Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Methods Cell Biol ; 180: 49-67, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37890932

RESUMEN

Transforming growth factor ß (TGFß) is exquisitely regulated under physiological conditions but its activity is highly dysregulated in cancer. All cells make TGFß and have receptors for the ligand, which is sequestered in the extracellular matrix in a latent form. Ionizing radiation elicits rapid release of TGFß from these stores, so-called activation, over a wide range of doses and exposures, including low dose (<1Gy) whole-body irradiation, creating an extraordinarily potent signal in the irradiated tissue or tumor. Hence, accurate evaluation of TGFß activity is complicated because of its ubiquitous distribution as a latent complex. Here we describe conditions for assays that reveal TGFß activity in situ using either tissue preparations or functional imaging.


Asunto(s)
Neoplasias , Factor de Crecimiento Transformador beta , Humanos , Transducción de Señal , Neoplasias/radioterapia , Matriz Extracelular
2.
Clin Cancer Res ; 27(6): 1778-1791, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33402361

RESUMEN

PURPOSE: Women treated with radiotherapy before 30 years of age have increased risk of developing breast cancer at an early age. Here, we sought to investigate mechanisms by which radiation promotes aggressive cancer. EXPERIMENTAL DESIGN: The tumor microenvironment (TME) of breast cancers arising in women treated with radiotherapy for Hodgkin lymphoma was compared with that of sporadic breast cancers. To investigate radiation effects on carcinogenesis, we analyzed tumors arising from Trp53-null mammary transplants after irradiation of the target epithelium or host using immunocompetent and incompetent mice, some of which were treated with aspirin. RESULTS: Compared with age-matched specimens of sporadic breast cancer, radiation-preceded breast cancers (RP-BC) were characterized by TME rich in TGFß, cyclooxygenase 2, and myeloid cells, indicative of greater immunosuppression, even when matched for triple-negative status. The mechanism by which radiation impacts TME construction was investigated in carcinomas arising in mice bearing Trp53-null mammary transplants. Immunosuppressive TMEs (iTME) were recapitulated in mice irradiated before transplantation, which implicated systemic immune effects. In nu/nu mice lacking adaptive immunity irradiated before Trp53-null mammary transplantation, cancers also established an iTME, which pointed to a critical role for myeloid cells. Consistent with this, irradiated mammary glands contained more macrophages and human cells cocultured with polarized macrophages underwent dysplastic morphogenesis mediated by IFNγ. Treating mice with low-dose aspirin for 6 months postirradiation prevented establishment of an iTME and resulted in less aggressive tumors. CONCLUSIONS: These data show that radiation acts via nonmutational mechanisms to promote markedly immunosuppressive features of aggressive, RP-BCs.


Asunto(s)
Neoplasias de la Mama/radioterapia , Inflamación/complicaciones , Neoplasias Inflamatorias de la Mama/patología , Macrófagos/inmunología , Radioterapia/efectos adversos , Microambiente Tumoral , Animales , Apoptosis , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Inflamación/patología , Neoplasias Inflamatorias de la Mama/etiología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Radiat Oncol Biol Phys ; 109(2): 527-539, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33007434

RESUMEN

PURPOSE: Transforming growth factor ß (TGFß) promotes cell survival by endorsing DNA damage repair and mediates an immunosuppressive tumor microenvironment. Thus, TGFß activation in response to radiation therapy is potentially targetable because it opposes therapeutic control. Strategies to assess this potential in the clinic are needed. METHODS AND MATERIALS: We evaluated positron emission tomography (PET) to image 89Zr -fresolimumab, a humanized TGFß neutralizing monoclonal antibody, as a means to detect TGFß activation in intracranial tumor models. Pathway activity of TGFß was validated by immunodetection of phosphorylated SMAD2 and the TGFß target, tenascin. The contribution of TGFß to radiation response was assessed by Kaplan-Meier survival analysis of mice bearing intracranial murine tumor models GL261 and SB28 glioblastoma and brain-adapted 4T1 breast cancer (4T1-BrA) treated with TGFß neutralizing monoclonal antibody, 1D11, and/or focal radiation (10 Gy). RESULTS: 89Zr-fresolimumab PET imaging detected engineered, physiological, and radiation-induced TGFß activation, which was confirmed by immunostaining of biological markers. GL261 glioblastoma tumors had a greater PET signal compared with similar-sized SB28 glioblastoma tumors, whereas the widespread PET signal of 4T1-BrA intracranial tumors was consistent with their highly dispersed histologic distribution. Survival of mice bearing intracranial tumors treated with 1D11 neutralizing antibody alone was similar to that of mice treated with control antibody, whereas 1D11 improved survival when given in combination with focal radiation. The extent of survival benefit of a combination of radiation and 1D11 was associated with the degree of TGFß activity detected by PET. CONCLUSIONS: This study demonstrates that 89Zr-fresolimumab PET imaging detects radiation-induced TGFß activation in tumors. Functional imaging indicated a range of TGFß activity in intracranial tumors, but TGFß blockade provided survival benefit only in the context of radiation treatment. This study provides further evidence that radiation-induced TGFß activity opposes therapeutic response to radiation.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/radioterapia , Tomografía de Emisión de Positrones , Factor de Crecimiento Transformador beta/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Factor de Crecimiento Transformador beta/inmunología
4.
Mol Cancer Ther ; 20(9): 1508-1520, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34210826

RESUMEN

Advanced peritoneal carcinomatosis including high-grade ovarian cancer has poor prognoses and a poor response rate to current checkpoint inhibitor immunotherapies; thus, there is an unmet need for effective therapeutics that would provide benefit to these patients. Here we present the preclinical development of SENTI-101, a cell preparation of bone marrow-derived mesenchymal stromal (also known as stem) cells (MSC), which are engineered to express two potent immune-modulatory cytokines, IL12 and IL21. Intraperitoneal administration of SENTI-101 results in selective tumor-homing and localized and sustained cytokine production in murine models of peritoneal cancer. SENTI-101 has extended half-life, reduced systemic distribution, and improved antitumor activity when compared with recombinant cytokines, suggesting that it is more effective and has lower risk of systemic immunotoxicities. Treatment of tumor-bearing immune-competent mice with a murine surrogate of SENTI-101 (mSENTI-101) results in a potent and localized immune response consistent with increased number and activation of antigen presenting cells, T cells and B cells, which leads to antitumor response and memory-induced long-term immunity. Consistent with this mechanism of action, co-administration of mSENTI-101 with checkpoint inhibitors leads to synergistic improvement in antitumor response. Collectively, these data warrant potential clinical development of SENTI-101 for patients with peritoneal carcinomatosis and high-grade ovarian cancer.Graphical abstract: SENTI-101 schematic and mechanism of actionSENTI-101 is a novel cell-based immunotherapeutic consisting of bone marrow-derived mesenchymal stromal cells (BM-MSC) engineered to express IL12 and IL21 intended for the treatment of peritoneal carcinomatosis including high-grade serous ovarian cancer. Upon intraperitoneal administration, SENTI-101 homes to peritoneal solid tumors and secretes IL12 and IL21 in a localized and sustained fashion. The expression of these two potent cytokines drives tumor infiltration and engagement of multiple components of the immune system: antigen-presenting cells, T cells, and B cells, resulting in durable antitumor immunity in preclinical models of cancer.


Asunto(s)
Interleucina-12/metabolismo , Interleucinas/metabolismo , Melanoma Experimental/inmunología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Neoplasias/inmunología , Neoplasias Peritoneales/inmunología , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/terapia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Imaging Biol ; 22(1): 105-114, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31065895

RESUMEN

PURPOSE: Tumor necrosis factor alpha (TNFα) drives inflammation and bone degradation in patients with rheumatoid arthritis (RA). Some RA patients experience a rapid clinical response to TNFα inhibitors such as certolizumab pegol (CZP) while other patients show limited to no response. Current methods for imaging RA have limited sensitivity and do not assist in the selection of patients most likely to respond to immune-mediated therapy. Herein, we developed a novel positron emission tomography (PET) radiotracer for immuno-PET imaging of TNFα in transgenic human TNFα-expressing mice. PROCEDURES: CZP was modified with p-isothiocyanatobenzyl-deferoxamine (DFO) and radiolabeled with Zr-89. The biological activity of [89Zr]DFO-CZP was evaluated by HPLC and binding assay using human recombinant TNFα (hTNFα). The feasibility of specific immuno-PET imaging of human TNFα was assessed in a transgenic mouse model of RA that expresses human TNFα. This model resembles the progression of RA in humans by maintaining lower levels of circulating hTNFα and exhibits chronic arthritis in the forepaw and hind paw joints. The dosimetry of [89Zr]DFO-CZP in humans was estimated using microPET/CT imaging in Sprague Dawley rats. RESULTS: [89Zr]DFO-CZP was isolated with radiolabeling yields of 85 ± 6 % (n = 5) and specific activities ranging from 74 to 185 MBq/mg (n = 5). Following size exclusion purification, the radiochemical purity of [89Zr]DFO-CZP was greater than 97 %. [89Zr]DFO-CZP retained high immunoreactivity with more than 95 % of the radioactivity shifted into higher molecular weight complexes. Images showed increasing uptake of the tracer in forepaw and hind paw joints with disease progression. No uptake was observed in the model previously administered with an excess amount of unmodified CZP and in normal control mice, demonstrating in vivo specific uptake of [89Zr]DFO-CZP. CONCLUSION: The feasibility of immuno-PET imaging of human TNFα with [89Zr]DFO-CZP has been demonstrated in a preclinical model of RA.


Asunto(s)
Artritis Experimental/patología , Artritis Reumatoide/patología , Certolizumab Pegol/farmacocinética , Inmunoconjugados/farmacocinética , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Radioisótopos/farmacocinética , Factor de Necrosis Tumoral alfa/metabolismo , Circonio/farmacocinética , Animales , Antirreumáticos/farmacocinética , Artritis Experimental/diagnóstico por imagen , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Artritis Reumatoide/diagnóstico por imagen , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Distribución Tisular
6.
Cancer Immunol Res ; 7(2): 306-320, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30538091

RESUMEN

Transforming growth factor ß (TGFß) is an effector of immune suppression and contributes to a permissive tumor microenvironment that compromises effective immunotherapy. We identified a correlation between TGFB1 and genes expressed by myeloid cells, but not granulocytes, in The Cancer Genome Atlas lung adenocarcinoma data, in which high TGFB1 expression was associated with poor survival. To determine whether TGFß affected cell fate decisions and lineage commitment, we studied primary cultures of CD14+ monocytes isolated from peripheral blood of healthy donors. We discovered that TGFß was a survival factor for CD14+ monocytes, which rapidly executed an apoptotic program in its absence. Continued exposure to TGFß in combination with granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin 6 (IL6) amplified HLA-DRlowCD14+CD11b+CD33+ myeloid-derived suppressor cells (MDSCs) at the expense of macrophage and dendritic cell (DC) differentiation. MDSCs generated in the presence of TGFß were more effective in suppressing T-cell proliferation and promoted the T regulatory cell phenotype. In contrast, inhibition of TGFß signaling using a small-molecule inhibitor of receptor kinase activity in CD14+ monocytes treated with GM-CSF and IL6 decreased MDSC differentiation and increased differentiation to proinflammatory macrophages and antigen-presenting DCs. The effect of autocrine and paracrine TGFß on myeloid cell survival and lineage commitment suggests that pharmacologic inhibition of TGFß-dependent signaling in cancer would favor antitumor immunity.


Asunto(s)
Comunicación Autocrina , Diferenciación Celular/inmunología , Inmunomodulación , Monocitos/inmunología , Monocitos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adenocarcinoma del Pulmón/etiología , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/mortalidad , Adenocarcinoma del Pulmón/patología , Presentación de Antígeno/inmunología , Biomarcadores , Supervivencia Celular/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Humanos , Monocitos/citología , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1/metabolismo
7.
PLoS One ; 10(3): e0119707, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25803850

RESUMEN

Glioblastoma (GBM) is the most prevalent adult brain tumor, with virtually no cure, and with a median overall survival of 15 months from diagnosis despite of the treatment. SNARE proteins mediate membrane fusion events in cells and are essential for many cellular processes including exocytosis and neurotransmission, intracellular trafficking and cell migration. Here we show that the blockade of the SNARE protein Syntaxin 1 (Stx1) function impairs GBM cell proliferation. We show that Stx1 loss-of-function in GBM cells, through ShRNA lentiviral transduction, a Stx1 dominant negative and botulinum toxins, dramatically reduces the growth of GBM after grafting U373 cells into the brain of immune compromised mice. Interestingly, Stx1 role on GBM progression may not be restricted just to cell proliferation since the blockade of Stx1 also reduces in vitro GBM cell invasiveness suggesting a role in several processes relevant for tumor progression. Altogether, our findings indicate that the blockade of SNARE proteins may represent a novel therapeutic tool against GBM.


Asunto(s)
Toxinas Botulínicas/farmacología , Proliferación Celular/efectos de los fármacos , Glioblastoma/fisiopatología , ARN Interferente Pequeño/farmacología , Sintaxina 1/antagonistas & inhibidores , Animales , Western Blotting , Bromodesoxiuridina , Línea Celular Tumoral , Citometría de Flujo , Glioblastoma/tratamiento farmacológico , Humanos , Lentivirus , Ratones , Invasividad Neoplásica/prevención & control , ARN Interferente Pequeño/genética , Estadísticas no Paramétricas , Transducción Genética/métodos
8.
Cancer Discov ; 4(10): 1230-41, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25084773

RESUMEN

UNLABELLED: In advanced cancer, including glioblastoma, the TGFß pathway acts as an oncogenic factor. Some tumors exhibit aberrantly high TGFß activity, and the mechanisms underlying this phenomenon are not well understood. We have observed that TGFß can induce TGFß2, generating an autocrine loop leading to aberrantly high levels of TGFß2. We identified cAMP-responsive element-binding protein 1 (CREB1) as the critical mediator of the induction of TGFß2 by TGFß. CREB1 binds to the TGFB2 gene promoter in cooperation with SMAD3 and is required for TGFß to activate transcription. Moreover, the PI3K-AKT and RSK pathways regulate the TGFß2 autocrine loop through CREB1. The levels of CREB1 and active phosphorylated CREB1 correlate with TGFß2 in glioblastoma. In addition, using patient-derived in vivo models of glioblastoma, we found that CREB1 levels determine the expression of TGFß2. Our results show that CREB1 can be considered a biomarker to stratify patients for anti-TGFß treatments and a therapeutic target in glioblastoma. SIGNIFICANCE: TGFß is considered a promising therapeutic target, and several clinical trials using TGFß inhibitors are generating encouraging results. Here, we discerned the molecular mechanisms responsible for the aberrantly high levels of TGFß2 found in certain tumors, and we propose biomarkers to predict the clinical response to anti-TGFß therapies.


Asunto(s)
Comunicación Autocrina , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Glioblastoma/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/mortalidad , Glioblastoma/patología , Xenoinjertos , Humanos , Ratones , Datos de Secuencia Molecular , Motivos de Nucleótidos , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Activación Transcripcional , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/química , Factor de Crecimiento Transformador beta2/genética
9.
Nat Commun ; 5: 4632, 2014 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-25130259

RESUMEN

Gliomas are the most common primary tumours affecting the adult central nervous system and respond poorly to standard therapy. Myc is causally implicated in most human tumours and the majority of glioblastomas have elevated Myc levels. Using the Myc dominant negative Omomyc, we previously showed that Myc inhibition is a promising strategy for cancer therapy. Here, we preclinically validate Myc inhibition as a therapeutic strategy in mouse and human glioma, using a mouse model of spontaneous multifocal invasive astrocytoma and its derived neuroprogenitors, human glioblastoma cell lines, and patient-derived tumours both in vitro and in orthotopic xenografts. Across all these experimental models we find that Myc inhibition reduces proliferation, increases apoptosis and remarkably, elicits the formation of multinucleated cells that then arrest or die by mitotic catastrophe, revealing a new role for Myc in the proficient division of glioma cells.


Asunto(s)
Astrocitoma/patología , Neoplasias Encefálicas/patología , Glioblastoma/patología , Glioma/patología , Mitosis/fisiología , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Apoptosis/fisiología , Astrocitoma/fisiopatología , Astrocitoma/terapia , Neoplasias Encefálicas/fisiopatología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Glioblastoma/fisiopatología , Glioblastoma/terapia , Glioma/fisiopatología , Glioma/terapia , Xenoinjertos , Humanos , Ratones , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Enzimas Activadoras de Ubiquitina/fisiología
10.
Nat Med ; 18(3): 429-35, 2012 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-22344298

RESUMEN

In advanced cancer, including glioblastoma, the transforming growth factor ß (TGF-ß) pathway acts as an oncogenic factor and is considered to be a therapeutic target. Using a functional RNAi screen, we identified the deubiquitinating enzyme ubiquitin-specific peptidase 15 (USP15) as a key component of the TGF-ß signaling pathway. USP15 binds to the SMAD7-SMAD specific E3 ubiquitin protein ligase 2 (SMURF2) complex and deubiquitinates and stabilizes type I TGF-ß receptor (TßR-I), leading to an enhanced TGF-ß signal. High expression of USP15 correlates with high TGF-ß activity, and the USP15 gene is found amplified in glioblastoma, breast and ovarian cancer. USP15 amplification confers poor prognosis in individuals with glioblastoma. Downregulation or inhibition of USP15 in a patient-derived orthotopic mouse model of glioblastoma decreases TGF-ß activity. Moreover, depletion of USP15 decreases the oncogenic capacity of patient-derived glioma-initiating cells due to the repression of TGF-ß signaling. Our results show that USP15 regulates the TGF-ß pathway and is a key factor in glioblastoma pathogenesis.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Transformación Celular Neoplásica/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Glioblastoma/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Células HEK293 , Humanos , Imagen por Resonancia Magnética , Ratones , Fosforilación , Pronóstico , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/genética , Ubiquitina , Ubiquitina-Proteína Ligasas/metabolismo , Proteasas Ubiquitina-Específicas
11.
Cancer Cell ; 18(6): 655-68, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21156287

RESUMEN

Glioma-initiating cells (GICs), also called glioma stem cells, are responsible for tumor initiation, relapse, and therapeutic resistance. Here, we show that TGF-ß inhibitors, currently under clinical development, target the GIC compartment in human glioblastoma (GBM) patients. Using patient-derived specimens, we have determined the gene responses to TGF-ß inhibition, which include inhibitors of DNA-binding protein (Id)-1 and -3 transcription factors. We have identified a cell population enriched for GICs that expresses high levels of CD44 and Id1 and tend to be located in a perivascular niche. The inhibition of the TGF-ß pathway decreases the CD44(high)/Id1(high) GIC population through the repression of Id1 and Id3 levels, therefore inhibiting the capacity of cells to initiate tumors. High CD44 and Id1 levels confer poor prognosis in GBM patients.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Receptores de Hialuranos/análisis , Proteína 1 Inhibidora de la Diferenciación/análisis , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , Glioblastoma/química , Glioblastoma/patología , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/genética , Ratones , Ratones SCID , Proteínas de Neoplasias/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA