Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
N Engl J Med ; 375(26): 2561-9, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-28029927

RESUMEN

A patient with recurrent multifocal glioblastoma received chimeric antigen receptor (CAR)-engineered T cells targeting the tumor-associated antigen interleukin-13 receptor alpha 2 (IL13Rα2). Multiple infusions of CAR T cells were administered over 220 days through two intracranial delivery routes - infusions into the resected tumor cavity followed by infusions into the ventricular system. Intracranial infusions of IL13Rα2-targeted CAR T cells were not associated with any toxic effects of grade 3 or higher. After CAR T-cell treatment, regression of all intracranial and spinal tumors was observed, along with corresponding increases in levels of cytokines and immune cells in the cerebrospinal fluid. This clinical response continued for 7.5 months after the initiation of CAR T-cell therapy. (Funded by Gateway for Cancer Research and others; ClinicalTrials.gov number, NCT02208362 .).


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Glioblastoma/terapia , Inmunoterapia Adoptiva , Recurrencia Local de Neoplasia/terapia , Receptores de Antígenos de Linfocitos T/uso terapéutico , Ingeniería Celular , Terapia Combinada , Humanos , Subunidad alfa2 del Receptor de Interleucina-13 , Masculino , Persona de Mediana Edad
2.
Mol Ther ; 26(1): 31-44, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29103912

RESUMEN

T cell immunotherapy is emerging as a powerful strategy to treat cancer and may improve outcomes for patients with glioblastoma (GBM). We have developed a chimeric antigen receptor (CAR) T cell immunotherapy targeting IL-13 receptor α2 (IL13Rα2) for the treatment of GBM. Here, we describe the optimization of IL13Rα2-targeted CAR T cells, including the design of a 4-1BB (CD137) co-stimulatory CAR (IL13BBζ) and a manufacturing platform using enriched central memory T cells. Utilizing orthotopic human GBM models with patient-derived tumor sphere lines in NSG mice, we found that IL13BBζ-CAR T cells improved anti-tumor activity and T cell persistence as compared to first-generation IL13ζ-CAR CD8+ T cells that had shown evidence for bioactivity in patients. Investigating the impact of corticosteroids, given their frequent use in the clinical management of GBM, we demonstrate that low-dose dexamethasone does not diminish CAR T cell anti-tumor activity in vivo. Furthermore, we found that local intracranial delivery of CAR T cells elicits superior anti-tumor efficacy as compared to intravenous administration, with intraventricular infusions exhibiting possible benefit over intracranial tumor infusions in a multifocal disease model. Overall, these findings help define parameters for the clinical translation of CAR T cell therapy for the treatment of brain tumors.


Asunto(s)
Glioblastoma/inmunología , Glioblastoma/metabolismo , Inmunoterapia Adoptiva , Subunidad alfa2 del Receptor de Interleucina-13/antagonistas & inhibidores , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Anticuerpos Antineoplásicos/inmunología , Antígenos CD19/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Citotoxicidad Inmunológica , Dextroanfetamina/farmacología , Modelos Animales de Enfermedad , Orden Génico , Ingeniería Genética , Vectores Genéticos/genética , Glioblastoma/mortalidad , Glioblastoma/terapia , Humanos , Inmunoterapia Adoptiva/métodos , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Ratones , Receptores Quiméricos de Antígenos/química , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nat Med ; 30(4): 1001-1012, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38454126

RESUMEN

Chimeric antigen receptor T cell (CAR-T) therapy is an emerging strategy to improve treatment outcomes for recurrent high-grade glioma, a cancer that responds poorly to current therapies. Here we report a completed phase I trial evaluating IL-13Rα2-targeted CAR-T cells in 65 patients with recurrent high-grade glioma, the majority being recurrent glioblastoma (rGBM). Primary objectives were safety and feasibility, maximum tolerated dose/maximum feasible dose and a recommended phase 2 dose plan. Secondary objectives included overall survival, disease response, cytokine dynamics and tumor immune contexture biomarkers. This trial evolved to evaluate three routes of locoregional T cell administration (intratumoral (ICT), intraventricular (ICV) and dual ICT/ICV) and two manufacturing platforms, culminating in arm 5, which utilized dual ICT/ICV delivery and an optimized manufacturing process. Locoregional CAR-T cell administration was feasible and well tolerated, and as there were no dose-limiting toxicities across all arms, a maximum tolerated dose was not determined. Probable treatment-related grade 3+ toxicities were one grade 3 encephalopathy and one grade 3 ataxia. A clinical maximum feasible dose of 200 × 106 CAR-T cells per infusion cycle was achieved for arm 5; however, other arms either did not test or achieve this dose due to manufacturing feasibility. A recommended phase 2 dose will be refined in future studies based on data from this trial. Stable disease or better was achieved in 50% (29/58) of patients, with two partial responses, one complete response and a second complete response after additional CAR-T cycles off protocol. For rGBM, median overall survival for all patients was 7.7 months and for arm 5 was 10.2 months. Central nervous system increases in inflammatory cytokines, including IFNγ, CXCL9 and CXCL10, were associated with CAR-T cell administration and bioactivity. Pretreatment intratumoral CD3 T cell levels were positively associated with survival. These findings demonstrate that locoregional IL-13Rα2-targeted CAR-T therapy is safe with promising clinical activity in a subset of patients. ClinicalTrials.gov Identifier: NCT02208362 .


Asunto(s)
Glioblastoma , Glioma , Receptores Quiméricos de Antígenos , Humanos , Recurrencia Local de Neoplasia , Glioma/terapia , Linfocitos T , Glioblastoma/terapia , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos
4.
Neoplasia ; 30: 100801, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35550513

RESUMEN

High-grade (WHO grades III-IV) glioma remains one of the most lethal human cancers. Adoptive transfer of tumor-targeting chimeric antigen receptor (CAR)-redirected T cells for high-grade glioma has revealed promising indications of anti-tumor activity, but objective clinical responses remain elusive for most patients. A significant challenge to effective immunotherapy is the highly heterogeneous structure of these tumors, including large variations in the magnitudes and distributions of target antigen expression, observed both within individual tumors and between patients. To obtain a more detailed understanding of immunotherapy target antigens within patient tumors, we immunochemically mapped at single cell resolution three clinically-relevant targets, IL13Rα2, HER2 and EGFR, on tumor samples drawn from a 43-patient cohort. We observed that within individual tumor samples, expression of these antigens was neither random nor uniform, but rather that they mapped into local neighborhoods - phenotypically similar cells within regions of cellular tumor - reflecting not well understood properties of tumor cells and their milieu. Notably, tumor cell neighborhoods of high antigen expression were not arranged independently within regions. For example, in cellular tumor regions, neighborhoods of high IL13Rα2 and HER2 expression appeared to be reciprocal to those of EGFR, while in areas of pseudopalisading necrosis, expression of IL13Rα2 and HER2, but not EGFR, appeared to reflect the radial organization of tumor cells around hypoxic cores. Other structural features affecting expression of immunotherapy target antigens remain to be elucidated. This structured but heterogeneous organization of antigen expression in high grade glioma is highly permissive for antigen escape, and combinatorial antigen targeting is a commonly suggested potential mitigating strategy. Deeper understanding of antigen expression within and between patient tumors will enhance optimization of combination immunotherapies, the most immediate clinical application of the observations presented here being the importance of including (wild-type) EGFR as a target antigen.


Asunto(s)
Glioblastoma , Glioma , Subunidad alfa2 del Receptor de Interleucina-13 , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Glioma/tratamiento farmacológico , Glioma/terapia , Humanos , Inmunoterapia , Inmunoterapia Adoptiva , Subunidad alfa2 del Receptor de Interleucina-13/genética , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Stem Cells Int ; 2021: 5549381, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34122556

RESUMEN

As the success of stem cell-based therapies is contingent on efficient cell delivery to damaged areas, neural stem cells (NSCs) have promising therapeutic potential because they inherently migrate to sites of central nervous system (CNS) damage. To explore the possibility of NSC-based therapy after traumatic brain injury (TBI), isoflurane-anesthetized adult male rats received a controlled cortical impact (CCI) of moderate severity (2.8 mm deformation at 4 m/s) or sham injury (i.e., no cortical impact). Beginning 1-week post-injury, the rats were immunosuppressed and 1 × 106 human NSCs (LM-NS008.GFP.fLuc) or vehicle (VEH) (2% human serum albumen) were administered intranasally (IN) on post-operative days 7, 9, 11, 13, 15, and 17. To evaluate the spatial distributions of the LM-NSC008 cells, half of the rats were euthanized on day 25, one day after completion of the cognitive task, and the other half were euthanized on day 46. 1 mm thick brain sections were optically cleared (CLARITY), and volumes were imaged by confocal microscopy. In addition, LM-NSC008 cell migration to the TBI site by immunohistochemistry for human-specific Nestin was observed at day 39. Acquisition of spatial learning was assessed in a well-established Morris water maze task on six successive days beginning on post-injury day 18. IN administration of LM-NSC008 cells after TBI (TBI + NSC) significantly facilitated spatial learning relative to TBI + VEH rats (p < 0.05) and had no effect on sham + NSC rats. Overall, these data indicate that IN-administered LM-NSC008 cells migrate to sites of TBI damage and that their presence correlates with cognitive improvement. Future studies will expand on these preliminary findings by evaluating other LM-NSC008 cell dosing paradigms and evaluating mechanisms by which LM-NSC008 cells contribute to cognitive recovery.

6.
Sci Transl Med ; 12(533)2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32132216

RESUMEN

Although chimeric antigen receptor (CAR) T cells have demonstrated signs of antitumor activity against glioblastoma (GBM), tumor heterogeneity remains a critical challenge. To achieve broader and more effective GBM targeting, we developed a peptide-bearing CAR exploiting the GBM-binding potential of chlorotoxin (CLTX). We find that CLTX peptide binds a great proportion of tumors and constituent tumor cells. CAR T cells using CLTX as the targeting domain (CLTX-CAR T cells) mediate potent anti-GBM activity and efficiently target tumors lacking expression of other GBM-associated antigens. Treatment with CLTX-CAR T cells resulted in tumor regression in orthotopic xenograft GBM tumor models. CLTX-CAR T cells do not exhibit observable off-target effector activity against normal cells or after adoptive transfer into mice. Effective targeting by CLTX-CAR T cells requires cell surface expression of matrix metalloproteinase-2. Our results pioneer a peptide toxin in CAR design, expanding the repertoire of tumor-selective CAR T cells with the potential to reduce antigen escape.


Asunto(s)
Glioblastoma , Venenos de Escorpión , Animales , Línea Celular Tumoral , Glioblastoma/terapia , Inmunoterapia Adoptiva , Metaloproteinasa 2 de la Matriz , Ratones , Receptores de Antígenos de Linfocitos T , Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Stem Cells ; 26(6): 1575-86, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18339768

RESUMEN

The utility of neural stem cells (NSCs) has extended beyond regenerative medicine to targeted gene delivery, as NSCs possess an inherent tropism to solid tumors, including invasive gliomas. However, for optimal clinical implementation, an understanding of the molecular events that regulate NSC tumor tropism is needed to ensure their safety and to maximize therapeutic efficacy. We show that human NSC lines responded to multiple tumor-derived growth factors and that hepatocyte growth factor (HGF) induced the strongest chemotactic response. Gliomatropism was critically dependent on c-Met signaling, as short hairpin RNA-mediated ablation of c-Met significantly attenuated the response. Furthermore, inhibition of Ras-phosphoinositide 3-kinase (PI3K) signaling impaired the migration of human neural stem cells (hNSCs) toward HGF and other growth factors. Migration toward tumor cells is a highly regulated process, in which multiple growth factor signals converge on Ras-PI3K, causing direct modification of the cytoskeleton. The signaling pathways that regulate hNSC migration are similar to those that promote unregulated glioma invasion, suggesting shared cellular mechanisms and responses. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Neoplasias Encefálicas/enzimología , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Quimiotaxis , Glioma/enzimología , Humanos , Riñón , Neuronas/enzimología , Neuronas/fisiología , Células Madre/enzimología , Células Madre/fisiología , Proteína de Unión al GTP rac1/metabolismo
10.
PLoS One ; 13(8): e0199967, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30071048

RESUMEN

BACKGROUND: Preclinical studies indicate that neural stem cells (NSCs) can limit or reverse central nervous system (CNS) damage through delivery of therapeutic agents for cell regeneration. Clinical translation of cell-based therapies raises concerns about long-term stability, differentiation and fate, and absence of tumorigenicity of these cells, as well as manufacturing time required to produce therapeutic cells in quantities sufficient for clinical use. Allogeneic NSC lines are in growing demand due to challenges inherent in using autologous stem cells, including production costs that limit availability to patients. METHODS/PRINCIPAL FINDINGS: We demonstrate the long-term stability of L-MYC immortalized human NSCs (LM-NSC008) cells in vivo, including engraftment, migration, and absence of tumorigenicity in mouse brains for up to nine months. We also examined the distributions of engrafted LM-NSC008 cells within brain, and present computational techniques to analyze NSC migration characteristics in relation to intrinsic brain structures. CONCLUSIONS/SIGNIFICANCE: This computational analysis of NSC distributions following implantation provides proof-of-concept for the development of computational models that can be used clinically to predict NSC migration paths in patients. Previously, models of preferential migration of malignant tumor cells along white matter tracts have been used to predict their final distributions. We suggest that quantitative measures of tissue orientation and white matter tracts determined from MR images can be used in a diffusion tensor imaging tractography-like approach to describe the most likely migration routes and final distributions of NSCs administered in a clinical setting. Such a model could be very useful in choosing the optimal anatomical locations for NSC administration to patients to achieve maximum therapeutic effects.


Asunto(s)
Encéfalo/citología , Células-Madre Neurales/trasplante , Animales , Encéfalo/patología , Movimiento Celular , Células Cultivadas , ADN/aislamiento & purificación , ADN/metabolismo , Femenino , Humanos , Imagenología Tridimensional , Masculino , Ratones , Ratones Endogámicos NOD , Nestina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Sustancia Blanca/citología , Sustancia Blanca/patología
11.
J Clin Invest ; 113(4): 582-90, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14966567

RESUMEN

Cerebellar ataxia, a devastating neurological disease, may be initiated by hyperexcitability of deep cerebellar nuclei (DCN) secondary to loss of inhibitory input from Purkinje neurons that frequently degenerate in this disease. This mechanism predicts that intrinsic DCN hyperexcitability would cause ataxia in the absence of upstream Purkinje degeneration. We report the generation of a transgenic (Tg) model that supports this mechanism of disease initiation. Small-conductance calcium-activated potassium (SK) channels, regulators of firing frequency, were silenced in the CNS of Tg mice with the dominant-inhibitory construct SK3-1B-GFP. Transgene expression was restricted to the DCN within the cerebellum and was detectable beginning on postnatal day 10, concomitant with the onset of cerebellar ataxia. Neurodegeneration was not evident up to the sixth month of age. Recordings from Tg DCN neurons revealed loss of the apamin-sensitive after-hyperpolarization current (IAHP) and increased spontaneous firing through SK channel suppression, indicative of DCN hyperexcitability. Spike duration and other electrogenic conductance were unaffected. Thus, a purely electrical alteration is sufficient to cause cerebellar ataxia, and SK openers such as the neuroprotective agent riluzole may reduce neuronal hyperexcitability and have therapeutic value. This dominant-inhibitory strategy may help define the in vivo role of SK channels in other neuronal pathways.


Asunto(s)
Ataxia Cerebelosa/fisiopatología , Núcleos Cerebelosos/fisiopatología , Neuronas/patología , Neuronas/fisiología , Animales , Apamina/metabolismo , Ataxia Cerebelosa/patología , Núcleos Cerebelosos/citología , Núcleos Cerebelosos/patología , Electrofisiología , Técnicas In Vitro , Ratones , Ratones Transgénicos , Actividad Motora , Canales de Potasio/genética , Canales de Potasio/metabolismo , Regiones Promotoras Genéticas , Células de Purkinje/citología , Células de Purkinje/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transgenes
12.
Stem Cells Transl Med ; 6(6): 1522-1532, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28481046

RESUMEN

Engineered neural stem cells (NSCs) intrinsically migrating to brain tumors offer a promising mechanism for local therapeutic delivery. However, difficulties in quantitative assessments of NSC migration and in estimates of tumor coverage by diffusible therapeutics have impeded development and refinement of NSC-based therapies. To address this need, we developed techniques by which conventional serial-sectioned formalin-fixed paraffin-embedded (FFPE) brains can be analyzed in their entirety across multiple test animals. We considered a conventional human glioblastoma model: U251 glioma cells orthotopically engrafted in immunodeficient mice receiving intracerebral (i.c.) or intravenous (i.v.) administrations of NSCs expressing a diffusible enzyme to locally catalyze chemotherapeutic formation. NSC migration to tumor sites was dose-dependent, reaching 50%-60% of total administered NSCs for the i.c route and 1.5% for the i.v. route. Curiously, the most efficient NSC homing was seen with smaller NSC doses, implying existence of rate-limiting process active during administration and/or migration. Predicted tumor exposure to a diffusing therapeutic (assuming a 50 µm radius of action) could reach greater than 50% of the entire tumor volume for i.c. and 25% for i.v. administration. Within individual sections, coverage of tumor area could be as high as 100% for i.c. and 70% for i.v. routes. Greater estimated therapeutic coverage was observed for larger tumors and for larger tumor regions in individual sections. Overall, we have demonstrated a framework within which investigators may rationally evaluate NSC migration to, and integration into, brain tumors, and therefore enhance understanding of mechanisms that both promote and limit this therapeutic modality. Stem Cells Translational Medicine 2017;6:1522-1532.


Asunto(s)
Neoplasias Encefálicas/terapia , Movimiento Celular , Glioma/terapia , Células-Madre Neurales/citología , Trasplante de Células Madre/métodos , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones SCID , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante
13.
Stem Cell Reports ; 7(3): 483-495, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27546534

RESUMEN

Pre-clinical studies indicate that neural stem cells (NSCs) can limit or reverse CNS damage through direct cell replacement, promotion of regeneration, or delivery of therapeutic agents. Immortalized NSC lines are in growing demand due to the inherent limitations of adult patient-derived NSCs, including availability, expandability, potential for genetic modifications, and costs. Here, we describe the generation and characterization of a new human fetal NSC line, immortalized by transduction with L-MYC (LM-NSC008) that in vitro displays both self-renewal and multipotent differentiation into neurons, oligodendrocytes, and astrocytes. These LM-NSC008 cells were non-tumorigenic in vivo, and migrated to orthotopic glioma xenografts in immunodeficient mice. When administered intranasally, LM-NSC008 distributed specifically to sites of traumatic brain injury (TBI). These data support the therapeutic development of immortalized LM-NSC008 cells for allogeneic use in TBI and other CNS diseases.


Asunto(s)
Diferenciación Celular/genética , Autorrenovación de las Células/genética , Expresión Génica , Genes myc , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/terapia , Movimiento Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Xenoinjertos , Humanos , Ratones , Células-Madre Neurales/patología , Trasplante de Células Madre , Transcriptoma , Transducción Genética , Transgenes
14.
J Neurosci ; 22(20): 8992-9004, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12388606

RESUMEN

The hyperpolarization-activated excitatory current I(h) shapes rhythmic firing and other components of excitability in differentiating neurons, and may thus influence activity-dependent CNS development. We therefore studied developmental changes in I(h) and underlying hyperpolarization-activated cyclic nucleotide-gated (HCN) channel subunits in pyramidal neurons of neonatal mouse hippocampus using electrophysiological and immunofluorescence approaches. I(h) conductance (at -80 mV) tripled in CA3 neurons and quintupled in CA1 neurons between postnatal day 1 (P1) and P20; parallel changes in membrane area resulted in current density maxima at P5 in CA3 and P10 in CA1. Concurrently, I(h) activation times fell sevenfold in CA3 and 10-fold in CA1. A computational model indicates that a decrease in I(h) activation time will increase the rhythmic firing rate. Two mechanisms contributed to more rapid I(h) activation at P20 in CA3 and CA1 neurons: a fall in the intrinsic time constants of two kinetic components, tau(fast) and tau(slow), to 35-40% (at -90 mV) of their P1 values, and a preferential increase in fast component amplitude and contribution to I(h) (from approximately 35% to approximately 74% of total). HCN1, HCN2, and HCN4 immunoreactivities showed independent temporal and spatial developmental patterns. HCN1 immunoreactivity was low at P1 and P5 and increased by P20. HCN2 immunoreactivity was detected at P1 and increased steadily up to P20. HCN4 immunoreactivity was initially low and showed a small increase by P20. We suggest that developmental increases in I(h) amplitude and activation rate reflect changes in the number and underlying structure of I(h) channels, and that I(h) maturation may shape rhythmic activity important for hippocampal circuit maturation.


Asunto(s)
Canales Iónicos/metabolismo , Proteínas del Tejido Nervioso , Células Piramidales/metabolismo , Factores de Edad , Animales , Diferenciación Celular/fisiología , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Estimulación Eléctrica , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Técnicas In Vitro , Canales Iónicos/antagonistas & inhibidores , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Técnicas de Placa-Clamp , Periodicidad , Canales de Potasio , Subunidades de Proteína , Células Piramidales/citología , Células Piramidales/efectos de los fármacos , Pirimidinas/farmacología
15.
Artículo en Inglés | MEDLINE | ID: mdl-26654402

RESUMEN

The depletion of stem cell pools and the accumulation of senescent cells in animal tissues are linked to aging. Planarians are invertebrate flatworms and are unusual in that their stem cells, called neoblasts, are constantly replacing old and dying cells. By eliminating neoblasts in worms via irradiation, the biological principles of aging are exposed in the absence of wound healing and regeneration, making planaria a powerful tool for aging research.

16.
Mol Cancer Res ; 13(4): 636-50, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25652587

RESUMEN

UNLABELLED: Jumonji domain-containing protein 3 (JMJD3/KDM6B) demethylates lysine 27 on histone H3 (H3K27me3), a repressive epigenetic mark controlling chromatin organization and cellular senescence. To better understand the functional consequences of JMJD3 its expression was investigated in brain tumor cells. Querying patient expression profile databases confirmed JMJD3 overexpression in high-grade glioma. Immunochemical staining of two glioma cell lines, U251 and U87, indicated intrinsic differences in JMJD3 expression levels that were reflected in changes in cell phenotype and variations associated with cellular senescence, including senescence-associated ß-galactosidase (SA-ß-gal) activity and the senescence-associated secretory phenotype (SASP). Overexpressing wild-type JMJD3 (JMJD3wt) activated SASP-associated genes, enhanced SA-ß-gal activity, and induced nuclear blebbing. Conversely, overexpression of a catalytically inactive dominant negative mutant JMJD3 (JMJD3mut) increased proliferation. In addition, a large number of transcripts were identified by RNA-seq as altered in JMJD3 overexpressing cells, including cancer- and inflammation-related transcripts as defined by Ingenuity Pathway Analysis. These results suggest that expression of the SASP in the context of cancer undermines normal tissue homeostasis and contributes to tumorigenesis and tumor progression. These studies are therapeutically relevant because inflammatory cytokines have been linked to homing of neural stem cells and other stem cells to tumor loci. IMPLICATIONS: This glioma study brings together actions of a normal epigenetic mechanism (JMJD3 activity) with dysfunctional activation of senescence-related processes, including secretion of SASP proinflammatory cytokines and stem cell tropism toward tumors.


Asunto(s)
Neoplasias Encefálicas/patología , Senescencia Celular , Glioma/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Epigénesis Genética , Glioma/patología , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Clasificación del Tumor , Células-Madre Neurales/inmunología , Tropismo
17.
Clin Cancer Res ; 21(18): 4062-72, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26059190

RESUMEN

PURPOSE: A first-in-human pilot safety and feasibility trial evaluating chimeric antigen receptor (CAR)-engineered, autologous primary human CD8(+) cytotoxic T lymphocytes (CTL) targeting IL13Rα2 for the treatment of recurrent glioblastoma (GBM). EXPERIMENTAL DESIGN: Three patients with recurrent GBM were treated with IL13(E13Y)-zetakine CD8(+) CTL targeting IL13Rα2. Patients received up to 12 local infusions at a maximum dose of 10(8) CAR-engineered T cells via a catheter/reservoir system. RESULTS: We demonstrate the feasibility of manufacturing sufficient numbers of autologous CTL clones expressing an IL13(E13Y)-zetakine CAR for redirected HLA-independent IL13Rα2-specific effector function for a cohort of patients diagnosed with GBM. Intracranial delivery of the IL13-zetakine(+) CTL clones into the resection cavity of 3 patients with recurrent disease was well-tolerated, with manageable temporary brain inflammation. Following infusion of IL13-zetakine(+) CTLs, evidence for transient anti-glioma responses was observed in 2 of the patients. Analysis of tumor tissue from 1 patient before and after T-cell therapy suggested reduced overall IL13Rα2 expression within the tumor following treatment. MRI analysis of another patient indicated an increase in tumor necrotic volume at the site of IL13-zetakine(+) T-cell administration. CONCLUSIONS: These findings provide promising first-in-human clinical experience for intracranial administration of IL13Rα2-specific CAR T cells for the treatment of GBM, establishing a foundation on which future refinements of adoptive CAR T-cell therapies can be applied.


Asunto(s)
Neoplasias Encefálicas/terapia , Linfocitos T CD8-positivos/inmunología , Glioblastoma/terapia , Inmunoterapia Adoptiva/métodos , Subunidad alfa2 del Receptor de Interleucina-13/uso terapéutico , Receptores de Antígenos de Linfocitos T/uso terapéutico , Adulto , Anciano , Encéfalo/patología , Neoplasias Encefálicas/inmunología , Linfocitos T CD8-positivos/citología , Estudios de Factibilidad , Femenino , Glioblastoma/inmunología , Glioma/inmunología , Glioma/terapia , Antígenos HLA/química , Humanos , Inflamación , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Seguridad del Paciente , Proyectos Piloto , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/inmunología , Recurrencia , Resultado del Tratamiento , Adulto Joven
18.
Neurosci Lett ; 350(2): 73-6, 2003 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-12972156

RESUMEN

We used high-density cDNA microarray analysis to examine changes in the gene expression profile of the hippocampus of C57BL/6 mice following intraperitoneal injection of lipopolysaccharide (LPS). Three hours after injection, the greatest increase in RNA expression was found for an expressed sequence tag subsequently identified as the Cdkn1a gene, coding for the cyclin-dependent kinase inhibitor p21(Waf1/Cip1). Northern blot hybridization confirmed the induction of Cdkn1a mRNA in the central nervous system (CNS), and also revealed similar increases in kidney, liver and heart. Induction of Cdkn1a expression was transient, reaching maximal levels in the CNS 3-6 h after LPS administration, and returning to untreated levels by 24 h. Combined use of laser capture microdissection and quantitative reverse transcription-polymerase chain reaction showed that there was a similar change in Cdkn1a expression for the pyramidal cell layer as for total hippocampus.


Asunto(s)
Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Ciclinas/genética , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Inflamación/etiología , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/biosíntesis , ARN Mensajero/genética
19.
PLoS One ; 9(5): e95123, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24787244

RESUMEN

The interleukin-13 receptor alpha2 (IL13Rα2) is a cell surface receptor that is over-expressed by a subset of high-grade gliomas, but not expressed at significant levels by normal brain tissue. For both malignant and non-malignant cells, IL13Rα2 surface expression is reported to be induced by various cytokines such as IL-4 or IL-13 and tumor necrosis factor (TNF). Our group has developed a therapeutic platform to target IL13Rα2-positive brain tumors by engineering human cytotoxic T lymphocytes (CTLs) to express the IL13-zetakine chimeric antigen receptor. We therefore sought to investigate the potential of cytokine stimulation to induce IL13Rα2 cell surface expression, and thereby increase susceptibility to IL13Rα2-specific T cell killing. In the course of these experiments, we unexpectedly found that the commercially available putative IL13Rα2-specific monoclonal antibody B-D13 recognizes cytokine-induced VCAM-1 on glioblastoma. We provide evidence that the induced receptor is not IL13Rα2, because its expression does not consistently correlate with IL13Rα2 mRNA levels, it does not bind IL-13, and it is not recognized by IL13-zetakine CTL. Instead we demonstrate by immunoprecipitation experiments and mass spectrometry that the antigen recognized by the B-D13 antibody following cytokine stimulation is VCAM-1, and that VCAM-1, but not IL13Rα2, is induced on glioma cells by TNF alone or in combination with IL-13 or IL-4. Further evaluation of several commercial B-D13 antibodies revealed that B-D13 is bi-specific, recognizing both IL13Rα2 and VCAM-1. This binding is non-overlapping based on soluble receptor competition experiments, and mass spectrometry identifies two distinct heavy and light chain species, providing evidence that the B-D13 reagent is di-clonal. PE-conjugation of the B-D13 antibody appears to disrupt IL13Rα2 recognition, while maintaining VCAM-1 specificity. While this work calls into question previous studies that have used the B-D13 antibody to assess IL13Rα2 expression, it also suggests that TNF may have significant effects on glioma biology by up-regulating VCAM-1.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Citocinas/metabolismo , Glioma/patología , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Especificidad de Anticuerpos , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Citocinas/biosíntesis , Progresión de la Enfermedad , Epítopos/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/inmunología , Humanos , Subunidad alfa2 del Receptor de Interleucina-13/genética , Terapia Molecular Dirigida , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T Citotóxicos/inmunología , Molécula 1 de Adhesión Celular Vascular/genética
20.
PLoS One ; 8(10): e77769, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204956

RESUMEN

A major challenge for successful immunotherapy against glioma is the identification and characterization of validated targets. We have taken a bioinformatics approach towards understanding the biological context of IL-13 receptor α2 (IL13Rα2) expression in brain tumors, and its functional significance for patient survival. Querying multiple gene expression databases, we show that IL13Rα2 expression increases with glioma malignancy grade, and expression for high-grade tumors is bimodal, with approximately 58% of WHO grade IV gliomas over-expressing this receptor. By several measures, IL13Rα2 expression in patient samples and low-passage primary glioma lines most consistently correlates with the expression of signature genes defining mesenchymal subclass tumors and negatively correlates with proneural signature genes as defined by two studies. Positive associations were also noted with proliferative signature genes, whereas no consistent associations were found with either classical or neural signature genes. Probing the potential functional consequences of this mesenchymal association through IPA analysis suggests that IL13Rα2 expression is associated with activation of proinflammatory and immune pathways characteristic of mesenchymal subclass tumors. In addition, survival analyses indicate that IL13Rα2 over-expression is associated with poor patient prognosis, a single gene correlation ranking IL13Rα2 in the top ~1% of total gene expression probes with regard to survival association with WHO IV gliomas. This study better defines the functional consequences of IL13Rα2 expression by demonstrating association with mesenchymal signature gene expression and poor patient prognosis. It thus highlights the utility of IL13Rα2 as a therapeutic target, and helps define patient populations most likely to respond to immunotherapy in present and future clinical trials.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Glioma/genética , Subunidad alfa2 del Receptor de Interleucina-13/genética , Recurrencia Local de Neoplasia/genética , Transcriptoma , Adulto , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Femenino , Citometría de Flujo , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Glioma/mortalidad , Glioma/patología , Humanos , Masculino , Mesodermo/metabolismo , Persona de Mediana Edad , Clasificación del Tumor , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia , Células Tumorales Cultivadas , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA