Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cell Rep ; 42(8): 112927, 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37537841

RESUMEN

Tumor relapse is linked to rapid chemoresistance and represents a bottleneck for cancer therapy success. Engagement of a reduced proliferation state is a non-mutational mechanism exploited by cancer cells to bypass therapy-induced cell death. Through combining functional pulse-chase experiments in engineered cells and transcriptomic analyses, we identify DPPA3 as a master regulator of slow-cycling and chemoresistant phenotype in colorectal cancer (CRC). We find a vicious DPPA3-HIF1α feedback loop that downregulates FOXM1 expression via DNA methylation, thereby delaying cell-cycle progression. Moreover, downregulation of HIF1α partially restores a chemosensitive proliferative phenotype in DPPA3-overexpressing cancer cells. In cohorts of CRC patient samples, DPPA3 overexpression acts as a predictive biomarker of chemotherapeutic resistance that subsequently requires reduction in its expression to allow metastatic outgrowth. Our work demonstrates that slow-cycling cancer cells exploit a DPPA3/HIF1α axis to support tumor persistence under therapeutic stress and provides insights on the molecular regulation of disease progression.

3.
J Vis Exp ; (192)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36847362

RESUMEN

Over the last decade, more sophisticated preclinical colorectal cancer (CRC) models have been established using patient-derived cancer cells and 3D tumoroids. Since patient derived tumor organoids can retain the characteristics of the original tumor, these reliable preclinical models enable cancer drug screening and the study of drug resistance mechanisms. However, CRC related death in patients is mostly associated with the presence of metastatic disease. It is therefore essential to evaluate the efficacy of anti-cancer therapies in relevant in vivo models that truly recapitulate the key molecular features of human cancer metastasis. We have established an orthotopic model based on the injection of CRC patient-derived cancer cells directly into the cecum wall of mice. These tumor cells develop primary tumors in the cecum that metastasize to the liver and lungs, which is frequently observed in patients with advanced CRC. This CRC mouse model can be used to evaluate drug responses monitored by microcomputed tomography (µCT), a clinically relevant small-scale imaging method that can easily identify primary tumors or metastases in patients. Here, we describe the surgical procedure and the required methodology to implant patient-derived cancer cells in the cecum wall of immunodeficient mice.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Humanos , Ratones , Animales , Microtomografía por Rayos X , Neoplasias Colorrectales/patología , Ciego/patología , Implantación del Embrión , Modelos Animales de Enfermedad
4.
Clin Cancer Res ; 27(3): 889-902, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33257429

RESUMEN

PURPOSE: Extracellular matrix (ECM) component hyaluronan (HA) facilitates malignant phenotypes of glioblastoma (GBM), however, whether HA impacts response to GBM immunotherapies is not known. Herein, we investigated whether degradation of HA enhances oncolytic virus immunotherapy for GBM. EXPERIMENTAL DESIGN: Presence of HA was examined in patient and murine GBM. Hyaluronidase-expressing oncolytic adenovirus, ICOVIR17, and its parental virus, ICOVIR15, without transgene, were tested to determine if they increased animal survival and modulated the immune tumor microenvironment (TME) in orthotopic GBM. HA regulation of NF-κB signaling was examined in virus-infected murine macrophages. We combined ICOVIR17 with PD-1 checkpoint blockade and assessed efficacy and determined mechanistic contributions of tumor-infiltrating myeloid and T cells. RESULTS: Treatment of murine orthotopic GBM with ICOVIR17 increased tumor-infiltrating CD8+ T cells and macrophages, and upregulated PD-L1 on GBM cells and macrophages, leading to prolonged animal survival, compared with control virus ICOVIR15. High molecular weight HA inhibits adenovirus-induced NF-κB signaling in macrophages in vitro, linking HA degradation to macrophage activation. Combining ICOVIR17 with anti-PD-1 antibody further extended the survival of GBM-bearing mice, achieving long-term remission in some animals. Mechanistically, CD4+ T cells, CD8+ T cells, and macrophages all contributed to the combination therapy that induced tumor-associated proinflammatory macrophages and tumor-specific T-cell cytotoxicity locally and systemically. CONCLUSIONS: Our studies are the first to show that immune modulatory ICOVIR17 has a dual role of mediating degradation of HA within GBM ECM and subsequently modifying the immune landscape of the TME, and offers a mechanistic combination immunotherapy with PD-L1/PD-1 blockade that remodels innate and adaptive immune cells.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Hialuronoglucosaminidasa/genética , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Femenino , Glioblastoma/genética , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Inmunoterapia/métodos , Ratones , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Clin Invest ; 129(4): 1407-1418, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30829653

RESUMEN

Oncolytic virotherapy (OVT) is a promising approach in which WT or engineered viruses selectively replicate and destroy tumor cells while sparing normal ones. In the last two decades, different oncolytic viruses (OVs) have been modified and tested in a number of preclinical studies, some of which have led to clinical trials in cancer patients. These clinical trials have revealed several critical limitations with regard to viral delivery, spread, resistance, and antiviral immunity. Here, we focus on promising research strategies that have been developed to overcome the aforementioned obstacles. Such strategies include engineering OVs to target a broad spectrum of tumor cells while evading the immune system, developing unique delivery mechanisms, combining other immunotherapeutic agents with OVT, and using clinically translatable mouse tumor models to potentially translate OVT more readily into clinical settings.


Asunto(s)
Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia , Viroterapia Oncolítica , Virus Oncolíticos/metabolismo , Animales , Humanos , Ratones , Neoplasias Experimentales/patología
6.
Theranostics ; 8(12): 3380-3391, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29930736

RESUMEN

Reporter gene systems can serve as therapy targets. However, the therapeutic use of reporters has been limited by the challenges of transgene delivery to a majority of cancer cells. This study specifically assesses the efficacy of targeting human somatostatin receptor subtype 2 (hSSTR2) with peptide receptor radionuclide therapy (PRRT) when a small subpopulation of cells bears the transgene. Methods: The hSSTR2 transgene was delivered to A549 and Panc-1tumors using the lentiviral vector, LV-hSSTR2-IRES-GFP or murine mesenchymal stem cells (mMSC)s using a retroviral vector. SSTR2 expression was assessed using Western blot and correlated to GFP fluorescence and 68Ga-DOTATOC uptake. Wild type (WT), transduced (TD), and mixed population A549 or Panc-1 xenografts were implanted in nude mice. Separate groups with A549WT and Panc-1WT tumors received intratumoral injection of SSTR2-expressing mMSCs. Tumor-bearing mice were treated with 90Y-DOTATOC or saline and evaluated with 68Ga-DOTATOC PET before and after treatment. Results: Cell studies showed a strong correlation between 68Ga-DOTATOC uptake and SSTR2 expression in A549 (p < 0.004) and Panc-1 cells (p < 0.01). 68Ga-DOTATOC PET SUVmean was 8- and 5-fold higher in TD compared to WT A549 and Panc-1 tumors, respectively (p < 0.001). After 90Y-DOTATOC treatment, 100% TD and mixed population TD xenografts showed growth cessation while the WT xenografts did not. A549WT and Panc-1WT tumors with SSTR2-expressing mMSCs treated with 90Y-DOTATOC showed significantly lower tumor volumes compared to controls (p < 0.05). 68Ga-DOTATOC PET SUVmean of treated TD tumors monotonically declined and was significantly lower than that of non-treated xenografts. Conclusions: We showed that SSTR2 delivery to a small population of cells in tumor in conjunction with PRRT is effective in tumor growth cessation. The availability of various transgene delivery methods for hSSTR2 and radiotherpaeutic somatostatin analogs highlights the direct translational potential of this paradigm in the treatment of various cancers.


Asunto(s)
Biomarcadores de Tumor/análisis , Terapia Molecular Dirigida/métodos , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/tratamiento farmacológico , Tomografía de Emisión de Positrones/métodos , Receptores de Somatostatina/metabolismo , Nanomedicina Teranóstica/métodos , Células A549 , Animales , Antineoplásicos/administración & dosificación , Modelos Animales de Enfermedad , Genes Reporteros , Xenoinjertos , Humanos , Células Madre Mesenquimatosas , Ratones Desnudos , Trasplante de Neoplasias , Octreótido/administración & dosificación , Octreótido/análogos & derivados , Radiofármacos/administración & dosificación , Resultado del Tratamiento
7.
J Clin Invest ; 128(9): 3887-3905, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-29944140

RESUMEN

Dormant or slow-cycling tumor cells can form a residual chemoresistant reservoir responsible for relapse in patients, years after curative surgery and adjuvant therapy. We have adapted the pulse-chase expression of H2BeGFP for labeling and isolating slow-cycling cancer cells (SCCCs). SCCCs showed cancer initiation potential and enhanced chemoresistance. Cells at this slow-cycling status presented a distinctive nongenetic and cell-autonomous gene expression profile shared across different tumor types. We identified TET2 epigenetic enzyme as a key factor controlling SCCC numbers, survival, and tumor recurrence. 5-Hydroxymethylcytosine (5hmC), generated by TET2 enzymatic activity, labeled the SCCC genome in carcinomas and was a predictive biomarker of relapse and survival in cancer patients. We have shown the enhanced chemoresistance of SCCCs and revealed 5hmC as a biomarker for their clinical identification and TET2 as a potential drug target for SCCC elimination that could extend patients' survival.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Dioxigenasas , Resistencia a Antineoplásicos/genética , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Neoplasias/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Recurrencia , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Clin Cancer Res ; 23(22): 7047-7058, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28912136

RESUMEN

Purpose: Despite tumor resection being the first-line clinical care for glioblastoma (GBM) patients, nearly all preclinical immune therapy models intend to treat established GBM. Characterizing cytoreductive surgery-induced immune response combined with the administration of immune cytokines has the potential of offering a new treatment paradigm of immune therapy for GBMs.Experimental Design: We developed syngeneic orthotopic mouse GBM models of tumor resection and characterized the immune response of intact and resected tumors. We also created a highly secretable variant of immune cytokine IFNß to enhance its release from engineered mouse mesenchymal stem cells (MSC-IFNß) and assessed whether surgical resection of intracranial GBM tumor significantly enhanced the antitumor efficacy of targeted on-site delivery of encapsulated MSC-IFNß.Results: We show that tumor debulking results in substantial reduction of myeloid-derived suppressor cells (MDSC) and simultaneous recruitment of CD4/CD8 T cells. This immune response significantly enhanced the antitumor efficacy of locally delivered encapsulated MSC-IFNß via enhanced selective postsurgical infiltration of CD8 T cells and directly induced cell-cycle arrest in tumor cells, resulting in increased survival of mice. Utilizing encapsulated human MSC-IFNß in resected orthotopic tumor xenografts of patient-derived GBM, we further show that IFNß induces cell-cycle arrest followed by apoptosis, resulting in increased survival in immunocompromised mice despite their absence of an intact immune system.Conclusions: This study demonstrates the importance of syngeneic tumor resection models in developing cancer immunotherapies and emphasizes the translational potential of local delivery of immunotherapeutic agents in treating cancer. Clin Cancer Res; 23(22); 7047-58. ©2017 AACR.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/inmunología , Interferón beta/genética , Células Madre/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Interferón beta/metabolismo , Ratones , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase S del Ciclo Celular/genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Brain ; 138(Pt 6): 1710-21, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25910782

RESUMEN

Characterizing clinically relevant brain metastasis models and assessing the therapeutic efficacy in such models are fundamental for the development of novel therapies for metastatic brain cancers. In this study, we have developed an in vivo imageable breast-to-brain metastasis mouse model. Using real time in vivo imaging and subsequent composite fluorescence imaging, we show a widespread distribution of micro- and macro-metastasis in different stages of metastatic progression. We also show extravasation of tumour cells and the close association of tumour cells with blood vessels in the brain thus mimicking the multi-foci metastases observed in the clinics. Next, we explored the ability of engineered adult stem cells to track metastatic deposits in this model and show that engineered stem cells either implanted or injected via circulation efficiently home to metastatic tumour deposits in the brain. Based on the recent findings that metastatic tumour cells adopt unique mechanisms of evading apoptosis to successfully colonize in the brain, we reasoned that TNF receptor superfamily member 10A/10B apoptosis-inducing ligand (TRAIL) based pro-apoptotic therapies that induce death receptor signalling within the metastatic tumour cells might be a favourable therapeutic approach. We engineered stem cells to express a tumour selective, potent and secretable variant of a TRAIL, S-TRAIL, and show that these cells significantly suppressed metastatic tumour growth and prolonged the survival of mice bearing metastatic breast tumours. Furthermore, the incorporation of pro-drug converting enzyme, herpes simplex virus thymidine kinase, into therapeutic S-TRAIL secreting stem cells allowed their eradication post-tumour treatment. These studies are the first of their kind that provide insight into targeting brain metastasis with stem-cell mediated delivery of pro-apoptotic ligands and have important clinical implications.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/terapia , Neoplasias de la Mama/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular , Femenino , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Simplexvirus/enzimología , Simplexvirus/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Timidina Quinasa/uso terapéutico
11.
Mol Ther ; 23(1): 108-18, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25352242

RESUMEN

Despite the proven safety of oncolytic viruses (OV) in clinical trials for glioblastoma (GBM), their efficacy has been hindered by suboptimal spreading within the tumor. We show that hyaluronan or hyaluronic acid (HA), an important component of extracellular matrix (ECM), is highly expressed in a majority of tumor xenografts established from patient-derived GBM lines that present both invasive and nodular phenotypes. Intratumoral injection of a conditionally replicating adenovirus expressing soluble hyaluronidase (ICOVIR17) into nodular GBM, mediated HA degradation and enhanced viral spread, resulting in a significant antitumor effect and mice survival. In an effort to translate OV-based therapeutics into clinical settings, we encapsulated human adipose-derived mesenchymal stem cells (MSC) loaded with ICOVIR17 in biocompatible synthetic extracellular matrix (sECM) and tested their efficacy in a clinically relevant mouse model of GBM resection. Compared with direct injection of ICOVIR17, sECM-MSC loaded with ICOVIR17 resulted in a significant decrease in tumor regrowth and increased mice survival. This is the first report of its kind revealing the expression of HA in GBM and the role of OV-mediated HA targeting in clinically relevant mouse model of GBM resection and thus has clinical implications.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Hialuronoglucosaminidasa/genética , Células Madre Mesenquimatosas/metabolismo , Viroterapia Oncolítica/métodos , Proteínas Virales/genética , Adenoviridae/metabolismo , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Expresión Génica , Glioblastoma/genética , Glioblastoma/mortalidad , Glioblastoma/patología , Células HEK293 , Humanos , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Inyecciones Intralesiones , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/virología , Ratones , Ratones Desnudos , Análisis de Supervivencia , Proteínas Virales/metabolismo , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Natl Cancer Inst ; 106(6): dju090, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24838834

RESUMEN

BACKGROUND: The current treatment regimen for malignant glioblastoma multiforme (GBM) is tumor resection followed by chemotherapy and radiation therapy. Despite the proven safety of oncolytic herpes simplex virus (oHSV) in clinical trials for GBMs, its efficacy is suboptimal mainly because of insufficient viral spread after tumor resection. METHODS: Human mesenchymal stem cells (MSC) were loaded with oHSV (MSC-oHSV), and their fate was explored by real-time imaging in vitro and in vivo. Using novel diagnostic and armed oHSV mutants and real-time multimodality imaging, the efficacy of MSC-oHSV and its proapoptotic variant, oHSV-TRAIL encapsulated in biocompatible synthetic extracellular matrix (sECM), was tested in different mouse GBM models, which more accurately reflect the current clinical settings of malignant, resistant, and resected tumors. All statistical tests were two-sided. RESULTS: MSC-oHSVs effectively produce oHSV progeny, which results in killing of GBMs in vitro and in vivo mediated by a dynamic process of oHSV infection and tumor destruction. sECM-encapsulated MSC-oHSVs result in statistically significant increased anti-GBM efficacy compared with direct injection of purified oHSV in a preclinical model of GBM resection, resulting in prolonged median survival in mice (P < .001 with Gehan-Breslow-Wilcoxin test). To supersede resistant tumors, MSC loaded with oHSV-TRAIL effectively induce apoptosis-mediated killing and prolonged median survival in mice bearing oHSV- and TRAIL-resistant GBM in vitro (P < .001 with χ(2) contingency test). CONCLUSIONS: Human MSC loaded with different oHSV variants provide a platform to translate oncolytic virus therapies to clinics in a broad spectrum of GBMs after resection and could also have direct implications in different cancer types.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Células Madre Mesenquimatosas/virología , Viroterapia Oncolítica/métodos , Simplexvirus , Animales , Femenino , Humanos , Ratones , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Stem Cells ; 31(8): 1706-14, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23389839

RESUMEN

Therapeutically engineered stem cells (SC) are emerging as an effective tumor-targeted approach for different cancer types. However, the assessment of the long-term fate of therapeutic SC post-tumor treatment is critical if such promising therapies are to be translated into clinical practice. In this study, we have developed an efficient SC-based therapeutic strategy that simultaneously allows killing of tumor cells and assessment and eradication of SC after treatment of highly malignant glioblastoma multiforme (GBM). Mesenchymal stem cells (MSC) engineered to co-express the prodrug converting enzyme, herpes simplex virus thymidine kinase (HSV-TK) and a potent and secretable variant of tumor necrosis factor apoptosis-inducing ligand (S-TRAIL) induced caspase-mediated GBM cell death and showed selective MSC sensitization to the prodrug ganciclovir (GCV). A significant decrease in tumor growth and a subsequent increase in survival were observed when mice bearing highly aggressive GBM were treated with MSC coexpressing S-TRAIL and HSV-TK. Furthermore, the systemic administration of GCV post-tumor treatment selectively eliminated therapeutic MSC expressing HSV-TK in vitro and in vivo, which was monitored in real time by positron emission-computed tomography imaging using 18F-FHBG, a substrate for HSV-TK. These findings demonstrate the development and validation of a novel therapeutic strategy that has implications in translating SC-based therapies in cancer patients.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Animales , Apoptosis/fisiología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/cirugía , Línea Celular Tumoral , Supervivencia Celular/fisiología , Ganciclovir/administración & dosificación , Ganciclovir/farmacocinética , Ingeniería Genética/métodos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/cirugía , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Simplexvirus/enzimología , Simplexvirus/genética , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética
14.
J Neurooncol ; 111(2): 153-61, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23242736

RESUMEN

The lack of relevant pre-clinical animal models incorporating the clinical scenario of Glioblastoma multiforme (GBM) resection and recurrence has contributed significantly to the inability to successfully treat GBM. A multi-modality imaging approach that allows real-time assessment of tumor resection during surgery and non-invasive detection of post-operative tumor volumes is urgently needed. In this study, we report the development and implementation of an optical imaging and magnetic resonance imaging (MRI) approach to guide GBM resection during surgery and track tumor recurrence at multiple resolutions in mice. Intra-operative fluorescence-guided surgery allowed real-time monitoring of intracranial tumor removal and led to greater than 90 % removal of established intracranial human GBM. The fluorescent signal clearly delineated tumor margins, residual tumor, and correlated closely with the clinically utilized fluorescence surgical marker 5-aminolevulinic acid/porphyrin. Post-operative non-invasive optical imaging and MRI confirmed near-complete tumor removal, which was further validated by immunohistochemistry (IHC). Longitudinal non-invasive imaging and IHC showed rapid recurrence of multi-focal tumors that exhibited a faster growth rate and altered blood-vessel density compared to non-resected tumors. Surgical tumor resection significantly extended long-term survival, however mice ultimately succumbed to the recurrent GBM. This multi-modality imaging approach to GBM resection and recurrence in mice should provide an important platform for investigating multiple aspects of GBM and ultimately evaluating novel therapeutics.


Asunto(s)
Neoplasias Encefálicas/cirugía , Glioblastoma/cirugía , Análisis de Varianza , Animales , Línea Celular Tumoral , Imagen de Difusión por Resonancia Magnética , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Humanos , Antígeno Ki-67/metabolismo , Mediciones Luminiscentes , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Ther ; 18(11): 1960-71, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20808288

RESUMEN

Oncolytic adenoviruses are promising anticancer agents due to their ability to self-amplify at the tumor mass. However, tumor stroma imposes barriers difficult to overcome by these agents. Transgene expression is a valuable strategy to counteract these limitations and to enhance antitumor activity. For this purpose, the genetic backbone in which the transgene is inserted should be optimized to render transgene expression compatible with the adenovirus replication cycle and to keep genome size within the encapsidation size limit. In order to design a potent and selective oncolytic adenovirus that keeps intact all the viral functions with minimal increase in genome size, we inserted palindromic E2F-binding sites into the endogenous E1A promoter. The insertion of these sites controlling E1A-Δ24 results in a low systemic toxicity profile in mice. Importantly, the E2F-binding sites also increased the cytotoxicity and the systemic antitumor activity relative to wild-type adenovirus in all cancer models tested. The low toxicity and the increased potency results in improved antitumor efficacy after systemic injection and increased survival of mice carrying tumors. Furthermore, the constrained genome size of this backbone allows an efficient and potent expression of transgenes, indicating that this virus holds promise for overcoming the limitations of oncolytic adenoviral therapy.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Terapia Genética , Neoplasias/terapia , Virus Oncolíticos/fisiología , Regiones Promotoras Genéticas/genética , Proteína de Retinoblastoma/fisiología , Proteínas E1A de Adenovirus/metabolismo , Animales , Western Blotting , Efecto Citopatogénico Viral , Factor de Transcripción E2F1/metabolismo , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/genética , Neoplasias/virología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transgenes/fisiología , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Mol Cancer Ther ; 8(11): 3098-107, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19887556

RESUMEN

Thymidine kinase (TK)-mediated suicide gene therapy has been considered for the treatment of pancreatic cancer. However, despite a bystander effect, the proportion of transduced tumor cells has proven too low to result in efficacy. We propose the use of a drug-selectable marker (MDR1) to enrich TK-expressing cells using chemotherapy. This enrichment or positive selection phase may increase the efficacy of suicide gene therapy. To test this strategy, we generated stable NP18MDR/TK-GFP transfectants and showed docetaxel resistance in vivo. Mixed tumors of MDR/TK-expressing cells and parental NP18 cells were established and docetaxel was used to increase the proportion of TK-expressing cells. After this positive selection phase, suicide gene therapy with ganciclovir was applied. Upon positive selection, the proportion of TK-expressing cells increased from 4% to 22%. Subsequent suicide gene therapy was more effective compared with a control group without positive selection. Starting with 10% of TK-expressing cells the positive-negative selection strategy completely inhibited tumor growth. Taken together, these results suggest that a positive-negative selection strategy based on MDR and TK genes represents an efficient way to increase the proportion of TK-expressing cells in the tumor and the efficacy of TK-mediated suicide gene therapy.


Asunto(s)
Ganciclovir/farmacología , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Taxoides/farmacología , Timidina Quinasa/genética , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/terapia , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Terapia Combinada , Docetaxel , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias Pancreáticas/tratamiento farmacológico , Timidina Quinasa/biosíntesis , Timidina Quinasa/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Hum Gene Ther ; 20(7): 728-38, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19281300

RESUMEN

A major obstacle for the efficacy of cancer gene therapy is the need to transduce a high proportion of tumor cells with genes that directly or indirectly cause their death. During the formation of certain organs, cells compete among themselves to colonize the whole tissue. We reasoned that cell competition could be used to increase the proportion of cells that become transfected in a tumor. For this, a transgene that provides a selective advantage to the transfected cells should be used. If the same gene conferred a suicide mechanism the tumor could be eradicated after a period of selection. Bystander effect of transfected cells over neighboring nonmodified cells may eliminate tumors even with incomplete replacement of tumor cells. To test this strategy a competitive advantage was provided to colon cancer cells, using a gene encoding a fusion protein of dihydrofolate reductase (DHFR) and thymidine kinase (TK). DHFR confers resistance to methotrexate (MTX) and TK confers sensitivity to ganciclovir (GCV). Modified cells were also transduced with green fluorescent protein and parental cells with red fluorescent protein. In vitro and in vivo experiments were performed, using various proportions of modified cells and applying positive selection with MTX followed by negative selection with GCV. In vitro, cell competition was evident. Under MTX treatment, tumor cells transfected with the DHFR-TK fusion gene efficiently replaced the parental cells (from 0.1 to 90% in 35 days). After this positive selection period, negative selection with GCV eliminated the transfected cells. In vivo, positive selection was also achieved and resulted in a statistically significant therapeutic effect.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Antineoplásicos/farmacología , Efecto Espectador/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Ganciclovir/farmacología , Ganciclovir/uso terapéutico , Proteínas Fluorescentes Verdes/metabolismo , Células HCT116 , Humanos , Proteínas Luminiscentes/metabolismo , Metotrexato/farmacología , Metotrexato/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidina Quinasa/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Fluorescente Roja
18.
Cancer Res ; 68(21): 8928-37, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18974137

RESUMEN

Genetic bioselection of a mutagenized Ad5wt stock in human tumor xenografts led us to isolate AdT1, a mutant displaying a large-plaque phenotype in vitro and an enhanced systemic antitumor activity in vivo. AdT1 phenotype correlates with an increased progeny release without affecting total viral yield in different human tumors and cancer-associated fibroblasts. An approach combining hybrid Ad5/AdT1 recombinants and sequencing identified a truncating insertion in the endoplasmic reticulum retention domain of the E3/19K protein (445A mutation) which relocates the protein to the plasma membrane and is responsible for AdT1's enhanced release. E3/19K-445A phenotype does not correlate with the protein's ability to interact with MHC-I or induce apoptosis. Intracellular calcium measurement revealed that the 445A mutation induces extracellular Ca(2+) influx, deregulating intracellular Ca(2+) homeostasis and inducing membrane permeabilization, a viroporin-like function. E3/19K-445A mutants also display enhanced antitumoral activity when injected both intratumorally and systemically in different models in vivo. Our results indicate that the inclusion of mutation 445A in tumor-selective adenoviruses would be a very powerful tool to enhance their antitumor efficacy.


Asunto(s)
Adenoviridae/fisiología , Mutación , Neoplasias/virología , Viroterapia Oncolítica , Adenoviridae/genética , Animales , Western Blotting , Línea Celular , Humanos , Ratones , Mutagénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...