Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 16(15)2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39123450

RESUMEN

Pancreatic cancer has one of the worst prognoses among all malignancies and few available treatment options. Patient-derived xenografts can be used to develop personalized therapy for pancreatic cancer. Endoscopic ultrasound fine-needle aspiration (EUS-FNA) may provide a powerful alternative to surgery for obtaining sufficient tissue for the establishment of patient-derived xenografts. In this study, EUS-FNA samples were obtained for 30 patients referred to the Ottawa Hospital, Ottawa, Ontario, Canada. These samples were used for xenotransplantation in NOD-SCID mice and for genetic analyses. The gene expression of pancreatic-cancer-relevant genes in xenograft tumors was examined by immunohistochemistry. Targeted sequencing of both the patient-derived tumors and xenograft tumors was performed. The xenografts' susceptibility to oncolytic virus infection was studied by infecting xenograft-derived cells with VSV∆51-GFP. The xenograft take rate was found to be 75.9% for passage 1 and 100% for passage 2. Eighty percent of patient tumor samples were successfully sequenced to a high depth for 42 cancer genes. Xenograft histological characteristics and marker expression were maintained between passages. All tested xenograft samples were susceptible to oncoviral infection. We found that EUS-FNA is an accessible, minimally invasive technique that can be used to acquire adequate pancreatic cancer tissue for the generation of patient-derived xenografts and for genetic sequencing.

2.
Arch Cancer Biol Ther ; 1(1): 18-21, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32728673

RESUMEN

BACKGROUND: We have recently introduced a modification of the seminal Simeoni model for tumor growth, the modification entailing the incorporation of delay differential equations into its formulation. We found that the modification was competitive with the Simeoni construct in modeling mammary tumor growth under cisplatin treatment in an animal model. METHODS: In our original study, we had two cohorts of animals: untreated, and treatment with bolus injection of cisplatin on day 0. We here explore how modifications in the cisplatin dosing scheme affect tumor growth in our model. RESULTS: We found that modest fractionation dosing schemes have little ultimate impact on tumor growth. In contrast, metronomic dosing schemes seem quite efficacious, and might yield effective control over tumor progression. CONCLUSIONS: With regard to cisplatin as single agent chemotherapy, a minimum level of drug for a prolonged period of time seems more critical than rapid achievement of a very high dose for a shorter time frame for deterring tumor growth or progression. Exploration of tumor dose schedules with mathematical models can provide valuable insights into potentially effective therapeutic regimens.

3.
BMC Cancer ; 20(1): 226, 2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32183732

RESUMEN

BACKGROUND: Simeoni and colleagues introduced a compartmental model for tumor growth that has proved quite successful in modeling experimental therapeutic regimens in oncology. The model is based on a system of ordinary differential equations (ODEs), and accommodates a lag in therapeutic action through delay compartments. There is some ambiguity in the appropriate number of delay compartments, which we examine in this note. METHODS: We devised an explicit delay differential equation model that reflects the main features of the Simeoni ODE model. We evaluated the original Simeoni model and this adaptation with a sample data set of mammary tumor growth in the FVB/N-Tg(MMTVneu)202Mul/J mouse model. RESULTS: The experimental data evinced tumor growth heterogeneity and inter-individual diversity in response, which could be accommodated statistically through mixed models. We found little difference in goodness of fit between the original Simeoni model and the delay differential equation model relative to the sample data set. CONCLUSIONS: One should exercise caution if asserting a particular mathematical model uniquely characterizes tumor growth curve data. The Simeoni ODE model of tumor growth is not unique in that alternative models can provide equivalent representations of tumor growth.


Asunto(s)
Neoplasias Mamarias Experimentales/patología , Modelos Biológicos , Algoritmos , Animales , Femenino , Ratones
4.
Eur J Nucl Med Mol Imaging ; 47(8): 2026-2035, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31729540

RESUMEN

PURPOSE: Multidrug resistance (MDR) impedes cancer treatment. Two efflux transporters from the ATP-binding cassette (ABC) family, ABCB1 and ABCG2, may contribute to MDR by restricting the entry of therapeutic drugs into tumor cells. Although a higher expression of these transporters has been correlated with an unfavorable response to chemotherapy, transporter expression does not necessarily correlate with function. In this study, we characterized the pharmacological properties of [18F]AVT-011, a new PET radiotracer for imaging transporter-mediated MDR in tumors. METHODS: AVT-011 was radiolabeled with 18F and evaluated with PET imaging in preclinical models. Transport of [18F]AVT-011 by ABCB1 and/or ABCG2 was assessed by measuring its uptake in the brains of wild-type, Abcb1a/b-/-, and Abcg2-/- mice at baseline and after administration of the ABCB1 inhibitor tariquidar (n = 5/group). Metabolism and biodistribution of [18F]AVT-011 were also measured. To measure ABCB1 function in tumors, we performed PET experiments using both [18F]AVT-011 and [18F]FDG in mice bearing orthotopic breast tumors (n = 7-10/group) expressing clinically relevant levels of ABCB1. RESULTS: At baseline, brain uptake was highest in Abcb1a/b-/- mice. After tariquidar administration, brain uptake increased 3-fold and 8-fold in wild-type and Abcg2-/- mice, respectively, but did not increase further in Abcb1a/b-/- mice. At 30 min after injection, the radiotracer was > 90% in its parent form and had highest uptake in organs of the hepatobiliary system. Compared with that in drug-sensitive tumors, uptake of [18F]AVT-011 was 32% lower in doxorubicin-resistant tumors with highest ABCB1 expression and increased by 40% with tariquidar administration. Tumor uptake of [18F]FDG did not significantly differ among groups. CONCLUSION: [18F]AVT-011 is a dual ABCB1/ABCG2 substrate radiotracer that can quantify transporter function at the blood-brain barrier and in ABCB1-expressing tumors, making it potentially suitable for clinical imaging of ABCB1-mediated MDR in tumors.


Asunto(s)
Resistencia a Múltiples Medicamentos , Tomografía de Emisión de Positrones , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Ratones , Distribución Tisular
5.
Sci Transl Med ; 10(422)2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29298865

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive disease for which treatment options are limited and associated with severe toxicities. Immunotherapeutic approaches like immune checkpoint inhibitors (ICIs) are a potential strategy, but clinical trials have demonstrated limited success in this patient cohort. Clinical studies using ICIs have revealed that patients with preexisting anticancer immunity are the most responsive. Given that oncolytic viruses (OVs) induce antitumor immunity, we investigated their use as an ICI-sensitizing approach. Using a therapeutic model that mimics the course of treatment for women with newly diagnosed TNBC, we demonstrate that early OV treatment coupled with surgical resection provides long-term benefits. OV therapy sensitizes otherwise refractory TNBC to immune checkpoint blockade, preventing relapse in most of the treated animals. We suggest that OV therapy in combination with immune checkpoint blockade warrants testing as a neoadjuvant treatment option in the window of opportunity between TNBC diagnosis and surgical resection.


Asunto(s)
Viroterapia Oncolítica/métodos , Neoplasias de la Mama Triple Negativas/terapia , Femenino , Humanos , Terapia Neoadyuvante/métodos , Virus Oncolíticos/fisiología
6.
Sci Rep ; 7(1): 5153, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28698590

RESUMEN

Tudor domain containing protein 3 (TDRD3) is a modular protein identified based on its ability to recognize methylated arginine motifs through its Tudor domain. We have previously shown that TDRD3 localizes to cytoplasmic stress granules, a structure shown to promote survival upon treatment with chemotherapeutic drugs in cancer cells. Here, we report TDRD3 as a novel regulator of cell proliferation and invasion in breast cancer cells. Our study also demonstrates that TDRD3 depletion inhibits tumor formation and metastasis to the lung in vivo. Furthermore, we show that TDRD3 regulates the expression of a number of key genes associated with promotion of breast cancer tumorigenesis and disease progression. Strikingly, we report that TDRD3 regulates some of these key targets at the level of translation. These findings provide the first experimental demonstration of a functional role for TDRD3 in promoting breast cancer development and progression, and identify TDRD3 as a potential new therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Pulmonares/secundario , Proteínas/genética , Proteínas/metabolismo , Regulación hacia Arriba , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células MCF-7 , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Biosíntesis de Proteínas
7.
J Nucl Med ; 58(2): 221-227, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27635026

RESUMEN

Oncolytic virus (OV) therapy has emerged as a novel tool in our therapeutic arsenals for fighting cancer. As a live biologic agent, OV has the ability to target and selectively amplify at the tumor sites. We have reported that a vaccinia-based OV (Pexa-Vec) has shown good efficacy in preclinical models and in clinical trials. To give an additional tool to clinicians to allow both treatment of the tumor and improved visualization of tumor margins, we developed new viral-based platforms with 2 specific gene reporters. METHODS: We incorporated the human sodium iodide symporter (hNIS) and the human somatostatin receptor 2 (hSSR2) in the vaccinia-based OV and tested viral constructs for their abilities to track and treat tumor development in vivo. RESULTS: Early and high-level expression of hNIS is detrimental to the recombinant virus, leading to the aggregation of hNIS protein and early cell death. Putting hNIS under a late synthetic promoter allowed a higher functional expression of the protein and much stronger 123I or 99Tc uptake. In vivo, the hNIS-containing virus infected and amplified in the tumor site, showing a better efficacy than the parental virus. The hNIS expression at the tumor site allowed for the imaging of viral infection and tumor regression. Similarly, hSSR2-containing OV vaccinia infected and lysed cancer cells. CONCLUSION: When tumor-bearing mice were given hNIS- and hSSR2-containing OV, 99Tc and 111In signals coalesced at the tumor, highlighting the power of using these viruses for tumor diagnosis and treatment.


Asunto(s)
Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/terapia , Viroterapia Oncolítica/métodos , Receptores de Somatostatina/genética , Simportadores/genética , Virus Vaccinia/fisiología , Animales , Línea Celular Tumoral , Femenino , Genes Reporteros/genética , Humanos , Ratones , Ratones Desnudos , Neoplasias Experimentales/virología , Virus Oncolíticos/fisiología , Tomografía de Emisión de Positrones/métodos , Recombinación Genética/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Nanomedicina Teranóstica/métodos , Resultado del Tratamiento , Regulación hacia Arriba/genética
8.
Mol Ther Oncolytics ; 3: 16027, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27909702

RESUMEN

The systemic delivery of therapeutic viruses, such as oncolytic viruses or vaccines, is limited by the generation of neutralizing antibodies. While pseudotyping of rhabdoviruses with the lymphocytic choriomeningitis virus glycoprotein has previously allowed for multiple rounds of delivery in mice, this strategy has not translated to other animal models. For the first time, we provide experimental evidence that antibodies generated against the lymphocytic choriomeningitis virus glycoprotein mediate robust complement-dependent viral neutralization via activation of the classical pathway. We show that this phenotype can be capitalized upon to deliver maraba virus pseudotyped with the lymphocytic choriomeningitis virus glycoprotein in a Fischer rat model in the face of neutralizing antibody through the use of complement modulators. This finding changes the understanding of the humoral immune response to arenaviruses, and also describes methodology to deliver viral vectors to their therapeutic sites of action without the interference of neutralizing antibody.

9.
Breast Cancer Res ; 18(1): 83, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27503504

RESUMEN

BACKGROUND: Breast cancer is the most common malignant disease amongst Western women. The lack of treatment options for patients with chemotherapy-resistant or recurrent cancers is pushing the field toward the rapid development of novel therapies. The use of oncolytic viruses is a promising approach for the treatment of disseminated diseases like breast cancer, with the first candidate recently approved by the Food and Drug Administration for use in patients. In this report, we demonstrate the compatibility of oncolytic virotherapy and chemotherapy using various murine breast cancer models. This one-two punch has been explored in the past by several groups with different viruses and drugs and was shown to be a successful approach. Our strategy is to combine Paclitaxel, one of the most common drugs used to treat patients with breast cancer, and the oncolytic Rhabdovirus Maraba-MG1, a clinical trial candidate in a study currently recruiting patients with late-stage metastatic cancer. METHODS: We used the EMT6, 4 T1 and E0771 murine breast cancer models to evaluate in vitro and in vivo the effects of co-treatment with MG1 and Paclitaxel. Treatment-induced cytotoxicity was assessed and plaque assays, flow cytometry, microscopy and immunocytochemistry analysis were performed to quantify virus production and transgene expression. Orthotopically implanted tumors were measured during and after treatment to evaluate efficacy and Kaplan-Meier survival curves were generated. RESULTS: Our data demonstrate not only the compatibility of the treatments, but also their synergistic cytopathic activity. With Paclitaxel, EMT6 and 4 T1 tumors demonstrated increased virus production both in vitro and in vivo. Our results also show that Paclitaxel does not impair the safety profile of the virus treatment. Importantly, when combined, MG1 and the drug controlled tumor growth and prolonged survival. CONCLUSIONS: The combination of MG1 and Paclitaxel improved efficacy in all of the breast cancer models we tested and thus is a promising alternative approach for the treatment of patients with refractory breast cancer. Our strategy has potential for rapid translation to the clinic, given the current clinical status of both agents.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Paclitaxel/uso terapéutico , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Humanos , Interferón beta/farmacología , Ratones , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Paclitaxel/administración & dosificación , Carga Tumoral/efectos de los fármacos , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Ther Oncolytics ; 3: 16001, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27119116

RESUMEN

Oncolytic viruses are known to stimulate the antitumor immune response by specifically replicating in tumor cells. This is believed to be an important aspect of the durable responses observed in some patients and the field is rapidly moving toward immunotherapy. As a further means to engage the immune system, we engineered a virus, vesicular stomatitis virus (VSV), to encode the proinflammatory cytokine interferon-γ. We used the 4T1 mammary adenocarcinoma as well as other murine tumor models to characterize immune responses in tumor-bearing animals generated by treatment with our viruses. The interferon-γ-encoding virus demonstrated greater activation of dendritic cells and drove a more profound secretion of proinflammatory cytokines compared to the parental virus. From a therapeutic point of view, the interferon-γ virus slowed tumor growth, minimized lung tumors, and prolonged survival in several murine tumor models. The improved efficacy was lost in immunocompromized animals; hence the mechanism appears to be T-cell-mediated. Taken together, these results demonstrate the ability of oncolytic viruses to act as immune stimulators to drive antitumor immunity as well as their potential for targeted gene therapy.

11.
ILAR J ; 57(1): 73-85, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27034397

RESUMEN

The preclinical optimization and validation of novel treatments for cancer therapy requires the use of laboratory animals. Although in vitro experiments using tumor cell lines and ex vivo treatment of patient tumor samples provide a remarkable first-line tool for the initial study of tumoricidal potential, tumor-bearing animals remain the primary option to study delivery, efficacy, and safety of therapies in the context of a complete tumor microenvironment and functional immune system. In this review, we will describe the use of murine tumor models for oncolytic virotherapy using vesicular stomatitis virus. We will discuss studies using immunocompetent and immunodeficient models with respect to toxicity and therapeutic treatments, as well as the various techniques and tools available to study cancer therapy with Rhabdoviruses.


Asunto(s)
Viroterapia Oncolítica/métodos , Animales , Ratones , Neoplasias/terapia , Neoplasias/virología , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral/fisiología
12.
PLoS One ; 11(3): e0151516, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26986751

RESUMEN

Adenoviruses (Ads) are used in numerous preclinical and clinical studies for delivery of anti-cancer therapeutic genes. Unfortunately, Ad has a poor ability to distribute throughout a tumor mass after intratumoral injection, and infects cells primarily within the immediate area of the injection tract. Thus, Ad-encoded transgene expression is typically limited to only a small percentage of cells within the tumor. One method to increase the proportion of the tumor impacted by Ad is through expression of fusogenic proteins. Infection of a single cell with an Ad vector encoding a fusogenic protein should lead to syncytium formation with adjacent cells, effectively spreading the effect of Ad and Ad-encoded therapeutic transgenes to a greater percentage of the tumor mass. Moreover, syncytium formation can be cytotoxic, suggesting that such proteins may be effective sole therapeutics. We show that an early region 1 (E1)-deleted Ad expressing reptilian reovirus p14 fusion-associated small transmembrane (FAST) protein caused extensive cell fusion in the replication-permissive 293 cell line and at high multiplicity of infection in non-permissive human lung adenocarcinoma A549 cells in vitro. FAST protein expression in the A549 cancer cell line led to a loss of cellular metabolic activity and membrane integrity, which correlated with induction of apoptosis. However, in an A549 xenograft CD-1 nude mouse cancer model, Ad-mediated FAST gene delivery did not induce detectable cell fusion, reduce tumor burden nor enhance mouse survival compared to controls. Taken together, our results show that, although AdFAST can enhance cancer cell killing in vitro, it is not effective as a sole therapeutic in the A549 tumor model in vivo.


Asunto(s)
Adenoviridae/genética , Apoptosis/genética , Neoplasias/genética , Proteínas Virales de Fusión/genética , Animales , Western Blotting , Fusión Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Células HEK293 , Humanos , Ratones Desnudos , Microscopía Fluorescente , Neoplasias/patología , Neoplasias/terapia , Reoviridae/genética , Proteínas Virales de Fusión/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
13.
Cancer Cell ; 28(2): 210-24, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26212250

RESUMEN

Oncolytic viruses designed to attack malignant cells can in addition infect and destroy tumor vascular endothelial cells. We show here that this expanded tropism of oncolytic vaccinia virus to the endothelial compartment is a consequence of VEGF-mediated suppression of the intrinsic antiviral response. VEGF/VEGFR2 signaling through Erk1/2 and Stat3 leads to upregulation, nuclear localization, and activation of the transcription repressor PRD1-BF1/Blimp1. PRD1-BF1 does not contribute to the mitogenic effects of VEGF, but directly represses genes involved in type I interferon (IFN)-mediated antiviral signaling. In vivo suppression of VEGF signaling diminishes PRD1-BF1/Blimp1 expression in tumor vasculature and inhibits intravenously administered oncolytic vaccinia delivery to and consequent spread within the tumor.


Asunto(s)
Neoplasias/virología , Virus Oncolíticos/fisiología , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Ratones Endogámicos C57BL , Microscopía Fluorescente , Neoplasias/irrigación sanguínea , Neoplasias/terapia , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/virología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Interferencia de ARN , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Activación Transcripcional/efectos de los fármacos , Virus Vaccinia/fisiología
14.
Nat Med ; 21(5): 530-6, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25894825

RESUMEN

Tumors are complex ecosystems composed of networks of interacting 'normal' and malignant cells. It is well recognized that cytokine-mediated cross-talk between normal stromal cells, including cancer-associated fibroblasts (CAFs), vascular endothelial cells, immune cells, and cancer cells, influences all aspects of tumor biology. Here we demonstrate that the cross-talk between CAFs and cancer cells leads to enhanced growth of oncolytic virus (OV)-based therapeutics. Transforming growth factor-ß (TGF-ß) produced by tumor cells reprogrammed CAFs, dampened their steady-state level of antiviral transcripts and rendered them sensitive to virus infection. In turn, CAFs produced high levels of fibroblast growth factor 2 (FGF2), initiating a signaling cascade in cancer cells that reduced retinoic acid-inducible gene I (RIG-I) expression and impeded the ability of malignant cells to detect and respond to virus. In xenografts derived from individuals with pancreatic cancer, the expression of FGF2 correlated with the susceptibility of the cancer cells to OV infection, and local application of FGF2 to resistant tumor samples sensitized them to virotherapy both in vitro and in vivo. An OV engineered to express FGF2 was safe in tumor-bearing mice, showed improved therapeutic efficacy compared to parental virus and merits consideration for clinical testing.


Asunto(s)
Fibroblastos/metabolismo , Virus Oncolíticos/metabolismo , Microambiente Tumoral , Anciano , Animales , Antivirales/química , Línea Celular Tumoral , Chlorocebus aethiops , Técnicas de Cocultivo , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Microscopía Fluorescente , Persona de Mediana Edad , Trasplante de Neoplasias , Viroterapia Oncolítica/métodos , Neoplasias Ováricas/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Vero
15.
Mol Ther ; 23(6): 1066-1076, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25807289

RESUMEN

Oncolytic viruses (OVs) have shown promising clinical activity when administered by direct intratumoral injection. However, natural barriers in the blood, including antibodies and complement, are likely to limit the ability to repeatedly administer OVs by the intravenous route. We demonstrate here that for a prototype of the clinical vaccinia virus based product Pexa-Vec, the neutralizing activity of antibodies elicited by smallpox vaccination, as well as the anamnestic response in hyperimmune virus treated cancer patients, is strictly dependent on the activation of complement. In immunized rats, complement depletion stabilized vaccinia virus in the blood and led to improved delivery to tumors. Complement depletion also enhanced tumor infection when virus was directly injected into tumors in immunized animals. The feasibility and safety of using a complement inhibitor, CP40, in combination with vaccinia virus was tested in cynomolgus macaques. CP40 pretreatment elicited an average 10-fold increase in infectious titer in the blood early after the infusion and prolonged the time during which infectious virus was detectable in the blood of animals with preexisting immunity. Capitalizing on the complement dependence of antivaccinia antibody with adjunct complement inhibitors may increase the infectious dose of oncolytic vaccinia virus delivered to tumors in virus in immune hosts.


Asunto(s)
Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Virus Vaccinia/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Línea Celular Tumoral , Chlorocebus aethiops , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Estudios de Factibilidad , Femenino , Células HeLa , Humanos , Inyecciones Intralesiones , Macaca fascicularis/inmunología , Masculino , Neoplasias/sangre , Neoplasias/terapia , Pruebas de Neutralización , Piridonas/inmunología , Piridonas/farmacología , Ratas , Ratas Endogámicas F344 , Vacuna contra Viruela/sangre , Vacuna contra Viruela/inmunología , Vacunación , Células Vero
16.
Nat Commun ; 6: 6410, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25817275

RESUMEN

In this study, we show that several microtubule-destabilizing agents used for decades for treatment of cancer and other diseases also sensitize cancer cells to oncolytic rhabdoviruses and improve therapeutic outcomes in resistant murine cancer models. Drug-induced microtubule destabilization leads to superior viral spread in cancer cells by disrupting type I IFN mRNA translation, leading to decreased IFN protein expression and secretion. Furthermore, microtubule-destabilizing agents specifically promote cancer cell death following stimulation by a subset of infection-induced cytokines, thereby increasing viral bystander effects. This study reveals a previously unappreciated role for microtubule structures in the regulation of the innate cellular antiviral response and demonstrates that unexpected combinations of approved chemotherapeutics and biological agents can lead to improved therapeutic outcomes.


Asunto(s)
Efecto Espectador/efectos de los fármacos , Citocinas/efectos de los fármacos , Interferón Tipo I/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Viroterapia Oncolítica , Virus Oncolíticos , ARN Mensajero/efectos de los fármacos , Infecciones por Rhabdoviridae/inmunología , Moduladores de Tubulina/farmacología , Albendazol/farmacología , Animales , Bencimidazoles/farmacología , Efecto Espectador/inmunología , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Colchicina/farmacología , Citocinas/inmunología , Células HT29 , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Nocodazol/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , Rhabdoviridae , Células Vero , Vinblastina/análogos & derivados , Vinblastina/farmacología , Vinorelbina
17.
Oncotarget ; 6(5): 3013-32, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25605249

RESUMEN

Recent evidence points to the protein arginine methyltransferase (PRMT) family of enzymes playing critical roles in cancer. PRMT7 has been identified in several gene expression studies to be associated with increased metastasis and decreased survival in breast cancer patients. However, this has not been extensively studied. Here we report that PRMT7 expression is significantly upregulated in both primary breast tumour tissues and in breast cancer lymph node metastases. We have demonstrated that reducing PRMT7 levels in invasive breast cancer cells using RNA interference significantly decreased cell invasion in vitro and metastasis in vivo. Conversely, overexpression of PRMT7 in non-aggressive MCF7 cells enhanced their invasiveness. Furthermore, we show that PRMT7 induces the expression of matrix metalloproteinase 9 (MMP9), a well-known mediator of breast cancer metastasis. Importantly, we significantly rescued invasion of aggressive breast cancer cells depleted of PRMT7 by the exogenous expression of MMP9. Our results demonstrate that upregulation of PRMT7 in breast cancer may have a significant role in promoting cell invasion through the regulation of MMP9. This identifies PRMT7 as a novel and potentially significant biomarker and therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama/enzimología , Movimiento Celular , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ganglios Linfáticos/enzimología , Ganglios Linfáticos/patología , Metástasis Linfática , Metaloproteinasa 9 de la Matriz/genética , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Proteína-Arginina N-Metiltransferasas/genética , Interferencia de ARN , Transducción de Señal , Transfección , Regulación hacia Arriba
18.
Cancer Res ; 74(13): 3567-78, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24812275

RESUMEN

As cancer treatment tools, oncolytic viruses (OV) have yet to realize what some see as their ultimate clinical potential. In this study, we have engineered a chimeric vesicular stomatitis virus (VSV) that is devoid of its natural neurotoxicity while retaining potent oncolytic activity. The envelope glycoprotein (G) of VSV was replaced with a variant glycoprotein of the lymphocytic choriomeningitis virus (LCMV-GP), creating a replicating therapeutic, rVSV(GP), that is benign in normal brain but can effectively eliminate brain cancer in multiple preclinical tumor models in vivo. Furthermore, it can be safely administered systemically to mice and displays greater potency against a spectrum of human cancer cell lines than current OV candidates. Remarkably, rVSV(GP) escapes humoral immunity, thus, for the first time, allowing repeated systemic OV application without loss of therapeutic efficacy. Taken together, rVSV(GP) offers a considerably improved OV platform that lacks several of the major drawbacks that have limited the clinical potential of this technology to date.


Asunto(s)
Antígenos Virales/genética , Glioblastoma/terapia , Glicoproteínas/genética , Glicoproteínas de Membrana/genética , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética , Animales , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antivirales/inmunología , Línea Celular Tumoral , Cricetinae , Femenino , Vectores Genéticos , Humanos , Evasión Inmune , Inmunidad Humoral , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Ingeniería de Proteínas
19.
Mol Ther ; 22(7): 1320-1332, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24695102

RESUMEN

This study characterizes the ability of novel oncolytic rhabdoviruses (Maraba MG1) to boost natural killer (NK) cell activity. Our results demonstrate that MG1 activates NK cells via direct infection and maturation of conventional dendritic cells. Using NK depletion and conventional dendritic cells ablation studies in vivo, we established that both are required for MG1 efficacy. We further explored the efficacy of attenuated MG1 (nonreplicating MG1-UV(2min) and single-cycle replicating MG1-Gless) and demonstrated that these viruses activate conventional dendritic cells, although to a lesser extent than live MG1. This translates to equivalent abilities to remove tumor metastases only at the highest viral doses of attenuated MG1. In tandem, we characterized the antitumor ability of NK cells following preoperative administration of live and attenuated MG1. Our results demonstrates that a similar level of NK activation and reduction in postoperative tumor metastases was achieved with equivalent high viral doses concluding that viral replication is important, but not necessary for NK activation. Biochemical characterization of a panel of UV-inactivated MG1 (2-120 minutes) revealed that intact viral particle and target cell recognition are essential for NK cell-mediated antitumor responses. These findings provide mechanistic insight and preclinical rationale for safe perioperative virotherapy to effectively reduce metastatic disease following cancer surgery.


Asunto(s)
Células Dendríticas/citología , Células Asesinas Naturales/citología , Melanoma/terapia , Rhabdoviridae/fisiología , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Viroterapia Oncolítica/métodos
20.
Mol Ther ; 22(6): 1188-1197, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24569832

RESUMEN

Oncolytic viruses (OVs) and bacteria share the property of tumor-selective replication following systemic administration. In the case of nonpathogenic bacteria, tumor selectivity relates to their ability to grow extracellularly within tumor stroma and is therefore ideally suited to restricting the production of bacterially produced therapeutic agents to tumors. We have previously shown the ability of the type 1 interferon antagonist B18R to enhance the replication and spread of vesicular stomatitis virus (VSV) by overcoming related cellular innate immunity. In this study, we utilized nonpathogenic bacteria (E. coli) expressing B18R to facilitate tumor-specific production of B18R, resulting in a microenvironment depleted of bioactive antiviral cytokine, thus "preconditioning" the tumor to enhance subsequent tumor destruction by the OV. Both in vitro and in vivo infection by VSVΔ51 was greatly enhanced by B18R produced from E. coli. Moreover, a significant increase in therapeutic efficacy resulted from intravenous (i.v.) injection of bacteria to tumor-bearing mice 5 days prior to i.v. VSVΔ51 administration, as evidenced by a significant reduction in tumor growth and increased survival in mice. Our strategy is the first example where two such diverse microorganisms are rationally combined and demonstrates the feasibility of combining complementary microorganisms to improve therapeutic outcome.


Asunto(s)
Carcinoma Pulmonar de Lewis/patología , Escherichia coli/genética , Virus Oncolíticos/genética , Vesiculovirus/genética , Proteínas Virales/metabolismo , Animales , Carcinoma Pulmonar de Lewis/microbiología , Carcinoma Pulmonar de Lewis/terapia , Carcinoma Pulmonar de Lewis/virología , Línea Celular Tumoral , Escherichia coli/metabolismo , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacología , Células HT29 , Humanos , Inyecciones Intravenosas , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Vesiculovirus/fisiología , Proteínas Virales/genética , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA