Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 11(1): 4997, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-33020472

RESUMEN

Despite a deeper molecular understanding, human glioblastoma remains one of the most treatment refractory and fatal cancers. It is known that the presence of macrophages and microglia impact glioblastoma tumorigenesis and prevent durable response. Herein we identify the dual function cytokine IL-33 as an orchestrator of the glioblastoma microenvironment that contributes to tumorigenesis. We find that IL-33 expression in a large subset of human glioma specimens and murine models correlates with increased tumor-associated macrophages/monocytes/microglia. In addition, nuclear and secreted functions of IL-33 regulate chemokines that collectively recruit and activate circulating and resident innate immune cells creating a pro-tumorigenic environment. Conversely, loss of nuclear IL-33 cripples recruitment, dramatically suppresses glioma growth, and increases survival. Our data supports the paradigm that recruitment and activation of immune cells, when instructed appropriately, offer a therapeutic strategy that switches the focus from the cancer cell alone to one that includes the normal host environment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioma/metabolismo , Glioma/patología , Interleucina-33/metabolismo , Animales , Neoplasias Encefálicas/mortalidad , Carcinogénesis , Núcleo Celular/metabolismo , Citocinas/metabolismo , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/patología , Glioma/mortalidad , Humanos , Inflamación , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones SCID , Microglía , Análisis de Supervivencia , Linfocitos T/metabolismo , Linfocitos T/patología , Microambiente Tumoral/inmunología
2.
Proc Natl Acad Sci U S A ; 116(38): 19098-19108, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31471491

RESUMEN

Glioblastoma multiforme (GBM) is the most deadly brain tumor, and currently lacks effective treatment options. Brain tumor-initiating cells (BTICs) and orthotopic xenografts are widely used in investigating GBM biology and new therapies for this aggressive disease. However, the genomic characteristics and molecular resemblance of these models to GBM tumors remain undetermined. We used massively parallel sequencing technology to decode the genomes and transcriptomes of BTICs and xenografts and their matched tumors in order to delineate the potential impacts of the distinct growth environments. Using data generated from whole-genome sequencing of 201 samples and RNA sequencing of 118 samples, we show that BTICs and xenografts resemble their parental tumor at the genomic level but differ at the mRNA expression and epigenomic levels, likely due to the different growth environment for each sample type. These findings suggest that a comprehensive genomic understanding of in vitro and in vivo GBM model systems is crucial for interpreting data from drug screens, and can help control for biases introduced by cell-culture conditions and the microenvironment in mouse models. We also found that lack of MGMT expression in pretreated GBM is linked to hypermutation, which in turn contributes to increased genomic heterogeneity and requires new strategies for GBM treatment.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Genómica/métodos , Glioblastoma/patología , Células Madre Neoplásicas/patología , Microambiente Tumoral/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Neoplasias Encefálicas/genética , Estudios de Casos y Controles , Proliferación Celular , Metilación de ADN , Resistencia a Antineoplásicos , Femenino , Perfilación de la Expresión Génica , Glioblastoma/genética , Humanos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Transcriptoma , Células Tumorales Cultivadas , Secuenciación Completa del Genoma , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Res ; 78(17): 5023-5037, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29945963

RESUMEN

Glioblastoma (GBM) carries a dismal prognosis and inevitably relapses despite aggressive therapy. Many members of the Eph receptor tyrosine kinase (EphR) family are expressed by GBM stem cells (GSC), which have been implicated in resistance to GBM therapy. In this study, we identify several EphRs that mark a therapeutically targetable GSC population in treatment-refractory, recurrent GBM (rGBM). Using a highly specific EphR antibody panel and CyTOF (cytometry by time-of-flight), we characterized the expression of all 14 EphR in primary and recurrent patient-derived GSCs to identify putative rGBM-specific EphR. EPHA2 and EPHA3 coexpression marked a highly tumorigenic cell population in rGBM that was enriched in GSC marker expression. Knockdown of EPHA2 and EPHA3 together led to increased expression of differentiation marker GFAP and blocked clonogenic and tumorigenic potential, promoting significantly higher survival in vivo Treatment of rGBM with a bispecific antibody against EPHA2/A3 reduced clonogenicity in vitro and tumorigenic potential of xenografted recurrent GBM in vivo via downregulation of AKT and ERK and increased cellular differentiation. In conclusion, we show that EPHA2 and EPHA3 together mark a GSC population in rGBM and that strategic cotargeting of EPHA2 and EPHA3 presents a novel and rational therapeutic approach for rGBM.Significance: Treatment of rGBM with a novel bispecific antibody against EPHA2 and EPHA3 reduces tumor burden, paving the way for the development of therapeutic approaches against biologically relevant targets in rGBM. Cancer Res; 78(17); 5023-37. ©2018 AACR.


Asunto(s)
Efrina-A2/genética , Glioblastoma/genética , Recurrencia Local de Neoplasia/genética , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Efrina-A2/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/radioterapia , Humanos , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/radioterapia , Células Madre Neoplásicas/patología , Pronóstico , Radiación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor EphA3 , Receptores de la Familia Eph/antagonistas & inhibidores , Receptores de la Familia Eph/genética , Temozolomida/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Cell Biol ; 216(11): 3655-3675, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28877995

RESUMEN

Axon degeneration is an early event and pathological in neurodegenerative conditions and nerve injuries. To discover agents that suppress neuronal death and axonal degeneration, we performed drug screens on primary rodent neurons and identified the pan-kinase inhibitor foretinib, which potently rescued sympathetic, sensory, and motor wt and SOD1 mutant neurons from trophic factor withdrawal-induced degeneration. By using primary sympathetic neurons grown in mass cultures and Campenot chambers, we show that foretinib protected neurons by suppressing both known degenerative pathways and a new pathway involving unliganded TrkA and transcriptional regulation of the proapoptotic BH3 family members BimEL, Harakiri,and Puma, culminating in preservation of mitochondria in the degenerative setting. Foretinib delayed chemotherapy-induced and Wallerian axonal degeneration in culture by preventing axotomy-induced local energy deficit and preserving mitochondria, and peripheral Wallerian degeneration in vivo. These findings identify a new axon degeneration pathway and a potentially clinically useful therapeutic drug.


Asunto(s)
Anilidas/farmacología , Lesiones por Aplastamiento/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Receptor trkA/antagonistas & inhibidores , Nervio Ciático/efectos de los fármacos , Neuropatía Ciática/tratamiento farmacológico , Degeneración Walleriana , Fibras Adrenérgicas/efectos de los fármacos , Fibras Adrenérgicas/enzimología , Fibras Adrenérgicas/patología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Axones/efectos de los fármacos , Axones/enzimología , Axones/patología , Células Cultivadas , Lesiones por Aplastamiento/enzimología , Lesiones por Aplastamiento/genética , Lesiones por Aplastamiento/patología , Citoprotección , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Genotipo , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/enzimología , Mitocondrias/patología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/enzimología , Neuronas Motoras/patología , Mutación , Neuronas/enzimología , Neuronas/patología , Fenotipo , Fosforilación , Ratas Sprague-Dawley , Receptor trkA/genética , Receptor trkA/metabolismo , Nervio Ciático/enzimología , Nervio Ciático/lesiones , Nervio Ciático/patología , Neuropatía Ciática/enzimología , Neuropatía Ciática/genética , Neuropatía Ciática/patología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/enzimología , Células Receptoras Sensoriales/patología , Transducción de Señal , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Factores de Tiempo , Transcripción Genética
5.
Oncotarget ; 7(37): 59360-59376, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27449082

RESUMEN

Glioblastoma (GBM) is the most lethal and aggressive adult brain tumor, requiring the development of efficacious therapeutics. Towards this goal, we screened five genetically distinct patient-derived brain-tumor initiating cell lines (BTIC) with a unique collection of small molecule epigenetic modulators from the Structural Genomics Consortium (SGC). We identified multiple hits that inhibited the growth of BTICs in vitro, and further evaluated the therapeutic potential of EZH2 and HDAC inhibitors due to the high relevance of these targets for GBM. We found that the novel SAM-competitive EZH2 inhibitor UNC1999 exhibited low micromolar cytotoxicity in vitro on a diverse collection of BTIC lines, synergized with dexamethasone (DEX) and suppressed tumor growth in vivo in combination with DEX. In addition, a unique brain-penetrant class I HDAC inhibitor exhibited cytotoxicity in vitro on a panel of BTIC lines and extended survival in combination with TMZ in an orthotopic BTIC model in vivo. Finally, a combination of EZH2 and HDAC inhibitors demonstrated synergy in vitro by augmenting apoptosis and increasing DNA damage. Our findings identify key epigenetic modulators in GBM that regulate BTIC growth and survival and highlight promising combination therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Ensayos de Selección de Medicamentos Antitumorales/métodos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Piridonas/uso terapéutico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Dexametasona/uso terapéutico , Sinergismo Farmacológico , Quimioterapia Combinada , Epigénesis Genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Ratones SCID , Terapia Molecular Dirigida , Piridonas/farmacología , Bibliotecas de Moléculas Pequeñas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Clin Cancer Res ; 22(15): 3860-75, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27006494

RESUMEN

PURPOSE: Glioblastoma is one of the most lethal cancers in humans, and with existing therapy, survival remains at 14.6 months. Current barriers to successful treatment include their infiltrative behavior, extensive tumor heterogeneity, and the presence of a stem-like population of cells, termed brain tumor-initiating cells (BTIC) that confer resistance to conventional therapies. EXPERIMENTAL DESIGN: To develop therapeutic strategies that target BTICs, we focused on a repurposing approach that explored already-marketed (clinically approved) drugs for therapeutic potential against patient-derived BTICs that encompass the genetic and phenotypic heterogeneity of glioblastoma observed clinically. RESULTS: Using a high-throughput in vitro drug screen, we found that montelukast, clioquinol, and disulfiram (DSF) were cytotoxic against a large panel of patient-derived BTICs. Of these compounds, disulfiram, an off-patent drug previously used to treat alcoholism, in the presence of a copper supplement, showed low nanomolar efficacy in BTICs including those resistant to temozolomide and the highly infiltrative quiescent stem-like population. Low dose DSF-Cu significantly augmented temozolomide activity in vitro, and importantly, prolonged in vivo survival in patient-derived BTIC models established from both newly diagnosed and recurrent tumors. Moreover, we found that in addition to acting as a potent proteasome inhibitor, DSF-Cu functionally impairs DNA repair pathways and enhances the effects of DNA alkylating agents and radiation. These observations suggest that DSF-Cu inhibits proteasome activity and augments the therapeutic effects of DNA-damaging agents (temozolomide and radiation). CONCLUSIONS: DSF-Cu should be considered as an adjuvant therapy for the treatment of patients with glioblastoma in both newly diagnosed and recurrent settings. Clin Cancer Res; 22(15); 3860-75. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Cobre/farmacología , Dacarbazina/análogos & derivados , Disulfiram/farmacología , Glioblastoma/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Reparación del ADN , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Perfilación de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Temozolomida , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
ACS Med Chem Lett ; 4(11): 1102-7, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24900612

RESUMEN

The STAT3 gene is abnormally active in glioblastoma (GBM) and is a critically important mediator of tumor growth and therapeutic resistance in GBM. Thus, for poorly treated brain cancers such as gliomas, astrocytomas, and glioblastomas, which harbor constitutively activated STAT3, a STAT3-targeting therapeutic will be of significant importance. Herein, we report a most potent, small molecule, nonphosphorylated STAT3 inhibitor, 31 (SH-4-54) that strongly binds to STAT3 protein (K D = 300 nM). Inhibitor 31 potently kills glioblastoma brain cancer stem cells (BTSCs) and effectively suppresses STAT3 phosphorylation and its downstream transcriptional targets at low nM concentrations. Moreover, in vivo, 31 exhibited blood-brain barrier permeability, potently controlled glioma tumor growth, and inhibited pSTAT3 in vivo. This work, for the first time, demonstrates the power of STAT3 inhibitors for the treatment of BTSCs and validates the therapeutic efficacy of a STAT3 inhibitor for GBM clinical application.

8.
Cancer Res ; 71(4): 1385-95, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21303981

RESUMEN

Neuroblastoma (NB) is an often fatal pediatric tumor of neural crest origin. We previously isolated NB tumor-initiating cells (NB TIC) from bone marrow metastases that resemble cancer stem cells and form metastatic NB in immunodeficient animals with as few as ten cells. To identify signaling pathways important for the survival and self-renewal of NB TICs and potential therapeutic targets, we screened a small molecule library of 143 protein kinase inhibitors, including 33 in clinical trials. Cytostatic or cytotoxic drugs were identified that targeted PI3K (phosphoinositide 3-kinase)/Akt, PKC (protein kinase C), Aurora, ErbB2, Trk, and Polo-like kinase 1 (PLK1). Treatment with PLK1 siRNA or low nanomolar concentrations of BI 2536 or BI 6727, PLK1 inhibitors in clinical trials for adult malignancies, were cytotoxic to TICs whereas only micromolar concentrations of the inhibitors were cytotoxic for normal pediatric neural stem cells. Furthermore, BI 2536 significantly inhibited TIC tumor growth in a therapeutic xenograft model, both as a single agent and in combination with irinotecan, an active agent for relapsed NB. Our findings identify candidate kinases that regulate TIC growth and survival and suggest that PLK1 inhibitors are an attractive candidate therapy for metastatic NB.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Proteínas de Ciclo Celular/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Neuroblastoma/prevención & control , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/análisis , Algoritmos , Animales , Neoplasias Encefálicas/patología , Células Cultivadas , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/patología , Neuroblastoma/patología , Inhibidores de Proteínas Quinasas/análisis , Pteridinas/farmacología , Pteridinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
9.
Blood ; 117(4): 1146-55, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21088134

RESUMEN

We have recently reported that CD8(+) T-cell memory maintenance after immunization with recombinant human adenovirus type 5 (rHuAd5) is dependent upon persistent transgene expression beyond the peak of the response. In this report, we have further investigated the location and nature of the cell populations responsible for this sustained response. The draining lymph nodes were found to be important for primary expansion but not for memory maintenance, suggesting that antigen presentation through a nonlymphoid source was required. Using bone marrow chimeric mice, we determined that antigen presentation by nonhematopoietic antigen-presenting cells (APCs) was sufficient for maintenance of CD8(+) T-cell numbers. However, antigen presentation by this mechanism alone yielded a memory population that displayed alterations in phenotype, cytokine production and protective capacity, indicating that antigen presentation through both hematopoietic and nonhematopoietic APCs ultimately defines the memory CD8(+) T-cell response produced by rHuAd5. These results shed new light on the immunobiology of rHuAd5 vectors and provide evidence for a mechanism of CD8(+) T-cell expansion and memory maintenance that relies upon both hematopoietic and nonhematopoietic APCs.


Asunto(s)
Adenovirus Humanos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunización , Memoria Inmunológica/fisiología , Vacunas Virales/inmunología , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Cultivadas , Femenino , Sistema Hematopoyético/inmunología , Humanos , Inmunización/métodos , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Viroterapia Oncolítica/métodos , Vacunas Sintéticas/inmunología
10.
EMBO Mol Med ; 2(9): 371-84, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20721990

RESUMEN

Neuroblastoma (NB) is the most deadly extra-cranial solid tumour in children necessitating an urgent need for effective and less toxic treatments. One reason for the lack of efficacious treatments may be the inability of existing drugs to target the tumour-initiating or cancer stem cell population responsible for sustaining tumour growth, metastases and relapse. Here, we describe a strategy to identify compounds that selectively target patient-derived cancer stem cell-like tumour-initiating cells (TICs) while sparing normal paediatric stem cells (skin-derived precursors, SKPs) and characterize two therapeutic candidates. DECA-14 and rapamycin were identified as NB TIC-selective agents. Both compounds induced TIC death at nanomolar concentrations in vitro, significantly reduced NB xenograft tumour weight in vivo, and dramatically decreased self-renewal or tumour-initiation capacity in treated tumours. These results demonstrate that differential drug sensitivities between TICs and normal paediatric stem cells can be exploited to identify novel, patient-specific and potentially less toxic therapies.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Decualinio/análogos & derivados , Células Madre Neoplásicas/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Sirolimus/uso terapéutico , Bibliotecas de Moléculas Pequeñas/química , Animales , Apoptosis , Decualinio/química , Decualinio/uso terapéutico , Transporte de Electrón , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Mitocondrias/genética , Mitocondrias/metabolismo , Células Madre Neoplásicas/metabolismo , Neuroblastoma/genética , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Clin Cancer Res ; 16(18): 4572-82, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20651058

RESUMEN

PURPOSE: Neuroblastoma (NB) is an aggressive tumor of the developing peripheral nervous system that remains difficult to cure in the advanced stages. The poor prognosis for high-risk NB patients is associated with common disease recurrences that fail to respond to available therapies. NB tumor-initiating cells (TICs), isolated from metastases and primary tumors, may escape treatment and contribute to tumor relapse. New therapies that target the TICs may therefore prevent or treat tumor recurrences. EXPERIMENTAL DESIGN: We undertook a system-level characterization of NB TICs to identify potential drug targets against recurrent NB. We used next-generation RNA sequencing and/or human exon arrays to profile the transcriptomes of 11 NB TIC lines from six NB patients, revealing genes that are highly expressed in the TICs compared with normal neural crest-like cells and unrelated cancer tissues. We used gel-free two-dimensional liquid chromatography coupled to shotgun tandem mass spectrometry to confirm the presence of proteins corresponding to the most abundant TIC-enriched transcripts, thereby providing validation to the gene expression result. RESULTS: Our study revealed that genes in the BRCA1 signaling pathway are frequently misexpressed in NB TICs and implicated Aurora B kinase as a potential drug target for NB therapy. Treatment with a selective AURKB inhibitor was cytotoxic to NB TICs but not to the normal neural crest-like cells. CONCLUSION: This work provides the first high-resolution system-level analysis of the transcriptomes of 11 primary human NB TICs and identifies a set of candidate NB TIC-enriched transcripts for further development as therapeutic targets.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Madre Neoplásicas/patología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Aurora Quinasa B , Aurora Quinasas , Cromatografía Liquida/métodos , Evaluación Preclínica de Medicamentos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Análisis por Micromatrices , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/metabolismo , Neuroblastoma/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Biología de Sistemas/métodos , Espectrometría de Masas en Tándem/métodos , Terapias en Investigación/métodos , Terapias en Investigación/tendencias , Estudios de Validación como Asunto
12.
J Virol ; 83(23): 12027-36, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19759135

RESUMEN

Previous studies determined that the CD8(+) T-cell response elicited by recombinant adenovirus exhibited a protracted contraction phase that was associated with long-term presentation of antigen. To gain further insight into this process, a doxycycline-regulated adenovirus was constructed to enable controlled extinction of transgene expression in vivo. We investigated the impact of premature termination of transgene expression at various time points (day 3 to day 60) following immunization. When transgene expression was terminated before the maximum response had been attained, overall expansion was attenuated, yielding a small memory population. When transgene expression was terminated between day 13 and day 30, the memory population was not sustained, demonstrating that the early memory population was antigen dependent. Extinction of transgene expression at day 60 had no obvious impact on memory maintenance, indicating that maintenance of the memory population may ultimately become independent of transgene expression. Premature termination of antigen expression had significant but modest effects on the phenotype and cytokine profile of the memory population. These results offer new insights into the mechanisms of memory CD8(+) T-cell maintenance following immunization with a recombinant adenovirus.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Expresión Génica , Memoria Inmunológica , Ovalbúmina/inmunología , Vacunas Virales/inmunología , Adenoviridae/genética , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Vacunas Virales/genética
13.
Cancer Res ; 69(9): 3979-85, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19383917

RESUMEN

Tumors that recur following surgical resection of melanoma are typically metastatic and associated with poor prognosis. Using the murine B16F10 melanoma and a robust antimelanoma vaccine, we evaluated immunization as a tool to improve tumor-free survival following surgery. We investigated the utility of vaccination in both neoadjuvant and adjuvant settings. Surprisingly, neoadjuvant vaccination was far superior and provided approximately 100% protection against tumor relapse. Neoadjuvant vaccination was associated with enhanced frequencies of tumor-specific T cells within the tumor and the tumor-draining lymph nodes following resection. We also observed increased infiltration of antigen-specific T cells into the area of surgery. This method should be amenable to any vaccine platform and can be readily extended to the clinic.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Melanoma Experimental/terapia , Recurrencia Local de Neoplasia/prevención & control , Adenoviridae/genética , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Femenino , Humanos , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/cirugía , Ratones , Ratones Endogámicos C57BL , Terapia Neoadyuvante , Recurrencia Local de Neoplasia/inmunología , Linfocitos T/inmunología
14.
Cancer Res ; 67(13): 6459-67, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616707

RESUMEN

Immunization of mice with human dopachrome tautomerase (hDCT) provides greater protection against melanoma than immunization with the murine homologue (mDCT). We mapped the CD8(+) and CD4(+) T-cell epitopes in both proteins to better understand the mechanisms of the enhanced protection. The dominant CD8(+) T-cell epitopes were fully conserved between both proteins, yet immunization with hDCT produced frequencies of CD8(+) T cells that were 5- to 10-fold higher than immunization with mDCT. This difference was not intrinsic to the two proteins because comparable frequencies of CD8(+) T cells were elicited by both antigens in DCT-deficient mice. Strikingly, only hDCT elicited a significant level of specific CD4(+) T cells in wild-type (WT) mice. The murine protein was not devoid of CD4(+) T-cell epitopes because immunization of DCT-deficient mice with mDCT resulted in robust CD4(+) T-cell immunity directed against two epitopes that were not identified in WT mice. These results suggested that the reduced immunogenicity of mDCT in WT mice may be a function of insufficient CD4(+) T-cell help. To address this possibility, the dominant CD4(+) T-cell epitope from hDCT was introduced into mDCT. Immunization with the mutated mDCT evoked CD8(+) T-cell frequencies and protective immunity comparable with hDCT. These results reveal a novel mechanism by which xenoantigens overcome tolerance. Our data also suggest that immunologic tolerance is more stringent for CD4(+) T cells than CD8(+) T cells, providing a mechanism of peripheral tolerance where autoreactive CD8(+) T cells fail to be activated due to a lack of autoreactive CD4(+) T cells specific for the same antigen.


Asunto(s)
Antígenos Heterófilos/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Inmunoterapia/métodos , Secuencia de Aminoácidos , Animales , Epítopos/química , Femenino , Tolerancia Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Linfocitos T/metabolismo
15.
Expert Rev Vaccines ; 6(3): 347-56, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17542750

RESUMEN

Recombinant adenovirus vaccines show great promise for generating protective immunity against infectious agents and tumors. Our studies have identified several interesting biological features of the adenovirus vector that influence the T-cell response. Notably, we have demonstrated that following immunization with adenovirus vaccines, the transgene antigen remains available to the system for a longer period than would be expected, resulting in a T-cell population with a sustained effector phenotype. The implications of these observations with regards to the utility of adenovirus vaccines are discussed.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos , Vacunas Sintéticas/inmunología , Animales , Presentación de Antígeno , Vacunas contra el Cáncer/inmunología , Humanos , Vacunas Virales/inmunología
16.
Mol Ther ; 15(5): 997-1006, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17375073

RESUMEN

We have investigated the role of CD4(+) T cells in the development of the CD8(+) T-cell response after immunization with recombinant adenovirus (rAd). In the absence of CD4(+) T cells, the "unhelped" CD8(+) T-cell population exhibited a reduction in primary expansion and long-term survival that appeared to be due to inadequate priming of naïve T cells. There were few functional or phenotypic differences between the helped and unhelped CD8(+) T-cell populations with the exception of O-glycosylated CD43, a marker of effector cells, which was augmented on the unhelped CD8(+) T-cell population. In some cases, the unhelped CD8(+) T-cell population exhibited reduced ability to control virus infection; however, this appeared to be a function of the reduced frequency of antigen-specific CD8(+) T cells. Most notably, the unhelped CD8(+) T-cell population exhibited no defect in secondary expansion. These results provide insight into the role of CD4(+) T cells during the primary CD8(+) T-cell response generated by rAd vaccines and identify potential benefits and issues that must be considered when using adenovirus vaccines under conditions where CD4(+) T-cell function may be limiting, such as vaccination of human immunodeficiency virus patients.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas Virales/inmunología , Adenoviridae/genética , Animales , Antígenos CD4/genética , Antígenos CD4/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Citometría de Flujo , Inmunización/métodos , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
17.
Cell Immunol ; 250(1-2): 55-67, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18313652

RESUMEN

Virus-based recombinant vaccines have proven highly effective at generating protective CD8+ T cell responses. Multiple vector platforms are available, however, little is known about the relative influence of the different vectors on the transgene-specific CD8+ T cell population. To address this question, we compared several characteristics of the CD8+ T cell response elicited by recombinant adenovirus (rAd) and vaccinia virus (rVV). We found that following rAd immunization the transgene-specific CD8+ T cell response peaked around day 12 and was larger and more sustained than the response produced by rVV. In addition, the CD8+ T cell response generated by rAd was directed primarily against the transgene, whereas the CD8+ T cell response produced by rVV principally targeted the vector backbone. In addition, we also observed that transgene selection also impacted on the magnitude of the CD8+ T cell response elicited by both vectors. Despite differences in the magnitude of the anti-transgene CD8+ T cell response, both vectors elicited CD8+ T cell populations with similar cytokine production, functional avidity and cytolytic activity. In addition, plasmid priming prior to immunization with either rAd or rVV only impacted the magnitude of the transgene gene specific CD8+ T cell response. Our study demonstrates that both vector and transgene selection can influence the magnitude of the CD8+ T cell response, but they do not influence functionality.


Asunto(s)
Adenoviridae/genética , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/genética , Proteínas Recombinantes/genética , Virus Vaccinia/inmunología , Animales , Antígenos/genética , Linfocitos T CD8-positivos/virología , Células Cultivadas , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/biosíntesis , Transgenes/genética , Virus Vaccinia/genética , Proteínas Virales/genética , Proteínas Virales/inmunología
18.
J Immunol ; 176(1): 200-10, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16365411

RESUMEN

We have previously reported that the CD8+ T cell response elicited by recombinant adenovirus vaccination displayed a delayed contraction in the spleen. In our current study, we demonstrate that this unusual kinetic is a general phenomenon observed in multiple tissues. Phenotypic analysis of transgene-specific CD8+ T cells present 30 days postimmunization with recombinant adenovirus revealed a population with evidence of partial exhaustion, suggesting that the cells had been chronically exposed to Ag. Although Ag expression could no longer be detected 3 wk after immunization, examination of Ag presentation within the draining lymph nodes demonstrated that APCs were loaded with Ag peptide for at least 40 days postimmunization, suggesting that Ag remains available to the system for a prolonged period, although the exact source of this Ag remains to be determined. At 60 days postimmunization, the CD8+ T cell population continued to exhibit a phenotype consistent with partially exhausted effector memory cells. Nonetheless, these CD8+ T cells conferred sterilizing immunity against virus challenge 7-12 wk postimmunization, suggesting that robust protective immunity can be provided by CD8+ T cells with an exhausted phenotype. These data demonstrate that prolonged exposure to Ag may not necessarily impair protective immunity and prompt a re-evaluation of the impact of persistent exposure to Ag on T cell function.


Asunto(s)
Adenoviridae/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/inmunología , Adenoviridae/genética , Traslado Adoptivo , Animales , Antígenos Virales/inmunología , Proteínas del Huevo/inmunología , Femenino , Citometría de Flujo , Vectores Genéticos/administración & dosificación , Inmunofenotipificación , Ganglios Linfáticos/inmunología , Ratones , Ovalbúmina/inmunología , Fragmentos de Péptidos , Factores de Tiempo , Transgenes
19.
Mol Ther ; 13(2): 270-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16297666

RESUMEN

We have examined the efficacy of vaccination with recombinant adenovirus under conditions of extreme leukopenia in lethally irradiated mice reconstituted with autologous bone marrow. The expansion of antigen-specific CD8(+) T cells following immunization of lethally irradiated hosts paralleled the recovery of total CD8(+) T cells. Surprisingly, the numbers of antigen-specific CD8(+) T cells in lethally irradiated mice beyond 6 weeks postimmunization were comparable to the numbers found in nonirradiated controls. CD8(+) T cells elicited in the lethally irradiated hosts were functionally indistinguishable from those elicited in normal hosts. Antigen expression and presentation persisted for a longer period of time in the draining lymph nodes of irradiated mice compared to those of nonirradiated animals, suggesting that antigen presentation mechanisms were intact during the reconstitution period. Experiments employing allogeneic bone marrow demonstrated that radioresistant host antigen-presenting cells were responsible for antigen presentation during the process of immune reconstitution. These results demonstrate clear compatibility of adenovirus vaccines and cytotoxic therapy. Furthermore, these observations provide novel insights into the mechanisms of CD8(+) T cell activation following adenovirus immunization.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Leucopenia/inmunología , Leucopenia/terapia , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/virología , Células Dendríticas/inmunología , Femenino , Leucopenia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Quimera por Radiación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Irradiación Corporal Total
20.
Hum Gene Ther ; 15(2): 179-88, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14975190

RESUMEN

Adenoviruses (Ads) have shown great utility as vectors for the delivery of genes to mammalian cells, partly because of their ability to infect a wide range of different cell types independent of the replicative state of the cell. However, Ads do not transduce mature muscle efficiently because of low levels of the natural viral primary receptor, the coxsackie virus and adenovirus receptor, on the surface of adult muscle cells. In this study, we have addressed whether incorporation of polylysine [p(K)] or arginine-glycine-aspartic acid (RGD) placed in the H-I loop of the adenoviral fiber protein can improve helper-dependent Ad vector (hdAd) transduction of mature muscle cells. We show that incorporation of the p(K) motif into the fiber of early region 1 (E1)-deleted Ad results in enhanced transduction of undifferentiated and differentiated C2C12 cells relative to a virus, containing a wild-type fiber (12- and 21-fold enhancement, respectively). Incorporation of the RGD motif resulted in only a 60-70% increase in transduction efficiency in these cells. The two fiber modifications were then incorporated into helper viruses for use in the Cre-lox system for generating hdAd, and the resulting retargeted Ad vectors, which encoded the beta-galactosidase reporter gene (beta-Gal), demonstrated enhanced transduction of C2C12 cells in culture. Although hdAdpK also showed enhanced infection of mature mouse muscle in vivo, hdAdRGD did not. All hdAd vectors elicited only minor anti-Ad immune responses, compared with an E1-deleted control vector, but each vector elicited strong anti-beta-Gal immunoreactivity. Our results demonstrate that hdAd with modified cell tropism can be generated efficiently and, in the case of polylysine-modified hdAd, can lead to improved transduction of adult muscle cells in vivo.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Virus Helper/genética , Músculo Esquelético/metabolismo , Transducción Genética/métodos , Adenoviridae/inmunología , Animales , Proteínas de la Cápside/genética , Vectores Genéticos/inmunología , Humanos , Ratones , Oligopéptidos/química , Polilisina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...