Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(29): e2401420121, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38995966

RESUMEN

Cerebral (Aß) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aß/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aß/pTau, however, appears to vary depending on the animal model. Our prior work suggested that antigen-specific memory CD8 T ("hiT") cells act upstream of Aß/pTau after brain injury. Here, we examine whether hiT cells influence sporadic AD-like pathophysiology upstream of Aß/pTau. Examining neuropathology, gene expression, and behavior in our hiT mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. We identify an age-related factor acting upstream of Aß/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD.


Asunto(s)
Enfermedad de Alzheimer , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/genética , Animales , Linfocitos T CD8-positivos/inmunología , Ratones , Humanos , Placa Amiloide/patología , Placa Amiloide/inmunología , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Encéfalo/patología , Encéfalo/inmunología , Masculino , Interferón gamma/metabolismo , Interferón gamma/inmunología , Envejecimiento/inmunología , Memoria Inmunológica , Células T de Memoria/inmunología , Perforina/metabolismo , Perforina/genética , Femenino
2.
Cancer Immunol Immunother ; 63(9): 911-24, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24893855

RESUMEN

BACKGROUND: Cancer vaccines reproducibly cure laboratory animals and reveal encouraging trends in brain tumor (glioma) patients. Identifying parameters governing beneficial vaccine-induced responses may lead to the improvement of glioma immunotherapies. CD103(+) CD8 T cells dominate post-vaccine responses in human glioma patients for unknown reasons, but may be related to recent thymic emigrant (RTE) status. Importantly, CD8 RTE metrics correlated with beneficial immune responses in vaccinated glioma patients. METHODS: We show by flow cytometry that murine and human CD103(+) CD8 T cells respond better than their CD103(-) counterparts to tumor peptide-MHC I (pMHC I) stimulation in vitro and to tumor antigens on gliomas in vivo. RESULTS: Glioma responsive T cells from mice and humans both exhibited intrinsic de-sialylation-affecting CD8 beta. Modulation of CD8 T cell sialic acid with neuraminidase and ST3Gal-II revealed de-sialylation was necessary and sufficient for promiscuous binding to and stimulation by tumor pMHC I. Moreover, de-sialylated status was required for adoptive CD8 T cells and lymphocytes to decrease GL26 glioma invasiveness and increase host survival in vivo. Finally, increased tumor ST3Gal-II expression correlated with clinical vaccine failure in a meta-analysis of high-grade glioma patients. CONCLUSIONS: Taken together, these findings suggest that de-sialylation of CD8 is required for hyper-responsiveness and beneficial anti-glioma activity by CD8 T cells. Because CD8 de-sialylation can be induced with exogenous enzymes (and appears particularly scarce on human T cells), it represents a promising target for clinical glioma vaccine improvement.


Asunto(s)
Antígenos CD/inmunología , Neoplasias Encefálicas/terapia , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/farmacología , Células Dendríticas/inmunología , Glioma/terapia , Cadenas alfa de Integrinas/inmunología , Animales , Antígenos CD/metabolismo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/inmunología , Femenino , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/terapia , Glioma/inmunología , Glioma/metabolismo , Humanos , Inmunoterapia Adoptiva/métodos , Cadenas alfa de Integrinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuraminidasa/metabolismo , Neuraminidasa/farmacología , Sialiltransferasas/metabolismo , Sialiltransferasas/farmacología , beta-Galactosida alfa-2,3-Sialiltransferasa
3.
BMC Cancer ; 14: 920, 2014 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-25481245

RESUMEN

BACKGROUND: Human Hematopoietic Signal peptide-containing Secreted 1 (hHSS1) is a truly novel protein, defining a new class of secreted factors. We have previously reported that ectopic overexpression of hHSS1 has a negative modulatory effect on cell proliferation and tumorigenesis in glioblastoma model systems. Here we have used microarray analysis, screened glioblastoma samples in The Cancer Genome Atlas (TCGA), and studied the effects of hHSS1 on glioma-derived cells and endothelial cells to elucidate the molecular mechanisms underlying the anti-tumorigenic effects of hHSS1. METHODS: Gene expression profiling of human glioma U87 and A172 cells overexpressing hHSS1 was performed. Ingenuity® iReport™ and Ingenuity Pathway Analysis (IPA) were used to analyze the gene expression in the glioma cells. DNA content and cell cycle analysis were performed by FACS, while cell migration, cell invasion, and effects of hHSS1 on HUVEC tube formation were determined by transwell and matrigel assays. Correlation was made between hHSS1 expression and specific genes in glioblastoma samples in the TCGA database. RESULTS: We have clarified the signaling and metabolic pathways (i.e. role of BRCA1 in DNA damage response), networks (i.e. cell cycle) and biological processes (i.e. cell division process of chromosomes) that result from hHSS1effects upon glioblastoma growth. U87-overexpressing hHSS1 significantly decreased the number of cells in the G0/G1 cell cycle phase, and significantly increased cells in the S and G2/M phases (P < 0.05). U87-overexpressing hHSS1 significantly lost their ability to migrate (P < 0.001) and to invade (P < 0.01) through matrigel matrix. hHSS1-overexpression significantly decreased migration of A172 cells (P < 0.001), inhibited A172 tumor-induced migration and invasion of HUVECs (P < 0.001), and significantly inhibited U87 tumor-induced invasion of HUVECs (P < 0.001). Purified hHSS1 protein inhibited HUVEC tube formation. TCGA database revealed significant correlation between hHSS1 and BRCA2 (r = -0.224, P < 0.0005), ADAMTS1 (r = -0.132, P <0.01) and endostatin (r = 0.141, P < 0.005). CONCLUSIONS: hHSS1-overexpression modulates signaling pathways involved in tumorigenesis. hHSS1 inhibits glioma-induced cell cycle progression, cell migration, invasion and angiogenesis. Our data suggest that hHSS1 is a potential therapeutic for malignant glioblastoma possessing significant antitumor and anti-angiogenic activity.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/metabolismo , Proteínas/metabolismo , Transducción de Señal , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Biología Computacional , Daño del ADN , Bases de Datos de Ácidos Nucleicos , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Glioma/patología , Humanos , Proteínas de la Membrana , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Reproducibilidad de los Resultados
4.
bioRxiv ; 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38328072

RESUMEN

Cerebral (Aß) plaque and (pTau) tangle deposition are hallmarks of Alzheimer's disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aß/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aß/fibrillar pTau, however, appears to vary depending on the animal model used. Our prior work suggested that antigen-specific memory CD8 T (" hi T") cells act upstream of Aß/pTau after brain injury. Here we examine whether hi T cells influence sporadic AD-like pathophysiology upstream of Aß/pTau. Examining neuropathology, gene expression, and behavior in our hi T mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. Our work is the first to identify an age-related factor acting upstream of Aß/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD. Significance Statement: This study changes our view of Alzheimer's Disease (AD) initiation and progression. Mutations promoting cerebral beta-amyloid (Aß) deposition guarantee rare genetic forms of AD. Thus, the prevailing hypothesis has been that Aß is central to initiation and progression of all AD, despite contrary animal and patient evidence. We show that age-related T cells generate neurodegeneration with compelling features of AD in mice, with distinct T cell functions required for pathological initiation and neurodegenerative progression. Knowledge from these mice was applied to successfully predict previously unknown features of human AD and generate novel tools for its clinical management.

5.
Cancer Immunol Immunother ; 62(1): 125-35, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22847020

RESUMEN

BACKGROUND: This study evaluated the safety and immune responses to an autologous dendritic cell vaccine pulsed with class I peptides from tumor-associated antigens (TAA) expressed on gliomas and overexpressed in their cancer stem cell population (ICT-107). METHODS: TAA epitopes included HER2, TRP-2, gp100, MAGE-1, IL13Rα2, and AIM-2. HLA-A1- and/or HLA-A2-positive patients with glioblastoma (GBM) were eligible. Mononuclear cells from leukapheresis were differentiated into dendritic cells, pulsed with TAA peptides, and administered intradermally three times at two-week intervals. RESULTS: Twenty-one patients were enrolled with 17 newly diagnosed (ND-GBM) and three recurrent GBM patients and one brainstem glioma. Immune response data on 15 newly diagnosed patients showed 33 % responders. TAA expression by qRT-PCR from fresh-frozen tumor samples showed all patient tumors expressed at least three TAA, with 75 % expressing all six. Correlations of increased PFS and OS with quantitative expression of MAGE1 and AIM-2 were observed, and a trend for longer survival was observed with gp100 and HER2 antigens. Target antigens gp100, HER1, and IL13Rα2 were downregulated in recurrent tumors from 4 HLA-A2+ patients. A decrease in or absence of CD133 expression was seen in five patients who underwent a second resection. At a median follow-up of 40.1 months, six of 16 ND-GBM patients showed no evidence of tumor recurrence. Median PFS in newly diagnosed patients was 16.9 months, and median OS was 38.4 months. CONCLUSIONS: Expression of four ICT-107 targeted antigens in the pre-vaccine tumors correlated with prolonged overall survival and PFS in ND-GBM patients. The goal of targeting tumor antigens highly expressed on glioblastoma cancer stem cells is supported by the observation of decreased or absent CD133 expression in the recurrent areas of gadolinium-enhanced tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Epítopos/inmunología , Glioblastoma/terapia , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Células Dendríticas/trasplante , Femenino , Glioblastoma/inmunología , Glioblastoma/mortalidad , Glioblastoma/patología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
J Alzheimers Dis ; 93(3): 919-921, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37212123

RESUMEN

Smith and Ashford present a compelling hypothesis on evolution of APOE alleles, namely that ɛ4 prevalence is mediated by immune selection pressure against enteric pathogens. While the ɛ3 allele is more prevalent today, it outcompetedɛ4 only relatively recently, as immune selection pressure for more effective immune responses to such pathogens was alleviated with transition to agrarian from hunter-gatherer lifestyles. Smith and Ashford's hypothesis is intriguing in itself, but the implications for APOE ɛ4 function in Alzheimer's disease are even more so and encourage greater focus on specific aspects of immunity in accounting for both ɛ4-mediated and general Alzheimer's disease risk.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/genética , Alelos , Prevalencia , Apolipoproteínas E/genética , Apolipoproteína E4/genética , Genotipo
7.
Oncogene ; 42(25): 2088-2098, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37161052

RESUMEN

The promise of adaptive cancer immunotherapy in treating highly malignant tumors such as glioblastoma multiforme (GBM) can only be realized through expanding its benefits to more patients. Alleviating various modes of immune suppression has so far failed to achieve such expansion, but exploiting endogenous immune enhancers among mutated cancer genes could represent a more direct approach to immunotherapy improvement. We found that Isocitrate Dehydrogenase-1 (IDH1), which is commonly mutated in gliomas, enhances glioma vaccine efficacy in mice and discerns long from short survivors after vaccine therapy in GBM patients. Extracellular IDH1 directly enhanced T cell responses to multiple tumor antigens, and prolonged experimental glioma cell lysis. Moreover, IDH1 specifically bound to and exhibited sialidase activity against CD8. By contrast, mutant IDH1R132H lacked sialidase activity, delayed killing in glioma cells, and decreased host survival after immunotherapy. Overall, our findings identify IDH1 as an immunotherapeutic enhancer that mediates the known T cell-enhancing reaction of CD8 desialylation. This uncovers a new axis for immunotherapeutic improvement in GBM and other cancers, reveals novel physiological and molecular functions of IDH1, and hints at an unexpectedly direct link between lytic T cell function and metabolic activity in target cells.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Ratones , Animales , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Ácido N-Acetilneuramínico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/metabolismo , Neuraminidasa , Glioma/genética , Glioma/terapia , Glioma/metabolismo , Glioblastoma/genética , Glioblastoma/terapia , Linfocitos T CD8-positivos/metabolismo , Inmunoterapia , Mutación
8.
J Neurooncol ; 102(2): 197-211, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20680400

RESUMEN

The completion of the Human Genome Project resulted in discovery of many unknown novel genes. This feat paved the way for the future development of novel therapeutics for the treatment of human disease based on novel biological functions and pathways. Towards this aim, we undertook a bioinformatics analysis of in-house microarray data derived from purified hematopoietic stem cell populations. This effort led to the discovery of HSS1 (Hematopoietic Signal peptide-containing Secreted 1) and its splice variant HSM1 (Hematopoietic Signal peptide-containing Membrane domain-containing 1). HSS1 gene is evolutionarily conserved across species, phyla and even kingdoms, including mammals, invertebrates and plants. Structural analysis showed no homology between HSS1 and known proteins or known protein domains, indicating that it was a truly novel protein. Interestingly, the human HSS1 (hHSS1) gene is located at chromosome 19q13.33, a genomic region implicated in various cancers, including malignant glioma. Stable expression of hHSS1 in glioma-derived A172 and U87 cell lines greatly reduced their proliferation rates compared to mock-transfected cells. hHSS1 expression significantly affected the malignant phenotype of U87 cells both in vitro and in vivo. Further, preliminary immunohistochemical analysis revealed an increase in hHSS1/HSM1 immunoreactivity in two out of four high-grade astrocytomas (glioblastoma multiforme, WHO IV) as compared to low expression in all four low-grade diffuse astrocytomas (WHO grade II). High-expression of hHSS1 in high-grade gliomas was further supported by microarray data, which indicated that mesenchymal subclass gliomas exclusively up-regulated hHSS1. Our data reveal that HSS1 is a truly novel protein defining a new class of secreted factors, and that it may have an important role in cancer, particularly glioma.


Asunto(s)
Neoplasias Encefálicas/patología , Cromosomas Humanos Par 19/genética , Glioblastoma/patología , Proteínas/metabolismo , Proteínas Supresoras de Tumor/genética , Secuencia de Aminoácidos , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias Encefálicas/genética , Clonación Molecular , Perfilación de la Expresión Génica , Biblioteca de Genes , Glioblastoma/genética , Glicosilación , Humanos , Técnicas para Inmunoenzimas , Masculino , Proteínas de la Membrana , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Tasa de Supervivencia , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Clin Neurosci ; 74: 187-193, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32169363

RESUMEN

High grade gliomas are associated with poor prognosis and high mortality. Conventional treatments and management of high grade gliomas have shown little improvement in 5-year overall survival. This phase I trial evaluated the safety, immunogenicity, and potential synergy of surgical resection with Gliadel Wafer implantation, followed by autologous tumor lysate-pulsed dendritic cell (DC) vaccine in patients with malignant glioma. Primary end points of this study were safety and surrogate markers of immunogenicity, overall survival, and progression free survival. Following surgical resection, Gliadel Wafers were placed along the resection cavity. Patients subsequently received intradermal injections of autologous tumor lysate-pulsed DC vaccines 3 times at 2 week intervals. Treatment response was evaluated clinically and through MRI at regular intervals. Twenty-eight patients received Gliadel Wafers and DC vaccination: 11 newly diagnosed (8 glioblastoma [GBM], 2 anaplastic astrocytoma [AA], and 1 anaplastic oligodendroglioma [AO]) and 17 recurrent (15 GBMs, 1 AA, and 1 AO) high grade gliomas. Immunogenicity data was collected for 20 of the 28 patients. Five of 20 patients showed elevated IFN-γ responses following vaccination. Median progression-free survival and overall survival for all GBM patients in the trial from the start of vaccination were 3.6 months and 16.9 months respectively. Comparisons between vaccine responders and non-vaccine responders were not statistically significant. Adjuvant autologous dendritic cells pulsed with tumor-lysate following resection and Gliadel Wafer placement is safe, elicits modest immunogenicity and shows similar clinical outcomes in patients who had DC vaccination in previous studies.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Carmustina/uso terapéutico , Ácidos Decanoicos/uso terapéutico , Células Dendríticas/trasplante , Glioma/terapia , Poliésteres/uso terapéutico , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Antineoplásicos/uso terapéutico , Terapia Combinada/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Vacunación/métodos
10.
Front Neurol ; 11: 557269, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33424735

RESUMEN

The incidence of autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD), which frequently co-occur, are both rising. The causes of ASD and ADHD remain elusive, even as both appear to involve perturbation of the gut-brain-immune axis. CD103 is an integrin and E-cadherin receptor most prominently expressed on CD8 T cells that reside in gut, brain, and other tissues. CD103 deficiency is well-known to impair gut immunity and resident T cell function, but it's impact on neurodevelopmental disorders has not been examined. We show here that CD8 T cells influence neural progenitor cell function, and that CD103 modulates this impact both directly and potentially by controlling CD8 levels in brain. CD103 knockout (CD103KO) mice exhibited a variety of behavioral abnormalities, including superior cognitive performance coupled with repetitive behavior, aversion to novelty and social impairment in females, with hyperactivity with delayed learning in males. Brain protein markers in female and male CD103KOs coincided with known aspects of ASD and ADHD in humans, respectively. Surprisingly, CD103 deficiency also decreased age-related cognitive decline in both sexes, albeit by distinct means. Together, our findings reveal a novel role for CD103 in brain developmental function, and identify it as a unique factor linking ASD and ADHD etiology. Our data also introduce a new animal model of combined ASD and ADHD with associated cognitive benefits, and reveal potential therapeutic targets for these disorders and age-related cognitive decline.

11.
Mech Ageing Dev ; 191: 111351, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32910956

RESUMEN

Mitigating effects of aging on human health remains elusive because aging impacts multiple systems simultaneously, and because experimental animals exhibit critical aging differences relative to humans. Separation of aging into discrete processes may identify targetable drivers of pathology, particularly when applied to human-specific features. Gradual homeostatic expansion of CD8 T cells dominantly alters their function in aging humans but not in mice. Injecting T cells into athymic mice induces rapid homeostatic expansion, but its relevance to aging remains uncertain. We hypothesized that homeostatic expansion of T cells injected into T-deficient hosts models physiologically relevant CD8 T cell aging in young mice, and aimed to analyze age-related T cell phenotype and tissue pathology in such animals. Indeed, we found that such injection conferred uniform age-related phenotype, genotype, and function to mouse CD8 T cells, heightened age-associated tissue pathology in young athymic hosts, and humanized amyloidosis after brain injury in secondary wild-type recipients. This validates a model conferring a human-specific aging feature to mice that identifies targetable drivers of tissue pathology. Similar examination of independent aging features should promote systematic understanding of aging and identify additional targets to mitigate its effects on human health.


Asunto(s)
Envejecimiento/inmunología , Amiloidosis/inmunología , Lesiones Encefálicas/inmunología , Linfocitos T CD8-positivos/inmunología , Senescencia Celular/inmunología , Envejecimiento/genética , Amiloidosis/genética , Animales , Senescencia Celular/genética , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos
12.
Mol Cancer Ther ; 18(3): 718-725, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30755456

RESUMEN

A cell culture platform that enables ex vivo tissue growth from patients or patient-derived xenograft (PDX) models and assesses sensitivity to approved therapies (e.g., temozolomide) in a clinically relevant time frame would be very useful in translational research and personalized medicine. Here, we present a novel three-dimensional (3D) ECM hydrogel system, VersaGel, for assaying ex vivo growth and therapeutic response with standard image microscopy. Specifically, multicellular spheroids deriving from either 5 patients with glioblastoma (GBM) or a renal cell carcinoma (RCC) PDX model were incorporated into VersaGel and treated with temozolomide and several other therapies, guided by the most recent advances in GBM treatment. RCC ex vivo tissue displayed invasive phenotypes in conditioned media. For the GBM patient tumor testing, all five clinical responses were predicted by the results of our 3D-temozolomide assay. In contrast, the MTT assay found no response to temozolomide regardless of the clinical outcome, and moreover, basement membrane extract failed to predict the 2 patient responders. Finally, 1 patient was tested with repurposed drugs currently being administered in GBM clinical trials. Interestingly, IC50s were lower than C max for crizotinib and chloroquine, but higher for sorafenib. In conclusion, a novel hydrogel platform, VersaGel, enables ex vivo tumor growth of patient and PDX tissue and offers insight into patient response to clinically relevant therapies. We propose a novel 3D hydrogel platform, VersaGel, to grow ex vivo tissue (patient and PDX) and assay therapeutic response using time-course image analysis.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Esferoides Celulares/efectos de los fármacos , Temozolomida/farmacología , Anciano , Animales , Carcinoma de Células Renales/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Femenino , Glioblastoma/patología , Humanos , Hidrogeles/farmacología , Masculino , Ratones , Supervivencia sin Progresión , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oncogene ; 24(33): 5226-34, 2005 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-15897911

RESUMEN

Tyrosinase-related protein (TRP)-2 is not only expressed on glioma cells, but is naturally processed and presented by their surface MHC molecules and is recognized by TRP-2-specific cytotoxic T cells. After active immunotherapy, we detected TRP-2-specific cytotoxic T lymphocyte (CTL) activity in patients' peripheral blood mononuclear cells (PBMC). Tumor cells from postvaccination resections showed significantly lower TRP-2 expression and higher sensitivity to carboplatin and temozolomide than those autologous cell lines from prevaccination resections in two patients who demonstrated CTL response to TRP-2. One of two patients underwent treatment with temozolomide after recurrence and responded dramatically. TRP-2-transfected cell line (TRP-2-U373) resulted in significant drug resistance to carboplatin and temozolomide compared to wild-type U-373 (W-U373). There was no significant difference, however, in the mRNA expression of other common drug resistance related proteins, such as BCRP-1, MGMT, MDR-1, MRP-1 and MRP-3, after TRP-2 transfection. TRP-2-U373 tumor cells were immunoselected by a TRP-2-specific CTL line. The immunoselected cells (IS-TRP-2-U373) demonstrated significantly increased sensitivity to carboplatin and temozolomide compared to TRP-2-U373. For the first time, we provide evidence that immunological targeting of tumor-associated antigen TRP-2 significantly increases sensitivity to chemotherapy.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos/inmunología , Glioma/inmunología , Glioma/terapia , Oxidorreductasas Intramoleculares/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos de Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carboplatino/farmacología , Terapia Combinada , Daño del ADN , Dacarbazina/farmacología , Interacciones Farmacológicas , Humanos , Inmunoterapia Activa , Temozolomida , Transfección , Resultado del Tratamiento , Células Tumorales Cultivadas
14.
Cancer Res ; 64(14): 4973-9, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15256471

RESUMEN

The primary goal of this Phase I study was to assess the safety and bioactivity of tumor lysate-pulsed dendritic cell (DC) vaccination to treat patients with glioblastoma multiforme and anaplastic astrocytoma. Adverse events, survival, and cytotoxicity against autologous tumor and tumor-associated antigens were measured. Fourteen patients were thrice vaccinated 2 weeks apart with autologous DCs pulsed with tumor lysate. Peripheral blood mononuclear cells were differentiated into phenotypically and functionally confirmed DCs. Vaccination with tumor lysate-pulsed DCs was safe, and no evidence of autoimmune disease was noted. Ten patients were tested for the development of cytotoxicity through a quantitative PCR-based assay. Six of 10 patients demonstrated robust systemic cytotoxicity as demonstrated by IFN-gamma expression by peripheral blood mononuclear cells in response to tumor lysate after vaccination. Using HLA-restricted tetramer staining, we identified a significant expansion in CD8+ antigen-specific T-cell clones against one or more of tumor-associated antigens MAGE-1, gp100, and HER-2 after DC vaccination in four of nine patients. A significant CD8+ T-cell infiltrate was noted intratumorally in three of six patients who underwent reoperation. The median survival for patients with recurrent glioblastoma multiforme in this study (n = 8) was 133 weeks. This Phase I study demonstrated the feasibility, safety, and bioactivity of an autologous tumor lysate-pulsed DC vaccine for patients with malignant glioma. We demonstrate for the first time the ability of an active immunotherapy strategy to generate antigen-specific cytotoxicity in brain tumor patients.


Asunto(s)
Astrocitoma/terapia , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Glioblastoma/terapia , Inmunoterapia Adoptiva/métodos , Adulto , Astrocitoma/inmunología , Neoplasias Encefálicas/inmunología , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/uso terapéutico , Epítopos de Linfocito T/inmunología , Femenino , Glioblastoma/inmunología , Humanos , Inmunoterapia Adoptiva/efectos adversos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/terapia , Linfocitos T Citotóxicos/inmunología
15.
Cancer Res ; 64(14): 4980-6, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15256472

RESUMEN

It has recently been demonstrated that malignant glioma cells express certain known tumor-associated antigens, such as HER-2, gp100, and MAGE-1. To further determine the possible utilization of these antigens for glioma immunotherapy and as surrogate markers for specific tumor antigen cytotoxicity, we characterized the presence of mRNA and protein expression in 43 primary glioblastoma multiforme (GBM) cell lines and 7 established human GBM cell lines. HER-2, gp100, and MAGE-1 mRNA expression was detected in 81.4%, 46.5%, and 39.5% of the GBM primary cell lines, respectively. Using immunoreactive staining analysis by flow cytometry, HER-2, gp100, and MAGE-1 protein expression was detected in 76%, 45%, and 38% of the GBM primary cell lines, respectively. HLA-A1-restricted epitope specific for MAGE-1 peptide (EADPTGHSY) CTL clone B07 and HLA-A2-restricted epitope specific for HER-2 peptide (KIFGSLAFL) CTL clone A05 and gp100 peptide (ITDQVPFSV) CTL clone CK3H6 were used in this study. The specificity of CTL clone was verified by HLA/peptide tetramer staining. Three CTL clones could efficiently recognize GBM tumor cells in an antigen-specific and MHC class I-restricted manner. IFN-gamma treatment can dramatically increase MHC class I expression of GBM tumor cells and significantly increase CTL recognition of tumor cells. Treatment with the DNA hypomethylating agent 5-aza-2'-deoxycytidine induced and up-regulated the mRNA expression of MAGE-1 and epitope presentation by autologous MHC. These data indicate that HER-2, gp100, and MAGE-1 could be used as tumor antigen targets for surrogate assays for antigen-specific CTLs or to develop antigen-specific active immunotherapy strategies for glioma patients.


Asunto(s)
Azacitidina/análogos & derivados , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas de Neoplasias/inmunología , Receptor ErbB-2/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Presentación de Antígeno , Antígenos de Neoplasias , Azacitidina/inmunología , Azacitidina/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Metilación de ADN , Decitabina , Epítopos de Linfocito T/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Interferón gamma/inmunología , Interferón gamma/farmacología , Antígenos Específicos del Melanoma , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/genética , Antígeno gp100 del Melanoma
17.
Front Biosci ; 10: 2861-81, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15970541

RESUMEN

The dismal prognoses suffered by malignant primary brain tumor (glioma) patients remain unchanged over the past two decades despite significant improvements in the treatment of distinct tumors. Immunotherapy, and vaccine therapy in particular, represents a promising experimental approach to treat malignant gliomas, but major challenges still remain to render vaccination clinically effective. These challenges include diminishing the risk of pathologic autoimmunity, and identifying the cellular basis of clinical vaccine benefits. Addressing such challenges should eventually help increase the proportion of patients experiencing clinical vaccine benefits. Recent studies in glioma patients have characterized tumor antigens on human gliomas, identified some of the immune cells involved in beneficial anti-glioma immunity, and examined how gliomas may be altered by sub-lethal immune influences. This has provided a glimpse of the strength to which immunity influences glioma clinical outcome, and resurrects hope that clinically effective vaccines to treat these tumors is within reach. Insight into the complex dynamics of immune-tumor interactions promises to extend this reach by delineating mechanisms of immune synergy with other forms of treatment.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Glioma/terapia , Linfocitos T/inmunología , Antígenos Virales de Tumores/inmunología , Neoplasias Encefálicas/inmunología , Vacunas contra el Cáncer/efectos adversos , Glioma/inmunología , Humanos , Inmunoterapia
18.
Curr Opin Mol Ther ; 7(1): 35-47, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15732528

RESUMEN

The dismal prognoses suffered by malignant primary brain tumor (glioma) patients remain unchanged over the past two decades, despite significant improvements in the treatment of distinct tumors. Dendritic cell-based vaccine therapies represent a promising experimental approach to treat malignant gliomas, but major challenges still remain to render vaccination more effective. These challenges include diminishing the risk of pathological autoimmunity, identifying the cellular basis of clinical vaccine benefits and increasing the proportion of patients experiencing such benefits. Over the past three years, studies in glioma patients have characterized tumor antigens on human gliomas, identified some of the immune cells involved in beneficial anti-glioma immunity and examined how gliomas may be altered by sub-lethal immune influences, providing a glimpse of the strength with which immunity influences glioma clinical outcome. This progress promises to improve clinical vaccines for glioma, and perhaps for other cancers.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Glioma/tratamiento farmacológico , Animales , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Glioma/inmunología , Glioma/mortalidad , Humanos , Ratones , Sobrevida , Timo/inmunología
19.
Clin Cancer Res ; 10(16): 5316-26, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15328167

RESUMEN

PURPOSE: Although the development of immune-based therapies for various cancers including malignant glioma has been heralded with much hope and optimism, objective clinical improvements in most vaccinated cancer patients have not been realized. To broaden the search for vaccine-induced benefits, we examined synergy of vaccines with conventional chemotherapy. EXPERIMENTAL DESIGN: Survival and progression times were analyzed retrospectively in 25 vaccinated (13 with and 12 without subsequent chemotherapy) and 13 nonvaccinated de novo glioblastoma (GBM) patients receiving chemotherapy. Immune responsiveness and T-cell receptor excision circle (TREC) content within CD8+ T cells (CD8+ TRECs) was determined in vaccinated patients. RESULTS: Vaccinated patients receiving subsequent chemotherapy exhibited significantly longer times to tumor recurrence after chemotherapy relative to their own previous recurrence times, as well as significantly longer postchemotherapy recurrence times and survival relative to patients receiving isolated vaccination or chemotherapy. Patients exhibiting objective (>50%) tumor regression, extremely rare in de novo GBM, were also confined to the vaccine + chemotherapy group. Prior tumor behavior, demographic factors, other treatment variables, distribution of vaccine responders, and patients with high CD8+ TRECs all failed to account for these differences in clinical outcome. Within all GBM patients receiving post-vaccine chemotherapy, however, CD8+ TRECs predicted significantly longer chemotherapeutic responses, revealing a strong link between the predominant T-cell effectors in GBM and tumor chemosensitivity. CONCLUSIONS: We propose that therapeutic vaccination synergizes with subsequent chemotherapy to elicit tangible clinical benefits for GBM patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Vacunas contra el Cáncer/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/inmunología , Adulto , Anciano , Neoplasias Encefálicas/mortalidad , Craneotomía , Cartilla de ADN , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Glioblastoma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Estudios Retrospectivos , Análisis de Supervivencia , Linfocitos T/inmunología , Resultado del Tratamiento
20.
Cancer Gene Ther ; 9(1): 9-15, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11916248

RESUMEN

Interleukin 12 (IL-12) isa cytokine that promotesan antitumor Th1-type pattern of differentiation in mature naïveT cells. Despite its therapeutic success in multiple animal models of cancer, the utility of systemically administered recombinant cytokine has been limited by its toxicity. This has encouraged the development of local IL-12 delivery systems through gene transfer. To determine the effect of local adenoviral delivery of IL- 12 on glioma immunogenicity, mice bearing GL-26 gliomas in the right corpus striatum were treated with direct intratumoral administration of AdmIL-12, AdLacZ, or normal saline. Survival was significantly prolonged in AdmIL-12-treated animals and immunohistochemistry demonstrated robust CD4+ and CD8+ T-cell infiltration in these mice compared to the two control groups. Glioma-infiltrating T lymphocytes from mice that received AdmIL-12 also demonstrated relatively increased, albeit statistically nonsignificant tumor killing. Based on IL-12's known ability to enhance Th1-type cytotoxic antitumor immune responses, we postulate our findings to be a result of localized induction of tumor immunity.


Asunto(s)
Adenovirus Humanos/genética , Neoplasias Encefálicas/terapia , Glioma/terapia , Interleucina-12/genética , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Terapia Genética , Vectores Genéticos , Glioma/inmunología , Glioma/patología , Humanos , Técnicas para Inmunoenzimas , Operón Lac/fisiología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Tasa de Supervivencia , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA