Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Hum Reprod ; 26(9): 2344-52, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21705369

RESUMEN

BACKGROUND: Endometriosis is still a highly underdiagnosed disease, and the current medical and surgical treatment of endometriosis is associated with a high recurrence rate. This study investigates the use of derivatives of the human antibody F8, specific to the alternatively spliced extra-domain A of fibronectin (Fn), for the imaging and treatment of endometriosis. METHODS: Immunohistochemistry and immunofluorescence was used to evaluate antigen expression in endometriotic tissue of human endometriosis and of a syngeneic mouse model of the disease. The in vivo targeting performance of a fluorescent derivative of the F8 antibody was assessed by imaging mice with endometriosis using a near-infrared fluorescence imager, 24 h following i.v. injection of the antibody conjugate. Furthermore, the mouse model was used for therapy experiments using two recombinant F8-based immunocytokines [F8-interleukin-10 (IL10) and F8-IL2] or saline for the treatment groups. RESULTS: A very strong vascular expression of splice isoforms of Fn and of tenascin-C was observed in human endometriotic lesions by immunohistochemistry and immunofluorescence techniques. After i.v. administration, a selective accumulation of the F8 antibody in endometriotic lesions could be observed in a syngeneic mouse model. These targeting data were used as a basis for therapy experiments with a pro-inflammatory (F8-IL2) and an anti-inflammatory (F8-IL10) cytokine fusion protein of the F8 antibody. The average lesion size in the F8-IL10 treatment group was clearly reduced compared with the saline control group and with the F8-IL2 group, for which no therapeutic effects were observed. CONCLUSIONS: The F8 antibody targets endometriotic lesions in vivo in a mouse model of endometriosis and may be used for the non-invasive imaging of the disease and for the pharmacodelivery of anti-inflammatory cytokines, such as IL10.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Endometriosis/tratamiento farmacológico , Interleucina-10/uso terapéutico , Empalme Alternativo , Animales , Anticuerpos Monoclonales Humanizados , Citocinas/sangre , Endometriosis/patología , Femenino , Fibronectinas/genética , Fibronectinas/inmunología , Humanos , Interleucina-2/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Espectroscopía Infrarroja Corta
2.
Exp Dermatol ; 20(8): 685-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21649738

RESUMEN

We have investigated the staining patterns of primary and metastatic melanoma lesions using F8, L19 and F16. These three clinical-stage antibodies are currently being studied in clinical trials for the pharmacodelivery of cytokines or therapeutic radionuclides to neoplastic sites in patients with cancer. Frozen sections of 24 primary and 29 metastatic melanoma lesions were stained, using immunofluorescence procedures, with biotinylated preparations of the F8, L19 and F16 antibodies, which are specific to the alternatively spliced extra domain A and extra domain B domains of fibronectin and A1 domain of tenascin-C, respectively. Blood vessels were costained using von Willebrand factor-specific antibodies. In primary cutaneous melanoma lesions, F16 and F8 (but not L19) strongly stained the basal lamina at the interface between epidermis and dermis, with a strikingly complementary pattern. By contrast, metastatic melanoma lesions displayed a strong and diffuse pattern of immunoreactivity with all three antibodies. It was found that the extracellular matrix in melanoma undergoes extensive remodelling during the transition from primary to metastatic lesions. The intense staining of metastatic melanoma lesions by the F8, L19 and F16 antibodies provides a strong rationale for the use of these antibodies and their derivatives for the treatment of melanoma patients and possibly for the personalized choice of the best performing antibody in individual patients.


Asunto(s)
Fibronectinas/metabolismo , Melanoma/patología , Melanoma/secundario , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/secundario , Piel/metabolismo , Tenascina/metabolismo , Anticuerpos Antineoplásicos/inmunología , Anticuerpos Antineoplásicos/metabolismo , Estudios de Casos y Controles , Dermis/metabolismo , Epidermis/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Melanoma/metabolismo , Isoformas de Proteínas/metabolismo , Neoplasias Cutáneas/metabolismo , Tenascina/inmunología
3.
J Urol ; 184(6): 2540-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21030045

RESUMEN

PURPOSE: We investigated the therapeutic action of F8-IL2, a fusion protein consisting of the F8 antibody specific to the alternatively spliced extradomain-A of fibronectin, in diabody format and of human interleukin-2 in the Caki-1 (ATCC®) model of human renal cell carcinoma grafted subcutaneously in nude mice. MATERIALS AND METHODS: F8-IL2 was cloned, expressed in CHO cells and purified to homogeneity. This immunocytokine was administered alone or combined with 3 standard drugs commonly used as therapy for kidney cancer, including sunitinib, sorafenib and interferon-α, in 2 sets of doses and treatment schedules. RESULTS: Neither F8-IL2 nor any other therapeutic agent cured tumor bearing mice when used as a single agent. The best therapeutic results were observed for the combination of sunitinib with F8-IL2 in a continuous administration schedule, which yielded a 28% cure rate and substantial tumor growth retardation. CONCLUSIONS: Considering that recombinant interleukin-2 based immunocytokines are now being investigated in several clinical trials in patients with cancer alone or combined with chemotherapy our preclinical results provide a motivation to study F8-IL2 combined with sunitinib in clinical trials in patients with kidney cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Modelos Animales de Enfermedad , Indoles/uso terapéutico , Interleucina-2/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Pirroles/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Sunitinib
5.
J Invest Dermatol ; 133(3): 751-758, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23096716

RESUMEN

Systemic high-dose IL2 promotes long-term survival in a subset of metastatic melanoma patients, but this treatment is accompanied by severe toxicities. The immunocytokine L19-IL2, in which IL2 is fused to the human L19 antibody capable of selective accumulation on tumor neovasculature, has recently shown encouraging clinical activity in patients with metastatic melanoma. In this study, we have investigated the therapeutic performance of L19-IL2, administered systemically in combination with a murine anti-CTLA-4 antibody or with a second clinical-stage immunocytokine (L19-TNF) in two syngeneic immunocompetent mouse models of cancer. We observed complete tumor eradications when L19-IL2 was used in combination with CTLA-4 blockade. Interestingly, mice cured from F9 tumors developed new lesions when rechallenged with tumor cells after therapy, whereas mice cured from CT26 tumors were resistant to tumor rechallenge. Similarly, L19-IL2 induced complete remissions when administered in a single intratumoral injection in combination with L19-TNF, whereas the two components did not lead to cures when administered as single agents. These findings provide a rationale for combination trials in melanoma, as the individual therapeutic agents have been extensively studied in clinical trials, and the antigen recognized by the L19 antibody has an identical sequence in mouse and man.


Asunto(s)
Anticuerpos Antiidiotipos/uso terapéutico , Antígeno CTLA-4/antagonistas & inhibidores , Neoplasias del Colon/tratamiento farmacológico , Proteínas Recombinantes de Fusión/uso terapéutico , Teratocarcinoma/tratamiento farmacológico , Animales , Anticuerpos Antiidiotipos/administración & dosificación , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Humanos , Inyecciones Intralesiones , Ipilimumab , Melanoma/tratamiento farmacológico , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Teratocarcinoma/patología
6.
Arthritis Res Ther ; 15(5): R138, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-24289726

RESUMEN

INTRODUCTION: Etanercept is a fusion protein consisting of the soluble portion of the p75-tumor necrosis factor receptor (TNFR) and the Fc fragment of human IgG1, which is often used for the treatment of patients with rheumatoid arthritis. F8-IL10 is a human immunocytokine based on the F8 antibody and interleukin-10, which is currently being investigated in rheumatoid arthritis with promising clinical results. We have aimed at expressing murine versions of these two fusion proteins, in order to assess their pharmaceutical performance in the collagen-induced model of rheumatoid arthritis in the mouse. METHODS: Two fusion proteins (termed muTNFR-Fc and F8-muIL10) were cloned, expressed in chinese hamster ovary (CHO) cells, purified and characterized. Biological activity of muTNFR-Fc was assessed by its ability to inhibit TNF-induced killing of mouse fibroblasts, while F8-muIL10 was characterized in terms of muIL10 activity, of binding affinity to the cognate antigen of F8, the alternatively-spliced EDA domain of fibronectin, by quantitative biodistribution analysis and in vivo imaging. The therapeutic activity of both fusion proteins was investigated in a collagen-induced mouse model of arthritis. Mouse plasma was analyzed for anti-drug antibody formation and cytokine levels were determined by bead-based multiplex technology. The association of F8-IL10 proteins with blood cells was studied in a centrifugation assay with radiolabeled protein. RESULTS: Both fusion proteins exhibited excellent purity and full biological activity in vitro. In addition, F8-muIL10 was able to localize on newly-formed blood vessels in vivo. When used in a murine model of arthritis, the two proteins inhibited arthritis progression. The activity of muTNFR-Fc was tested alone and in combination with F8-huIL10. The chimeric version of F8-IL10 was not better then the fully human fusion protein and showed similar generation of mouse anti-fusion protein antibodies. Incubation studies of F8-muIL10 and F8-huIL10 with blood revealed that only the fully human fusion protein is not associated with cellular components at concentrations as low as 0.2 µg/ml, thus facilitating its extravasation from blood vessels. CONCLUSIONS: The described products may represent useful research tools for the study of the anti-arthritic properties of TNF blockade and of IL10-based immunocytokines in syngeneic immunocompetent models of arthritis.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Artritis Experimental/tratamiento farmacológico , Inmunoglobulina G/farmacología , Interleucina-10/farmacología , Animales , Anticuerpos/sangre , Anticuerpos/inmunología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Antirreumáticos/inmunología , Antirreumáticos/farmacología , Artritis Experimental/inmunología , Artritis Experimental/patología , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Citocinas/sangre , Progresión de la Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Etanercept , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inyecciones Intravenosas , Interleucina-10/genética , Interleucina-10/inmunología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Resultado del Tratamiento
7.
Cancer Res ; 72(7): 1814-24, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22392081

RESUMEN

The selective delivery of bioactive agents to tumors reduces toxicity and enhances the efficacy of anticancer therapies. In this study, we show that the antibody F8, which recognizes perivascular and stromal EDA-fibronectin (EDA-Fn), when conjugated to interleukin-2 (F8-IL2) can effectively inhibit the growth of EDA-Fn-expressing melanomas in combination with paclitaxel. We obtained curative effects with paclitaxel administered before the immunocytokine. Coadministration of paclitaxel increased the uptake of F8 in xenografted melanomas, enhancing tumor perfusion and permeability. Paclitaxel also boosted the recruitment of F8-IL2-induced natural killer (NK) cells to the tumor, suggesting a host response as part of the observed therapeutic benefit. In support of this likelihood, NK cell depletion impaired the antitumor effect of paclitaxel plus F8-IL2. Importantly, this combination reduced both the tumor burden and the number of pulmonary metastatic nodules. The combination did not cause cumulative toxicity. Together, our findings offer a preclinical proof that by acting on the tumor stroma paclitaxel potentiates the antitumor activity elicited by a targeted delivery of IL2, thereby supporting the use of immunochemotherapy in the treatment of metastatic melanoma.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Fibronectinas/análisis , Interleucina-2/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Paclitaxel/uso terapéutico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Permeabilidad Capilar , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Humanos , Interleucina-2/administración & dosificación , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/química , Melanoma Experimental/patología , Ratones , Paclitaxel/administración & dosificación , Isoformas de Proteínas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Integr Biol (Camb) ; 3(4): 468-78, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21225052

RESUMEN

The antibody-mediated targeted delivery of cytokines, growth factors and immunomodulators offers great potential for the therapy of cancer and other serious conditions. Interferon-alpha has long been used in the clinic for the treatment of patients with certain malignancies or with viral disease. Promising anticancer activity has recently been reported for two fusion proteins consisting of immunoglobulins bearing the interferon-alpha polypeptide at the C-terminal end of the molecule. Here we describe the design, production and characterization of a novel immunocytokine, in which murine interferon-alpha2 was sequentially fused with the tumor-targeting antibody fragment scFv(F8), specific to the alternatively-spliced EDA domain of fibronectin. The resulting fusion protein (F8-IFNa) could be produced to homogeneity and was shown to retain both antigen binding activity and interferon-alpha activity. Biodistribution studies in tumor-bearing mice with radioiodinated protein preparations confirmed the ability of F8-IFNa to selectively localize at the tumor site. However, using two different murine models of cancer (F9 teratocarcinomas and Cloudman S91 melanomas in immunocompetent mice), we could not detect a striking superiority for the therapeutic performance of F8-IFNa as compared to KSF-IFNa, a fusion protein of irrelevant specificity in the mouse which was used as negative control. In the paper, we present hypotheses why the antibody-based pharmacodelivery of interferon-alpha fails to eradicate tumors, in contrast to the situation observed by our group for other immunocytokines, which benefit from a selective localization at the tumor site.


Asunto(s)
Inmunoconjugados/uso terapéutico , Interferón-alfa/administración & dosificación , Interferón-alfa/uso terapéutico , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Anticuerpos de Cadena Única/inmunología , Estructuras Animales/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Vasos Sanguíneos/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Fibronectinas/inmunología , Humanos , Inmunoconjugados/genética , Inmunoconjugados/inmunología , Inmunoconjugados/farmacocinética , Inmunoconjugados/farmacología , Interferón-alfa/genética , Leucocitos Mononucleares/patología , Melanoma/irrigación sanguínea , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones de la Cepa 129 , Muramidasa/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Anticuerpos de Cadena Única/genética , Teratocarcinoma/irrigación sanguínea , Teratocarcinoma/tratamiento farmacológico , Teratocarcinoma/metabolismo , Teratocarcinoma/patología , Distribución Tisular , Resultado del Tratamiento
9.
Head Neck Oncol ; 3: 25, 2011 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-21548989

RESUMEN

BACKGROUND: The antibody-based targeted delivery of bioactive molecules to tumour vasculature is an attractive avenue to concentrate therapeutic agents at cancer sites, while sparing normal organs. L19, F8 and F16 are three fully human monoclonal antibodies, specific to splice isoforms of fibronectin and tenascin-C, which bind to sites of active tissue remodeling and which are currently in Phase I and II clinical trials as radio-immunoconjugates and immunocytokines in patients with cancer and arthritis.In this article, we report the first comparative analysis of expression patterns for the extra domains EDB and EDA of fibronectin and A1 of tenascin-C in both primary and metastatic head and neck cancer lesions. METHODS: We performed a comparative immunofluorescence analysis with the L19, F8 and F16 antibodies in 40 freshly frozen human head and neck cancer specimens. RESULTS: On average, F8 and F16 exhibited similar staining intensities, which were typically stronger than L19. Interestingly, some specimens exhibited striking differences in staining by the three antibodies. CONCLUSIONS: These results suggests that an individualized treatment procedure (e.g., choice of L19, F8 or F16 based on immuno-PET or immunofluorescence procedure) may represent the most logical avenue for offering the best possible antibody to any given patient.


Asunto(s)
Anticuerpos/análisis , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Inmunohistoquímica/métodos , Anticuerpos/metabolismo , Anticuerpos/uso terapéutico , Afinidad de Anticuerpos , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Técnica del Anticuerpo Fluorescente , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunoconjugados/uso terapéutico , Inmunoterapia/métodos , Metástasis de la Neoplasia
10.
Clin Cancer Res ; 17(24): 7732-42, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22028492

RESUMEN

PURPOSE: L19-IL2 is an immunocytokine composed of an antibody fragment specific to the EDB domain of fibronectin, a tumor angiogenesis marker, and of human interleukin-2 (IL2). L19-IL2 delivers IL2 to the tumor site exploiting the selective expression of EDB on newly formed blood vessels. Previously, the recommended dose of L19-IL2 monotherapy was defined as 22.5 million international units (Mio IU) IL2 equivalents. In this study, safety and clinical activity of L19-IL2 in combination with dacarbazine were assessed in patients with metastatic melanoma. EXPERIMENTAL DESIGN: The first 10 studied patients received escalating doses of L19-IL2 on days 1, 3, and 5 in combination with 1 g/m(2) of dacarbazine on day 1 of a 3-weekly therapy cycle. Subsequently, 22 patients received L19-IL2 at recommended dose plus dacarbazine. Up to six treatment cycles were given, followed by a maintenance regimen with biweekly L19-IL2. RESULTS: The recommended dose of L19-IL2 in combination with dacarbazine was defined as 22.5 Mio IU. Toxicity was manageable and reversible, with no treatment-related deaths. Twenty-nine patients were evaluable for efficacy according to Response Evaluation Criteria in Solid Tumors (RECIST). In a centralized radiology analysis, eight of 29 (28%) patients achieved a RECIST-confirmed objective response, including a complete response still ongoing 21 months after treatment beginning. The 12-month survival rate and median overall survival of the recommended dose-treated patients (n = 26) were 61.5% and 14.1 months, respectively. CONCLUSIONS: The repeated administration of L19-IL2 in combination with dacarbazine is safe and shows encouraging signs of clinical activity in patients with metastatic melanoma. This combination therapy is currently evaluated in a randomized phase II trial with patients with metastatic melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Melanoma/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Estudios de Cohortes , Dacarbazina/administración & dosificación , Dacarbazina/efectos adversos , Dacarbazina/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Fatiga/inducido químicamente , Femenino , Fiebre/inducido químicamente , Humanos , Masculino , Melanoma/metabolismo , Melanoma/mortalidad , Tasa de Depuración Metabólica , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/farmacocinética , Análisis de Supervivencia , Tasa de Supervivencia , Resultado del Tratamiento
11.
Arthritis Res Ther ; 11(5): R142, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19781067

RESUMEN

INTRODUCTION: In this article, we present a comparative immunohistochemical evaluation of four clinical-stage antibodies (L19, F16, G11 and F8) directed against splice isoforms of fibronectin and of tenascin-C for their ability to stain synovial tissue alterations in rheumatoid arthritis patients. Furthermore we have evaluated the therapeutic potential of the most promising antibody, F8, fused to the anti-inflammatory cytokine interleukin (IL) 10. METHODS: F8-IL10 was produced and purified to homogeneity in CHO cells and shown to comprise biological active antibody and cytokine moieties by binding assays on recombinant antigen and by MC/9 cell proliferation assays. We have also characterized the ability of F8-IL10 to inhibit arthritis progression in the collagen-induced arthritis mouse model. RESULTS: The human antibody F8, specific to the extra-domain A of fibronectin, exhibited the strongest and most homogenous staining pattern in synovial biopsies and was thus selected for the development of a fully human fusion protein with IL10 (F8-IL10, also named DEKAVIL). Following radioiodination, F8-IL10 was able to selectively target arthritic lesions and tumor neo-vascular structures in mice, as evidenced by autoradiographic analysis and quantitative biodistribution studies. The subcutaneous administration route led to equivalent targeting results when compared with intravenous administration and was thus selected for the clinical development of the product. F8-IL10 potently inhibited progression of established arthritis in the collagen-induced mouse model when tested alone and in combination with methotrexate. In preparation for clinical trials in patients with rheumatoid arthritis, F8-IL10 was studied in rodents and in cynomolgus monkeys, revealing an excellent safety profile at doses tenfold higher than the planned starting dose for clinical phase I trials. CONCLUSIONS: Following the encouraging preclinical results presented in this paper, clinical trials with F8-IL10 will now elucidate the therapeutic potential of this product and whether the targeted delivery of IL10 potentiates the anti-arthritic action of the cytokine in rheumatoid arthritis patients.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Artritis Experimental/tratamiento farmacológico , Inmunoterapia/métodos , Interleucina-10/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Especificidad de Anticuerpos , Artritis Experimental/patología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Células CHO , Cricetinae , Cricetulus , Citocinas/sangre , Citocinas/inmunología , Sistemas de Liberación de Medicamentos , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Inmunosupresores/farmacología , Interleucina-10/efectos adversos , Interleucina-10/inmunología , Macaca fascicularis , Masculino , Metotrexato/farmacología , Ratones , Ratones Endogámicos DBA , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/inmunología , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA