Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros











Intervalo de año de publicación
1.
Ren Fail ; 45(2): 2279642, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37942512

RESUMEN

BACKGROUND: Focal segmental glomerulosclerosis (FSGS) is an important cause of refractory nephrotic syndrome (NS) in children and adults. Urinary CD80 is elevated in some patients with primary FSGS, however, its clinical value is not fully clarified. This study aims to evaluate the clinical and pathological significance of urinary CD80 in patients with primary FSGS. METHODS: Sixty-one adult patients with biopsy-proven primary FSGS, with standard treatment and long-term follow up, were enrolled retrospectively. Urinary CD80, on the day of kidney biopsy, was measured using commercial ELISA kits and adjusted by urinary creatinine excretion. Their associations with clinical and pathological parameters were investigated. RESULTS: Urinary CD80 was detectable in 30/61 (49.2%) patients, who presented with a higher level of proteinuria (10.7 vs. 5.8 g/24h; p = 0.01), a lower level of serum albumin (19.3 ± 3.9 vs. 24.2 ± 8.2 g/L; p = 0.005), a higher prevalence of hematuria (70.0 vs. 38.7%; p = 0.01), and showed a lower percentage of segmental glomerulosclerosis lesion [4.8 (3.7-14.0) vs. 9.1 (5.6-21.1) %; p = 0.06]. The cumulative relapse rate was remarkably high in these patients (log-rank, p = 0.001). Multivariate analysis identified that the elevated urinary CD80 was an independent risk factor for steroid-dependent NS (OR 8.81, 95% CI 1.41-54.89; p = 0.02) and relapse (HR, 2.87; 95% CI 1.29-6.38; p = 0.01). CONCLUSIONS: The elevated urinary CD80 is associated with mild pathological change and steroid-dependent cases of primary FSGS adults, which indicates these patients are more similar to minimal change disease (MCD) in clinicopathological features.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Nefrosis Lipoidea , Síndrome Nefrótico , Niño , Adulto , Humanos , Nefrosis Lipoidea/complicaciones , Glomeruloesclerosis Focal y Segmentaria/patología , Estudios Retrospectivos , Antígeno B7-1/uso terapéutico , Antígeno B7-1/orina , Síndrome Nefrótico/etiología , Recurrencia , Esteroides/uso terapéutico
2.
Cytometry B Clin Cytom ; 90(2): 128-40, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26332491

RESUMEN

BACKGROUND: Receptor occupancy (RO) assays measure drug target engagement, and are used as pharmacodynamic (PD) biomarkers. RO assays are commonly performed by flow cytometry and often require multiplexing for assessment of multiple PD biomarkers when specimen volumes are limited. We present multiplexed RO assays for an IGF1R-EGFR bispecific antibody (Bs-Ab) and a CTLA4-Ig recombinant fusion protein to demonstrate key considerations for accurate RO assessment. METHODS: RO in cynomolgus monkeys was determined in whole blood using flow cytometry. Free and total receptors were measured using anti-receptor fluorescence-labeled detection reagents, competitive and noncompetitive to drug, respectively. RESULTS: RO of IGF1R was examined as PD for Bs-Ab, since IGF1R was expressed on blood cells. Multiplexed measurements of free and total IGF1R showed that IGF1R expression measured by total receptor was highly variable, impacting interpretation of free-IGF1R. Normalization of free-over-total IGF1R measurements compensated for variability of receptor expression allowing for accurate RO assessment. RO of CTLA4-Ig, a recombinant fusion protein targeting CD80 and CD86 receptors, was multiplexed to simultaneously measure target engagements for both receptors. Both RO methods demonstrated specificity of receptor measurements without cross-reactivity to each other in multiplexed formats. RO methods were used for evaluation of PD activity of Bs-Ab and CTLA4-Ig in cynomolgus monkeys. In both cases, RO results showed dose-dependent target engagement, corresponding well to the pharmacokinetics. CONCLUSIONS: Multiplexed RO methods allowed accurate assessment of PD activity for Bs-Ab and CTLA4-Ig, facilitating development of these biopharmaceuticals from preclinical to clinical stages.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Receptores ErbB/inmunología , Citometría de Flujo , Receptores de Somatomedina/inmunología , Anticuerpos Biespecíficos/uso terapéutico , Antígeno B7-1/inmunología , Antígeno B7-1/uso terapéutico , Biomarcadores , Antígeno CTLA-4/inmunología , Receptores ErbB/uso terapéutico , Humanos , Inmunoconjugados/inmunología , Inmunosupresores/inmunología , Inmunosupresores/farmacocinética , Inmunosupresores/uso terapéutico , Receptor IGF Tipo 1 , Receptores de Somatomedina/uso terapéutico , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico
3.
Reumatol. clín. (Barc.) ; 8(2): 78-83, mar.-abr. 2012. ilus
Artículo en Español | IBECS | ID: ibc-97840

RESUMEN

La interacción molecular doble y simultánea entre las células presentadoras de antígeno (CPA) y los linfocitos T es imprescindible para la activación óptima de la respuesta inmunitaria y requiere de la participación de dos grupos de receptores de membrana. El abatacept es una proteína de fusión que modula selectivamente una de estas dos vías, uniéndose a los receptores CD80 y CD86 de las CPA. De esta forma el fármaco inhibe la activación de las células T, bloqueando selectivamente la unión específica de los receptores CD80/CD86 al CD28 y como consecuencia inhibiendo la proliferación de las células T y la respuesta inmunitaria de las células B. Esta acción farmacológica se traduce en la normalización de los niveles de los mediadores inflamatorios en los enfermos con artritis reumatoide y en una respuesta clínica segura y eficaz. El abatacept, en combinación con metotrexato, evita la progresión de la lesión articular y mejora la función física en enfermos con artritis reumatoide (AU)


The double and simultaneous molecular interaction between antigen-presentig cells (APC) and T lymphocytes is essential for the optimal activation of the immunological response and requires the participation of two membrane receptor groups. Abatacept is a fusion protein that selectively modulates one of these two ways, by binding to CD80 and CD86 receptors on APC. In this way, the drug inhibits T cell activation, selectively blocking the specific interaction of CD80/CD86 receptors to CD28 and, therefore, inhibiting T cell proliferation and B cell immunological response. This pharmacological action results in the normalization of inflammatory mediators in rheumatoid arthritis patients and in a safe and efficacious clinical response. Abatacept in combination with methotrexate prevents the progression of joint damage and improves physical function in rheumatoid arthritis patients (AU)


Asunto(s)
Humanos , Masculino , Femenino , Antirreumáticos/metabolismo , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/epidemiología , Artritis Reumatoide/prevención & control , Linfocitos T , Histocompatibilidad , Antígenos de Histocompatibilidad/administración & dosificación , Biotecnología/métodos , Antígeno B7-1/biosíntesis , Antígeno B7-1/uso terapéutico , Antígenos CD28/uso terapéutico , Metotrexato/metabolismo , Metotrexato/uso terapéutico , Artritis Reumatoide/fisiopatología
4.
Rheumatol Int ; 31(4): 513-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20035333

RESUMEN

Programmed death ligand (PDL) is a new member of the B7 family of costimulatory molecules that specifically interacts with programmed death 1 (PD-1) expressed on activated T cells, B cells, and myeloid cells. Collagen II (CII)-induced arthritis (CIA) is an experimental model of arthritis that has been used to dissect the pathogenesis of human rheumatoid arthritis. In this study, we have investigated the effects of PDL-Ig on CIA. Administration of PDL-Ig significantly ameliorated the disease as assessed by clinical arthritis score and histology in the joints. Expression of proinflammatory cytokines, such as IL-17 and IL-23, in the serum was reduced by PDL-Ig treatment. These results showed a beneficial effect of PDL-Ig on CIA through anti-inflammatory actions and inhibition of cell proliferation in response to CII, suggesting that the PD-1-PDL pathway may be involved in the pathogenesis of CIA, and thus PDL-Ig may be a useful therapy for the improvement of human rheumatoid arthritis.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Antígeno B7-1/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Antígenos de Superficie/fisiología , Proteínas Reguladoras de la Apoptosis/fisiología , Artritis Experimental/inmunología , Artritis Experimental/patología , Proliferación Celular , Interleucina-17/biosíntesis , Interleucina-23/biosíntesis , Masculino , Ratones , Ratones Endogámicos DBA , Receptor de Muerte Celular Programada 1
5.
Clin Immunol ; 136(1): 30-41, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20346734

RESUMEN

Previous studies demonstrate that both membrane B7-H4 and B7-H4-Ig fusion protein could inhibit T-cell responses. In the present study, we explored the potential effect of B7-H4-Ig on liver injury in a hepatitis mouse model induced by concanavalin A (ConA). A B7-H4-Ig construct was introduced into animals by the hydrodynamic gene delivery approach. It was found that ectopic expression of B7-H4-Ig could inhibit ConA-induced elevation of serum levels of ALT and AST, suppress liver necrosis and even mortality of mice. Furthermore, we observed that pretreatment of B7-H4-Ig dramatically decreased serum levels and the expression of mRNA for IL-2, IFN-gamma and IL-4, but increased IL-10 in ConA-treated mice. Our results suggest that B7-H4-Ig may protect animals from liver injury induced by ConA, which could be associated with reduced serum levels for IL-2, IFN-gamma and IL-4 as well as enhanced IL-10 production.


Asunto(s)
Antígeno B7-1/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Concanavalina A/farmacología , Terapia Genética/métodos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Antígeno B7-1/sangre , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Técnicas de Transferencia de Gen , Humanos , Fragmentos Fc de Inmunoglobulinas/sangre , Interferón gamma/sangre , Interferón gamma/genética , Interleucina-10/sangre , Interleucina-10/genética , Interleucina-2/sangre , Interleucina-2/genética , Interleucina-4/sangre , Interleucina-4/genética , Leucocitos Mononucleares/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Necrosis/inducido químicamente , Necrosis/patología , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Supervivencia , Inhibidor 1 de la Activación de Células T con Dominio V-Set
6.
Cancer Biol Ther ; 8(15): 1440-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19556848

RESUMEN

The goal of vaccine-based cancer immunotherapy is to induce a tumor-specific immune response that ultimately reduces tumor burden. However, the immune system is often tolerant to antigens presented by the tumor, as the cancer originates from within a patient and is therefore recognized as self. This article reviews selected clinical strategies for overcoming this immune tolerance, and approaches to enhance generation of immunity to tumor-associated antigens by activating innate immunity, potentiating adaptive immunity, reducing immunosuppression, and enhancing tumor immunogenicity. Success in the field of cancer vaccines has yet to be fully realized, but intelligent choice of immunomodulators, tumor antigens and patient populations will likely lead to clinically relevant uses for cancer vaccines.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Antiinflamatorios no Esteroideos/uso terapéutico , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antígeno B7-1/inmunología , Antígeno B7-1/uso terapéutico , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Terapia Combinada , Citocinas/inmunología , Citocinas/uso terapéutico , Electrocoagulación , Femenino , Ultrasonido Enfocado de Alta Intensidad de Ablación , Humanos , Factores Inmunológicos/inmunología , Factores Inmunológicos/uso terapéutico , Masculino , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Neoplasias/cirugía , Autotolerancia , Subgrupos de Linfocitos T/inmunología , Receptores Toll-Like/agonistas , Escape del Tumor
7.
Leuk Lymphoma ; 50(3): 447-54, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19197726

RESUMEN

Whole-cell vaccines allow the induction of anti-tumor immune responses without the need to define tumor antigens. We wished to directly compare, for the first time, the capacity of B7-1, B7-2 and 4-1BB ligand (4-1BBL) costimulatory molecules to convert murine and human acute myeloid leukemia (AML) cells into whole vaccines. 32Dc-kit is a murine myeloid cell line, which develops an AML-like disease over a protracted period, emulating human AML disease development. 32Dc-kit cells were modified to express elevated levels of B7-1, B7-2 or 4-1BBL, and each led to tumor rejection, although only mice injected with 32Dc-kit/B7-2 cells were able to reject subsequent parental tumor cell challenge. T-cell deficient nude mice were able to reject the 32Dc-kit variants, but they could not reject parental cell challenge; however, we found no evidence of cytotoxic T lymphocyte or natural killer (NK) activity ex vivo suggesting that tumor cell killing was mediated by an immune response that could not be recapitulated using purified NK or T cells as lone effectors. In human allogeneic mixed lymphocyte reactions (MLRs), we found no single costimulatory molecule was more effective, suggesting that the induction of a universal anti-tumor response will require a combination of costimulatory molecules.


Asunto(s)
Vacunas contra el Cáncer , Inmunidad , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/terapia , Ligando 4-1BB/inmunología , Ligando 4-1BB/uso terapéutico , Animales , Antígeno B7-1/inmunología , Antígeno B7-1/uso terapéutico , Antígeno B7-2/inmunología , Antígeno B7-2/uso terapéutico , Línea Celular Tumoral , Humanos , Activación de Linfocitos , Prueba de Cultivo Mixto de Linfocitos , Ratones
8.
Clin Cancer Res ; 14(10): 3060-9, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18483372

RESUMEN

PURPOSE: Poxviral vectors have a proven safety record and can be used to incorporate multiple transgenes. Prior clinical trials with poxviral vaccines have shown that immunologic tolerance to self-antigens can be broken. Carcinoembryonic antigen (CEA) and MUC-1 are overexpressed in a substantial proportion of common solid carcinomas. The primary end point of this study was vaccine safety, with immunologic and clinical responses as secondary end points. EXPERIMENTAL DESIGN: We report here a pilot study of 25 patients treated with a poxviral vaccine regimen consisting of the genes for CEA and MUC-1, along with a triad of costimulatory molecules (TRICOM; composed of B7.1, intercellular adhesion molecule 1, and lymphocyte function-associated antigen 3) engineered into vaccinia (PANVAC-V) as a prime vaccination and into fowlpox (PANVAC-F) as a booster vaccination. RESULTS: The vaccine was well tolerated. Apart from injection-site reaction, no grade > or =2 toxicity was seen in more than 2% of the cycles. Immune responses to MUC-1 and/or CEA were seen following vaccination in 9 of 16 patients tested. A patient with clear cell ovarian cancer and symptomatic ascites had a durable (18-month) clinical response radiographically and biochemically, and one breast cancer patient had a confirmed decrease of >20% in the size of large liver metastasis. CONCLUSIONS: This vaccine strategy seems to be safe, is associated with both CD8 and CD4 immune responses, and has shown evidence of clinical activity. Further trials with this agent, either alone or in combination with immunopotentiating and other therapeutic agents, are warranted.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/uso terapéutico , Mucina-1/uso terapéutico , Neoplasias/terapia , Poxviridae/inmunología , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/uso terapéutico , Adulto , Anciano , Antígeno B7-1/inmunología , Antígeno B7-1/uso terapéutico , Antígenos CD58/inmunología , Antígenos CD58/uso terapéutico , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Femenino , Vectores Genéticos , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/uso terapéutico , Masculino , Persona de Mediana Edad , Mucina-1/inmunología , Neoplasias/inmunología , Proyectos Piloto , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico
9.
Handb Exp Pharmacol ; (181): 291-328, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18071951

RESUMEN

This chapter describes the generation of novel reagents for the treatment of cancer using fusion proteins constructed with natural ligands of the immune system. Immunotherapy is a powerful therapeutic modality that has not been fully harnessed for the treatment of cancer. We and others have hypothesized that if the proper immunoregulatory ligands can be targeted to the tumor, an effective immune response can be mounted to treat both established primary tumors and distant metastatic lesions. Though it is generally believed that immunotherapy has the potential to treat only residual disease, we offer evidence that this approach can, by itself, destroy large tumor masses and produce lasting remissions of experimental solid tumors. From these studies, three major classes of immune activators, namely, cytokines, chemokines, and costimulatory molecules, have been shown to generate antitumor responses in animal models. In addition, the reversal of immune tolerance by the deletion of T regulatory (Treg) cells has been shown to be equally important for effective immunotherapy. In an attempt to identify reagents that can provide an enhanced immune stimulation and treatment of cancer, our laboratory has developed a novel monoclonal antibody targeting approach, designated Tumor Necrosis Therapy (TNT), which utilizes stable intracellular antigens present in all cell types but which are only accessible in dead and/or dying cells. Since tumors contain necrotic and degenerating regions that account for 30-80% of the tumor mass, this targeting approach can be used to deliver therapeutic reagents to the core of tumors, a site abundant in tumor antigens. In our first set of reagents, a panel of cytokine fusion proteins was genetically engineered using monoclonal antibody chimeric TNT-3 (chTNT-3) directed against necrotic regions of tumors (single-stranded DNA) fused with IL-2, or GM-CSF, or TNFalphaa, or IFNgamma. Tested against different solid tumors, these reagents were found to mount an effective although transient immune response to tumor especially when used in combination. To improve upon these results, additional chTNT-3 fusion proteins using the liver-expression chemokine (LEC) and the costimulatory molecule B7.1 were constructed. Both of these reagents were found to work significantly better than the above cytokine fusion proteins due to their ability to stimulate multiple arms of the immune system deemed useful for cancer immunotherapy. Finally, the Tumor Necrosis Factor Superfamily (TNFSF) gene DC137L was used to generate chTNT-3 antibody (targeted) and soluble Fc (untargeted) fusion proteins. When used alone, both forms of costimulatory fusion proteins were found to produce in a s dose-dependent manner, complete regression of murine solid tumors. Evidence is presented to show that Treg cells play an important role in suppressing antitumor immunity since the deletion of these cells, when used in combination with LEC or costimulatory fusion proteins, produced profound and effective treatment with sustained memory. It is hoped that these data will further the preclinical development of soluble Fc and antibody based fusion proteins fro the immunotherapy of cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Quimiocinas/uso terapéutico , Citocinas/uso terapéutico , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Animales , Antígeno B7-1/inmunología , Antígeno B7-1/uso terapéutico , Quimiocinas/inmunología , Citocinas/inmunología , Humanos , Ligandos , Neoplasias/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Reguladores/efectos de los fármacos , Escape del Tumor , Factores de Necrosis Tumoral/inmunología , Factores de Necrosis Tumoral/uso terapéutico
10.
Best Pract Res Clin Haematol ; 20(3): 557-68, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17707840

RESUMEN

Gene therapy for patients with hematologic malignancies, particularly chronic lymphocytic leukemia (CLL), have focused on transducing primary leukemia cells with a virus vector to express immune-stimulating genes which can induce and propagate a productive and clinically significant immune response against the malignant cells. A variety of replication-defective vectors has been studied to transduce genes for cytokines and function-associated surface molecules. Active vaccines have been developed in vitro, and their activity has been confirmed in clinical trials. Ongoing work aims to optimize this strategy and to identify the appropriate and optimal patient groups in which to apply vaccine therapy. Clinical trials also have provided insight into unexpected alternative mechanisms through which these strategies might provide a clinical benefit.


Asunto(s)
Terapia Genética , Inmunoterapia Activa , Leucemia Linfocítica Crónica de Células B/terapia , Apoptosis/inmunología , Antígeno B7-1/uso terapéutico , Antígeno B7-2/uso terapéutico , Ligando de CD40/uso terapéutico , Ensayos Clínicos como Asunto , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Interleucina-12/uso terapéutico , Interleucina-2/uso terapéutico , Factor de Necrosis Tumoral alfa/uso terapéutico
11.
Transplantation ; 84(2): 223-30, 2007 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-17667814

RESUMEN

BACKGROUND: Activated T cells play a key role in allograft rejection. T cell activation requires signaling via the T cell receptor as well as costimulatory signals. Inducible costimulatory molecule (ICOS), with its ligand B7RP-1, is a recently discovered costimulatory molecule of the CD28 family. The role of this signaling pathway during the early phases of kidney allograft rejection is not clear so far. METHODS: Kidneys were orthotopically transplanted from BALB/c to C57BL/6 mice. Animals were assigned to five experimental groups: blocking anti-ICOS monoclonal antibody, ICOS fusion protein, anti-B7RP1 monoclonal antibody, B7RP-1 fusion protein, and control immunoglobulin G. RESULTS: Survival was significantly reduced in animals treated with ICOS monoclonal antibody (mAb) and B7RP-1 Fc as compared with controls. These animals had also a lower number of apoptotic graft infiltrating T cells, whereas the expression of intracellular interferon-gamma in CD3CD4 T cells was increased. Animals treated with ICOS Fc and B7RP-1 mAb had similar survival and numbers of apoptotic T cells as compared with controls. CONCLUSIONS: In summary, the blockade of ICOS with ICOS mAb or B7RP-1 Fc reduced the amount of apoptosis of infiltrating lymphocytes and resulted in continuous inflammatory processes with progressive tissue damage and graft failure.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/uso terapéutico , Antígeno B7-1/uso terapéutico , Rechazo de Injerto/tratamiento farmacológico , Trasplante de Riñón/inmunología , Animales , Anticuerpos Monoclonales , Antígenos de Diferenciación de Linfocitos T/inmunología , Apoptosis , Antígeno B7-1/inmunología , Complejo CD3/biosíntesis , Complejo CD3/genética , Inmunoadhesinas CD4/biosíntesis , Inmunoadhesinas CD4/genética , Modelos Animales de Enfermedad , Citometría de Flujo , Expresión Génica , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Supervivencia de Injerto , Etiquetado Corte-Fin in Situ , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Líquido Intracelular/metabolismo , Trasplante de Riñón/patología , Ligandos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Mensajero/genética , Linfocitos T/inmunología , Linfocitos T/patología
12.
Acta Biochim Pol ; 53(4): 807-13, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17117211

RESUMEN

In order to investigate the anti-tumor activity of a soluble B7-1/immunoglobulin G fusion protein and explore an effective method to eliminate immune escape of tumor cells, a recombinant vector encoding this fusion protein was constructed and constitutively expressed in Chinese hamster ovary cells. After purification with protein G affinity chromatography, the soluble fusion protein was tested for bioactivity. Results showed that the fusion protein could significantly increase the density of B7-1 molecules on WEHI-3 cells, a mouse leukemia cell line. Through allogeneic mixed lymphocyte tumor cultures, it was demonstrated that, with the presence of the first signal, it could also significantly enhance T cell activation and killing activity against WEHI-3 cells and interleukin-2 secretion by activated mouse T lymphocytes. The conclusion can be drawn that the soluble B7-IgG fusion protein has a potent capacity to generate or enhance anti-tumor immune response in vitro, and its clinical value deserves further investigation.


Asunto(s)
Antineoplásicos/farmacología , Antígeno B7-1/uso terapéutico , Inmunoglobulina G/uso terapéutico , Animales , Antígeno B7-1/genética , Línea Celular Tumoral , Citotoxicidad Inmunológica , Inmunoglobulina G/genética , Leucemia/patología , Activación de Linfocitos , Prueba de Cultivo Mixto de Linfocitos , Ratones , Proteínas Recombinantes de Fusión/uso terapéutico , Solubilidad
14.
Zhonghua Zhong Liu Za Zhi ; 27(7): 404-7, 2005 Jul.
Artículo en Chino | MEDLINE | ID: mdl-16188124

RESUMEN

OBJECTIVE: To investigate the synergistic effects of antisense HIF-1alpha gene therapy combined with B7-1-mediated immunotherapy on cancer treatment. METHODS: Antisense HIF-1alpha and B7-1 expression vector were constructed. Lymphoma cells EL-4 were injected subcutaneously into C57BL/6 mice and transplanted lymphomas were established. The mice received either antisense HIF-1alpha, B7-1, or a combinational agent, complexed with DOTAP cationic liposomes. The tumor growth in the mice was monitored. Expression of HIF-1alpha, B7-1 and VEGF were detected by immunohistochemistry and Western blotting. The tumor blood vessels were immunostained with CD31- antibodies and the tumor vascular density was assessed by light microscopy. RESULTS: Gene transfer of plasmid expressing the encoded antisense HIF-1alpha inhibited VEGF expression and reduced vascular density in the tumors, eradicated tumors in diameter smaller than 0.1 cm and only retarded the growth of larger tumors. Whereas combination of antisense HIF-1alpha gene therapy and B7-1 immunotherapy eradicated all tumors in diameter of 0.4 cm. CONCLUSION: Antisense HIF-1alpha blocks tumor hypoxia pathway by downregulating VEGF expression, reduction of vascular density and enhances B7-1-mediated immunotherapy. Strategies that target HIF-1 may have therapeutic potential in cancer treatment and are worthy of further studying.


Asunto(s)
Antígeno B7-1/uso terapéutico , Terapia Genética/métodos , Subunidad alfa del Factor 1 Inducible por Hipoxia/uso terapéutico , Linfoma/terapia , Oligonucleótidos Antisentido/uso terapéutico , Animales , Antígeno B7-1/genética , Técnicas de Transferencia de Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Linfoma/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/terapia , Oligonucleótidos Antisentido/genética
15.
J Immunol ; 175(6): 3715-23, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16148117

RESUMEN

The avidity of Ag-specific CTL is a critical determinant for clearing viral infection and eliminating tumor. Although previous studies have demonstrated that vaccines using enhanced costimulation will enhance the level and avidity of Ag-specific T cells from naive mice, there are conflicting data about the effects of vaccines using enhanced costimulation (vector or dendritic cell based) on the survival of memory T cells. In this study we have first extended previous observations that primary vaccination with a recombinant vaccinia virus (rV-) expressing a model Ag (LacZ) and a triad of T cell costimulatory molecules (B7-1, ICAM-1, and LFA-3 (designated TRICOM)) enhances the level and avidity of T cells from naive vaccinated C57BL/6 (Thy1.2) mice. Adoptive transfer of Thy1.1 memory CD8(+) T cells into naive Thy1.2 C57BL/6 mice was followed by booster vaccinations with a recombinant fowlpox (rF-)-expressing LacZ (rF-LacZ) or booster vaccinations with rF-LacZ/TRICOM. Analysis of levels of beta-galactosidase tetramer-positive T cells and functional assays (IFN-gamma expression and lytic activity) determined that booster vaccinations with rF-LacZ/TRICOM were superior to booster vaccinations with rF-LacZ in terms of both maintenance and enhanced avidity of memory CD8(+) T cells. Antitumor experiments using a self-Ag (carcinoembryonic Ag (CEA) vaccines in CEA transgenic mice bearing CEA-expressing tumors) also demonstrated that the use of booster vaccinations with vaccines bearing enhanced costimulatory capacity had superior antitumor effects. These studies thus have implications in the design of more effective vaccine strategies.


Asunto(s)
Memoria Inmunológica , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/inmunología , Animales , Antígeno B7-1/uso terapéutico , Antígenos CD58/uso terapéutico , Inmunización Secundaria/métodos , Molécula 1 de Adhesión Intercelular/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/terapia , Vacunas de ADN/uso terapéutico
16.
Curr Hematol Rep ; 4(4): 324-34, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16009048

RESUMEN

In recent years, several molecular mechanisms involved in promoting cancer cell survival and growth have been identified. These discoveries helped in designing and testing novel drugs that target specific cellular pathways. In this review, we focus on new molecular targets that are currently being explored for the treatment of non-Hodgkin's lymphoma and Hodgkin's lymphoma.


Asunto(s)
Linfoma/terapia , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales de Origen Murino , Antígenos de Neoplasias/uso terapéutico , Antígeno B7-1/uso terapéutico , División Celular/efectos de los fármacos , Humanos , Linfoma/genética , Linfoma/patología , Rituximab
17.
J Clin Invest ; 115(7): 1903-12, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15937544

RESUMEN

Immunotherapy for the treatment of metastatic melanoma remains a major clinical challenge. The melanoma microenvironment may lead to local T cell tolerance in part through downregulation of costimulatory molecules, such as B7.1 (CD80). We report the results from the first clinical trial, to our knowledge, using a recombinant vaccinia virus expressing B7.1 (rV-B7.1) for monthly intralesional vaccination of accessible melanoma lesions. A standard 2-dose-escalation phase I clinical trial was conducted with 12 patients. The approach was well tolerated with only low-grade fever, myalgias, and fatigue reported and 2 patients experiencing vitiligo. An objective partial response was observed in 1 patient and disease stabilization in 2 patients, 1 of whom is alive without disease 59 months following vaccination. All patients demonstrated an increase in postvaccination antibody and T cell responses against vaccinia virus. Systemic immunity was tested in HLA-A*0201 patients who demonstrated an increased frequency of gp100 and T cells specific to melanoma antigen recognized by T cells 1 (MART-1), also known as Melan-A, by ELISPOT assay following local rV-B7.1 vaccination. Local immunity was evaluated by quantitative real-time RT-PCR, which suggested that tumor regression was associated with increased expression of CD8 and IFN-gamma. The local delivery of vaccinia virus expressing B7.1 was well tolerated and represents an innovative strategy for altering the local tumor microenvironment in patients with melanoma.


Asunto(s)
Antígeno B7-1/genética , Terapia Genética , Inmunoterapia , Melanoma/terapia , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Adulto , Anciano , Antígenos de Neoplasias , Antígeno B7-1/uso terapéutico , Antígenos CD8/genética , Femenino , Expresión Génica , Antígenos HLA-A , Antígeno HLA-A2 , Humanos , Inyecciones Intralesiones , Interferón gamma/genética , Interleucina-10/genética , Antígeno MART-1 , Masculino , Melanoma/genética , Melanoma/inmunología , Melanoma/secundario , Persona de Mediana Edad , Proteínas de Neoplasias/inmunología , Linfocitos T/inmunología
18.
Cancer Gene Ther ; 11(3): 215-26, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14671675

RESUMEN

The activation of T cells plays a central role in antitumor immunity. In order to activate naïve T cells, two key signals are required. Signal one is provided through the T-cell receptor (TCR) while signal two is that of costimulation. The CD28:B7 molecules are one of the best-studied costimulatory pathways, thought to be the main mechanism through which primary T-cell stimulation occurs. However, a number of molecules have been identified which serve to amplify and diversify the T-cell response, following initial T-cell activation. These include the more recently described 4-1BB:4-1BB ligand (4-1BBL) molecules. 4-1BB:4-1BBL are a member of the TNFR:TNF ligand family, which are expressed on T cells and antigen-presenting cells (APCs), respectively. Therapies utilizing the 4-1BB:4-1BBL signaling pathway have been shown to have antitumor effects in a number of model systems. In this paper, we focus on the 4-1BB:4-1BBL costimulatory molecules. In particular, we will describe the structure and function of the 4-1BB molecule, its receptor and how 4-1BB:4-1BBL costimulation has and may be used for the immunotherapy of cancer.


Asunto(s)
Neoplasias/terapia , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ligando 4-1BB , Secuencia de Aminoácidos , Animales , Antígenos CD , Antígeno B7-1/metabolismo , Antígeno B7-1/uso terapéutico , Antígenos CD28/metabolismo , Vacunas contra el Cáncer/metabolismo , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Ratones , Datos de Secuencia Molecular , Neoplasias/inmunología , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/fisiología
19.
Leuk Lymphoma ; 44(10): 1775-84, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14692533

RESUMEN

Increasing evidence suggests a role for immunologic vaccination and therapy in the management of minimal residual myeloma. We have previously demonstrated a synergistic effect of combining the Th1 stimulating cytokine IL-12 with the co-stimulatory molecule CD80 in murine myeloma vaccination therapy. We reasoned that the efficacy of such treatment might be further improved by incorporating additional gene products which enhance the function of antigen presenting cells. Studies were therefore conducted with murine myeloma BM1 cells expressing Flt3L (membrane bound or soluble forms) or GM-CSF and the IL-12 x CD80 combination. Single agent and combined therapeutic approaches were explored. All gene-modified BM1 cells, except BM1/IL-12 x CD80, developed tumors when subcutaneously injected into BALB/c mice. As prophylactic tumor vaccines, the combined use of gene-modified BM1/sFlt3L+GM-CSF+IL-12 x CD80 was most effective, providing 100% protection against subsequent parental BM1 tumor challenge. By comparison, only partial protection was observed with any single gene-engineered tumor vaccine. Notably, IL-12 x CD80 coexpressing BM1 cell vaccines were the most effective therapeutic vaccine in a minimal disease model. Such protective vaccination was achieved by stimulation of lymphocyte proliferation and enhancement of cytotoxic lymphocyte activity.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Terapia Genética , Inmunoterapia , Mieloma Múltiple/prevención & control , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/uso terapéutico , Animales , Antígeno B7-1/metabolismo , Antígeno B7-1/uso terapéutico , Vacunas contra el Cáncer/metabolismo , División Celular , Terapia Combinada , Citotoxicidad Inmunológica , Femenino , Técnicas de Transferencia de Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Interleucina-12/metabolismo , Interleucina-12/uso terapéutico , Ligandos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Mieloma Múltiple/inmunología , Mieloma Múltiple/terapia , Retroviridae/genética , Bazo/inmunología , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas , Vacunación
20.
Cell Immunol ; 225(1): 53-63, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14643304

RESUMEN

Increasing evidence suggests that B7/CD28 interactions are important in clonal expansion and effector function of nai;ve CD4(+) T cells, whereas ICOS/GL50 interactions may optimize the responses of recently activated T(H) cells. In tumor models, it has been shown that engagement of ICOS, like CD28, by its ligands can be effective in enhancing tumor immunity. In this report, we have directly compared the in vivo efficacy of CD28 vs ICOS activation in the MethA fibrosarcoma and B16F1 melanoma tumor models. We studied the efficacy of systemic treatment of tumors with murine B7.2-IgG or GL50-IgG fusion proteins, and the therapeutic potential of B7.1 or GL50 vaccines given during various phases of the antitumor responses. In addition, we compare the efficacy of ICOS-ligand splice variants GL50 and GL50B in promoting tumor immunity. We find that each of these pathways is equally effective in promoting tumor immunity and that the efficacy of both GL50 and B7.1 vaccines is IFN-gamma but not IL-10 dependent. Our results suggest that CD28 or ICOS costimulation-based strategies may be equally efficacious as adjuvants to conventional cancer treatment.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Antígeno B7-1/inmunología , Antígenos CD28/inmunología , Neoplasias Experimentales/inmunología , Proteínas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Diferenciación de Linfocitos T/uso terapéutico , Antígeno B7-1/uso terapéutico , Antígenos CD28/uso terapéutico , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Memoria Inmunológica/inmunología , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Neoplasias Experimentales/terapia , Proteínas/uso terapéutico , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Linfocitos T Citotóxicos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA