Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Clin Dev Immunol ; 2012: 831090, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133490

RESUMEN

Malignant glioma, or glioblastoma, is the most common and lethal form of brain tumor with a median survival time of 15 months. The established therapeutic regimen includes a tripartite therapy of surgical resection followed by radiation and temozolomide (TMZ) chemotherapy, concurrently with radiation and then as an adjuvant. TMZ, a DNA alkylating agent, is the most successful antiglioma drug and has added several months to the life expectancy of malignant glioma patients. However, TMZ is also responsible for inducing lymphopenia and myelosuppression in malignant glioma patients undergoing chemotherapy. Although TMZ-induced lymphopenia has been attributed to facilitate antitumor vaccination studies by inducing passive immune response, in general lymphopenic conditions have been associated with poor immune surveillance leading to opportunistic infections in glioma patients, as well as disrupting active antiglioma immune response by depleting both T and NK cells. Deletion of O6-methylguanine-DNA-methyltransferase (MGMT) activity, a DNA repair enzyme, by temozolomide has been determined to be the cause of lymphopenia. Drug-resistant mutation of the MGMT protein has been shown to render chemoprotection against TMZ. The immune modulating role of TMZ during glioma chemotherapy and possible mechanisms to establish a strong TMZ-resistant immune response have been discussed.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Glioma/inmunología , Animales , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/inmunología , Antineoplásicos Alquilantes/uso terapéutico , Dacarbazina/efectos adversos , Dacarbazina/inmunología , Dacarbazina/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Factores Inmunológicos/efectos adversos , Factores Inmunológicos/inmunología , Factores Inmunológicos/uso terapéutico , Temozolomida
2.
Adv Exp Med Biol ; 746: 53-76, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22639159

RESUMEN

A major contributing factor to glioma development and progression is its ability to evade the immune system. This chapter will explore the mechanisms utilized by glioma to mediate immunosuppression and immune evasion. These include intrinsic mechanisms linked to its location within the brain and interactions between glioma cells and immune cells. Lack of recruitment of naïve effector immune cells perhaps accounts for most of the immune suppression mediated by these tumor cells. This is enhanced by increased recruitment of microglia which resemble immature antigen presenting cells that are unable to support T-cell mediated immunity. Furthermore, secreted factors like TGF-ß, COX-2 and IL-10, altered costimulatory molecules and inhibition of STAT-3 all contribute to the recruitment and expansion of regulatory T cells, which further modulate the immunosuppressive environment of glioma. In light of these findings, multiple immunotherapeutic treatment modalities are currently being explored.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioma/inmunología , Escape del Tumor/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Ciclooxigenasa 2/inmunología , Ciclooxigenasa 2/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
3.
Mol Ther ; 18(10): 1846-56, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20588259

RESUMEN

Oncolytic adenoviral virotherapy is an attractive treatment modality for cancer. However, following intratumoral injections, oncolytic viruses fail to efficiently migrate away from the injection site and are rapidly cleared by the immune system. We have previously demonstrated enhanced viral delivery and replicative persistence in vivo using human bone marrow-derived mesenchymal stem cells (MSCs) as delivery vehicles. In this study, we evaluated the immune response to adenovirus (Ad)-loaded MSCs using the semipermissive cotton rat (CR) model. First, we isolated MSCs from CR bone marrow aspirates. Real-time quantitative PCR analysis revealed that CR MSCs supported the replication of Ads in vitro. Moreover, we observed similar levels of suppression of T-cell proliferation in response to mitogenic stimulation, by MSCs alone and virus-loaded MSCs. Additionally, we found that MSCs suppressed the production of interferon-γ (IFN-γ) by activated T cells. In our in vivo model, CR MSCs enhanced the dissemination and persistence of Ad, compared to virus injection alone. Collectively, our data suggest that the use of MSCs as a delivery strategy for oncolytic Ad potentially offers a myriad of benefits, including improved delivery, enhanced dissemination, and increased persistence of viruses via suppression of the antiviral immune response.


Asunto(s)
Adenoviridae/inmunología , Células Madre Mesenquimatosas/inmunología , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Proliferación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos/inmunología , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratas , Sigmodontinae
4.
Adv Exp Med Biol ; 614: 65-72, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18290315

RESUMEN

Authors contributed equally to this manuscript Natural adjuvants, such as bacterial lipopolysaccharide (LPS), activate antigen presenting cells via Toll-like receptors and, indirectly, increase the survival of antigen-activated T cells. The molecular mechanisms leading to increased survival remain poorly defined. Because T cell clonal expansion leads to high energy demands, we hypothesized that increased glucose uptake and/or utilization in adjuvant-activated T cells could be important molecular event(s) that would lead to adjuvant-associated T cell survival advantage. Using a fluorescent analog of 2-deoxyglucose, 2-NBDG, we measured glucose accumulation and rate of uptake in T cells from mice treated with antigen in the absence or presence of LPS. Although adjuvant activated T cells increased the accumulation of 2-NBDG, the rate of uptake was unchanged compared to cells activated with only antigen. Furthermore, glucose transport inhibitors, cytochalasin B or phloretin, decreased the accumulation of glucose in adjuvant-treated T cells, but this decrease did not impair adjuvant-associated survival advantages. Together, these data indicate that increased glucose uptake through glucose transporters is not required for increased survival of activated T cells.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Glucosa/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocalasina B/farmacología , Relación Dosis-Respuesta a Droga , Enterotoxinas/farmacología , Glucosamina/análogos & derivados , Glucosamina/metabolismo , Glucosa/análisis , Glucosa/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Mediciones Luminiscentes , Ratones , Ratones Endogámicos , Floretina/farmacología , Factores de Tiempo
5.
Cancer Lett ; 433: 131-139, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29959057

RESUMEN

Successful remission in hematological cancers by CAR-T cell immunotherapy has yet to be replicated in solid tumors like GBM. A significant impediment of CAR-T immunotherapy in solid tumors is poor exposure of T cells to tumor antigens resulting in suboptimal CAR-T cell activation, which ultimately fails to induce a robust anti-tumor immune response. Costimulatory moieties in advanced-generation CARs, along with additional IL2 therapy has been shown to be insufficient to overcome this hurdle and have its cytotoxic limitations. GSK3 is constitutively active in naïve T cells and is transiently inactivated during T cell activation resulting in rapid T cell proliferation. Pharmacologic inhibition of GSK3 in GBM-specific CAR-T cells reduced FasL expression, increased T cell proliferation and reduced exhaustion by lowering PD-1 levels resulting in the development of CAR-T effector memory phenotype. Treatment with GSK3-inhibited CAR-T cells resulted in 100% tumor elimination during the tumor-rechallenge experiment in GBM-bearing animals and increased accumulation of memory CAR-T cells in secondary lymphoid organs. These adjuvant-like effects of GSK3 inhibition on activated CAR-T cells may be a valuable adjunct to a successful implementation of CAR-T immunotherapy against GBM and other solid tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Indoles/farmacología , Maleimidas/farmacología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/trasplante , Animales , Neoplasias Encefálicas/inmunología , Línea Celular Tumoral , Terapia Combinada , Glioblastoma/inmunología , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos , Masculino , Ratones , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Clin Cancer Res ; 24(4): 821-833, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29146721

RESUMEN

Purpose: We sought a novel approach against glioblastomas (GBM) focused on targeting signaling molecules localized in the tumor extracellular matrix (ECM). We investigated fibulin-3, a glycoprotein that forms the ECM scaffold of GBMs and promotes tumor progression by driving Notch and NFκB signaling.Experimental Design: We used deletion constructs to identify a key signaling motif of fibulin-3. An mAb (mAb428.2) was generated against this epitope and extensively validated for specific detection of human fibulin-3. mAb428.2 was tested in cultures to measure its inhibitory effect on fibulin-3 signaling. Nude mice carrying subcutaneous and intracranial GBM xenografts were treated with the maximum achievable dose of mAb428.2 to measure target engagement and antitumor efficacy.Results: We identified a critical 23-amino acid sequence of fibulin-3 that activates its signaling mechanisms. mAb428.2 binds to that epitope with nanomolar affinity and blocks the ability of fibulin-3 to activate ADAM17, Notch, and NFκB signaling in GBM cells. mAb428.2 treatment of subcutaneous GBM xenografts inhibited fibulin-3, increased tumor cell apoptosis, and enhanced the infiltration of inflammatory macrophages. The antibody reduced tumor growth and extended survival of mice carrying GBMs as well as other fibulin-3-expressing tumors. Locally infused mAb428.2 showed efficacy against intracranial GBMs, increasing tumor apoptosis and reducing tumor invasion and vascularization, which are enhanced by fibulin-3.Conclusions: To our knowledge, this is the first rationally developed, function-blocking antibody against an ECM target in GBM. Our results offer a proof of principle for using "anti-ECM" strategies toward more efficient targeted therapies for malignant glioma. Clin Cancer Res; 24(4); 821-33. ©2017 AACR.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/inmunología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/inmunología , Glioblastoma/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Ratones Desnudos , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos
7.
R I Med J (2013) ; 100(6): 39-42, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28564668

RESUMEN

Glioblastoma multiforme (GBM) is the most malignant of the primary central nervous system (CNS) neoplasms, accounting for nearly 80% of all primary brain tumors and is associated with high morbidity and mortality. Immunotherapy is proving to be a fertile ground for next-generation GBM therapy, with large translational research projects and clinical trials currently underway. One particularly promising area is the chimeric antigen receptors (CARs) in the context of lymphocyte adoptive cell therapy (ACT), which has achieved success in the treatment of hematological malignancies. In this review, we will discuss CARs and review current challenges facing their use in GBM therapy. [Full article available at http://rimed.org/rimedicaljournal-2017-06.asp].


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunoterapia , Proteínas de Fusión Oncogénica , Receptores de Antígenos , Humanos
8.
Immunobiology ; 210(9): 647-59, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16325488

RESUMEN

Immunological adjuvants help increase the number of T cells responding to an immunizing antigen. Part of the increase is due to promotion of survival of clonally expanded T cells in the face of waning antigen load and subsequent growth-factor withdrawal. The phosphatidylinositide-3 kinase (PI3-kinase)/Akt pathway is activated upon T cell stimulation and plays a critical role in clonal expansion by mediating several aspects of co-stimulation in a growth-factor-dependent manner. We hypothesized that adjuvants must either cause the PI3-kinase/Akt pathway to operate in the absence of growth-factor or to render T cells independent of continuous PI3-kinase signaling for their survival. To determine which is true, mice were treated with model antigen in the presence or absence of the natural adjuvant lipopolysaccharide (LPS). T cells from treated mice were assayed for their dependence on PI3-kinase signaling by measuring (i) levels of phosphorylated Akt, (ii) survival after culture in the presence of the PI3-kinase inhibitor LY294002, and (iii) the amount of glucose uptake upon ex vivo culture. The results show that although LPS treatment increased the induced PI3-kinase activity, the presence of PI3-kinase inhibitor did not affect glucose uptake or survival of T cells, an attribute the cells acquired within 4 h of LPS injection. Therefore, adjuvant-dependent survival effects do not require continuous PI3-kinase activity to occur, a finding that may explain how activated T cells survive antigen-withdrawal long enough to traffic from priming lymph nodes to sites of infection.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Glucosa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Animales , Transporte Biológico/efectos de los fármacos , Hipoxia de la Célula , Supervivencia Celular , Células Cultivadas , Medio de Cultivo Libre de Suero , Femenino , Lipopolisacáridos/farmacología , Ratones , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/enzimología , Linfocitos T/metabolismo , Factores de Tiempo
9.
Biomed Res Int ; 2014: 952128, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25247196

RESUMEN

Glioblastoma (GBM) is the most lethal primary brain tumor, and despite several refinements in its multimodal management, generally has very poor prognosis. Targeted immunotherapy is an emerging field of research that shows great promise in the treatment of GBM. One of the most extensively studied targets is the interleukin-13 receptor alpha chain variant 2 (IL13Rα2). Its selective expression on GBM, discovered almost two decades ago, has been a target for therapy ever since. Immunotherapeutic strategies have been developed targeting IL13Rα2, including monoclonal antibodies as well as cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-expressing T cells. Advanced therapeutic development has led to the completion of several clinical trials with promising outcomes. In this review, we will discuss the recent advances in the IL13Rα2-targeted immunotherapy and evaluate the most promising strategy for targeted GBM immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Glioblastoma/tratamiento farmacológico , Glioblastoma/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Animales , Medicina Basada en la Evidencia , Humanos , Inmunoterapia/métodos , Subunidad alfa2 del Receptor de Interleucina-13/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , Resultado del Tratamiento
10.
Neuro Oncol ; 16(10): 1304-12, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24723564

RESUMEN

Glioblastoma multiforme (GBM) remains one of the most lethal primary brain tumors despite surgical and therapeutic advancements. Targeted therapies of neoplastic diseases, including GBM, have received a great deal of interest in recent years. A highly studied target of GBM is interleukin-13 receptor α chain variant 2 (IL13Rα2). Targeted therapies against IL13Rα2 in GBM include fusion chimera proteins of IL-13 and bacterial toxins, nanoparticles, and oncolytic viruses. In addition, immunotherapies have been developed using monoclonal antibodies and cell-based strategies such as IL13Rα2-pulsed dendritic cells and IL13Rα2-targeted chimeric antigen receptor-modified T cells. Advanced therapeutic development has led to the completion of phase I clinical trials for chimeric antigen receptor-modified T cells and phase III clinical trials for IL-13-conjugated bacterial toxin, with promising outcomes. Selective expression of IL13Rα2 on tumor cells, while absent in the surrounding normal brain tissue, has motivated continued study of IL13Rα2 as an important candidate for targeted glioma therapy. Here, we review the preclinical and clinical studies targeting IL13Rα2 in GBM and discuss new advances and promising applications.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Subunidad alfa2 del Receptor de Interleucina-13/uso terapéutico , Animales , Neoplasias Encefálicas/inmunología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Exotoxinas/uso terapéutico , Glioblastoma/inmunología , Humanos , Interleucina-13/uso terapéutico , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Liposomas/uso terapéutico , Ratones , Terapia Molecular Dirigida/métodos , Proteínas Recombinantes de Fusión , Resultado del Tratamiento
11.
Clin Cancer Res ; 18(21): 5949-60, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22966020

RESUMEN

PURPOSE: Glioblastoma multiforme (GBM) remains highly incurable, with frequent recurrences after standard therapies of maximal surgical resection, radiation, and chemotherapy. To address the need for new treatments, we have undertaken a chimeric antigen receptor (CAR) "designer T cell" (dTc) immunotherapeutic strategy by exploiting interleukin (IL)13 receptor α-2 (IL13Rα2) as a GBM-selective target. EXPERIMENTAL DESIGN: We tested a second-generation IL13 "zetakine" CAR composed of a mutated IL13 extracellular domain linked to intracellular signaling elements of the CD28 costimulatory molecule and CD3ζ. The aim of the mutation (IL13.E13K.R109K) was to enhance selectivity of the CAR for recognition and killing of IL13Rα2(+) GBMs while sparing normal cells bearing the composite IL13Rα1/IL4Rα receptor. RESULTS: Our aim was partially realized with improved recognition of tumor and reduced but persisting activity against normal tissue IL13Rα1(+) cells by the IL13.E13K.R109K CAR. We show that these IL13 dTcs were efficient in killing IL13Rα2(+) glioma cell targets with abundant secretion of cytokines IL2 and IFNγ, and they displayed enhanced tumor-induced expansion versus control unmodified T cells in vitro. In an in vivo test with a human glioma xenograft model, single intracranial injections of IL13 dTc into tumor sites resulted in marked increases in animal survivals. CONCLUSIONS: These data raise the possibility of immune targeting of diffusely invasive GBM cells either via dTc infusion into resection cavities to prevent GBM recurrence or via direct stereotactic injection of dTcs to suppress inoperable or recurrent tumors. Systemic administration of these IL13 dTc could be complicated by reaction against normal tissues expressing IL13Ra1.


Asunto(s)
Glioblastoma/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD28/genética , Antígenos CD28/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Orden Génico , Glioblastoma/mortalidad , Glioblastoma/terapia , Humanos , Inmunoterapia Adoptiva/métodos , Interleucina-13/genética , Interleucina-13/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Mutación , Multimerización de Proteína , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Neuro Oncol ; 13(12): 1308-23, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21908444

RESUMEN

Glioblastoma multiforme (GBM) is a highly malignant brain tumor with an average survival time of 15 months. Previously, we and others demonstrated that CD4(+)FoxP3(+) regulatory T cells (Tregs) infiltrate human GBM as well as mouse models that recapitulate malignant brain tumors. However, whether brain tumor-resident Tregs are thymus-derived natural Tregs (nTregs) or induced Tregs (iTregs), by the conversion of conventional CD4(+) T cells, has not been established. To investigate this question, we utilized the i.c. implanted GL261 cell-based orthotopic mouse model, the RasB8 transgenic astrocytoma mouse model, and a human GBM tissue microarray. We demonstrate that Tregs in brain tumors are predominantly thymus derived, since thymectomy, prior to i.c. GL261 cell implantation, significantly decreased the level of Tregs in mice with brain tumors. Accordingly, most Tregs in human GBM and mouse brain tumors expressed the nTreg transcription factor, Helios. Interestingly, a significant effect of the brain tumor microenvironment on Treg lineage programming was observed, based on higher levels of brain tumor-resident Tregs expressing glucocorticoid-induced tumor necrosis factor receptor and CD103 and lower levels of Tregs expressing CD62L and CD45RB compared with peripheral Tregs. Furthermore, there was a higher level of nTregs in brain tumors that expressed the proliferative marker Ki67 compared with iTregs and conventional CD4(+) T cells. Our study demonstrates that future Treg-depleting therapies should aim to selectively target systemic rather than intratumoral nTregs in brain tumor-specific immunotherapeutic strategies.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Reguladores/inmunología , Timo/inmunología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Tasa de Supervivencia , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Timectomía , Timo/citología , Timo/metabolismo
13.
PLoS One ; 6(3): e18091, 2011 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-21464908

RESUMEN

BACKGROUND: Adenoviruses are often used as vehicles to mediate gene delivery for therapeutic purposes, but their research scope in hematological cells remains limited due to a narrow choice of host cells that express the adenoviral receptor (CAR). T cells, which are attractive targets for gene therapy of numerous diseases, remain resistant to adenoviral infection because of the absence of CAR expression. Here, we demonstrate that this resistance can be overcome when murine or human T cells are transduced with an adenovirus incorporating the RGD-fiber modification (Ad-RGD). METHODOLOGY/PRINCIPAL FINDING: A luciferase-expressing replication-deficient Ad-RGD infected 3-fold higher number of activated primary T cells than an adenovirus lacking the RGD-fiber modification in vitro. Infection with replication-competent Ad-RGD virus also caused increased cell cycling, higher E1A copy number and enriched hexon antigen expression in both human and murine T cells. Transduction with oncolytic Ad-RGD also resulted in higher titers of progeny virus and enhanced the killing of T cells. In vivo, 35-45% of splenic T cells were transduced by Ad-RGD. CONCLUSIONS: Collectively, our results prove that a fiber modified Ad-RGD successfully transduces and replicates in primary T cells of both murine and human origin.


Asunto(s)
Adenoviridae/fisiología , Oligopéptidos/metabolismo , Linfocitos T/virología , Transducción Genética , Replicación Viral , Proteínas E1A de Adenovirus/genética , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/virología , Ciclo Celular , Supervivencia Celular , Células Cultivadas , Dosificación de Gen , Humanos , Cadenas beta de Integrinas/metabolismo , Integrina beta3/metabolismo , Ratones , Ratones Endogámicos C57BL , Virus Oncolíticos/fisiología , Linfocitos T/citología , Volumetría
14.
Neurosurg Clin N Am ; 21(1): 201-14, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19944979

RESUMEN

Glioblastoma multiforme (GBM) is the most common and lethal primary malignant brain tumor. The traditional treatments for GBM, including surgery, radiation, and chemotherapy, only modestly improve patient survival. Therefore, immunotherapy has emerged as a novel therapeutic modality. Immunotherapeutic strategies exploit the immune system's ability to recognize and mount a specific response against tumor cells, but not normal cells. Current immunotherapeutic approaches for glioma can be divided into 3 categories: immune priming (active immunotherapy), immunomodulation (passive immunotherapy), and adoptive immunotherapy. Immune priming sensitizes the patient's immune cells to tumor antigens using various vaccination protocols. In the case of immunomodulation, strategies are aimed at reducing suppressive cytokines in the tumor microenvironment or using immune molecules to specifically target tumor cells. Adoptive immunotherapy involves harvesting the patient's immune cells, followed by ex vivo activation and expansion before reinfusion. This article provides an overview of the interactions between the central nervous system and the immune system, and discusses the challenges facing current immunotherapeutic strategies.


Asunto(s)
Neoplasias Encefálicas/terapia , Ensayos Clínicos como Asunto , Glioma/terapia , Inmunoterapia , Proyectos de Investigación , Glioblastoma/terapia , Humanos , Inmunización Pasiva , Inmunoterapia/métodos , Inmunoterapia Activa , Inmunoterapia Adoptiva
15.
PLoS One ; 5(10): e15390, 2010 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-21060663

RESUMEN

BACKGROUND: Recently, CD4(+)IL-17A(+) T helper 17 (Th17) cells were identified and reported in several diseased states, including autoimmunity, infection and various peripheral nervous system tumors. However, the presence of Th17 in glia-derived tumors of the central nervous system has not been studied. METHODOLOGY/PRINCIPAL FINDINGS: In this report, we demonstrate that mRNA expression for the Th17 cell cytokine IL-17A, as well as Th17 cells, are present in human glioma. The mRNA expression for IL-17A in glioma was recapitulated in an immunocompetent mouse model of malignant glioma. Furthermore, the presence of Th17 cells was confirmed in both human and mouse glioma. Interestingly, some Th17 cells present in mouse glioma co-expressed the Th1 and Th2 lineage markers, IFN-γ and IL-4, respectively, but predominantly co-expressed the Treg lineage marker FoxP3. CONCLUSIONS: These data confirm the presence of Th17 cells in glia-derived CNS tumors and provide the rationale for further investigation into the role of Th17 cells in malignant glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Interleucina-17/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Secuencia de Bases , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Glioma/inmunología , Glioma/patología , Ratones , Reacción en Cadena de la Polimerasa
16.
Neoplasia ; 12(10): 837-47, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20927322

RESUMEN

Glioblastoma multiforme is the most common and lethal primary brain tumor. Glioma progression depends on the rapid proliferation of tumor cells accompanied by an acute immunosuppressive environment, facilitated mainly by tumor infiltration of regulatory T cells (Tregs). In this study, we characterize the role of fibronectin, a high-molecular weight extracellular matrix glycoprotein secreted by tumor cells, in controlling glioma progression and in mediating immunosuppression. Fibronectin binds to membrane-spanning integrin receptors and plays an important role in cell signaling, in defining cellular shape, in mobility, and in regulating the cell cycle. We found that inhibition of fibronectin expression in glioma cells, using short hairpin RNA-mediated silencing of gene expression, delayed cell proliferation in vitro. This delayed growth is explained, in part, by the observed reduced expression of integrin ß(1) fibronectin receptor, which was restored by the inhibition of proteosomal activity. In our analysis of the downstream signaling targets of integrin ß(1), we demonstrated reduced phosphorylation of Src kinase and STAT-3. We also observed reduced survivin expression that induced a three-fold increased accumulation of fibronectin-knockdown cells in the G(2)/M phase. In an experimental animal model, the fibronectin knockdown tumors had a mean survival advantage of 23 days over wild-type tumors. Moreover, brain samples of animals bearing fibronectin-knockdown tumors showed delayed Treg recruitment. Collectively, we propose that fibronectin is a key mediator of glioma progression because its inhibition delays both tumor progression and immunosuppression.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/terapia , Modelos Animales de Enfermedad , Fibronectinas/genética , Fibronectinas/metabolismo , Glioma/terapia , ARN Interferente Pequeño/genética , Animales , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , División Celular/fisiología , Proliferación Celular , Inhibidores de Cisteína Proteinasa/farmacología , Citometría de Flujo , Fase G2/fisiología , Glioma/genética , Glioma/metabolismo , Humanos , Técnicas para Inmunoenzimas , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Leupeptinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Survivin , Linfocitos T Reguladores , Células Tumorales Cultivadas , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
17.
PLoS One ; 5(3): e9750, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20305783

RESUMEN

BACKGROUND: Glioblastoma multiforme is the most lethal brain tumor with limited therapeutic options. Antigens expressed on the surface of malignant cells are potential targets for antibody-mediated gene/drug delivery. PRINCIPAL FINDINGS: In this study, we investigated the ability of genetically modified human mesenchymal stem cells (hMSCs) expressing a single-chain antibody (scFv) on their surface against a tumor specific antigen, EGFRvIII, to enhance the therapy of EGFRvIII expressing glioma cells in vivo. The growth of U87-EGFRvIII was specifically delayed in co-culture with hMSC-scFvEGFRvIII. A significant down-regulation was observed in the expression of pAkt in EGFRvIII expressing glioma cells upon culture with hMSC-scFvEGFRvIII vs. controls as well as in EGFRvIII expressing glioma cells from brain tumors co-injected with hMSC-scFvEGFRvIII in vivo. hMSC expressing scFvEGFRvIII also demonstrated several fold enhanced retention in EGFRvIII expressing flank and intracranial glioma xenografts vs. control hMSCs. The growth of U87-EGFRvIII flank xenografts was inhibited by 50% in the presence of hMSC-scFvEGFRvIII (p<0.05). Moreover, animals co-injected with U87-EGFRvIII and hMSC-scFvEGFRvIII intracranially showed significantly improved survival compared to animals injected with U87-EGFRvIII glioma cells alone or with control hMSCs. This survival was further improved when the same animals received an additional dosage of hMSC-scFvEGFRvIII two weeks after initial tumor implantation. Of note, EGFRvIII expressing brain tumors co-injected with hMSCs had a lower density of CD31 expressing blood vessels in comparison with control tumors, suggesting a possible role in tumor angiogenesis. CONCLUSIONS/SIGNIFICANCE: The results presented in this study illustrate that genetically modified MSCs may function as a novel therapeutic vehicle for malignant brain tumors.


Asunto(s)
Neoplasias Encefálicas/patología , Receptores ErbB/genética , Glioma/patología , Células Madre Mesenquimatosas/citología , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Citometría de Flujo/métodos , Glioma/metabolismo , Glioma/terapia , Humanos , Ratones , Trasplante de Neoplasias , Neovascularización Patológica , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis
18.
J Immunol ; 178(10): 6083-91, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17475833

RESUMEN

Activated T cell death (ATCD) after peak clonal expansion is required for effective homeostasis of the immune system. Using a mouse model of T cell clonal expansion and contraction, we found that regulation of the proapoptotic kinase glycogen synthase kinase (GSK)-3beta plays a decisive role in determining the extent to which T cells are eliminated after activation. Involvement of GSK-3beta in ATCD was tested by measuring T cell survival after GSK-3beta inhibition, either ex vivo with chemical and pharmacological inhibitors or in vivo by retroviral expression of a dominant-negative form of GSK-3. We also measured amounts of inactivating phosphorylation of GSK-3beta (Ser9) in T cells primed in the presence or absence of LPS. Our results show that GSK-3beta activity is required for ATCD and that its inhibition promoted T cell survival. Adjuvant treatment in vivo maintained GSK-3beta (Ser9) phosphorylation in activated T cells, whereas with adjuvant-free stimulation it peaked and then decayed as the cells became susceptible to ATCD. We conclude that the duration of GSK-3beta inactivation determines activated T cell survival and that natural adjuvant stimulation decreases the severity of clonal contraction in part by keeping a critical proapoptotic regulatory factor, GSK-3beta, inactivated.


Asunto(s)
Adyuvantes Inmunológicos/fisiología , Apoptosis/inmunología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Femenino , Glucógeno Sintasa Quinasa 3/fisiología , Glucógeno Sintasa Quinasa 3 beta , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA