Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 47(1): 159-170.e10, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723548

RESUMEN

Clearance of pathogens or tumor cells by antibodies traditionally requires both Fab and Fc domains of IgG. Here, we show the Fc domain of IgG alone mediates recognition and clearance of herpes simplex virus (HSV1)-infected cells. The human natural killer (NK) cell surface is naturally coated with IgG bound by its Fc domain to the Fcγ receptor CD16a. NK cells utilize the Fc domain of bound IgG to recognize gE, an HSV1-encoded glycoprotein that also binds the Fc domain of IgG but at a site distinct from CD16a. The bridge formed by the Fc domain between the HSV1-infected cell and the NK cell results in NK cell activation and lysis of the HSV1-infected cell in the absence of HSV1-specific antibody in vitro and prevents fatal HSV1 infection in vivo. This mechanism also explains how bacterial IgG-binding proteins regulate NK cell function and may be broadly applicable to Fcγ-receptor-bearing cells.


Asunto(s)
Anticuerpos Antivirales/metabolismo , Herpes Simple/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Células Asesinas Naturales/inmunología , Simplexvirus/inmunología , Animales , Anticuerpos Antivirales/inmunología , Células Cultivadas , Citotoxicidad Inmunológica , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Unión Proteica , Agregación de Receptores , Receptores de IgG/metabolismo , Transducción de Señal , Proteínas Virales/inmunología
2.
Mol Ther ; 30(6): 2153-2162, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35143960

RESUMEN

Cancer is a disease caused by loss of regulatory processes that control the cell cycle, resulting in increased proliferation. The loss of control can deregulate both tumor suppressors and oncogenes. Apart from cell intrinsic gene mutations and environmental factors, infection by cancer-causing viruses also induces changes that lead to malignant transformation. This can be caused by both expression of oncogenic viral proteins and also by changes in cellular genes and proteins that affect the epigenome. Thus, these epigenetic modifiers are good therapeutic targets, and several epigenetic inhibitors are approved for the treatment of different cancers. In addition to small molecule drugs, biological therapies, such as antibodies and viral therapies, are also increasingly being used to treat cancer. An HSV-1-derived oncolytic virus is currently approved by the US FDA and the European Medicines Agency. Similarly, an adenovirus-based therapeutic is approved for use in China for some cancer types. Because viruses can affect cellular epigenetics, the interaction of epigenome-targeting drugs with oncogenic and oncolytic viruses is a highly significant area of investigation. Here, we will review the current knowledge about the impact of using epigenetic drugs in tumors positive for oncogenic viruses or as therapeutic combinations with oncolytic viruses.


Asunto(s)
Histonas , Neoplasias , Virus Oncogénicos , Virus Oncolíticos , Histonas/genética , Humanos , Neoplasias/genética , Neoplasias/terapia , Virus Oncogénicos/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética
3.
Pharmacol Rev ; 72(3): 639-667, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32554488

RESUMEN

Cancer and organ injury-such as that occurring in the perioperative period, including acute lung injury, myocardial infarction, and acute gut injury-are among the leading causes of death in the United States and impose a significant impact on quality of life. MicroRNAs (miRNAs) have been studied extensively during the last two decades for their role as regulators of gene expression, their translational application as diagnostic markers, and their potential as therapeutic targets for disease treatment. Despite promising preclinical outcomes implicating miRNA targets in disease treatment, only a few miRNAs have reached clinical trials. This likely relates to difficulties in the delivery of miRNA drugs to their targets to achieve efficient inhibition or overexpression. Therefore, understanding how to efficiently deliver miRNAs into diseased tissues and specific cell types in patients is critical. This review summarizes current knowledge on various approaches to deliver therapeutic miRNAs or miRNA inhibitors and highlights current progress in miRNA-based disease therapy that has reached clinical trials. Based on ongoing advances in miRNA delivery, we believe that additional therapeutic approaches to modulate miRNA function will soon enter routine medical treatment of human disease, particularly for cancer or perioperative organ injury. SIGNIFICANCE STATEMENT: MicroRNAs have been studied extensively during the last two decades in cancer and organ injury, including acute lung injury, myocardial infarction, and acute gut injury, for their regulation of gene expression, application as diagnostic markers, and therapeutic potentials. In this review, we specifically emphasize the pros and cons of different delivery approaches to modulate microRNAs, as well as the most recent exciting progress in the field of therapeutic targeting of microRNAs for disease treatment in patients.


Asunto(s)
MicroARNs/genética , Neoplasias/genética , Heridas y Lesiones/genética , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Humanos , MicroARNs/biosíntesis , MicroARNs/sangre , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología , Heridas y Lesiones/terapia
4.
Ann Neurol ; 89(5): 926-941, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33675084

RESUMEN

OBJECTIVE: Brain arteriovenous malformations (bAVMs) are a leading cause of hemorrhagic stroke and neurological deficits in children and young adults, however, no pharmacological intervention is available to treat these patients. Although more than 95% of bAVMs are sporadic without family history, the pathogenesis of sporadic bAVMs is largely unknown, which may account for the lack of therapeutic options. KRAS mutations are frequently observed in cancer, and a recent unprecedented finding of these mutations in human sporadic bAVMs offers a new direction in the bAVM research. Using a novel adeno-associated virus targeting brain endothelium (AAV-BR1), the current study tested if endothelial KRASG12V mutation induces sporadic bAVMs in mice. METHODS: Five-week-old mice were systemically injected with either AAV-BR1-GFP or -KRASG12V . At 8 weeks after the AAV injection, bAVM formation and characteristics were addressed by histological and molecular analyses. The effect of MEK/ERK inhibition on KRASG12V -induced bAVMs was determined by treatment of trametinib, a US Food and Drug Administration (FDA)-approved MEK/ERK inhibitor. RESULTS: The viral-mediated KRASG12V overexpression induced bAVMs, which were composed of a tangled nidus mirroring the distinctive morphology of human bAVMs. The bAVMs were accompanied by focal angiogenesis, intracerebral hemorrhages, altered vascular constituents, neuroinflammation, and impaired sensory/cognitive/motor functions. Finally, we confirmed that bAVM growth was inhibited by trametinib treatment. INTERPRETATION: Our innovative approach using AAV-BR1 confirms that KRAS mutations promote bAVM development via the MEK/ERK pathway, and provides a novel preclinical mouse model of bAVMs which will be useful to develop a therapeutic strategy for patients with bAVM. ANN NEUROL 2021;89:926-941.


Asunto(s)
Endotelio Vascular , Malformaciones Arteriovenosas Intracraneales/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Cognición , Dependovirus/genética , Encefalitis/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Regulación de la Expresión Génica/genética , Humanos , Malformaciones Arteriovenosas Intracraneales/complicaciones , Malformaciones Arteriovenosas Intracraneales/psicología , Hemorragias Intracraneales/etiología , Hemorragias Intracraneales/genética , Imagen por Resonancia Magnética , Ratones , Mutación/genética , Neovascularización Patológica/etiología , Neovascularización Patológica/genética , Desempeño Psicomotor , Piridonas/farmacología , Pirimidinonas/farmacología
5.
BMC Cancer ; 22(1): 400, 2022 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-35418059

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is the most common high-grade malignant brain tumour in adults and arises from the glial cells in the brain. The prognosis of treated GBM remains very poor with 5-year survival rates of 5%, a figure which has not improved over the last few decades. Currently, there is a modest 14-month overall median survival in patients undergoing maximum safe resection plus adjuvant chemoradiotherapy. HOX gene dysregulation is now a widely recognised feature of many malignancies. METHODS: In this study we have focused on HOX gene dysregulation in GBM as a potential therapeutic target in a disease with high unmet need. RESULTS: We show significant dysregulation of these developmentally crucial genes and specifically that HOX genes A9, A10, C4 and D9 are strong candidates for biomarkers and treatment targets for GBM and GBM cancer stem cells. We evaluated a next generation therapeutic peptide, HTL-001, capable of targeting HOX gene over-expression in GBM by disrupting the interaction between HOX proteins and their co-factor, PBX. HTL-001 induced both caspase-dependent and -independent apoptosis in GBM cell lines. CONCLUSION: In vivo biodistribution studies confirmed that the peptide was able to cross the blood brain barrier. Systemic delivery of HTL-001 resulted in improved control of subcutaneous murine and human xenograft tumours and improved survival in a murine orthotopic model.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Adulto , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Genes Homeobox , Glioblastoma/tratamiento farmacológico , Glioblastoma/terapia , Humanos , Ratones , Péptidos/genética , Distribución Tisular
6.
Curr Issues Mol Biol ; 41: 381-468, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32938804

RESUMEN

The therapeutic promise of oncolytic viruses (OVs) rests on their ability to both selectively kill tumor cells and induce anti-tumor immunity. The potential of tumors to be recognized and eliminated by an effective anti-tumor immune response has been spurred on by the discovery that immune checkpoint inhibition can overcome tumor-specific cytotoxic T cell (CTL) exhaustion and provide durable responses in multiple tumor indications. OV-mediated tumor destruction is now recognized as a powerful means to assist in the development of anti-tumor immunity for two important reasons: (i) OVs, through the elicitation of an anti-viral response and the production of type I interferon, are potent stimulators of inflammation and can be armed with transgenes to further enhance anti-tumor immune responses; and (ii) lytic activity can promote the release of tumor-associated antigens (TAAs) and tumor neoantigens that function as in situ tumor-specific vaccines to elicit adaptive immunity. Oncolytic herpes simplex viruses (oHSVs) are among the most widely studied OVs for the treatment of solid malignancies, and Amgen's oHSV Imlygic® for the treatment of melanoma is the only OV approved in major markets. Here we describe important biological features of HSV that make it an attractive OV, clinical experience with HSV-based vectors, and strategies to increase applicability to cancer treatment.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Virus Oncolíticos/inmunología , Simplexvirus/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/inmunología , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos T Citotóxicos/inmunología
7.
Proc Natl Acad Sci U S A ; 115(19): 4927-4932, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686060

RESUMEN

In the present work, we investigated the role of natural killer (NK) cells in combination therapy with oncolytic virus (OV) and bortezomib, a proteasome inhibitor. NK cells display rapid and potent immunity to metastatic and hematological cancers, and they overcome immunosuppressive effects of tumor microenvironment. We developed a mathematical model to address the question of how the density of NK cells affects the growth of the tumor. We found that the antitumor efficacy increases when the endogenous NKs are depleted and also when exogenous NK cells are injected into the tumor. These predictions were validated by our in vivo and in vitro experiments.


Asunto(s)
Bortezomib/uso terapéutico , Neoplasias Hematológicas , Células Asesinas Naturales/inmunología , Modelos Inmunológicos , Viroterapia Oncolítica , Microambiente Tumoral , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Humanos , Células Asesinas Naturales/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Células Vero
8.
Mol Ther ; 25(7): 1544-1555, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28109960

RESUMEN

Targeted inhibition of oncogenic miRNA-21 has been proposed to treat glioblastoma by rescuing tumor suppressors, PTEN and PDCD4. However, systemic delivery of anti-miR-21 sequences requires a robust and efficient delivery platform to successfully inhibit this druggable target. Three-way-junction (3WJ)-based RNA nanoparticles (RNP), artificially derived from pRNA of bacteriophage phi29 DNA packaging motor, was recently shown to target glioblastoma. Here, we report that multi-valent folate (FA)-conjugated 3WJ RNP constructed to harbor anti-miR-21 LNA sequences (FA-3WJ-LNA-miR21) specifically targeted and delivered anti-miR-21 LNA and knocked down miR-21 expression in glioblastoma cells in vitro and in vivo with favorable biodistribution. Systemically injected FA-3WJ-LNA-miR21 RNP efficiently rescued PTEN and PDCD4, resulting in glioblastoma cell apoptosis and tumor growth regression. Overall survival rate was also significantly improved by FA-3WJ-LNA-miR21 RNP. These results are indicative of the clinical benefit of FA-3WJ RNP-based gene therapy for the successful targeted therapy of developing and even recurring glioblastoma.


Asunto(s)
Antagomirs/farmacología , Neoplasias Encefálicas/terapia , Regulación Neoplásica de la Expresión Génica , Glioblastoma/terapia , MicroARNs/antagonistas & inhibidores , Nanopartículas/administración & dosificación , Animales , Antagomirs/química , Antagomirs/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Portadores de Fármacos , Femenino , Receptores de Folato Anclados a GPI/genética , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/química , Ácido Fólico/metabolismo , Glioblastoma/genética , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Nanopartículas/química , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Gene Med ; 19(3)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28087981

RESUMEN

BACKGROUND: Malignant gliomas (glioblastomas; GBMs) are extremely aggressive and have a median survival of approximately 15 months. Current treatment modalities, which include surgical resection, radiation and chemotherapy, have done little to prolong the lives of GBM patients. Chondroitin sulfate proteoglycans (CSPG) are critical for cell-cell and cell-extracellular matrix (ECM) interactions and are implicated in glioma growth and invasion. Chondroitinase (Chase) ABC is a bacterial enzyme that cleaves chondroitin sulfate disaccharide chains from CSPGs in the tumor ECM. Wild-type Chase ABC has limited stability and/or activity in mammalian cells; therefore, we created a mutant humanized version (Chase M) with enhanced function in mammalian cells. METHODS: We hypothesized that disruption of cell-cell and cell-ECM interactions by ChaseM and temozolomide (TMZ) will enhance the chemotherapeutic availability and sensitivity of glioma cells. RESULTS: Utilizing primary patient-derived neurospheres, we found that ChaseM decreases glioma neurosphere aggregation in vitro. Furthermore, an oncolytic HSV-1 virus expressing secreted ChaseM (OV-ChaseM) enhanced viral spread and glioma cell killing compared to OV-Control, in vitro. OV-ChaseM plus TMZ combinatorial treatment resulted in a significant synergistic enhancement of glioma cell killing accompanied by an increase in apoptotic cell death. Intracellular flow cytometric analysis revealed a significant reduction in the phosphorylation of the pro-survival AKT protein following OV-ChaseM plus TMZ treatment. Lastly, in nude mice bearing intracranial GBM30 glioma xenografts, intratumoral OV-ChaseM plus TMZ (10 mg/kg by oral gavage) combination therapy resulted in a significant (p < 0.02) enhancement of survival compared to each individual treatment alone. CONCLUSIONS: These data reveal that OV-ChaseM enhances glioma cell viral susceptibility and sensitivity to TMZ.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Condroitina ABC Liasa/genética , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Glioblastoma/genética , Alelos , Sustitución de Aminoácidos , Animales , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Chlorocebus aethiops , Condroitina ABC Liasa/metabolismo , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Activación Enzimática , Expresión Génica , Vectores Genéticos/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Herpesvirus Humano 1/genética , Humanos , Ratones , Mutación , Viroterapia Oncolítica , Temozolomida , Transducción Genética , Resultado del Tratamiento , Carga Tumoral , Células Tumorales Cultivadas , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
10.
PLoS Genet ; 10(10): e1004652, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25340791

RESUMEN

Metastasis is the principal cause of cancer death and occurs through multiple, complex processes that involve the concerted action of many genes. A number of studies have indicated that the Fragile Histidine Triad (FHIT) gene product, FHIT, functions as a tumor suppressor in a variety of common human cancers. Although there are suggestions of a role for FHIT loss in progression of various cancers, a role for such loss in metastasis has not been defined. Here, via in vivo and in vitro assays, we reveal that the enforced expression of FHIT significantly suppresses metastasis, accompanied by inhibition of the epithelial-mesenchymal transition (EMT), a process involved in metastasis through coordinate modulation of EMT-related genes. Specifically, miR-30c, a FHIT-upregulated microRNA, contributes to FHIT function in suppression of EMT and metastasis by directly targeting metastasis genes Metadherin (MTDH), High-mobility group AT-hook 2 (HMGA2), and the mesenchymal markers, Vimentin (VIM) and Fibronectin (FN1), in human lung cancer. Finally, we demonstrate that the expression pattern of FHIT and miR-30c is inversely correlated with that of MTDH and HMGA2 in normal tissue, non-metastatic and metastatic tumors, serving as a potential biomarker for metastasis in lung cancer.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Transición Epitelial-Mesenquimal/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Ácido Anhídrido Hidrolasas/biosíntesis , Moléculas de Adhesión Celular , Línea Celular Tumoral , Fibronectinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Proteína HMGA2/biosíntesis , Humanos , Neoplasias Pulmonares/patología , Proteínas de la Membrana , MicroARNs/biosíntesis , Metástasis de la Neoplasia , Proteínas de Neoplasias/biosíntesis , Proteínas de Unión al ARN , Vimentina/biosíntesis
11.
Biomed Microdevices ; 18(2): 38, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27106026

RESUMEN

EPR (electron paramagnetic resonance) based biological oximetry is a powerful tool that accurately and repeatedly measures tissue oxygen levels. In vivo determination of oxygen in tissues is crucial for the diagnosis and treatment of a number of diseases. Here, we report the first successful fabrication and remarkable properties of nanofiber sensors for EPR-oximetry applications. Lithium octa-n-butoxynaphthalocyanine (LiNc- BuO), an excellent paramagnetic oxygen sensor, was successfully encapsulated in 300-500 nm diameter fibers consisting of a core of polydimethylsiloxane (PDMS) and a shell of polycaprolactone (PCL) by electrospinning. This core-shell nanosensor (LiNc-BuO-PDMS-PCL) shows a linear dependence of linewidth versus oxygen partial pressure (pO2). The nanofiber sensors have response and recovery times of 0.35 s and 0.55 s, respectively, these response and recovery times are ~12 times and ~218 times faster than those previously reported for PDMS-LiNc-BuO chip sensors. This greater responsiveness is likely due to the high porosity and excellent oxygen permeability of the nanofibers. Electrospinning of the structurally flexible PDMS enabled the fabrication of fibers having tailored spin densities. Core-shell encapsulation ensures the non-exposure of embedded LiNc-BuO and mitigates potential biocompatibility concerns. In vitro evaluation of the fiber performed under exposure to cultured cells showed that it is both stable and biocompatible. The unique combination of biocompatibility due to the PCL 'shell,' the excellent oxygen transparency of the PDMS core, and the excellent oxygen-sensing properties of LiNc-BuO makes LiNc-BuO-PDMS-PCL platform promising for long-term oximetry and repetitive oxygen measurements in both biological systems and clinical applications.


Asunto(s)
Fenómenos Magnéticos , Nanofibras/química , Oximetría/instrumentación , Animales , Células CHO , Cricetinae , Cricetulus , Dimetilpolisiloxanos/química , Ensayo de Materiales , Oxígeno/análisis , Poliésteres/química , Porfirinas/química , Presión , Factores de Tiempo
12.
J Neurooncol ; 126(3): 377-84, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26542029

RESUMEN

The five-year survival rate for patients with malignant glioma is less than 10%. Despite aggressive chemo/radiotherapy these tumors have remained resistant to almost every interventional strategy evaluated in patients. Resistance to these agents is attributed to extrinsic mechanisms such as the tumor microenvironment, poor drug penetration, and tumoral heterogeneity. In addition, genetic and molecular examination of these tumors has revealed defective apoptotic regulation, enhanced pro-survival autophagy signaling, and a propensity for necrosis that aids in the adaptation to environmental stress and resistance to treatment. The combination of extrinsic and intrinsic hallmarks in glioma contributes to the multifaceted resistance to traditional anti-tumor agents. Here we describe the biology of the disease relevant to therapeutic resistance, with a specific focus on molecular deregulation of cell death pathways. Emerging studies investigating the targeting of these pathways including BH3 mimetics and autophagy inhibitors that are being evaluated in both the preclinical and clinical settings are discussed. This review highlights the pathways exploited by glioblastoma cells that drive their hallmark pro-survival predisposition and makes therapy development such a challenge.


Asunto(s)
Apoptosis , Neoplasias Encefálicas/patología , Glioma/patología , Humanos
13.
Mol Ther ; 23(1): 99-107, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25200130

RESUMEN

Glioblastoma multiforme (GBM) is an aggressive brain cancer for which there is no effective treatment. Oncolytic HSV vectors (oHSVs) are attenuated lytic viruses that have shown promise in the treatment of human GBM models in animals, but their efficacy in early phase patient trials has been limited. Instead of attenuating the virus with mutations in virulence genes, we engineered four copies of the recognition sequence for miR-124 into the 3'UTR of the essential ICP4 gene to protect healthy tissue against lytic virus replication; miR-124 is expressed in neurons but not in glioblastoma cells. Following intracranial inoculation into nude mice, the miR-124-sensitive vector failed to replicate or show overt signs of pathogenesis. To address the concern that this safety feature may reduce oncolytic activity, we inserted the miR-124 response elements into an unattenuated, human receptor (EGFR/EGFRvIII)-specific HSV vector. We found that miR-124 sensitivity did not cause a loss of treatment efficiency in an orthotopic model of primary human GBM in nude mice. These results demonstrate that engineered miR-124 responsiveness can eliminate off-target replication by unattenuated oHSV without compromising oncolytic activity, thereby providing increased safety.


Asunto(s)
Regiones no Traducidas 3' , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/genética , MicroARNs/genética , Viroterapia Oncolítica/métodos , Animales , Secuencia de Bases , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Cromosomas Artificiales Bacterianos/química , Cromosomas Artificiales Bacterianos/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Células HEK293 , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/metabolismo , Inyecciones Intraventriculares , Ratones , Ratones Desnudos , MicroARNs/metabolismo , Datos de Secuencia Molecular , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Bull Math Biol ; 77(6): 984-1012, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25758754

RESUMEN

Oncolytic virus (OV) is a genetically engineered virus that can selectively replicate in and kill tumor cells while not harming normal cells. OV therapy has been explored as a treatment for numerous cancers including glioblastoma, an aggressive and devastating brain tumor. Experiments show that extracellular matrix protein CCN1 limits OV therapy of glioma by orchestrating an antiviral response and enhancing the proinflammatory activation and migration of macrophages. Neutralizing CCN1 by antibody has been demonstrated to improve OV spread and tends to increase the time to disease progression. In this paper, we develop a mathematical model to investigate the effects of CCN1 on the treatment of glioma with oncolytic herpes simplex virus. We show that numerical simulations of the model are in agreement with the experimental results and then use the model to explore the anti-tumor effects of combining antibodies with OV therapy. Model simulations suggest that the macrophage content of the tumor is a critical factor to the success of OV therapy and to the reduction in tumor volume gained with the CCN1 antibody.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Viroterapia Oncolítica , Animales , Anticuerpos Neutralizantes/administración & dosificación , Neoplasias Encefálicas/metabolismo , Proteína 61 Rica en Cisteína/antagonistas & inhibidores , Proteína 61 Rica en Cisteína/metabolismo , Glioma/metabolismo , Glioma/patología , Humanos , Activación de Macrófagos , Macrófagos/patología , Conceptos Matemáticos , Ratones , Modelos Biológicos
15.
Mol Ther ; 22(9): 1678-87, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24895995

RESUMEN

Glioblastoma is a devastating disease, and there is an urgent need to develop novel therapies, such as oncolytic HSV1 (OV) to effectively target tumor cells. OV therapy depends on tumor-specific replication leading to destruction of neoplastic tissues. Host responses that curtail virus replication limit its efficacy in vivo. We have previously shown that cysteine-rich 61 protein (CCN1) activates a type 1 IFN antiviral defense response in glioblastoma cells. Incorporating TCGA data, we found CCN1 expression to be a negative prognostic factor for glioblastoma patients. Based on this, we used neutralizing antibodies against CCN1 to investigate its effect on OV therapy. Use of an anti-CCN1 antibody in mice bearing glioblastomas treated with OV led to enhanced virus expression along with reduced immune cell infiltration. OV-induced CCN1 increases macrophage migration toward infected glioblastoma cells by directly binding macrophages and also by enhancing the proinflammatory activation of macrophages inducing MCP-1 expression in glioblastoma cells. Activation of macrophages by CCN1 also increases viral clearance. Neutralization of integrin αMß2 reversed CCN1-induced macrophage activation and migration, and reduced MCP-1 expression by glioblastoma cells. Our findings reveal that CCN1 plays a novel role in pathogen clearance; increasing macrophage infiltration and activation resulting in increased virus clearance in tumors.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Proteína 61 Rica en Cisteína/genética , Proteína 61 Rica en Cisteína/metabolismo , Glioblastoma/inmunología , Herpesvirus Humano 1/genética , Macrófagos/metabolismo , Animales , Anticuerpos Monoclonales/uso terapéutico , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Femenino , Vectores Genéticos/administración & dosificación , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Activación de Macrófagos , Ratones , Trasplante de Neoplasias , Virus Oncolíticos/genética
16.
Proc Natl Acad Sci U S A ; 109(14): 5316-21, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22431589

RESUMEN

MicroRNAs (miRNAs) are increasingly implicated in regulating cancer initiation and progression. In this study, two miRNAs, miR-25 and -32, are identified as p53-repressed miRNAs by p53-dependent negative regulation of their transcriptional regulators, E2F1 and MYC. However, miR-25 and -32 result in p53 accumulation by directly targeting Mdm2 and TSC1, which are negative regulators of p53 and the mTOR (mammalian target of rapamycin) pathway, respectively, leading to inhibition of cellular proliferation through cell cycle arrest. Thus, there is a recurrent autoregulatory circuit involving expression of p53, E2F1, and MYC to regulate the expression of miR-25 and -32, which are miRNAs that, in turn, control p53 accumulation. Significantly, overexpression of transfected miR-25 and -32 in glioblastoma multiforme cells inhibited growth of the glioblastoma multiforme cells in mouse brain in vivo. The results define miR-25 and -32 as positive regulators of p53, underscoring their role in tumorigenesis in glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , MicroARNs/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Encefálicas/patología , Ciclo Celular , Proliferación Celular , Factor de Transcripción E2F1/fisiología , Glioblastoma/patología , Humanos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas c-myc/fisiología , Transcripción Genética , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
17.
J Biol Chem ; 288(31): 22248-56, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23782696

RESUMEN

Brain-specific angiogenesis inhibitor-1 (BAI1) is an adhesion G protein-coupled receptor that has been studied primarily for its anti-angiogenic and anti-tumorigenic properties. We found that overexpression of BAI1 results in activation of the Rho pathway via a Gα(12/13)-dependent mechanism, with truncation of the BAI1 N terminus resulting in a dramatic enhancement in receptor signaling. This constitutive activity of the truncated BAI1 mutant also resulted in enhanced downstream phosphorylation of ERK as well as increased receptor association with ß-arrestin2 and increased ubiquitination of the receptor. To gain insights into the regulation of BAI1 signaling, we screened the C terminus of BAI1 against a proteomic array of PDZ domains to identify novel interacting partners. These screens revealed that the BAI1 C terminus interacts with a variety of PDZ domains from synaptic proteins, including MAGI-3. Removal of the BAI1 PDZ-binding motif resulted in attenuation of receptor signaling to Rho but had no effect on ERK activation. Conversely, co-expression with MAGI-3 was found to potentiate signaling to ERK by constitutively active BAI1 in a manner that was dependent on the PDZ-binding motif of the receptor. Biochemical fractionation studies revealed that BAI1 is highly enriched in post-synaptic density fractions, a finding consistent with our observations that BAI1 can interact with PDZ proteins known to be concentrated in the post-synaptic density. These findings demonstrate that BAI1 is a synaptic receptor that can activate both the Rho and ERK pathways, with the N-terminal and C-terminal regions of the receptor playing key roles in the regulation of BAI1 signaling activity.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Densidad Postsináptica/metabolismo , Transducción de Señal , Proteínas Angiogénicas/fisiología , Animales , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Ratones , Dominios PDZ , Unión Proteica , Receptores Acoplados a Proteínas G
18.
Mol Ther ; 21(8): 1517-25, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23732993

RESUMEN

Saposin C-dioleoylphosphatidylserine (SapC-DOPS) nanovesicles are a nanotherapeutic which effectively target and destroy cancer cells. Here, we explore the systemic use of SapC-DOPS in several models of brain cancer, including glioblastoma multiforme (GBM), and the molecular mechanism behind its tumor-selective targeting specificity. Using two validated spontaneous brain tumor models, we demonstrate the ability of SapC-DOPS to selectively and effectively cross the blood-brain tumor barrier (BBTB) to target brain tumors in vivo and reveal the targeting to be contingent on the exposure of the anionic phospholipid phosphatidylserine (PtdSer). Increased cell surface expression of PtdSer levels was found to correlate with SapC-DOPS-induced killing efficacy, and tumor targeting in vivo was inhibited by blocking PtdSer exposed on cells. Apart from cancer cell killing, SapC-DOPS also exerted a strong antiangiogenic activity in vitro and in vivo. Interestingly, unlike traditional chemotherapy, hypoxic cells were sensitized to SapC-DOPS-mediated killing. This study emphasizes the importance of PtdSer exposure for SapC-DOPS targeting and supports the further development of SapC-DOPS as a novel antitumor and antiangiogenic agent for brain tumors.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Nanopartículas/administración & dosificación , Fosfatidilserinas/química , Saposinas/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Hipoxia de la Célula , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Masculino , Ratones , Nanopartículas/química , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Saposinas/administración & dosificación , Saposinas/química , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Ther ; 21(3): 561-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23070115

RESUMEN

Glioblastoma multiforme (GBM) remains an untreatable human brain malignancy. Despite promising preclinical studies using oncolytic herpes simplex virus (oHSV) vectors, efficacy in patients has been limited by inefficient virus replication in tumor cells. This disappointing outcome can be attributed in part to attenuating mutations engineered into these viruses to prevent replication in normal cells. Alternatively, retargeting of fully replication-competent HSV to tumor-associated receptors has the potential to achieve tumor specificity without impairment of oncolytic activity. Here, we report the establishment of an HSV retargeting system that relies on the combination of two engineered viral glycoproteins, gD and gB, to mediate highly efficient HSV infection exclusively through recognition of the abundantly expressed epidermal growth factor receptor (EGFR) on glioblastoma cells. We demonstrate efficacy in vitro and in a heterotopic tumor model in mice. Evidence for systemically administered virus homing to the tumor mass is presented. Treatment of orthotopic primary human GBM xenografts demonstrated prolonged survival with up to 73% of animals showing a complete response as confirmed by magnetic resonance imaging. Our study describes an approach to HSV retargeting that is effective in a glioma model and may be applicable to the treatment of a broad range of tumor types.


Asunto(s)
Receptores ErbB/metabolismo , Glioblastoma/terapia , Viroterapia Oncolítica/métodos , Simplexvirus/genética , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Cricetinae , Femenino , Vectores Genéticos , Células HT29 , Humanos , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Plásmidos , Recombinación Genética , Simplexvirus/fisiología , Resultado del Tratamiento , Células Vero , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Mol Ther ; 21(5): 1014-23, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23481323

RESUMEN

Understanding the host response to oncolytic viruses is important to maximize their antitumor efficacy. Despite robust cytotoxicity and high virus production of an oncolytic herpes simplex virus (oHSV) in cultured human sarcoma cells, intratumoral (ITu) virus injection resulted in only mild antitumor effects in some xenograft models, prompting us to characterize the host inflammatory response. Virotherapy induced an acute neutrophilic infiltrate, a relative decrease of ITu macrophages, and a myeloid cell-dependent upregulation of host-derived vascular endothelial growth factor (VEGF). Anti-VEGF antibodies, bevacizumab and r84, the latter of which binds VEGF and selectively inhibits binding to VEGF receptor-2 (VEGFR2) but not VEGFR1, enhanced the antitumor effects of virotherapy, in part due to decreased angiogenesis but not increased virus production. Neither antibody affected neutrophilic infiltration but both partially mitigated virus-induced depletion of macrophages. Enhancement of virotherapy-mediated antitumor effects by anti-VEGF antibodies could largely be recapitulated by systemic depletion of CD11b(+) cells. These data suggest the combined effect of oHSV virotherapy and anti-VEGF antibodies is in part due to modulation of a host inflammatory reaction to virus. Our data provide strong preclinical support for combined oHSV and anti-VEGF antibody therapy and suggest that understanding and counteracting the innate host response may help enable the full antitumor potential of oncolytic virotherapy.


Asunto(s)
Vectores Genéticos/inmunología , Células Mieloides/inmunología , Neoplasias/inmunología , Virus Oncolíticos/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacología , Bevacizumab , Antígeno CD11b/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Células Mieloides/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Neovascularización Patológica/terapia , Viroterapia Oncolítica , Sarcoma/inmunología , Sarcoma/metabolismo , Sarcoma/terapia , Simplexvirus/inmunología , Células del Estroma/metabolismo , Células del Estroma/virología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/inmunología , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA