanopartículas/toxicidade , Vírus da Floresta de Semliki/genética , Espectroscopia de Infravermelho com Transformada de Fourier , Tirosina/química , Células Vero/efeitos dos fármacos , Células Vero/virologia
9.
PLoS Pathog ; 7(11): e1002383, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22114558

RESUMO

Among the four non-structural proteins of alphaviruses the function of nsP3 is the least well understood. NsP3 is a component of the viral replication complex, and composed of a conserved aminoterminal macro domain implicated in viral RNA synthesis, and a poorly conserved carboxyterminal region. Despite the lack of overall homology we noted a carboxyterminal proline-rich sequence motif shared by many alphaviral nsP3 proteins, and found it to serve as a preferred target site for the Src-homology 3 (SH3) domains of amphiphysin-1 and -2. Nsp3 proteins of Semliki Forest (SFV), Sindbis (SINV), and Chikungunya viruses all showed avid and SH3-dependent binding to amphiphysins. Upon alphavirus infection the intracellular distribution of amphiphysin was dramatically altered and colocalized with nsP3. Mutations in nsP3 disrupting the amphiphysin SH3 binding motif as well as RNAi-mediated silencing of amphiphysin-2 expression resulted in impaired viral RNA replication in HeLa cells infected with SINV or SFV. Infection of Balb/c mice with SFV carrying an SH3 binding-defective nsP3 was associated with significantly decreased mortality. These data establish SH3 domain-mediated binding of nsP3 with amphiphysin as an important host cell interaction promoting alphavirus replication.


Assuntos
Alphavirus/metabolismo , Proteínas do Tecido Nervoso/metabolismo , RNA Viral/biossíntese , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Vírus Chikungunya/genética , Células HeLa , Humanos , Ligantes , Camundongos , Vírus da Floresta de Semliki/genética , Sindbis virus/genética , Domínios de Homologia de src/genética
10.
Mol Ther ; 20(8): 1529-39, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22434140

RESUMO

In our recent study, replicative alphaviral vector VA7 was found to be effective against orthotopic human U87-glioma xenografts in an athymic mouse model eradicating the tumors with single intravenous (i.v.) injection. Here, we tested the efficacy of VA7 in immunocompetent orthotopic GL261 and CT-2A glioma models of C57BL/6 mouse in vivo. The cell lines were susceptible to VA7 infection in vitro, but GL261 infection was highly restricted in confluent cell cultures, and mouse interferon-ß (IFNß) pretreatment prevented the replication of VA7 in both cell lines. When mice bearing orthotopic GL261 or CT-2A tumors were administered neurotropic VA7, either i.v. or intracranially (i.c.), the vector was unable to infect the tumor and no survival benefit was achieved. Pretreatments with immunosuppressive cyclophosphamide (CPA) and rapamycin markedly lowered serum-neutralizing antibodies (NAbs) but had no effect on tumor infection or survival. Intracranial GL261 tumors were refractory also in athymic C57BL/6 mice, which have serious defects in their adaptive immunity. Implanted VA7-infected GL261 cells formed tumors with only slightly delayed kinetics and without improving survival thus excluding the participation of physical barriers and indicating robust host IFN action. Mouse and human IFNß do not seem be species cross-reactive, which might limit the translational relevance of xenograft models in oncolytic virotherapy.


Assuntos
Alphavirus/genética , Glioma/tratamento farmacológico , Glioma/terapia , Interferon beta/uso terapêutico , Terapia Viral Oncolítica/métodos , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Interferon beta/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cytotherapy ; 11(6): 726-37, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19878059

RESUMO

BACKGROUND AIMS: Mesenchymal stromal cells (MSC) have been used in a wide variety of pre-clinical experiments and in an increasing number of human clinical trials. Although many of these studies have shown different levels of engraftment, the exact fate of MSC after transplantation and the tissue response to their engraftment have not been investigated in detail. In the present work we studied the distribution of human MSC in a rat hind limb ischemic injury model immediately after transplantation and also analyzed the recipient tissue response to transplanted cells. METHODS: We tracked the in vivo fate of the transplanted MSC utilizing bioluminescence imaging, fluorescence microscopy and gene/protein expression analysis in a rat hind limb ischemia model. We also monitored the viability of transplanted cells by graft versus recipient expression analysis and determined the angiogenic and proliferative effect of transplantation by histologic staining. RESULTS: According to imaging analysis only a small portion of cells persisted for an extended period of time at the site of injury. Interestingly, recipient versus graft expression studies showed increased synthesis of rat-origin angiogenic factors and no human-origin mRNA or protein synthesis in transplanted tissues. More importantly, despite the lack of robust engraftment or growth factor secretion the transplantation procedure exerted a significant pro-angiogenic and pro-proliferative effect, which was mediated by angiogenic and mitogenic signaling pathways. CONCLUSIONS: Our results show an immediate temporal tissue effect in response to MSC transplantation that may represent a novel indirect paracrine mechanism for the beneficial effects of cell transplantation observed in injured tissues.


Assuntos
Extremidade Inferior/irrigação sanguínea , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica , Traumatismo por Reperfusão/cirurgia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Expressão Gênica/fisiologia , Reação Enxerto-Hospedeiro/imunologia , Reação Hospedeiro-Enxerto/imunologia , Humanos , Extremidade Inferior/patologia , Extremidade Inferior/cirurgia , Masculino , Células-Tronco Mesenquimais/citologia , Ratos , Regeneração , Traumatismo por Reperfusão/patologia , Transdução de Sinais/fisiologia , Transdução Genética
12.
Int J Cancer ; 123(7): 1704-11, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18651570

RESUMO

We evaluated the therapeutic potential of the replication competent vector VA7-EGFP, which is based on the avirulent Semliki Forest virus (SFV) strain A7 (74) carrying the EGFP marker gene in an orthotopic lung cancer tumor model in nude mice. We have previously shown that this oncolytic vector destroys tumor cells efficiently in vitro and in vivo (in subcutaneous tumor model). Tumor growth in animals with orthotopically implanted adenocarcinoma cells (A549) were monitored during the study with small animal CT. We show that locally administered virotherapy with VA7-EGFP increased survival rate in experimental lung cancer significantly (p < 0.001) comparable to results obtained with the second generation conditionally replicating adenoviral vector Ad5-Delta24TK-GFP, used for comparison. The limited efficacy in systemically administered oncolytic viruses is the essential problem in oncolytic virotherapy and also in this study we were not able to elicit significant response with systemic administration route. Despite the fact that tumor microenvironment in orthotopic lung cancer is more optimal, viruses failed to home to the tumors and were unable to initiate efficient intratumoral replication. Clearly, the efficacy of virotherapy is influenced by many factors such as the route of virus administration, immunological and physiological barriers and cancer cell-specific features (IFN-responsiveness).


Assuntos
Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Vírus da Floresta de Semliki/fisiologia , Replicação Viral , Animais , Linhagem Celular Tumoral , Efeito Citopatogênico Viral , Feminino , Camundongos , Camundongos Nus
13.
Cancer Res ; 66(14): 7185-94, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849565

RESUMO

Oncolytic viruses have gained attention as a novel form of cancer treatment. Many viral vectors in use today have been rendered safe by deletion of genes encoding viral structural proteins, thus making them unable to spread beyond the first infected cells. Hence, such replication-deficient constructs may lack efficacy. Here, we analyzed the oncolytic potential of the replication-competent vector VA7-EGFP, based on the avirulent Semliki Forest virus (SFV) strain A7(74), to kill cancer cells in culture as well as to target s.c. human melanoma xenografts in severe combined immunodeficient (SCID) mice. VA7-EGFP was able to infect most cancer cell lines studied, leading to complete lysis of the cells within 72 hours after infection. In SCID mice grafted with A2058 human melanoma, marked regression of the xenografts was observed following a single injection of 10(6) plaque-forming units of virus given either i.p., i.v., or intratumorally. Histologic analysis revealed the presence of virus not only in all treated tumors but also in the brains of the treated mice, causing progressing neuropathology beginning at day 16 after infection. Following initial oncolysis, clusters of viable tumor cells were observed embedded in connective tissue, and at later stages, encapsulated tumor nodules had formed. Infection of melanoma cells from explant cultures of these nodules revealed that a portion of the cells were resistant to virus. To be eligible for use in virotherapy, the ability of avirulent SFV to spread within tumor tissue may have to be improved and the biological safety of the virus may have to be addressed thoroughly in higher animals.


Assuntos
Melanoma/terapia , Melanoma/virologia , Terapia Viral Oncolítica/métodos , Vírus da Floresta de Semliki/fisiologia , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Cricetinae , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Melanoma/patologia , Camundongos , Camundongos SCID , Vírus da Floresta de Semliki/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Lett ; 254(2): 178-216, 2007 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-17383089

RESUMO

Oncolytic virotherapy is a promising form of gene therapy for cancer, employing nature's own agents to find and destroy malignant cells. The purpose of this review is to provide an introduction to this very topical field of research and to point out some of the current observations, insights and ideas circulating in the literature. We have strived to acknowledge as many different oncolytic viruses as possible to give a broader picture of targeting cancer using viruses. Some of the newest additions to the panel of oncolytic viruses include the avian adenovirus, foamy virus, myxoma virus, yaba-like disease virus, echovirus type 1, bovine herpesvirus 4, Saimiri virus, feline panleukopenia virus, Sendai virus and the non-human coronaviruses. Although promising, virotherapy still faces many obstacles that need to be addressed, including the emergence of virus-resistant tumor cells.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Animais , Modelos Animais de Doenças , Terapia Genética/métodos , Humanos , Neoplasias/patologia , Neoplasias/virologia , Vírus Oncolíticos/classificação , Replicação Viral
15.
Transl Oncol ; 10(5): 772-779, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28797937

RESUMO

BACKGROUND: Oncolytic adenoviruses show promise in targeting gliomas because they do not replicate in normal brain cells. However, clinical responses occur only in a subset of patients. One explanation could be the heterogenic expression level of virus receptors. Another contributing factor could be variable activity of tumor antiviral defenses in different glioma subtypes. METHODS: We established a collection of primary low-passage cell lines from different glioma subtypes (3 glioblastomas, 3 oligoastrocytomas, and 2 oligodendrogliomas) and assessed them for receptor expression and sensitivity to human adenovirus (HAd) serotypes 3, 5, and 11p. To gauge the impact of antiviral defenses, we also compared the infectivity of the oncolytic adenoviruses in interferon (IFN)-pretreated cells with IFN-sensitive Semliki Forest virus (SFV). RESULTS: Immunostaining revealed generally low expression of HAd5 receptor CAR in both primary tumors and derived cell lines. HAd11p receptor CD46 levels were maintained at moderate levels in both primary tumor samples and derived cell lines. HAd3 receptor DSG-2 was reduced in the cell lines compared to the tumors. Yet, at equal multiplicities of infection, the oncolytic potency of HAd5 in vitro in tumor-derived cells was comparable to HAd11p, whereas HAd3 lysed fewer cells than either of the other two HAd serotypes in 72 hours. IFN blocked replication of SFV, while HAds were rather unaffected. CONCLUSIONS: Adenovirus receptor levels on glioma-derived cell lines did not correlate with infection efficacy and may not be a relevant indicator of clinical oncolytic potency. Adenovirus receptor analysis should be preferentially performed on biopsies obtained perioperatively.

16.
Viral Immunol ; 17(2): 287-97, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15279706

RESUMO

Tissue expression in viral infection of immunological effector molecules may depend on virus structure or replication or both. We analyzed cytokine mRNA expression in the central nervous system (CNS) of Balb/c mice during viral infection with Semliki Forest virus (SFV) clones, which varied either in structure or virulence or both. Highly neurovirulent SFV4 effectively induced IFN-gamma, TNF-alpha, IL-6 and TGF-beta, but its avirulent derivative V4-opal with nsP3 arginine-476 to opal mutation, elicited only weak induction of these cytokines. Structurally different, avirulent rA774, obtained by cloning from avirulent SFV A7(74) strain, did not induce synthesis of proinflammatory Th1 or Th2 cytokines in murine CNS, but increased synthesis of TGF-beta transcripts. In contrast, structurally identical but moderately virulent rA774-arg virus with sense codon at opal position in nsP3, markedly stimulated synthesis of IFN-gamma, TNF-alpha, and IL-10 transcripts, without, however, reaching the levels elicited by lethal SFV4. The rA774-arg clone was more potent in attracting peripheral immune cells into the CNS than the completely avirulent strains. In conclusion, induction of proinflammatory cytokine mRNA in the CNS by SFV infection seemed to correlate with the rate of viral replication and was not significantly influenced by the virus envelope or nonstructural protein primary structure. The results also have relevance for development of CNS gene therapy vectors as SFV4 and A774 display differences in CNS infection characteristics.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Citocinas/metabolismo , Expressão Gênica , Vírus da Floresta de Semliki/fisiologia , Animais , Linhagem Celular , Citocinas/genética , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/metabolismo , RNA Viral/biossíntese , Vírus da Floresta de Semliki/genética , Proteínas Virais/biossíntese , Replicação Viral
17.
Biomedicines ; 2(2): 163-194, 2014 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-28548066

RESUMO

Oncolytic virotherapy has advanced since the days of its conception but therapeutic efficacy in the clinics does not seem to reach the same level as in animal models. One reason is premature oncolytic virus clearance in humans, which is a reasonable assumption considering the immune-stimulating nature of the oncolytic agents. However, several studies are beginning to reveal layers of restriction to oncolytic virotherapy that are present before an adaptive neutralizing immune response. Some of these barriers are present constitutively halting infection before it even begins, whereas others are raised by minute cues triggered by virus infection. Indeed, we and others have noticed that delivering viruses to tumors may not be the biggest obstacle to successful therapy, but instead the physical make-up of the tumor and its capacity to mount antiviral defenses seem to be the most important efficacy determinants. In this review, we summarize the constitutive and innate barriers to oncolytic virotherapy and discuss strategies to overcome them.

18.
J Med Chem ; 56(3): 1064-73, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23286196

RESUMO

Luciferase reporter assays are commonly used in high-throughput screening methods. Here, we report new firefly luciferase (FLuc) inhibitors based on 5-benzyl-3-phenyl-4,5-dihydroisoxazoles and 5-benzyl-3-phenyl-1,4,2-dioxazoles, which showed up as "false positives" in a luciferase reporter gene-based assay for nuclear receptor antagonists. The inhibition was shown to be noncompetitive for both natural enzyme substrates (d-luciferin and ATP) and selective to FLuc and proven to arise from a direct interaction between the enzyme and the inhibitor. Of the 63 evaluated compounds, 28 showed significantly better inhibition potency than the well-known inhibitor resveratrol (IC(50) = 59 nM), with five compounds having distinctly subnanomolar IC(50) values. The most efficient compounds inhibited the luminescence at concentrations lower than (1)/(100) in comparison to resveratrol (lowest IC(50) = 0.26 nM) and can thus be considered to belong to the most potent FLuc inhibitors reported thus far. Overall, the novel inhibitors form a unique molecular library for structure-activity relationship (SAR) analyses.


Assuntos
Azóis/química , Azóis/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Luciferases/antagonistas & inibidores , Animais , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Vaga-Lumes/enzimologia , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Modelos Moleculares
19.
PLoS One ; 5(1): e8603, 2010 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20066051

RESUMO

BACKGROUND: VA7 is a neurotropic alphavirus vector based on an attenuated strain of Semliki Forest virus. We have previously shown that VA7 exhibits oncolytic activity against human melanoma xenografts in immunodeficient mice. The purpose of this study was to determine if intravenously administered VA7 would be effective against human glioma. METHODOLOGY/PRINCIPAL FINDINGS: In vitro, U87, U251, and A172 human glioma cells were infected and killed by VA7-EGFP. In vivo, antiglioma activity of VA7 was tested in Balb/c nude mice using U87 cells stably expressing firefly luciferase in subcutaneous and orthotopic tumor models. Intravenously administered VA7-EGFP completely eradicated 100% of small and 50% of large subcutaneous U87Fluc tumors. A single intravenous injection of either VA7-EGFP or VA7 expressing Renilla luciferase (VA7-Rluc) into mice bearing orthotopic U87Fluc tumors caused a complete quenching of intracranial firefly bioluminescence and long-term survival in total 16 of 17 animals. In tumor-bearing mice injected with VA7-Rluc, transient intracranial and peripheral Renilla bioluminescence was observed. Virus was well tolerated and no damage to heart, liver, spleen, or brain was observed upon pathological assessment at three and ninety days post injection, despite detectable virus titers in these organs during the earlier time point. CONCLUSION: VA7 vector is apathogenic and can enter and destroy brain tumors in nude mice when administered systemically. This study warrants further elucidation of the mechanism of tumor destruction and attenuation of the VA7 virus.


Assuntos
Alphavirus/genética , Neoplasias Encefálicas/prevenção & controle , Vetores Genéticos , Glioma/prevenção & controle , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante Heterólogo
20.
Endocrinology ; 150(11): 5125-34, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19797403

RESUMO

Calcium entry is one of the main regulators of intracellular signaling. Here, we have described the importance of sphingosine, sphingosine kinase 1 (SK1), and sphingosine 1-phosphate (S1P) in regulating calcium entry in thyroid FRTL-5 cells. In cells incubated with the phosphatase inhibitor calyculin A, which evokes calcium entry without mobilizing sequestered intracellular calcium, sphingosine inhibited calcium entry in a concentration-dependent manner. Furthermore, inhibiting SK1 or the ATP-binding cassette ABCC1 multidrug transporter attenuated calcium entry. The addition of exogenous S1P restored calcium entry. Neither sphingosine nor inhibition of SK1 attenuated thapsigargin-evoked calcium entry. Blocking S1P receptor 2 or phospholipase C attenuated calcium entry, whereas blocking S1P receptor 3 did not. Overexpression of wild-type SK1, but not SK2, enhanced calyculin-evoked calcium entry compared with mock-transfected cells, whereas calcium entry was decreased in cells transfected with the dominant-negative G82D SK1 mutant. Exogenous S1P restored calcium entry in G82D cells. Our results suggest that the calcium entry pathway is blocked by sphingosine and that activation of SK1 and the production of S1P, through an autocrine mechanism, facilitate calcium entry through activation of S1P receptor 2. This is a novel mechanism by which the sphingosine-S1P rheostat regulates cellular calcium homeostasis.


Assuntos
Comunicação Autócrina , Cálcio/metabolismo , Lisofosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Glândula Tireoide/metabolismo , Animais , Linhagem Celular , Ratos , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA