Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Vaccine ; 40(33): 4682-4685, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35840470

RESUMEN

Previous studies reporting the response to SARS-CoV-2 mRNA vaccination in alloHSCT recipients used serological and/or cellular assays, but no study has evaluated vaccine-induced neutralizing antibodies. We prospectively studied 28 alloHSCT recipients who received two BNT162b2 doses. Two patients groups were defined according to time from alloHSCT and immunosuppressive treatment, and had different baseline immunologic status. Study end-point was the evaluation of humoral and cellular responses one month after the second vaccine. All patients seroconverted. Anti-S IgG levels and neutralizing antibodies percentages were not significantly different between both groups. Using IFNγ ELISpot assay, five patients showed a strong increase, without correlation with the humoral response. Using flow cytometry lymphocyte proliferation assay, 14 patients exhibited responding T cells, without difference between both groups or correlation with anti-S IgG levels. A few low serological responders had a detectable CD4 + T cell proliferative response. This finding should be confirmed in a larger cohort.


Asunto(s)
COVID-19 , Trasplante de Células Madre Hematopoyéticas , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacuna BNT162 , COVID-19/prevención & control , Humanos , Inmunidad Humoral , Inmunoglobulina G , SARS-CoV-2 , Vacunación
2.
Blood ; 139(12): 1820-1832, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-34905599

RESUMEN

Cutaneous T-cell lymphomas (CTCLs) are rare malignancies involving primarily the skin. Responses to treatment are usually short-lived in advanced CTCL. The determinants of long-term CTCL control are unclear. Mogamulizumab, an anti-human CCR4 antibody that acts by antibody-dependent cell cytotoxicity against CCR4+ CTCL tumor cells and peripheral memory blood regulatory T cells, has been associated with long-lasting remissions and immune adverse events. Here, we reported skin rashes in 32% of 44 patients with CTCL treated with mogamulizumab, associated with significantly higher overall survival (hazard ratio, 0.16; 0.04-0.73; P = .01). Rash occurred in patients with Sézary syndrome and was associated with longer time to progression. These rashes were characterized by a CD163+ granulomatous and/or CD8+ lichenoid skin infiltrate. High-throughput sequencing analysis of T-cell receptor ß genes in skin and blood flow cytometry confirmed the depletion of CTCL tumor cells, as well as the recruitment of new reactive T-cell clones in skin at the time of skin rash. CXCL9 and CXCL11, two macrophage-derived chemokines that recruit CXCR3+ T cells to skin, were overexpressed in skin rashes. A higher frequency of TIGIT+ and PD1+ exhausted reactive blood T cells was observed at baseline in patients with rash, and this frequency decreased with mogamulizumab treatment. These data are consistent with mogamulizumab-induced long-term immune CTCL control by activation of the macrophage and T-cell responses in patients with rash.


Asunto(s)
Exantema , Linfoma Cutáneo de Células T , Neoplasias Cutáneas , Anticuerpos Monoclonales Humanizados , Quimiocina CXCL11 , Quimiocina CXCL9 , Exantema/inducido químicamente , Humanos , Linfoma Cutáneo de Células T/tratamiento farmacológico , Linfoma Cutáneo de Células T/patología , Macrófagos/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Linfocitos T Reguladores
3.
Front Immunol ; 11: 579776, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329550

RESUMEN

Mechanisms driving acute graft-versus-host disease (aGVHD) onset in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are still poorly understood. To provide a detailed characterization of tissue-infiltrating T lymphocytes (TL) and search for eventual site-specific specificities, we developed a xenogeneic model of aGVHD in immunodeficient mice. Phenotypic characterization of xenoreactive T lymphocytes (TL) in diseased mice disclosed a massive infiltration of GVHD target organs by an original CD4+CD8+ TL subset. Immunophenotypic and transcriptional profiling shows that CD4+CD8+ TL comprise a major PD1+CD62L-/+ transitional memory subset (>60%) characterized by low level expression of cytotoxicity-related transcripts. CD4+CD8+ TL produce high IL-10 and IL-13 levels, and low IL-2 and IFN-γ, suggestive of regulatory function. In vivo tracking of genetically labeled CD4+ or CD8+ TL subsequently found that CD4+CD8+ TL mainly originate from chronically activated cytotoxic TL (CTL). On the other hand, phenotypic profiling of CD3+ TL from blood, duodenum or rectal mucosa in a cohort of allo-HSCT patients failed to disclose abnormal expansion of CD4+CD8+ TL independent of aGVHD development. Collectively, our results show that acquisition of surface CD4 by xenoreactive CD8+ CTL is associated with functional diversion toward a regulatory phenotype, but rule out a central role of this subset in the pathogenesis of aGVHD in allo-HSCT patients.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Linfocitos T Citotóxicos/inmunología , Animales , Citocinas/metabolismo , Femenino , Humanos , Memoria Inmunológica , Masculino , Ratones , Ratones SCID , Receptor de Muerte Celular Programada 1/metabolismo , Trasplante Heterólogo
5.
Proc Natl Acad Sci U S A ; 116(13): 6298-6307, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30846549

RESUMEN

Natural regulatory T cells (nTregs) ensure the control of self-tolerance and are currently used in clinical trials to alleviate autoimmune diseases and graft-versus-host disease after hematopoietic stem cell transfer. Based on CD39/CD26 markers, blood nTreg analysis revealed the presence of five different cell subsets, each representing a distinct stage of maturation. Ex vivo added microenvironmental factors, including IL-2, TGFß, and PGE2, direct the conversion from naive precursor to immature memory and finally from immature to mature memory cells, the latest being a no-return stage. Phenotypic and genetic characteristics of the subsets illustrate the structural parental maturation between subsets, which further correlates with the expression of regulatory factors. Regarding nTreg functional plasticity, both maturation stage and microenvironmental cytokines condition nTreg activities, which include blockade of autoreactive immune cells by cell-cell contact, Th17 and IL-10 Tr1-like activities, or activation of TCR-stimulating dendritic cell tolerization. Importantly, blood nTreg CD39/CD26 profile remained constant over a 2-y period in healthy persons but varied from person to person. Preliminary data on patients with autoimmune diseases or acute myelogenous leukemia illustrate the potential use of the nTreg CD39/CD26 profile as a blood biomarker to monitor chronic inflammatory diseases. Finally, we confirmed that naive conventional CD4 T cells, TCR-stimulated under a tolerogenic conditioned medium, could be ex vivo reprogrammed to FOXP3 lineage Tregs, and further found that these cells were exclusively committed to suppressive function under all microenvironmental contexts.


Asunto(s)
Microambiente Celular/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/fisiología , Apirasa/sangre , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Dinoprostona/metabolismo , Dipeptidil Peptidasa 4/sangre , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Leucemia Mieloide , Células Th17/inmunología , Factor de Crecimiento Transformador beta/metabolismo
6.
J Immunol Res ; 2019: 9141420, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30766892

RESUMEN

Dermatomyositis pathophysiology is complex. In recent years, medical research has identified molecules associated with disease activity. Besides providing insights into the driving mechanisms of dermatomyositis, these findings could provide potential biomarkers. Activity markers can be used to monitor disease activity in clinical trials and may also be useful in daily practice. This article reviews molecules that could be used as biomarkers for diagnosis and monitoring dermatomyositis disease activity.


Asunto(s)
Dermatomiositis/diagnóstico , Neoplasias/diagnóstico , Piel/efectos de los fármacos , Adulto , Biomarcadores/metabolismo , Humanos , Pronóstico , Piel/patología
9.
Blood ; 125(11): 1830-9, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25605369

RESUMEN

Interleukin 10 (IL-10)-producing B cells (regulatory B cells [Bregs]) regulate autoimmunity in mice and humans, and a regulatory role of IL-10-producing plasma cells has been described in mice. Dysfunction of B cells that maintain homeostasis may play a role in the pathogenesis of chronic graft-versus-host disease (cGVHD) after allogeneic stem cell transplantation. Here, we found a relation between decreased Breg frequencies and cGVHD severity. An impaired ability of B cells to produce IL-10, possibly linked to poor signal transducer and activator of transcription 3 and extracellular signal-regulated kinase phosphorylation, was found in patients with active cGVHD. IL-10 production was not confined to a single B-cell subset, but enriched in both the CD24(hi)CD27(+) and CD27(hi)CD38(hi) plasmablast B-cell compartments. In vitro plasmablast differentiation increased the frequency of IL-10-producing B cells. We confirmed that allogeneic transplant recipients had an impaired reconstitution of the memory B-cell pool. cGVHD patients had less CD24(hi)CD27(+) B cells and IL-10-producing CD24(hi)CD27(+) B cells. Patients with cGVHD had increased plasmablast frequencies but decreased IL-10-producing plasmablasts. These results suggest a role of CD24(hi)CD27(+) B-cell and plasmablast-derived IL-10 in the regulation of human cGVHD.


Asunto(s)
Linfocitos B Reguladores/inmunología , Antígeno CD24/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Adulto , Anciano , Animales , Linfocitos B Reguladores/metabolismo , Linfocitos B Reguladores/patología , Diferenciación Celular , Enfermedad Crónica , Femenino , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Interleucina-10/biosíntesis , Sistema de Señalización de MAP Quinasas , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Persona de Mediana Edad , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Estudios Prospectivos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Adulto Joven
10.
Methods Mol Biol ; 1190: 45-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25015272

RESUMEN

The analysis of human B cell populations of the blood relies on the expression of surface markers, mainly CD19, CD24, CD38, and CD27. According to these surface markers, three main B cell subsets can be identified in the blood: immature transitional B cells (CD19(+)CD24(high)CD38(high)), naïve B cells (CD19(+)CD24(int)CD38(int)) that have not encountered an antigen, and memory B cells (CD19(+)CD27(+)). To date, human B cells with regulatory functions have been essentially described within the CD24(high)CD38(high) transitional B cell subset. CD24(high)CD38(high) transitional B cells are able to produce interleukin 10 (IL-10) and to regulate in vitro Th1 and Th17 CD4(+) T cell activation. Here, we provide the methods to analyze and purify the CD24(high)CD38(high) transitional B cell subset for further in vitro experiments. We also provide a reliable method to detect B cell IL-10 production using intracellular cytokine staining.


Asunto(s)
ADP-Ribosil Ciclasa 1/análisis , Linfocitos B Reguladores/citología , Linfocitos B Reguladores/inmunología , Antígeno CD24/análisis , Inmunofenotipificación/métodos , ADP-Ribosil Ciclasa 1/inmunología , Antígeno CD24/inmunología , Separación Celular/métodos , Citometría de Flujo/métodos , Humanos , Interleucina-10/análisis , Interleucina-10/inmunología
11.
J Clin Invest ; 123(7): 2873-92, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23778140

RESUMEN

CD4⁺ T cells are critical regulators of immune responses, but their functional role in human breast cancer is relatively unknown. The goal of this study was to produce an image of CD4⁺ T cells infiltrating breast tumors using limited ex vivo manipulation to better understand the in vivo differences associated with patient prognosis. We performed comprehensive molecular profiling of infiltrating CD4⁺ T cells isolated from untreated invasive primary tumors and found that the infiltrating T cell subpopulations included follicular helper T (Tfh) cells, which have not previously been found in solid tumors, as well as Th1, Th2, and Th17 effector memory cells and Tregs. T cell signaling pathway alterations included a mixture of activation and suppression characterized by restricted cytokine/chemokine production, which inversely paralleled lymphoid infiltration levels and could be reproduced in activated donor CD4⁺ T cells treated with primary tumor supernatant. A comparison of extensively versus minimally infiltrated tumors showed that CXCL13-producing CD4⁺ Tfh cells distinguish extensive immune infiltrates, principally located in tertiary lymphoid structure germinal centers. An 8-gene Tfh signature, signifying organized antitumor immunity, robustly predicted survival or preoperative response to chemotherapy. Our identification of CD4⁺ Tfh cells in breast cancer suggests that they are an important immune element whose presence in the tumor is a prognostic factor.


Asunto(s)
Neoplasias de la Mama/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Antígenos CD/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Citocinas/genética , Citocinas/metabolismo , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transcriptoma
12.
Adv Immunol ; 115: 187-227, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22608260

RESUMEN

Anticytokine (AC) immune therapies derived from vaccine procedures aim at enhancing natural immune defense mechanisms ineffective to contain abnormally produced cytokines and counteract their pathogenic effects. Given their short half-life, cytokines, the production of which by effector immune cells (T and B lymphocytes, antigen-presenting cells (APCs), natural killer (NK) and endothelial cells) is inducible and controlled by negative feedback regulation, (1) exert locally their signaling to paracrine/autocrine target responder cells carrying high-affinity membrane receptors and (2) are commonly present at minimal concentration in the body fluid (lymph, serum). Aberrant signaling triggered by cytokines, uncontrolly released by effector immune cells or produced by cancer and other pathologic cells, contribute to the pathogenesis of chronic diseases including cancer, viral infections, allergy, and autoimmunity. To block these ectopic cytokine signaling and prevent their pathogenic effects, AC Abs supplied either by injections (passive AC immune therapy) or elicited by immunization with cytokine-derived immunogenes called Kinoids (active AC immune therapy) proved to be experimentally effective and safe. In this review, we detailed the rationale and the requirements for the use of AC immunotherapies in humans, the proof of efficacy of these medications in animal disease models, and their current clinical development and outcome, including adverse side effects they may generate. We particularly show that, to date, the benefit:risk ratio of AC immune therapies is highly positive.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Citocinas/antagonistas & inhibidores , Hipersensibilidad/terapia , Inmunización Pasiva/métodos , Inmunoterapia Activa/métodos , Neoplasias/terapia , Animales , Anticuerpos Neutralizantes/administración & dosificación , Comunicación Celular , Ensayos Clínicos como Asunto , Citocinas/inmunología , Modelos Animales de Enfermedad , Semivida , Humanos , Hipersensibilidad/inmunología , Neoplasias/inmunología , Medición de Riesgo , Resultado del Tratamiento
13.
Proc Natl Acad Sci U S A ; 108(47): 18995-9000, 2011 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-22065791

RESUMEN

Immune suppressive activities exerted by regulatory T-cell subsets have several specific functions, including self-tolerance and regulation of adaptive immune reactions, and their dysfunction can lead to autoimmune diseases and contribute to AIDS and cancer. Two functionally distinct regulatory T-cell subsets are currently identified in peripheral tissues: thymus-developed natural T regulatory cells (nTregs) controlling self-tolerance and antiinflammatory IL-10-secreting type 1 regulatory T cells (Tr1) derived from Ag-stimulated T cells, which regulate inflammation-dependent adaptive immunity and minimize immunopathology. We establish herein that cell contact-mediated nTreg regulatory function is inhibited by inflammation, especially in the presence of the complement C3b receptor (CD46). Instead, as with other T-cell subsets, the latter inflammatory conditions of stimulation skew nTreg differentiation to Tr1 cells secreting IL-10, an effect potentiated by IFN-α. The clinical relevance of these findings was verified in a study of 152 lupus patients, in which we showed that lupus nTreg dysfunction is not due to intrinsic defects but is rather induced by C3b stimulation of CD46 and IFN-α and that these immune components of inflammation are directly associated with active lupus. These results provide a rationale for using anti-IFN-α Ab immunotherapy in lupus patients.


Asunto(s)
Diferenciación Celular/inmunología , Activación de Complemento/inmunología , Inmunoterapia/métodos , Interferón-alfa/metabolismo , Lupus Eritematoso Sistémico/inmunología , Proteína Cofactora de Membrana/metabolismo , Linfocitos T Reguladores/inmunología , Anticuerpos Monoclonales , Complemento C3b/inmunología , Cartilla de ADN/genética , Citometría de Flujo , Humanos , Interferón-alfa/inmunología , Interleucina-10/inmunología , Leucocitos Mononucleares , Modelos Lineales , Proteína Cofactora de Membrana/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Proc Natl Acad Sci U S A ; 106(13): 5294-9, 2009 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19279210

RESUMEN

A major involvement of IFNalpha in the etiopathogenesis of systemic lupus erythematosus has been suggested by clinical observations, including the increase of serum levels of this cytokine in patients with active disease. Supporting this hypothesis, we have shown that expression of IFNalpha from a recombinant adenovirus (IFNalpha Adv) precipitates lupus manifestations in genetically susceptible New Zealand Black (NZB) x New Zealand White (NZW)F(1) mice (NZB/W) but not in BALB/c mice. In the present investigation, we have prepared an IFNalpha immunogen, termed IFNalpha kinoid, which, appropriately adjuvanted, induces transient neutralizing antibodies (Abs) but no cellular immune response to the cytokine and without apparent side effects. Using this preparation, we also showed that, in kinoid-vaccinated NZB/W mice, lupus manifestations, including proteinuria, histological renal lesions, and death triggered by IFNalpha Adv challenge were delayed/prevented as long as an effective threshold of anti-IFNalpha inhibitory capacity was present in the serum.


Asunto(s)
Formación de Anticuerpos , Interferón-alfa/inmunología , Lupus Eritematoso Sistémico/terapia , Vacunas/uso terapéutico , Animales , Anticuerpos , Muerte , Modelos Animales de Enfermedad , Enfermedades Renales , Ratones , Ratones Endogámicos , Proteinuria , Especificidad de la Especie , Resultado del Tratamiento , Vacunas/inmunología
15.
Proc Natl Acad Sci U S A ; 104(8): 2837-42, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17301234

RESUMEN

Tumor growth depends on blood supply, requiring the development of new vessels, and vascular endothelial growth factor (VEGF) plays a central role in neoangiogenic processes. For this reason, VEGF represents a target for the development of new therapeutic antiangiogenic molecules. Clinical trials using anti-VEGF mAbs such as bevacizumab have validated the efficacy of this therapeutic approach but have also revealed adverse effects. Here we report that a VEGF-derived immunogen, consisting of a heterocomplex of a murine (m)VEGF and keyhole limpet hemocyanin, called "mVEGF kinoid," triggered a strong Ab immune response in mice. The anti-VEGF Abs inhibited both the proliferation of human umbilical vein endothelial cells cultured in the presence of mVEGF and the binding of mVEGF to its receptor-2 Flk-1. In mVEGF kinoid-immunized BALB/c mice challenged with syngeneic CT26 colorectal tumor cells, the number and size of lung metastases were significantly decreased. In human (h)VEGF kinoid-immunized BALB/c mice, high levels of serum Abs to hVEGF were present, and purified IgG from these mice decreased by > or =50% the tumor growth of human A673 rhabdomyosarcoma cells and HT29 colon carcinoma xenografted in Swiss nude and NOD/SCID mice, respectively. Tumor cell growth inhibition was similar to that observed in mice receiving therapeutic doses of bevacizumab. These experiments suggest that a therapeutic vaccine containing VEGF kinoid may represent a strategy for safely combating VEGF-dependent neovascularization and metastases occurring in malignant tumors.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Factor A de Crecimiento Endotelial Vascular/inmunología , Animales , Anticuerpos/aislamiento & purificación , Formación de Anticuerpos/inmunología , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Femenino , Células HT29 , Humanos , Sueros Inmunes , Inmunización , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias/prevención & control , Paclitaxel/uso terapéutico , Rabdomiosarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Proc Natl Acad Sci U S A ; 103(51): 19442-7, 2006 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-17158801

RESUMEN

The proinflammatory cytokine TNFalpha is a potent mediator of septic shock and a therapeutic target for chronic inflammatory pathologies including rheumatoid arthritis and Crohn's disease. As an alternative to anti-human TNFalpha (hTNFalpha) mAbs and other hTNFalpha blocker approved drugs, we developed an active anti-hTNFalpha immunotherapy, based on a vaccine comprised of a keyhole limpet hemocyanin-hTNFalpha heterocomplex immunogen (hTNFalpha kinoid) adjuvanted in incomplete Freund's adjuvant. In mice transgenic for hTNFalpha (TTg mice), hTNFalpha kinoid vaccination elicited high titers of Abs that neutralized hTNFalpha bioactivities but did not result in a cellular response to hTNFalpha. The vaccine was safe and effective in two experimental models. Kinoid-immunized but not control TTg mice resisted hTNFalpha-driven shock in one model and were prevented from spontaneous arthritis, inflammatory synovitis, and articular destruction in a second model. These data demonstrate an anti-cytokine induction of autoimmune protection against both acute and chronic hTNFalpha exposure. They show that active vaccination against a human cytokine can be achieved, and that the immune response can be effective and safe.


Asunto(s)
Anticuerpos Bloqueadores/biosíntesis , Inmunoterapia/métodos , Inflamación/prevención & control , Artropatías/prevención & control , Factor de Necrosis Tumoral alfa/inmunología , Vacunación/métodos , Análisis de Varianza , Animales , Anticuerpos Bloqueadores/análisis , Cromatografía en Gel , Electroforesis en Gel de Poliacrilamida , Hemocianinas/inmunología , Immunoblotting , Inflamación/inmunología , Artropatías/inmunología , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA