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










Base de dados
Intervalo de ano de publicação
1.
Vaccines (Basel) ; 11(12)2023 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-38140209

RESUMO

The development of effective cancer vaccines remains a significant challenge due to immune tolerance and limited clinical benefits. Oncolytic herpes simplex virus type 1 (oHSV-1) has shown promise as a cancer therapy, but efficacy is often limited in advanced cancers. In this study, we constructed and characterized a novel oHSV-1 virus (VG22401) expressing the human epidermal growth factor receptor 2 (HER2), a transmembrane glycoprotein overexpressed in many carcinomas. VG22401 exhibited efficient replication and HER2 payload expression in both human and mouse colorectal cancer cells. Mice immunized with VG22401 showed significant binding of serum anti-HER2 antibodies to HER2-expressing tumor cells, inducing antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Furthermore, mice primed with VG22401 and intratumorally boosted with the same virus showed enhanced antitumor efficacy in a bilateral syngeneic HER2(+) tumor model, compared to HER2-null backbone virus. This effect was accompanied by the induction of anti-HER2 T cell responses. Our findings suggest that peripheral priming with HER2-expressing oHSV-1 followed by an intratumoral boost with the same virus can significantly enhance antitumor immunity and efficacy, presenting a promising strategy for cancer immunotherapy.

2.
Mol Ther Oncolytics ; 28: 334-348, 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36938544

RESUMO

VG2025 is a recombinant oncolytic herpes simplex virus type 1 (HSV-1) that uses transcriptional and translational dual regulation (TTDR) of critical viral genes to enhance virus safety and promote tumor-specific virus replication without reducing virulence. The TTDR platform is based on transcriptional control of the essential HSV-1 immediate-early protein ICP27 using a tumor-specific carcinoembryonic antigen (CEA) promoter, coupled with translational control of the neurovirulence factor ICP34.5 using multiple microRNA (miR)-binding sites. VG2025 further incorporates IL-12 and the IL-15/IL-15 receptor alpha subunit complex to enhance the antitumor and immune stimulatory properties of oncolytic HSVs. The TTDR strategy was verified in vitro and shown to be highly selective. Strong in vivo antitumor efficacy was observed following both intratumoral and intravenous administration. Clear abscopal and immune memory effects were also evident, indicating a robust antitumor immune response. Gene expression profiling of treated tumors revealed increased immune cell infiltration and activation of multiple immune-signaling pathways when compared with the backbone virus. Absence of neurotoxicity was verified in mice and in rhesus monkeys. Taken together, the enhanced tumor clearance, excellent safety profile, and positive correlation between CEA levels and viral replication efficiency may provide an opportunity for using biomarker-based precision medicine in oncolytic virotherapy.

3.
Biomedicines ; 8(11)2020 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-33182232

RESUMO

Oncolytic virotherapy is a promising new tool for cancer treatment, but direct lytic destruction of tumor cells is not sufficient and must be accompanied by strong immune activation to elicit anti-tumor immunity. We report here the creation of a novel replication-competent recombinant oncolytic herpes simplex virus type 1 (VG161) that carries genes coding for IL-12, IL-15, and IL-15 receptor alpha subunit, along with a peptide fusion protein capable of disrupting PD-1/PD-L1 interactions. The VG161 virus replicates efficiently and exhibits robust cytotoxicity in multiple tumor cell lines. Moreover, the encoded cytokines and the PD-L1 blocking peptide work cooperatively to boost immune cell function. In vivo testing in syngeneic CT26 and A20 tumor models reveals superior efficacy when compared to a backbone virus that does not express exogenous genes. Intratumoral injection of VG161 induces abscopal responses in non-injected distal tumors and grants resistance to tumor re-challenge. The robust anti-tumor effect of VG161 is associated with T cell and NK cell tumor infiltration, expression of Th1 associated genes in the injection site, and increased frequency of splenic tumor-specific T cells. VG161 also displayed a superb safety profile in GLP acute and repeated injection toxicity studies performed using cynomolgus monkeys. Overall, we demonstrate that VG161 can induce robust oncolysis and stimulate a robust anti-tumor immune response without sacrificing safety.

4.
Prostate ; 78(6): 457-468, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29450905

RESUMO

BACKGROUND: Patients with advanced prostate cancer have limited curative options, therefore new treatments are needed. Mouse models play a pivotal role in the discovery and development of new treatments. In the present study, a TRAMP-derived Orthotopic Prostate Syngeneic (TOPS) mouse model was developed and found to provide a consistent means of monitoring tumor and metastatic responses to novel treatments. METHODS: The mouse TOPS model was generated using luciferase transduced TRAMP-C2 prostate cancer cells that were orthotopically injected into Bl6 mice by ultrasound guidance. Tumor growth and development was monitored using ultrasound and bioluminescence imaging. RESULTS: Tumors and metastases were consistently established and increases in tumor size correlated with increases in bioluminescence. In addition, when mice with an established tumor were castrated, tumor progression mirrored clinical progression. We further treated the TOPS model with an oncolytic Herpes Simplex virus and showed that we were able to monitor the therapeutic effect of the orthotopic tumor after virus treatment through IVIS imaging system. CONCLUSION: We have developed a powerful animal model to advance the current selection of effective treatments for patients with advanced prostate cancer.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Adenocarcinoma/tratamento farmacológico , Animais , Progressão da Doença , Masculino , Camundongos , Camundongos Transgênicos , Próstata/patologia , Neoplasias da Próstata/tratamento farmacológico
5.
Cancer Res ; 78(3): 718-730, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29118089

RESUMO

The first oncolytic virotherapy employing HSV-1 (oHSV-1) was approved recently by the FDA to treat cancer, but further improvements in efficacy are needed to eradicate challenging refractory tumors, such as glioblastomas (GBM). Microglia/macrophages comprising approximately 40% of a GBM tumor may limit virotherapeutic efficacy. Here, we show these cells suppress oHSV-1 growth in gliomas by internalizing the virus through phagocytosis. Internalized virus remained capable of expressing reporter genes while viral replication was blocked. Macrophage/microglia formed a nonpermissive OV barrier, preventing dissemination of oHSV-1 in the glioma mass. The deficiency in viral replication in microglial cells was associated with silencing of particular viral genes. Phosphorylation of STAT1/3 was determined to be responsible for suppressing oHSV-1 replication in macrophages/microglia. Treatment with the oxindole/imidazole derivative C16 rescued oHSV-1 replication in microglia/macrophages by inhibiting STAT1/3 activity. In the U87 xenograft model of GBM, C16 treatment overcame the microglia/macrophage barrier, thereby facilitating tumor regression without causing a spread of the virus to normal organs. Collectively, our results suggest a strategy to relieve a STAT1/3-dependent therapeutic barrier and enhance oHSV-1 oncolytic activity in GBM.Significance: These findings suggest a strategy to enhance the therapeutic efficacy of oncolytic virotherapy in glioblastoma. Cancer Res; 78(3); 718-30. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Macrófagos/patologia , Microglia/patologia , Terapia Viral Oncolítica , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virologia , Proliferação de Células , Feminino , Glioblastoma/metabolismo , Glioblastoma/virologia , Humanos , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Nus , Microglia/metabolismo , Microglia/virologia , Vírus Oncolíticos/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nat Rev Urol ; 13(6): 334-52, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27215429

RESUMO

Oncolytic virotherapy is a cancer treatment in which replication-competent viruses are used that specifically infect, replicate in and lyse malignant tumour cells, while minimizing harm to normal cells. Anecdotal evidence of the effectiveness of this strategy has existed since the late nineteenth century, but advances and innovations in biotechnological methods in the 1980s and 1990s led to a renewed interest in this type of therapy. Multiple clinical trials investigating the use of agents constructed from a wide range of viruses have since been performed, and several of these enrolled patients with urological malignancies. Data from these clinical trials and from preclinical studies revealed a number of challenges to the effectiveness of oncolytic virotherapy that have prompted the development of further sophisticated strategies. Urological cancers have a range of distinctive features, such as specific genetic mutations and cell surface markers, which enable improving both effectiveness and safety of oncolytic virus treatments. The strategies employed in creating advanced oncolytic agents include alteration of the virus tropism, regulating transcription and translation of viral genes, combination with chemotherapy, radiotherapy or gene therapy, arming viruses with factors that stimulate the immune response against tumour cells and delivery technologies to ensure that the viral agent reaches its target tissue.


Assuntos
Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Neoplasias Urológicas/genética , Neoplasias Urológicas/terapia , Animais , Antineoplásicos/administração & dosagem , Ensaios Clínicos como Assunto/métodos , Terapia Genética/métodos , Humanos , Neoplasias Urológicas/diagnóstico
7.
Oncotarget ; 7(19): 28658-69, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27070093

RESUMO

Oncolytic herpes simplex virus type 1 (oHSV-1) therapy is an emerging treatment modality that selectively destroys cancer. Here we report use of a glioma specific HSV-1 amplicon virus (SU4-124 HSV-1) to selectively target tumour cells. To achieve transcriptional regulation of the SU4-124 HSV-1 virus, the promoter for the essential HSV-1 gene ICP4 was replaced with a tumour specific survivin promoter. Translational regulation was achieved by incorporating 5 copies of microRNA 124 target sequences into the 3'UTR of the ICP4 gene. Additionally, a 5'UTR of rat fibroblast growth factor -2 was added in front of the viral ICP4 gene open reading frame. Our results confirmed enhanced expression of survivin and eIF4E in different glioma cells and increased micro-RNA124 expression in normal human and mouse brain tissue. SU4-124 HSV-1 had an increased ICP4 expression and virus replication in different glioma cells compared to normal neuronal cells. SU4-124 HSV-1 exerted a strong antitumour effect against a panel of glioma cell lines. Intracranial injection of SU4-124 HSV-1 did not reveal any sign of toxicity on day 15 after the injection. Moreover, a significantly enhanced antitumour effect with the intratumourally injected SU4-124 HSV-1 virus was demonstrated in mice bearing human glioma U87 tumours, whereas viral DNA was almost undetectable in normal organs. Our study indicates that incorporation of multiple cancer-specific regulators in an HSV-1 system significantly enhances both cancer specificity and oncolytic activity.


Assuntos
Glioma/terapia , Herpesvirus Humano 1/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Regiões 3' não Traduzidas/genética , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Fator 2 de Crescimento de Fibroblastos/genética , Glioma/genética , Glioma/virologia , Células HEK293 , Herpesvirus Humano 1/fisiologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Inibidoras de Apoptose/genética , Camundongos , Vírus Oncolíticos/fisiologia , Regiões Promotoras Genéticas/genética , Ratos , Survivina , Carga Tumoral/genética , Células Vero
8.
J Cell Physiol ; 231(6): 1350-63, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26529564

RESUMO

It has long been realized that hematopoietic cells may have the capacity to trans-differentiate into non-lymphohematopoietic cells under specific conditions. However, the mechanisms and the factors for hematopoietic cell trans-differentiation remain unknown. In an in vitro culture system, we found that using a conditioned medium from proliferating fibroblasts can induce a subset of hematopoietic cells to become adherent fibroblast-like cells (FLCs). FLCs are not fibroblasts nor other mesenchymal stromal cells, based on their expression of type-1 collagen, and other stromal cell marker genes. To identify the active factors in the conditioned medium, we cultured fibroblasts in a serum-free medium and collected it for further purification. Using the fractions from filter devices of different molecular weight cut-offs, and ammonium sulfate precipitation collected from the medium, we found the active fraction is a protein. We then purified this fraction by using fast protein liquid chromatography (FPLC) and identified it by mass spectrometer as macrophage colony-stimulating factor (M-CSF). The mechanisms of M-CSF-inducing trans-differentiation of hematopoietic cells seem to involve a tyrosine kinase signalling pathway and its known receptor. The FLCs express a number of stem cell markers including SSEA-1 and -3, OCT3/4, NANOG, and SOX2. Spontaneous and induced differentiation experiments confirmed that FLCs can be further differentiated into cell types of three germ layers. These data indicate that hematopoietic cells can be induced by M-CSF to dedifferentiate to multipotent stem cells. This study also provides a simple method to generate multipotent stem cells for clinical applications.


Assuntos
Tecido Adiposo/metabolismo , Transdiferenciação Celular , Fibroblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Leucócitos Mononucleares/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Comunicação Parácrina , Baço/metabolismo , Adipócitos/metabolismo , Adipogenia , Tecido Adiposo/citologia , Animais , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Fenótipo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais , Baço/citologia
9.
ISRN Pharmacol ; 2012: 307102, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22957270

RESUMO

Exposure of cancer cells to anticancer agents in cultures induces detachment of cells that are usually considered dead. These drug-induced detached cells (D-IDCs) may represent a clinical problem for chemotherapy since they may survive anoikis, enter the circulation, invade other tissues and resume proliferation, creating a metastasis, especially in tissues where the bioavailability of anticancer agents is not enough to eliminate all cancer cells. In this study we evaluated the antiproliferative effect of menadione : sodium orthovanadate (M : SO) combination on A549 lung cancer cells as well as the ability of M : SO to induce cell detachment. In addition, we followed the fate and chemosensitivity of M : SO-induced detached cells. Using transwell chambers, we found that a fraction of the M : SO-induced detached cells were viable and, furthermore, were able to migrate, re-attach, and resume proliferation when re-incubated in drug-free media. The total elimination of A549 detachment-resistant cells and M : SO-induced detached cells were successfully eliminated by equivalent M : SO concentration (17.5 µM : 17.5 µM). Thus, M : SO prevented cell migration. Similar results were obtained on DBTRG.05MG human glioma cells. Our data guarantee further studies to evaluate the in vivo occurrence of D-IDCs, their implications for invasiveness and metastasis and their sensitivity to anticancer drugs.

10.
Invest New Drugs ; 30(4): 1302-10, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21553345

RESUMO

Gliomas are the most common primary brain tumor, and their treatment is still a challenge. Here, we evaluated the antiproliferative effect of a novel combination of two potent oxidative stress enhancers: menadione (M) and sodium orthovanadate (SO). We observed both short-term and prolonged growth inhibitory effects of M or SO alone as well as in combination (M:SO) on DBTRG.05MG human glioma cells. A stronger antiproliferative effect was observed in the short-term proliferation assay with the M:SO combination compared to either investigated agent alone. In the long-term proliferation assay, a 10-day exposure to M:SO at concentrations of 10 µM:17.5 µM or 17.5 µM:10 µM was enough to kill 100% of the cells; no cell regrowth was observed after re-incubation in drug-free media. When used in combination, the single concentration of M and SO could be decreased by 2.5- to 5-fold of those used for each experimental drug alone and still obtain a similar antiproliferative effect. The underlying molecular mechanism was investigated by co-incubating M:SO with dithiothreitol (DTT) and genistein. Both substances partially neutralized the effects of the M:SO combination, showing additive effects. This observation suggests a role of oxidative stress and tyrosine kinase stimulation in the M:SO cytotoxic effect. Our results indicate that M:SO combination is an attractive alternative for glioma treatment that encourages further study. The neutralizing effects of genistein and DTT reveal a possibility for their use in the minimization of potential M:SO systemic toxicity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Vanadatos/uso terapêutico , Vitamina K 3/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ditiotreitol/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Genisteína/farmacologia , Humanos , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Fatores de Tempo , Vanadatos/farmacologia , Vitamina K 3/farmacologia
11.
Acta Biochim Pol ; 58(2): 265-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21643560

RESUMO

In most cells, the major intracellular redox buffer is glutathione (GSH) and its disulfide-oxidized (GSSG) form. The GSH/GSSG system maintains the intracellular redox balance and the essential thiol status of proteins by thiol disulfide exchange. Topoisomerases are thiol proteins and are a target of thiol-reactive substances. In this study, the inhibitory effect of physiological concentration of GSH and GSSG on topoisomerase IIα activity in vitro was investigated. GSH (0-10 mM) inhibited topoisomerase IIα in a concentration-dependent manner while GSSG (1-100 µM) had no significant effect. These findings suggest that the GSH/GSSG system could have a potential in vivo role in regulating topoisomerase IIα activity.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Glutationa/química , Inibidores da Topoisomerase II/química , Antígenos de Neoplasias , DNA Topoisomerases Tipo II , Ensaios Enzimáticos , Humanos , Oxirredução
12.
Invest New Drugs ; 29(6): 1314-20, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20625795

RESUMO

Menadione (Vitamin K3) has anti-tumoral effects against a wide range of cancer cells. Its potential toxicity to normal cells and narrow therapeutic range limit its use as single agent but in combination with radiation or other anti-neoplastic agents can be of therapeutic use. In this paper, we first evaluated the early (within 3 h) effect of menadione on ongoing DNA replication. In normal rat cerebral cortex mini-units menadione showed an age dependent anti-proliferative effect. In tissue mini-units prepared from newborn rats, menadione inhibited ongoing DNA replication with an IC (50) of approximately 10 µM but 50 µM had no effect on mini-units from prepared adult rat tissue. The effect of short (72 h) and prolonged exposure (1-2 weeks) to menadione alone in the DBTRG.05MG human glioma cells line and in combination with vitamin C was studied. After short period of exposure data show that menadione alone or in combination with vitamin C provided similar concentration-response curves (and IC(50) values). Prolonged exposure to these drugs was evaluated by their ability to kill 100% of glioma cells and prevent regrowth when cells are re-incubated in drug-free media. In this long-term assay, menadione:vitamin C at a ratio 1:100 showed higher anti-proliferative activity when compared to each drug alone and allowed to reduce each drug concentration between 2.5 to 5-fold. Similar anti-proliferative effect was demonstrated in 8 patient derived glioblastoma cell cultures. Our data should be able to encourage further advanced studies on animal models to evaluate the potential use of this combination therapy for glioma treatment.


Assuntos
Antineoplásicos/farmacologia , Ácido Ascórbico/farmacologia , Glioma/tratamento farmacológico , Vitamina K 3/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Antineoplásicos/administração & dosagem , Ácido Ascórbico/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Replicação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Glioma/patologia , Humanos , Concentração Inibidora 50 , Ratos , Fatores de Tempo , Vitamina K 3/administração & dosagem , Vitaminas/administração & dosagem , Vitaminas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...