Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
BMC Cancer ; 10: 486, 2010 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-20836854

RESUMO

BACKGROUND: Delivery of small interfering RNA (siRNA) to tumours remains a major obstacle for the development of RNA interference (RNAi)-based therapeutics. Following the promising pre-clinical and clinical results with the oncolytic herpes simplex virus (HSV) OncoVEX GM-CSF, we aimed to express RNAi triggers from oncolytic HSV, which although has the potential to improve treatment by silencing tumour-related genes, was not considered possible due to the highly oncolytic properties of HSV. METHODS: To evaluate RNAi-mediated silencing from an oncolytic HSV backbone, we developed novel replicating HSV vectors expressing short-hairpin RNA (shRNA) or artificial microRNA (miRNA) against the reporter genes green fluorescent protein (eGFP) and ß-galactosidase (lacZ). These vectors were tested in non-tumour cell lines in vitro and tumour cells that are moderately susceptible to HSV infection both in vitro and in mice xenografts in vivo. Silencing was assessed at the protein level by fluorescent microscopy, x-gal staining, enzyme activity assay, and western blotting. RESULTS: Our results demonstrate that it is possible to express shRNA and artificial miRNA from an oncolytic HSV backbone, which had not been previously investigated. Furthermore, oncolytic HSV-mediated delivery of RNAi triggers resulted in effective and specific silencing of targeted genes in tumour cells in vitro and tumours in vivo, with the viruses expressing artificial miRNA being comprehensibly more effective. CONCLUSIONS: This preliminary data provide the first demonstration of oncolytic HSV-mediated expression of shRNA or artificial miRNA and silencing of targeted genes in tumour cells in vitro and in vivo. The vectors developed in this study are being adapted to silence tumour-related genes in an ongoing study that aims to improve the effectiveness of oncolytic HSV treatment in tumours that are moderately susceptible to HSV infection and thus, potentially improve response rates seen in human clinical trials.


Assuntos
Inativação Gênica , Gliossarcoma/terapia , MicroRNAs/fisiologia , Terapia Viral Oncolítica , Interferência de RNA , RNA Interferente Pequeno/fisiologia , Simplexvirus/fisiologia , Animais , Western Blotting , Células Cultivadas , Cricetinae , Terapia Genética , Vetores Genéticos/administração & dosagem , Gliossarcoma/genética , Gliossarcoma/virologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Herpes Simples/genética , Herpes Simples/terapia , Herpes Simples/virologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
2.
Nucleic Acids Res ; 36(14): e86, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18583367

RESUMO

Considerable interest has been focused on inducing RNA interference (RNAi) in neurons to study gene function and identify new targets for disease intervention. Although small interfering RNAs (siRNAs) have been used to silence genes in neurons, in vivo delivery of RNAi remains a major challenge limiting its applications. We have developed a highly efficient method for in vivo gene silencing in dorsal root ganglia (DRG) using replication-defective herpes simplex viral (HSV-1) vectors. HSV-mediated delivery of short-hairpin RNA (shRNA) targeting reporter genes resulted in highly effective and specific silencing in neuronal and non-neuronal cells in culture and in the DRG of mice in vivo including in a transgenic mouse model. We further establish proof of concept by demonstrating in vivo silencing of the endogenous trpv1 gene. These data are the first to show silencing in DRG neurons in vivo by vector-mediated delivery of shRNA. Our results support the utility of HSV vectors for gene silencing in peripheral neurons and the potential application of this technology to the study of nociceptive processes and in pain gene target validation studies.


Assuntos
Herpesvirus Humano 1/genética , Neurônios/metabolismo , Interferência de RNA , RNA não Traduzido/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Gânglios Espinais/metabolismo , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , RNA não Traduzido/genética , Ratos , Canais de Cátion TRPV/genética
3.
J Immunother Cancer ; 8(2)2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32759235

RESUMO

BACKGROUND: The aggressive clinical behavior of poorly differentiated and anaplastic thyroid cancers (PDTC and ATC) has proven challenging to treat, and survival beyond a few months from diagnosis is rare. Although 30%-60% of these tumors contain mutations in the BRAF gene, inhibitors designed specifically to target oncogenic BRAF have shown limited and only short-lasting therapeutic benefits as single agents, thus highlighting the need for improved treatment strategies, including novel combinations. METHODS: Using a BRAFV600E-driven mouse model of ATC, we investigated the therapeutic efficacy of the combination of BRAF inhibition and oncolytic herpes simplex virus (oHSV). Analyses of samples from tumor-bearing mice were performed to immunologically characterize the effects of different treatments. These immune data were used to inform the incorporation of immune checkpoint inhibitors into triple combination therapies. RESULTS: We characterized the immune landscape in vivo following BRAF inhibitor treatment and detected only modest immune changes. We, therefore, hypothesized that the addition of oncolytic virotherapy to BRAF inhibition in thyroid cancer would create a more favorable tumor immune microenvironment, boost the inflammatory status of tumors and improve BRAF inhibitor therapy. First, we showed that thyroid cancer cells were susceptible to infection with oHSV and that this process was associated with activation of the immune tumor microenvironment in vivo. Next, we showed improved therapeutic responses when combining oHSV and BRAF inhibition in vivo, although no synergistic effects were seen in vitro, further confirming that the dominant effect of oHSV in this context was likely immune-mediated. Importantly, both gene and protein expression data revealed an increase in activation of T cells and natural killer (NK) cells in the tumor in combination-treated samples. The benefit of combination oHSV and BRAF inhibitor therapy was abrogated when T cells or NK cells were depleted in vivo. In addition, we showed upregulation of PD-L1 and CTLA-4 following combined treatment and demonstrated that blockade of the PD-1/PD-L1 axis or CTLA-4 further improved combination therapy. CONCLUSIONS: The combination of oHSV and BRAF inhibition significantly improved survival in a mouse model of ATC by enhancing immune-mediated antitumor effects, and triple combination therapies, including either PD-1 or CTLA-4 blockade, further improved therapy.


Assuntos
Terapia Viral Oncolítica/métodos , Proteínas Proto-Oncogênicas B-raf/metabolismo , Neoplasias da Glândula Tireoide/genética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Herpesvirus Humano 1/patogenicidade , Humanos , Masculino , Camundongos , Neoplasias da Glândula Tireoide/patologia
4.
Methods Mol Biol ; 542: 551-64, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19565922

RESUMO

A large number of oncolytic viral vectors are currently under clinical development for cancer therapy. Herpes simplex virus type 1 (HSV-1) has demonstrated particular promise in this field, showing genetically engineered selective tumor replication and cytotoxicity in a wide variety of tumor types, without damaging healthy tissues. Enhanced activity has been observed when a range of therapeutic genes has been inserted into various oncolytic HSV genomes. Here, we discuss methods used to develop and characterize an oncolytic HSV virus that combines expression of a highly potent prodrug activating gene (yeast cytosine deaminase/uracil phosphoribosyltransferase fusion [Fcy::Fur]) and the fusogenic glycoprotein from gibbon ape leukemia virus (GALV) for enhanced local tumor control.


Assuntos
Vetores Genéticos/genética , Glicoproteínas/metabolismo , Biologia Molecular/métodos , Neoplasias/terapia , Vírus Oncolíticos/genética , Pró-Fármacos/farmacologia , Simplexvirus/genética , Animais , Morte Celular , Linhagem Celular Tumoral , Clonagem Molecular , Humanos , Plasmídeos , Proteínas Recombinantes de Fusão , Saccharomyces cerevisiae
5.
J Immunother Cancer ; 7(1): 214, 2019 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-31399043

RESUMO

BACKGROUND: Oncolytic viruses preferentially replicate in tumors as compared to normal tissue and promote immunogenic cell death and induction of host systemic anti-tumor immunity. HSV-1 was chosen for further development as an oncolytic immunotherapy in this study as it is highly lytic, infects human tumor cells broadly, kills mainly by necrosis and is a potent activator of both innate and adaptive immunity. HSV-1 also has a large capacity for the insertion of additional, potentially therapeutic, exogenous genes. Finally, HSV-1 has a proven safety and efficacy profile in patients with cancer, talimogene laherparepvec (T-VEC), an oncolytic HSV-1 which expresses GM-CSF, being the only oncolytic immunotherapy approach that has received FDA approval. As the clinical efficacy of oncolytic immunotherapy has been shown to be further enhanced by combination with immune checkpoint inhibitors, developing improved oncolytic platforms which can synergize with other existing immunotherapies is a high priority. In this study we sought to further optimize HSV-1 based oncolytic immunotherapy through multiple approaches to maximize: (i) the extent of tumor cell killing, augmenting the release of tumor antigens and danger-associated molecular pattern (DAMP) factors; (ii) the immunogenicity of tumor cell death; and (iii) the resulting systemic anti-tumor immune response. METHODS: To sample the wide diversity amongst clinical strains of HSV-1, twenty nine new clinical strains isolated from cold sores from otherwise healthy volunteers were screened across a panel of human tumor cell lines to identify the strain with the most potent tumor cell killing ability, which was then used for further development. Following deletion of the genes encoding ICP34.5 and ICP47 to provide tumor selectivity, the extent of cell killing and the immunogenicity of cell death was enhanced through insertion of a gene encoding a truncated, constitutively highly fusogenic form of the envelope glycoprotein of gibbon ape leukemia virus (GALV-GP-R-). A number of further armed derivatives of this virus were then constructed intended to further enhance the anti-tumor immune response which was generated following fusion-enhanced, oncolytic virus replication-mediated cell death. These viruses expressed GMCSF, an anti-CTLA-4 antibody-like molecule, CD40L, OX40L and/or 4-1BB, each of which is expected to act predominantly at the site and time of immune response initiation. Expression of these proteins was confirmed by ELISA and/or western blotting. Immunogenic cell death was assessed by measuring the levels of HMGB1 and ATP from cell free supernatants from treated cells, and by measuring the surface expression of calreticulin. GALV-GP-R- mediated cell to cell fusion and killing was tested in a range of tumor cell lines in vitro. Finally, the in vivo therapeutic potential of these viruses was tested using human A549 (lung cancer) and MDA-MB-231(breast cancer) tumor nude mouse xenograft models and systemic anti-tumor effects tested using dual flank syngeneic 4434 (melanoma), A20 (lymphoma) mouse tumor models alone and in combination with a murine anti-PD1 antibody, and 9 L (gliosarcoma) tumors in rats. RESULTS: The twenty nine clinical strains of HSV-1 isolated and tested demonstrated a broad range of tumor cell killing abilities allowing the most potent strain to be identified which was then used for further development. Oncolytic ability was demonstrated to be further augmented by the expression of GALV-GP-R- in a range of tumor cell lines in vitro and in mouse xenograft models in nude mice. The expression of GALV-GP-R- was also demonstrated to lead to enhanced immunogenic cell death in vitro as confirmed by the increased release of HMGB1 and ATP and increased levels of calreticulin on the cell surface. Experiments using the rat 9 L syngeneic tumor model demonstrated that GALV-GP-R- expression increased abscopal uninjected (anenestic) tumor responses and data using mouse 4434 tumors demonstrated that virus treatment increased CD8+ T cell levels both in the injected and uninjected tumor, and also led to increased expression of PD-L1. A combination study using varying doses of a virus expressing GALV-GP-R- and mGM-CSF and an anti-murine PD1 antibody showed enhanced anti-tumor effects with the combination which was most evident at low virus doses, and also lead to immunological memory. Finally, treatment of mice with derivatives of this virus which additionally expressed anti-mCTLA-4, mCD40L, m4-1BBL, or mOX40L demonstrated enhanced activity, particularly in uninjected tumors. CONCLUSION: The new HSV-1 based platform described provides a potent and versatile approach to developing new oncolytic immunotherapies for clinical use. Each of the modifications employed was demonstrated to aid in optimizing the potential of the virus to both directly kill tumors and to lead to systemic therapeutic benefit. For clinical use, these viruses are expected to be most effective in combination with other anti-cancer agents, in particular PD1/L1-targeted immune checkpoint blockade. The first virus from this program (expressing GALV-GP-R- and hGM-CSF) has entered clinical development alone and in combination with anti-PD1 therapy in a number of tumor types (NCT03767348).


Assuntos
Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1/patogenicidade , Imunoterapia/métodos , Terapia Viral Oncolítica/métodos , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus
6.
Cancer Res ; 66(9): 4835-42, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16651439

RESUMO

We have previously developed an oncolytic herpes simplex virus-1 based on a clinical virus isolate, which was deleted for ICP34.5 to provide tumor selected replication and ICP47 to increase antigen presentation as well as tumor selective virus replication. A phase I/II clinical trial using a version of this virus expressing granulocyte macrophage colony-stimulating factor has shown promising results. The work reported here aimed to develop a version of this virus in which local tumor control was further increased through the combined expression of a highly potent prodrug activating gene [yeast cytosine deaminase/uracil phospho-ribosyltransferase fusion (Fcy::Fur)] and the fusogenic glycoprotein from gibbon ape leukemia virus (GALV), which it was hoped would aid the spread of the activated prodrug through the tumor. Viruses expressing the two genes individually or in combination were constructed and tested, showing (a) GALV and/or Fcy::Fur expression did not affect virus growth; (b) GALV expression causes cell fusion and increases the tumor cell killing at least 30-fold in vitro and tumor shrinkage 5- to 10-fold in vivo; (c) additional expression of Fcy::Fur combined with 5-fluorocytosine administration improves tumor shrinkage further. These results indicate, therefore, that the combined expression of the GALV protein and Fcy::Fur provides a highly potent oncolytic virus with improved capabilities for local tumor control. It is intended to enter the GALV/Fcy::Fur expressing virus into clinical development for the treatment of tumor types, such as pancreatic or lung cancer, where local control would be anticipated to be clinically advantageous.


Assuntos
Fibrossarcoma/terapia , Flucitosina/farmacocinética , Vírus da Leucemia do Macaco Gibão/genética , Glicoproteínas de Membrana/genética , Terapia Viral Oncolítica/métodos , Simplexvirus/fisiologia , Animais , Biotransformação , Fusão Celular , Terapia Combinada , Citosina Desaminase/biossíntese , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Fibrossarcoma/virologia , Flucitosina/farmacologia , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Humanos , Vírus da Leucemia do Macaco Gibão/metabolismo , Vírus da Leucemia do Macaco Gibão/fisiologia , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Pentosiltransferases/biossíntese , Pentosiltransferases/genética , Pentosiltransferases/metabolismo , Pró-Fármacos/farmacocinética , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Simplexvirus/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Immunother Cancer ; 6(1): 78, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-30081947

RESUMO

In this White Paper, we discuss the current state of microbial cancer therapy. This paper resulted from a meeting ('Microbial Based Cancer Therapy') at the US National Cancer Institute in the summer of 2017. Here, we define 'Microbial Therapy' to include both oncolytic viral therapy and bacterial anticancer therapy. Both of these fields exploit tumor-specific infectious microbes to treat cancer, have similar mechanisms of action, and are facing similar challenges to commercialization. We designed this paper to nucleate this growing field of microbial therapeutics and increase interactions between researchers in it and related fields. The authors of this paper include many primary researchers in this field. In this paper, we discuss the potential, status and opportunities for microbial therapy as well as strategies attempted to date and important questions that need to be addressed. The main areas that we think will have the greatest impact are immune stimulation, control of efficacy, control of delivery, and safety. There is much excitement about the potential of this field to treat currently intractable cancer. Much of the potential exists because these therapies utilize unique mechanisms of action, difficult to achieve with other biological or small molecule drugs. By better understanding and controlling these mechanisms, we will create new therapies that will become integral components of cancer care.


Assuntos
Bactérias , Terapia Biológica/métodos , Vetores Genéticos , Neoplasias/prevenção & controle , Neoplasias/terapia , Vírus , Animais , Bactérias/genética , Terapia Biológica/normas , Terapia Biológica/tendências , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Estudos Clínicos como Assunto , Terapia Combinada , Avaliação Pré-Clínica de Medicamentos , Engenharia Genética , Vetores Genéticos/genética , Humanos , Neoplasias/etiologia , Terapia Viral Oncolítica , Resultado do Tratamento , Vírus/genética
8.
Clin Cancer Res ; 12(22): 6853-62, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17121907

RESUMO

PURPOSE: Oncolytic herpes simplex virus type 1 (HSV-1) vectors show considerable promise as agents for cancer therapy. We have developed a novel recombinant HSV-1 virus (JS1/34.5-/47-) for purging of occult breast cancer cells from bone marrow of patients. Here, we evaluate the therapeutic efficacy of this oncolytic virus. EXPERIMENTAL DESIGN: Electron microscopy was used to determine whether human breast cancer and bone marrow cells are permissive for JS1/34.5-/47- infection. Subsequently, the biological effects of JS1/34.5-/47- infection on human breast cancer cells and bone marrow were established using cell proliferation and colony formation assays, and the efficiency of cell kill was evaluated. Finally, the efficiency of JS1/34.5-/47- purging of breast cancer cells was examined in cocultures of breast cancer cells with bone marrow as well as bone marrow samples from high-risk breast cancer patients. RESULTS: We show effective killing of human breast cancer cell lines with the JS1/34.5-/47- virus. Furthermore, we show that treatment with JS1/34.5-/47- can significantly inhibit the growth of breast cancer cell lines without affecting cocultured mononuclear hematopoietic cells. Finally, we have found that the virus is effective in destroying disseminated tumors cells in bone marrow taken from breast cancer patients, without affecting the hematopoietic contents in these samples. CONCLUSION: Collectively, our data show that the JS1/34.5-/47- virus can selectively target breast cancer cells while sparing hematopoietic cells, suggesting that JS1/34.5-/47- can be used to purge contaminating breast cancer cells from human bone marrow in the setting of autologous hematopoietic cell transplantation.


Assuntos
Adenocarcinoma/patologia , Medula Óssea/patologia , Neoplasias da Mama/patologia , Herpesvirus Humano 1 , Vírus Oncolíticos/fisiologia , Adenocarcinoma/virologia , Biomarcadores Tumorais/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/virologia , Neoplasias da Mama/terapia , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Citometria de Fluxo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 1/fisiologia , Humanos , Queratina-19/metabolismo , Glândulas Mamárias Humanas/patologia , Glândulas Mamárias Humanas/virologia , Terapia Viral Oncolítica/efeitos adversos
9.
Clin Cancer Res ; 12(22): 6737-47, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17121894

RESUMO

PURPOSE: To conduct a phase I clinical trial with a second-generation oncolytic herpes simplex virus (HSV) expressing granulocyte macrophage colony-stimulating factor (Onco VEXGM-CSF) to determine the safety profile of the virus, look for evidence of biological activity, and identify a dosing schedule for later studies. EXPERIMENTAL DESIGN: The virus was administered by intratumoral injection in patients with cutaneous or s.c. deposits of breast, head and neck and gastrointestinal cancers, and malignant melanoma who had failed prior therapy. Thirteen patients were in a single-dose group, where doses of 10(6), 10(7), and 10(8) plaque-forming units (pfu)/mL were tested, and 17 patients were in a multidose group testing a number of dose regimens. RESULTS: The virus was generally well tolerated with local inflammation, erythema, and febrile responses being the main side effects. The local reaction to injection was dose limiting in HSV-seronegative patients at 10(7) pfu/mL. The multidosing phase thus tested seroconverting HSV-seronegative patients with 10(6) pfu/mL followed by multiple higher doses (up to 10(8) pfu/mL), which was well tolerated by all patients. Biological activity (virus replication, local reactions, granulocyte macrophage colony-stimulating factor expression, and HSV antigen-associated tumor necrosis), was observed. The duration of local reactions and virus replication suggested that dosing every 2 to 3 weeks was appropriate. Nineteen of 26 patient posttreatment biopsies contained residual tumor of which 14 showed tumor necrosis, which in some cases was extensive, or apoptosis. In all cases, areas of necrosis also strongly stained for HSV. The overall responses to treatment were that three patients had stable disease, six patients had tumors flattened (injected and/or uninjected lesions), and four patients showed inflammation of uninjected as well as the injected tumor, which, in nearly all cases, became inflamed. CONCLUSIONS: Onco VEXGM-CSF is well tolerated and can be safely administered using the multidosing protocol described. Evidence of an antitumor effect was seen.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Simplexvirus , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antinucleares/sangue , Biópsia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Carcinoma/patologia , Carcinoma/terapia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Citocinas/sangue , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Masculino , Melanoma/patologia , Melanoma/terapia , Pessoa de Meia-Idade , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos , Proteínas Recombinantes , Simplexvirus/imunologia , Simplexvirus/metabolismo , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Resultado do Tratamento
10.
J Cereb Blood Flow Metab ; 26(9): 1176-88, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16421510

RESUMO

Occlusive cerebrovascular disease leads to brain ischemia that causes neurological deficits. Here we introduce a new strategy combining mesenchymal stromal cells (MSCs) and ex vivo hepatocyte growth factor (HGF) gene transferring with a multimutated herpes simplex virus type-1 vector in a rat transient middle cerebral artery occlusion (MCAO) model. Gene-transferred MSCs were intracerebrally transplanted into the rats' ischemic brains at 2 h (superacute) or 24 h (acute) after MCAO. Behavioral tests showed significant improvement of neurological deficits in the HGF-transferred MSCs (MSC-HGF)-treated group compared with the phosphate-buffered saline (PBS)-treated and MSCs-only-treated group. The significant difference of infarction areas on day 3 was detected only between the MSC-HGF group and the PBS group with the superacute treatment, but was detected among each group on day 14 with both transplantations. After the superacute transplantation, we detected abundant expression of HGF protein in the ischemic brain of the MSC-HGF group compared with others on day 1 after treatment, and it was maintained for at least 2 weeks. Furthermore, we determined that the increased expression of HGF was derived from the transferred HGF gene in gene-modified MSCs. The percentage of apoptosis-positive cells in the ischemic boundary zone (IBZ) was significantly decreased, while that of remaining neurons in the cortex of the IBZ was significantly increased in the MSC-HGF group compared with others. The present study shows that combined therapy is more therapeutically efficient than MSC cell therapy alone, and it may extend the therapeutic time window from superacute to acute phase.


Assuntos
Transplante de Medula Óssea/métodos , Terapia Genética , Fator de Crescimento de Hepatócito/genética , Herpesvirus Humano 1/genética , Acidente Vascular Cerebral/terapia , Células Estromais/transplante , Animais , Apoptose/fisiologia , Encéfalo/patologia , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Fator de Crescimento de Hepatócito/biossíntese , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média/patologia , Artéria Cerebral Média/fisiologia , Ratos , Ratos Wistar , Acidente Vascular Cerebral/patologia , Resultado do Tratamento
11.
Stroke ; 36(12): 2725-30, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16282547

RESUMO

BACKGROUND AND PURPOSE: Fibroblast growth factor-2 (FGF-2) administration and bone marrow stromal cell (MSC) transplantation could improve neurological deficits after occlusive cerebrovascular disease. In the present study, we examined the effects of neurological improvement after transient middle cerebral artery occlusion (MCAO) in rats by a novel therapeutic strategy with FGF-2 gene-transferred MSCs by the herpes simplex virus type 1 (HSV-1) vector. METHODS: Adult Wistar rats were anesthetized. Nonmodified MSCs, FGF-2-modified MSCs with HSV-1 1764/-4/pR19/ssIL2-FGF-2, or PBS was administered intracerebrally 24 hours after transient right MCAO. All animals underwent behavioral tests for 21 days, and the infarction volume with 2-3-5-triphenylterazolium was detected 3 days and 14 days after the MCAO. Three days and 7 days after the MCAO, the FGF-2 production in the ipsilateral hemisphere of the MCAO was measured with ELISA. Seven and 14 days after the MCAO, immunohistochemical staining for FGF-2 was applied. RESULTS: The stroke animals receiving FGF-2-modified MSCs demonstrated significant functional recovery compared with the other groups. Fourteen days after the MCAO, there was a significant reduction in infarction volume only in FGF-2-modified MSC-treated group. FGF-2 production in the FGF-2-modified MSC-treated brain was significantly higher compared with the other groups at 3 and 7 days after MCAO. Administrated FGF-2-modified MSCs strongly expressed the FGF-2 protein, which was proven by ELISA. CONCLUSIONS: Our data suggest that the FGF-2 gene-modified MSCs with the HSV-1 vector can contribute to remarkable functional recovery after stroke compared with MSCs transplantation alone.


Assuntos
Transplante de Medula Óssea , Fator 2 de Crescimento de Fibroblastos/metabolismo , Vetores Genéticos/administração & dosagem , Herpesvirus Humano 1/imunologia , Ataque Isquêmico Transitório/terapia , Células Estromais/transplante , Animais , Encéfalo/metabolismo , Células Cultivadas , Técnicas de Transferência de Genes , Herpesvirus Humano 1/genética , Imuno-Histoquímica , Ataque Isquêmico Transitório/metabolismo , Ratos , Ratos Wistar , Extratos de Tecidos/metabolismo
12.
Curr Opin Virol ; 13: 93-100, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26121656

RESUMO

Viruses have been suggested to be useful as anti-cancer agents since the early 20th century, although following the advent of chemotherapy and radiotherapy work largely stopped until the 1990s when a number of groups began to explore the use of engineered viruses. This overview summarizes the development of the field from the 1990s to the present day, an era when oncolytic viruses have now demonstrated clear clinical benefit to patients. The hurdles and challenges which needed to be overcome are discussed, and in particular the importance of the immune component in achieving a therapeutic effect is highlighted. Today, oncolytic therapy is generally thought of as an immunotherapy, the term 'oncolytic immunotherapy' having been widely adopted. With the advent of immuno-oncology drugs based on immune checkpoint blockade, a clear rationale for synergy between the two approaches, and initial pre-clinical and clinical data suggesting this to be the case, it might be expected that oncolytic immunotherapy combined with checkpoint blockade will provide a cornerstone of future cancer treatment.


Assuntos
Imunoterapia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Animais , Humanos , Neoplasias/imunologia , Neoplasias/virologia , Vírus Oncolíticos/genética
13.
J Clin Oncol ; 33(25): 2780-8, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26014293

RESUMO

PURPOSE: Talimogene laherparepvec (T-VEC) is a herpes simplex virus type 1-derived oncolytic immunotherapy designed to selectively replicate within tumors and produce granulocyte macrophage colony-stimulating factor (GM-CSF) to enhance systemic antitumor immune responses. T-VEC was compared with GM-CSF in patients with unresected stage IIIB to IV melanoma in a randomized open-label phase III trial. PATIENTS AND METHODS: Patients with injectable melanoma that was not surgically resectable were randomly assigned at a two-to-one ratio to intralesional T-VEC or subcutaneous GM-CSF. The primary end point was durable response rate (DRR; objective response lasting continuously ≥ 6 months) per independent assessment. Key secondary end points included overall survival (OS) and overall response rate. RESULTS: Among 436 patients randomly assigned, DRR was significantly higher with T-VEC (16.3%; 95% CI, 12.1% to 20.5%) than GM-CSF (2.1%; 95% CI, 0% to 4.5%]; odds ratio, 8.9; P < .001). Overall response rate was also higher in the T-VEC arm (26.4%; 95% CI, 21.4% to 31.5% v 5.7%; 95% CI, 1.9% to 9.5%). Median OS was 23.3 months (95% CI, 19.5 to 29.6 months) with T-VEC and 18.9 months (95% CI, 16.0 to 23.7 months) with GM-CSF (hazard ratio, 0.79; 95% CI, 0.62 to 1.00; P = .051). T-VEC efficacy was most pronounced in patients with stage IIIB, IIIC, or IVM1a disease and in patients with treatment-naive disease. The most common adverse events (AEs) with T-VEC were fatigue, chills, and pyrexia. The only grade 3 or 4 AE occurring in ≥ 2% of T-VEC-treated patients was cellulitis (2.1%). No fatal treatment-related AEs occurred. CONCLUSION: T-VEC is the first oncolytic immunotherapy to demonstrate therapeutic benefit against melanoma in a phase III clinical trial. T-VEC was well tolerated and resulted in a higher DRR (P < .001) and longer median OS (P = .051), particularly in untreated patients or those with stage IIIB, IIIC, or IVM1a disease. T-VEC represents a novel potential therapy for patients with metastatic melanoma.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Antineoplásicos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos dos fármacos , Herpesvirus Humano 1 , Imunoterapia/métodos , Melanoma/tratamento farmacológico , Melanoma/imunologia , Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Calafrios/induzido quimicamente , Fadiga/induzido quimicamente , Feminino , Febre/induzido quimicamente , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Injeções Intralesionais , Masculino , Melanoma/mortalidade , Melanoma/prevenção & controle , Melanoma/secundário , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Razão de Chances , Neoplasias Cutâneas/mortalidade , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/prevenção & controle , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento , Estados Unidos/epidemiologia
14.
BMC Neurosci ; 5: 9, 2004 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-15113454

RESUMO

BACKGROUND: Many changes in gene expression occur in distal stumps of injured nerves but the transcriptional control of these events is poorly understood. We have examined the expression of the transcription factors ATF3 and c-Jun by non-neuronal cells during Wallerian degeneration following injury to sciatic nerves, dorsal roots and optic nerves of rats and mice, using immunohistochemistry and in situ hybridization. RESULTS: Following sciatic nerve injury--transection or transection and reanastomosis--ATF3 was strongly upregulated by endoneurial, but not perineurial cells, of the distal stumps of the nerves by 1 day post operation (dpo) and remained strongly expressed in the endoneurium at 30 dpo when axonal regeneration was prevented. Most ATF3+ cells were immunoreactive for the Schwann cell marker, S100. When the nerve was transected and reanastomosed, allowing regeneration of axons, most ATF3 expression had been downregulated by 30 dpo. ATF3 expression was weaker in the proximal stumps of the injured nerves than in the distal stumps and present in fewer cells at all times after injury. ATF3 was upregulated by endoneurial cells in the distal stumps of injured neonatal rat sciatic nerves, but more weakly than in adult animals. ATF3 expression in transected sciatic nerves of mice was similar to that in rats. Following dorsal root injury in adult rats, ATF3 was upregulated in the part of the root between the lesion and the spinal cord (containing Schwann cells), beginning at 1 dpo, but not in the dorsal root entry zone or in the degenerating dorsal column of the spinal cord. Following optic nerve crush in adult rats, ATF3 was found in some cells at the injury site and small numbers of cells within the optic nerve displayed weak immunoreactivity. The pattern of expression of c-Jun in all types of nerve injury was similar to that of ATF3. CONCLUSION: These findings raise the possibility that ATF3/c-Jun heterodimers may play a role in regulating changes in gene expression necessary for preparing the distal segments of injured peripheral nerves for axonal regeneration. The absence of the ATF3 and c-Jun from CNS glia during Wallerian degeneration may limit their ability to support regeneration.


Assuntos
Sistema Nervoso Central/metabolismo , Neuroglia/metabolismo , Nervos Periféricos/metabolismo , Fatores de Transcrição/metabolismo , Degeneração Walleriana/metabolismo , Fator 3 Ativador da Transcrição , Animais , Sistema Nervoso Central/patologia , Dimerização , Feminino , Masculino , Camundongos , Compressão Nervosa , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Regeneração Nervosa/genética , Neuroglia/patologia , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Nervos Periféricos/patologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Raízes Nervosas Espinhais/metabolismo , Raízes Nervosas Espinhais/patologia , Regulação para Cima/genética , Degeneração Walleriana/patologia
15.
Brain Res Mol Brain Res ; 111(1-2): 91-103, 2003 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-12654509

RESUMO

Heat shock proteins are expressed in response to cellular stress and can protect cells from further stress and facilitate recovery. Heat shock protein 27 is of particular interest because it has been implicated in a range of protective roles including protein chaperoning, stabilising elements of the cytoskeleton and as an active inhibitor of apoptosis. In the present study, we have examined the potential of administration of exogenous HSP27 to confer protection against KA-induced neuronal cell death in vivo. We aimed to exploit the neurotropic specificity of herpes simplex virus-1 based virus vectors, which have been rendered replication-incompetent, to infect neurons of the hippocampus. The systemic administration of kainic acid, an analogue of glutamate, causes seizures resulting in neuronal damage and is an established animal model of epilepsy. Neuron loss is particularly prominent in the hippocampus and the mode of death is at least partly apoptotic in nature. We show that the overexpression of HSP27 in these neurons can significantly augment their survival following kainic acid administration. In contrast, injection of a control virus expressing beta-galactosidase does not confer protection. This is the first time that protection by exogenously expressed HSP27 has been demonstrated in an in vivo model of neuronal cell death.


Assuntos
Morte Celular/efeitos dos fármacos , Epilepsia/tratamento farmacológico , Vetores Genéticos/farmacologia , Proteínas de Choque Térmico/genética , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Estresse Fisiológico/tratamento farmacológico , Animais , Contagem de Células , Morte Celular/fisiologia , Modelos Animais de Doenças , Epilepsia/genética , Epilepsia/metabolismo , Vetores Genéticos/uso terapêutico , Proteínas de Choque Térmico/uso terapêutico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Ácido Caínico/farmacologia , Masculino , Degeneração Neural/genética , Degeneração Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/farmacologia , Ratos , Ratos Sprague-Dawley , Simplexvirus/genética , Simplexvirus/metabolismo , Simplexvirus/patogenicidade , Estresse Fisiológico/genética , Estresse Fisiológico/metabolismo , Resultado do Tratamento
16.
Biotechnol Prog ; 19(1): 209-15, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12573027

RESUMO

Despite continuous improvements in culturing and recovery techniques, high-titer stocks of purified disabled herpes simplex virus type-1 (HSV-1 DIS) vector for drug discovery and use in preclinical and clinical trials are currently difficult to achieve. Efforts to improve their centrifugal recovery have been addressed in this paper. The operation of a swing-out centrifuge rotor was assessed, and its operational conditions were defined for the recovery of viable HSV-1 DIS. 80% virus recovery was achieved after 90 min at 26000g. The 20% loss of virus was attributed to damage to the viral envelope by overcompaction of the pellet and impaction with the base of the centrifuge tube. Virus recovery was increased by a further 10% by using a fixed-angle centrifuge rotor operating at 26000g. Plaque assays of recovered HSV-1 DIS gave values on the order of 10(6) pfu/mL, compared to values typically above 10(9) pfu/mL obtained for the replication-competent HSV-1 viron.


Assuntos
Centrifugação/instrumentação , Herpesvirus Humano 1/isolamento & purificação , Rim/virologia , Animais , Separação Celular/instrumentação , Separação Celular/métodos , Células Cultivadas , Centrifugação/métodos , Cricetinae , Desenho de Equipamento , Análise de Falha de Equipamento , Herpesvirus Humano 1/fisiologia , Humanos , Rim/fisiologia , Projetos Piloto , Sensibilidade e Especificidade , Cultura de Vírus/instrumentação , Cultura de Vírus/métodos , Inativação de Vírus
17.
Oncolytic Virother ; 3: 11-20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-27512660

RESUMO

Oncolytic viruses that selectively lyse tumor cells with minimal damage to normal cells are a new area of therapeutic development in oncology. An attenuated herpesvirus encoding the granulocyte-macrophage colony stimulating factor (GM-CSF), known as talimogene laherparepvec (T-VEC), has been identified as an attractive oncolytic virus for cancer therapy based on preclinical tumor studies and results from early-phase clinical trials and a large randomized Phase III study in melanoma. In this review, we discuss the basic biology of T-VEC, describe the role of GM-CSF as an immune adjuvant, summarize the preclinical data, and report the outcomes of published clinical trials using T-VEC. The emerging data suggest that T-VEC is a safe and potentially effective antitumor therapy in malignant melanoma and represents the first oncolytic virus to demonstrate therapeutic activity against human cancer in a randomized, controlled Phase III study.

18.
Expert Opin Biol Ther ; 10(1): 89-103, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20420517

RESUMO

IMPORTANCE OF THE FIELD: Pain is a hugely important area of research attracting considerable academic and commercial interest. However, the application of RNA interference (RNAi) to the study of nociceptive processes and the development of new analgesics has been limited by the specific challenges associated with the delivery of RNAi triggers to the cell bodies of sensory neurons in the dorsal root ganglia (DRG). AREAS COVERED IN THIS REVIEW: In the past five years, delivery of small-interfering RNA (siRNA) to the DRG and spinal cord has achieved effective and specific silencing of targeted genes in various animal models of pain. However, delivery of short-hairpin RNA (shRNA) or artificial microRNA (miRNA) to sensory neurons in vivo has not been feasible using most delivery systems currently available. WHAT THE READER WILL GAIN: Replication-defective vectors based on herpes simplex virus (HSV), which are particularly efficient at targeting DRG neurons, have been recently engineered to express shRNA and artificial miRNA. Whilst silencing induced by siRNA is transient and requires relatively high doses of silencing triggers, HSV-mediated expression of shRNA/miRNA in sensory neurons allows silencing of targeted genes for at least one week following a single injection. TAKE HOME MESSAGE: The potential to use inducible or tissue-specific promoters and to simultaneously silence multiple gene targets, in addition to recent studies suggesting that artificial miRNAs may have improved safety profiles, hold clear advantages for the use of miRNA-based vectors for gene silencing in sensory neurons.


Assuntos
Inativação Gênica , Manejo da Dor , Interferência de RNA , Células Receptoras Sensoriais/metabolismo , Simplexvirus/genética , Animais , Terapia Genética , Humanos , MicroRNAs/fisiologia , Dor/genética , RNA Interferente Pequeno/farmacologia , Replicação Viral
19.
Arch Otolaryngol Head Neck Surg ; 136(2): 151-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20157061

RESUMO

OBJECTIVE: To determine if prodrug conversion of fluorocytosine to fluorouracil by an engineered herpes virus, OncoVEX(GALV/CD), enhances oncolytic therapy of head and neck squamous cell carcinoma. DESIGN: We assessed the ability of OncoVEX(GALV/CD) and OncoVEX(GFP) to infect, replicate within, and lyse 4 head and neck squamous cell carcinoma lines in vitro. The effects of adding fluorocytosine with OncoVEX(GALV/CD) were evaluated. RESULTS: Head and neck squamous cell carcinoma was permissive to green fluorescent protein expression in100% of cells by OncoVEX(GFP) at a multiplicity of infection of 1 after 48 hours and supported logarithmic viral replication. Virus caused more than 60% cell death 6 days after exposure to virus at a multiplicity of infection of 0.1 in 3 of the 4 cell lines. Fluorocytosine did not enhance cytotoxicity induced by OncoVEX(GALV/CD) at a multiplicity of infection of 0.1. However, for the least-sensitive SCC25 cell line, virus at a multiplicity of infection of 0.01 was cytotoxic to only 4% of cells after 6 days but was cytotoxic to 35% of cells with fluorocytosine. CONCLUSIONS: OncoVEX(GALV/CD) efficiently infects, replicates within, and lyses head and neck squamous cell carcinoma at relatively low viral doses. Prodrug conversion by cytosine deaminase did not enhance therapy at viral doses that cause efficient cytotoxicity but may have beneficial effects in less-sensitive cell lines at low viral doses.


Assuntos
Carcinoma de Células Escamosas/terapia , Flucitosina/uso terapêutico , Neoplasias de Cabeça e Pescoço/terapia , Herpesvirus Humano 1/genética , Terapia Viral Oncolítica , Pró-Fármacos/uso terapêutico , Linhagem Celular Tumoral , Citosina Desaminase/biossíntese , Técnicas de Transferência de Genes , Engenharia Genética , Vetores Genéticos , Humanos , Vírus da Leucemia do Macaco Gibão/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa