Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Bone Marrow Transplant ; 28(6): 619-21, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11607778

RESUMEN

A 54-year-old woman with peripheral T cell lymphoma in second complete remission (CR) received an autologous peripheral blood stem cell transplant (PBSCT). Antibiotic-resistant bloody diarrhea, and fever developed 110 days after transplant. Blood and stool cultures were negative. Skin rash was not observed. Barium enema and colonoscopy showed typical features of pancolonic-type ulcerative colitis (UC). Endoscopic biopsies confirmed the diagnosis of UC. Mesalazine and immunosuppressive therapy improved symptoms dramatically. We detected serum antibodies against synthetic tropomyosin (TM) peptide when UC was diagnosed. We postulate that autoimmunity including autoreactive anti-TM antibodies may be involved in the pathogenesis of UC after autologous PBSCT in this patient.


Asunto(s)
Colitis Ulcerosa/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Linfoma no Hodgkin/complicaciones , Autoanticuerpos/sangre , Autoinmunidad/inmunología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Femenino , Humanos , Linfoma no Hodgkin/terapia , Linfoma de Células T Periférico/complicaciones , Linfoma de Células T Periférico/terapia , Persona de Mediana Edad , Trasplante Autólogo/efectos adversos , Tropomiosina/inmunología
2.
Gene Ther ; 8(2): 149-56, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11313784

RESUMEN

We have previously reported that superoxide stimulates the motility of tumor cells and the administration of Cu-Zn superoxide dismutase (SOD) significantly suppresses metastasis. However, ideally, anti-metastatic therapy should be long-lasting, systemically effective and have low toxicity. The half-life of Cu-Zn SOD in plasma is so short that it cannot provide long-lasting effects. Therefore, in this study we have developed a gene therapy in a mouse model utilizing extracellular SOD (EC-SOD), which is the most prevalent SOD isoenzyme in extracellular fluids. We retrovirally transfected fibroblasts (syngeneic) with the EC-SOD gene and established EC-SOD-secreting fibroblasts. Inoculation of EC-SOD-secreting fibroblasts suppressed both artificial and spontaneous metastatic lung nodules in mouse metastasis models. These data indicate the feasibility of anti-metastatic gene therapy utilizing the EC-SOD gene.


Asunto(s)
Terapia Genética/métodos , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Superóxido Dismutasa/genética , Transducción Genética , Animales , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/secundario , Carcinoma Pulmonar de Lewis/terapia , Técnicas de Cultivo de Célula , División Celular , Medios de Cultivo , ADN Complementario/genética , Estudios de Factibilidad , Fibroblastos/trasplante , Expresión Génica , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos , Trasplante de Neoplasias , ARN Mensajero/genética , Sarcoma Experimental/patología , Sarcoma Experimental/secundario , Sarcoma Experimental/terapia , Superóxido Dismutasa/metabolismo
3.
Blood ; 97(4): 1123-30, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11159546

RESUMEN

Acute graft-versus-host diseases (GVHD) is a major cause of morbidity and mortality in patients undergoing allogeneic bone marrow transplantation (BMT). T helper 1 (Th1)-type cytokines such as interferon-gamma or tumor necrosis factor-alpha have been implicated in the pathogenesis of acute GVHD. TAK-603 is a new quinoline derivative, which is now in clinical trials for use as a disease-modifying antirheumatic drug. In preclinical studies, it inhibited delayed-type hypersensitivity, but not Arthus-type reaction, in mice, and selectively suppressed Th1 cytokine production. Thus, the present study was designed to investigate whether the Th1 inhibitor (TAK-603) ameliorates lethal acute GVHD in a mouse model. Administration of TAK-603 into BALB/c mice given 10 Gy total body irradiation followed by transplantation of bone marrow and spleen cells from C57BL/6 mice markedly reduced the mortality in association with minimal signs of GVHD pathology in the liver, intestine, and skin. TAK-603 reduced not only the production of Th1-type cytokines, but also the proportion of Th1 cells in CD4(+) helper T cells in this GVHD mouse model. These results suggest that TAK-603 could be a potent therapeutic agent for acute lethal GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Inmunosupresores/uso terapéutico , Quinolinas/uso terapéutico , Células TH1/efectos de los fármacos , Triazoles/uso terapéutico , Enfermedad Aguda , Administración Oral , Animales , Trasplante de Médula Ósea/efectos adversos , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/patología , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacología , Intestinos/patología , Hígado/patología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Quinolinas/administración & dosificación , Quinolinas/farmacología , Quimera por Radiación , Piel/patología , Bazo/trasplante , Células TH1/metabolismo , Trasplante Homólogo/efectos adversos , Triazoles/administración & dosificación , Triazoles/farmacología
4.
Int J Hyperthermia ; 14(3): 309-17, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9679710

RESUMEN

Endogenous tumour necrosis factor (enTNF) acts as a resistant factor against cytotoxicity of heat by induction of manganous superoxide dismutase (MnSOD), thereby scavenging reactive oxygen free radicals. On the other hand, it is also well known that heat shock proteins (HSPs), which are induced by heat-stress, behave as cytoprotecting factor against this stress. However, the relationship of these two resistant factors is not yet elucidated. In the present study we would therefore propose the possibility that enTNF enhances HSP72 expression. Heat-sensitive L-M (mouse tomourigenic fibroblast) cells, which normally do not express enTNF, were transfected with a nonsecretory-type human TNF expression vector to produce enTNF. Stable transfectants showed resistance to heat treatment and an increase of HSP72 expression. Conversely, when HeLa (human uterine cervical cancer) cells, which normally produce an appreciable amount of enTNF, were transfected with an antisense TNF mRNA expression vector to inhibit enTNF synthesis, their heat sensitivity was enhanced and HSP72 expression was reduced by half. In conclusion, these findings indicate that enTNF regulates heat-inducible HSP72 synthesis.


Asunto(s)
Proteínas de Choque Térmico/biosíntesis , Factor de Necrosis Tumoral alfa/fisiología , Animales , Células Cultivadas , Vectores Genéticos , Proteínas del Choque Térmico HSP72 , Células HeLa , Humanos , Hipertermia Inducida , Cinética , Ratones , ARN sin Sentido/genética , ARN Mensajero/genética , Transfección , Factor de Necrosis Tumoral alfa/genética
5.
Chemotherapy ; 43(6): 406-14, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9395854

RESUMEN

To elucidate the relationship between two distinct resistant factors, endogenous tumor necrosis factor (enTNF) and heat shock proteins (HSPs), against hyperthermia, we assessed whether enTNF enhances HSP72 expression. Although there was a variability in the sensitivity of pancreatic carcinoma cell lines to heat, enTNF and HSP72 expression as well as MnSOD activity correlated positively with heat resistance. When MIAPaCa-2 pancreatic carcinoma cells, which had the lowest enTNF expression and highest heat sensitivity, were transfected with a nonsecretory-human TNF gene (pTNF delta pro), intracellular manganous superoxide dismutase (MnSOD) activity, HSP72 expression, and heat resistance were significantly increased. Furthermore, in these cells, enTNF expression correlated with the binding activity of heat shock factor 1 (HSF 1) to an oligonucleotide containing the human heat shock element. These results indicate that enTNF participates in the intrinsic resistance against heat via induction of MnSOD, enhances HSF1-binding activity, and augments of HSP72 expression. Therefore, inhibition of enTNF expression in pancreatic carcinoma cells would improve the efficacy of hyperthermia for pancreatic carcinoma.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Hipertermia Inducida , Neoplasias Pancreáticas/terapia , Factor de Necrosis Tumoral alfa/fisiología , Proteínas del Choque Térmico HSP72 , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/biosíntesis , Humanos , Superóxido Dismutasa/metabolismo , Factores de Transcripción , Transfección , Células Tumorales Cultivadas
6.
Blood ; 89(7): 2472-9, 1997 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9116291

RESUMEN

We have previously reported that intracellular tumor necrosis factor (enTNF) is responsible for resistance, in established cell lines to doxorubicin (DOX), exogenous TNF, and heat stress by inducing manganous superoxide dismutase (MnSOD), thereby scavenging reactive oxygen free radicals. Leukemic cells from 19 patients (6 acute lymphoblastic leukemia, 13 acute myeloid leukemia) were examined for their sensitivity to DOX and TNF in relation to their enTNF expression and MnSOD activity. Sensitivity to DOX and the expression of enTNF or MnSOD activity were inversely correlated. In a case with acquired resistance to chemotherapy which included DOX, enTNF expression and MnSOD activity were increased. Furthermore, in 14 cases treated with a regimen including an anthracycline, 4 cases that failed to respond to chemotherapy showed relatively high amounts of enTNF expression. KG-1 (human acute myelogenous leukemia) cells transfected with a nonsecretory-type TNF expression vector (pTNF delta pro) showed resistance to DOX. A significant increase in MnSOD levels was also noted in the transfectants. TNF antisense cDNA was transfected into isolated leukemic cells from five patients. Sensitivity of the antisense transfectants to DOX was increased, approximately 1.4- to 2.5-fold. These results suggest that enTNF acts as a resistance factor against DOX in leukemia, and that enTNF may be useful as a predictor of DOX sensitivity in leukemia.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Leucemia/patología , Proteínas de Neoplasias/análisis , Células Madre Neoplásicas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/análisis , Enfermedad Aguda , Adulto , Anciano , Antibióticos Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores , ADN sin Sentido/farmacología , ADN Complementario/farmacología , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos , Femenino , Humanos , Leucemia/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/química , Proteínas Recombinantes de Fusión/metabolismo , Superóxido Dismutasa/análisis , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Insuficiencia del Tratamiento , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
7.
Cancer Res ; 52(22): 6258-62, 1992 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-1423271

RESUMEN

One of the mechanisms of cytotoxicity by tumor necrosis factor (TNF) is the induction of reactive oxygen molecules. Cells producing endogenous tumor necrosis factor (enTNF) show resistance to the cytotoxicity of exogenous TNF by scavenging the reactive oxygen molecules. The intracellular hydroxyl radical production is also known to be involved in the heat-induced cytotoxicity. In the present study, we therefore examined the possibility that enTNF may act as a protective protein against the heat-induced cytotoxicity in a manner similar to that of exogenous TNF. Heat-sensitive L-M (mouse tumorigenic fibroblast) cells, originally expressing no enTNF, were transfected with a human TNF expression vector to produce enTNF. The stable transfectants showed apparent resistance to heat treatment. Conversely, when HeLa (human uterine cervical cancer) cells, originally producing an appreciable amount of enTNF, were transfected with an antisense TNF mRNA expression vector to inhibit enTNF synthesis, their heat sensitivity was enhanced. Furthermore, L-M cells which were transfected with nonsecretory human TNF expression vector also acquired resistance to heat treatment. In these cells, heat resistance correlated well with expression of enTNF and intracellular levels of manganous superoxide dismutase. These results indicate that enTNF exerts its intracellular protective effect against the heat-induced cytotoxicity by scavenging reactive oxygen with induced manganous superoxide dismutase in a manner similar to that found in cells treated with exogenous TNF.


Asunto(s)
Hipertermia Inducida , Factor de Necrosis Tumoral alfa/fisiología , Animales , Células Cultivadas , Fibroblastos/citología , Vectores Genéticos/genética , Células HeLa , Calor , Humanos , Ratones , ARN sin Sentido/genética , ARN Mensajero/genética , Sensibilidad y Especificidad , Superóxido Dismutasa/metabolismo , Transfección , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología
8.
Jpn J Cancer Res ; 83(5): 540-5, 1992 May.
Artículo en Inglés | MEDLINE | ID: mdl-1319987

RESUMEN

We previously reported that recombinant human tumor necrosis factor (rhTNF) and hyperthermia had a synergistic effect against tumors, in vitro and in vivo. We have now investigated the mechanism of this synergy by measuring the lysosomal enzyme activity and hydroxyl radical production of L-M cells treated with rhTNF and/or hyperthermia. A synergistic activation of lysosomal enzyme and the induction of hydroxyl radical production in L-M cells treated with both rhTNF and hyperthermia was observed. A synergistic cytotoxic effect was observed when rhTNF and hyperthermia were combined, and was inhibited by the addition of a reactive oxygen scavenger, dimethyl sulfoxide or bipyridine. The results show that the augmenting effect of hyperthermia on lysosomal enzyme activation and induction of hydroxyl radical production by rhTNF plays an important role in the synergistic cytotoxic effect.


Asunto(s)
Hipertermia Inducida , Neoplasias Experimentales/terapia , Factor de Necrosis Tumoral alfa/farmacología , Fosfatasa Ácida/efectos de los fármacos , Fosfatasa Ácida/metabolismo , Animales , Terapia Combinada , Esquema de Medicación , Sinergismo Farmacológico , Glucuronidasa/efectos de los fármacos , Glucuronidasa/metabolismo , Humanos , Hidróxidos/metabolismo , Radical Hidroxilo , Lisosomas/efectos de los fármacos , Lisosomas/enzimología , Ratones , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Proteínas Recombinantes/farmacología , Estimulación Química , Células Tumorales Cultivadas
9.
Gastroenterol Jpn ; 26(6): 774-8, 1991 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1722471

RESUMEN

A case of unresectable hepatocellular carcinoma which responded favorably to combined therapy with tumor necrosis factor (TNF) and local hyperthermia is reported. A 58-year-old man was admitted to our hospital in June 1988 for treatment of hepatocellular carcinoma affecting S4 and S8. After three sessions of transcatheter arterial embolization (TAE) therapy, the serum alpha 1-fetoprotein level decreased, and a reduction in the size of the lesions was also noted. Thereafter, the patient received local hyperthermia once a week (60 minutes of irradiation from a Thermotron-RF8 at 1,100W), but the alpha 1-fetoprotein level increased again in February 1989. On examination, enlargement of the S8 lesion and a new nodule in S7 were recognized. Since TAE was contraindicated due to liver dysfunction, human recombinant TNF (1 x 10(6)U) was given by intravenous infusion together with local hyperthermia once a week. Eight sessions of the combined therapy reduced the serum alpha 1-fetoprotein level markedly (7,512.0 to 2,782.0 pg/ml) and after eighteen sessions, 58.1% regression of tumor size (partial response) on computed tomography scans was observed. This anecdotal case supports previous experimental evidence suggesting that TNF plus hyperthermia may be effective for treating unresectable hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/terapia , Hipertermia Inducida , Neoplasias Hepáticas/terapia , Factor de Necrosis Tumoral alfa/uso terapéutico , Quimioembolización Terapéutica , Terapia Combinada , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico , alfa-Fetoproteínas/análisis
10.
Cancer Res ; 48(3): 654-7, 1988 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-2825981

RESUMEN

Synergy in cytotoxic effect between recombinant human tumor necrosis factor and hyperthermia (incubation at 38.5 degrees C or 40 degrees C) was observed to occur against L-M (mouse tumorigenic fibroblast) cells and shown to be related to an accelerated turnover rate of recombinant human tumor necrosis factor-receptor complex under elevated temperatures rather than to changes in number of cell receptors or binding strength. However, no synergy in cytotoxic effect was observed to occur against human embryonic lung (HEL) cells. A clearly synergistic inhibition of metastatic tumor growth by combined administration of recombinant human tumor necrosis factor (300 units) and whole-body hyperthermia (40 degrees C, 30 min) was also observed in BALB/c mice previously given injections of 1 x 10(6) Meth-A (MH) cells/mouse via tail vein, neither of which alone resulted in significant inhibition.


Asunto(s)
Hipertermia Inducida , Neoplasias Experimentales/terapia , Células Tumorales Cultivadas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Neoplasias Pulmonares/secundario , Ratones , Metástasis de la Neoplasia , Neoplasias Experimentales/tratamiento farmacológico , Receptores de Superficie Celular/metabolismo , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Factores de Tiempo , Factor de Necrosis Tumoral alfa/farmacología
11.
Cancer Res ; 48(3): 650-3, 1988 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-3335027

RESUMEN

A synergistic increase in the cytotoxic effects of recombinant human tumor necrosis factor (rH-TNF) and hyperthermia was demonstrated both in vitro and in vivo. The cytotoxicity of rH-TNF against L-M cells in incubation for 12 h at 38.5 and 40 degrees C based on the concentration necessary for 50% cytotoxicity was, respectively, 125 and more than 500 times as high as in similar incubation at 37 degrees C. As observed 18 days after implantation of Meth-A fibrosarcoma cells in mice, single i.v. administration of rH-TNF at 1000 units/mouse resulted in complete cures in five mice when performed in combination with hyperthermia (40 degrees C), whereas rH-TNF alone in the same dose resulted in 27.1% inhibition of tumor growth and hyperthermia alone had no appreciable effect on tumor growth. The i.v. administration of rH-TNF three times at 100 or 300 units/mouse together with hyperthermia (40 degrees C) resulted in 41.2 and 89.0% tumor growth inhibition, respectively; similar administration without hyperthermia appeared to have little or no appreciable effect on tumor growth. The results suggest that combination therapy including rH-TNF and hyperthermia may be of value in the treatment of malignancy in human patients.


Asunto(s)
Hipertermia Inducida , Neoplasias Experimentales/terapia , Células Tumorales Cultivadas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Supervivencia Celular/efectos de los fármacos , Esquema de Medicación , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Factores de Tiempo , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/farmacología
12.
Blood ; 70(6): 1955-8, 1987 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3676519

RESUMEN

Serum transferrin receptors were measured by a sandwich radioimmunoassay procedure in patients with iron deficiency anemia, autoimmune hemolytic anemia and aplastic anemia. The mean circulating transferrin receptor concentration of normal subjects and patients with iron deficiency anemia, autoimmune hemolytic anemia and aplastic anemia are 253 +/- 82 ng/mL, 730 +/- 391 ng/mL, 1,426 +/- 1,079 ng/mL, and 182 +/- 39 ng/mL, respectively. The values for those with iron deficiency anemia and autoimmune hemolytic anemia were significantly higher than that of normal controls and the values for those with aplastic anemia were lower than that of normal controls. After iron supplementation in iron deficiency anemia, the serum transferrin receptor values increased twofold over those of pretreatment values. This increase parallels an increase in peripheral reticulocytes. Therefore, the number of circulating transferrin receptors in anemic patients may reflect the level of bone marrow erythropoiesis and is a potentially useful new index for red cell production.


Asunto(s)
Anemia/sangre , Eritropoyesis , Receptores de Transferrina/sangre , Transferrina/metabolismo , Anemia Aplásica/sangre , Anemia Hemolítica Autoinmune/sangre , Anemia Hipocrómica/sangre , Humanos , Reticulocitos/fisiología
13.
Gan To Kagaku Ryoho ; 14(6 Pt 2): 2089-97, 1987 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-3038032

RESUMEN

TNF is cytokine derived from macrophages and shows much promise for use in cancer therapy because of its marked antitumor effects and its high specificity to tumors. The clinical application (Phase I-II) of TNF has been started because human recombinant TNF (rH-TNF) can be produced on a large scale. In spite of notable antitumor effects, little is known concerning the mechanism of its cytotoxic action. In this article, the antitumor effects of rH-TNF against human and murine tumors, the mechanism of its action and the synergistic effects of rH-TNF in combination with IFN-gamma, various anticancer drugs or with hyperthermia are reviewed.


Asunto(s)
Glicoproteínas/uso terapéutico , Neoplasias/terapia , Línea Celular , Terapia Combinada , Glicoproteínas/farmacología , Humanos , Hipertermia Inducida , Neoplasias Pulmonares/terapia , Miosarcoma/terapia , Neoplasias/metabolismo , Neoplasias/patología , ARN Neoplásico/biosíntesis , Receptores de Superficie Celular/análisis , Receptores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa
14.
Gan To Kagaku Ryoho ; 14(5 Pt 1): 1286-91, 1987 May.
Artículo en Japonés | MEDLINE | ID: mdl-3579327

RESUMEN

Synergistic effects of hyperthermia on the cytotoxicity of TNF were examined, using L-M cells (mouse tumorigenic fibroblasts) and HEL cells (human embryonic lung cells) as targets. The sensitivity of L-M cells to TNF apparently increased when the culture temperature was raised from 37 degrees C to 38.5 degrees C or 40 degrees C, probably reflecting the increased turnover rate of the TNF-receptor complex. The anti-metastatic effects of a combined regimen of TNF (300U) and whole-body hyperthermia (40 degrees C, 30 min) was also examined in Balb/c mice injected with 1 X 10(6) Meth A F15 cells into a tail vein. No effects of TNF or hyperthermia alone were observed on artificial metastasis, while the combination of TNF and hyperthermia remarkably enhanced the inhibition of metastasis. These studies clearly suggest that combination of TNF with hyperthermia provides a significant synergistic effect against tumor.


Asunto(s)
Glicoproteínas/uso terapéutico , Hipertermia Inducida , Neoplasias Pulmonares/patología , Sarcoma Experimental/patología , Animales , Células Cultivadas , Terapia Combinada , Fibrosarcoma/patología , Glicoproteínas/farmacología , Humanos , Neoplasias Pulmonares/terapia , Ratones , Sarcoma Experimental/terapia , Factor de Necrosis Tumoral alfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA