Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Immunol ; 183(6): 3770-7, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19710474

RESUMEN

Rheumatoid arthritis (RA) is characterized by inflammation and cellular proliferation in the synovial lining of joints that result in cartilage and bone destruction. Although the etiology of RA is unclear, activated lymphocytes and proinflammatory molecules, in particular TNF superfamily members, have been implicated in the disease pathology. A TNF superfamily member, CD70, is found on activated lymphocytes and shown to be important in memory and effector responses of lymphocytes. CD70 is expressed at high levels on chronically activated T cells in patients with autoimmune disorders, including RA. The involvement of CD70 in the progression of RA, however, remains unknown. In this study, we report effects of targeting CD70 on disease pathogenesis by using an anti-mouse CD70 Ab in a murine model of collagen-induced arthritis (CIA). In addition to blocking CD70 binding to its receptor CD27, the anti-CD70 Ab used also engages Fc-dependent effector functions including Ab-dependent cellular cytotoxicity, phagocytosis, and complement fixation. Treatment of mice with anti-CD70 Ab both before the onset or after the established disease in CIA model resulted in marked improvements in disease severity and significant reduction in the production of autoantibodies. Histopathological analyses of the joints of mice revealed a substantial reduction of inflammation, and bone and cartilage destruction in response to the anti-CD70 Ab treatment. These results uncover a novel role for CD27-CD70 interactions in the regulation of in vivo inflammatory response leading to arthritis, and provide a molecular basis to support the rationale for anti-CD70 therapy for autoimmune and inflammatory diseases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Artritis Experimental/tratamiento farmacológico , Ligando CD27/antagonistas & inhibidores , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Animales , Artritis Experimental/patología , Autoanticuerpos/análisis , Huesos/efectos de los fármacos , Huesos/patología , Cartílago/efectos de los fármacos , Cartílago/patología , Progresión de la Enfermedad , Inflamación/prevención & control , Artropatías , Masculino , Ratones
2.
Cancer Sci ; 101(1): 224-30, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19799612

RESUMEN

Adult T-cell leukemia (ATL) is an aggressive malignancy of activated CD4(+) T cells associated with human T-cell leukemia virus type I (HTLV-I) infection. No conventional chemotherapy regimen has appeared successful in patients with ATL, thus establishing effective therapy is urgently required. In some cases, ATL tumor cells express CD30 on the cell surface, therefore, a therapy with mAb against CD30 would be beneficial. To investigate the effect of CD30-mediated therapy on ATL, we assessed SGN-30, a chimeric anti-CD30 mAb, and SGN-35, a monomethyl auristatin E-conjugated anti-CD30 mAb, in vitro and in vivo. Three HTLV-I-infected cell lines were co-cultured with SGN-30 or SGN-35, and the growth-inhibitory effects on the HTLV-I-infected cells were evaluated using an in vitro cell proliferation assay and cell cycle analysis. SGN-30 and SGN-35 showed growth-inhibitory activity against the HTLV-I-infected cell lines by apoptosis and/or cell growth arrest in vitro. To further investigate the effects of SGN-30 and SGN-35 on HTLV-I-infected cells in vivo, we used NOD/SCID mice subcutaneously engrafted with HTLV-I-infected cells. Both mAbs significantly inhibited the growth of HTLV-I-infected cell tumors in the NOD/SCID murine xenograft models. These data suggest that CD30-mediated therapy with SGN-30 or SGN-35 would be useful for patients with ATL.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Infecciones por HTLV-I/tratamiento farmacológico , Antígeno Ki-1/inmunología , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Humanos , Ratones
3.
Clin Cancer Res ; 14(19): 6171-80, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18809969

RESUMEN

PURPOSE: The antitubulin agent monomethyl auristatin F (MMAF) induces potent antitumor effects when conjugated via protease cleavable linkers to antibodies targeting internalizing, tumor-specific cell surface antigens. Humanized 1F6 (h1F6) is a humanized monoclonal antibody targeting CD70, a member of the tumor necrosis factor family that is expressed on hematologic malignancies and carcinomas. Here, we tested h1F6-maleimidocaproyl (mc) MMAF conjugates, consisting of an uncleavable mc linker, for their ability to interfere with the growth of CD70-positive carcinomas. EXPERIMENTAL DESIGN: To evaluate the optimal drug per antibody ratio, we conjugated either four or eight MMAF molecules to the cysteines that comprise the interchain disulfides of h1F6 and determined antitumor activities in vitro and in xenografted mice. The tumor types tested included glioblastoma, patient-derived renal cell carcinoma (RCC) cell isolates, and standard RCC tumor cell lines. RESULTS: All h1F6-mcMMAF conjugates potently interfered with the growth of all carcinomas in vitro and resulted in complete responses of RCC tumors implanted orthotopically or s.c. in mice. In vitro, h1F6-mcMMAF(8) was generally more potent than h1F6-mcMMAF(4). However, h1F6-mcMMAF(4) displayed equal or better efficacy than h1F6-mcMMAF(8) when administered to tumor-bearing mice. CONCLUSIONS: We showed that h1F6-mcMMAF conjugates inhibited the growth of human carcinomas and that increased drug loading, while improving potency in vitro, did not substantially affect the pharmacodynamic and pharmacokinetic properties in vivo. Based on these findings, h1F6-mcMMAF(4), designated SGN-75, has been identified as a potential antibody-drug conjugate for clinical development.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/química , Antineoplásicos/farmacología , Ligando CD27/química , Ligando CD27/inmunología , Oligopéptidos/farmacología , Animales , Anticuerpos Monoclonales Humanizados/química , Antineoplásicos/química , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Disulfuros/química , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Concentración 50 Inhibidora , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Ratones , Trasplante de Neoplasias , Oligopéptidos/química , Tubulina (Proteína)/química , Moduladores de Tubulina/química
4.
Adv Exp Med Biol ; 647: 174-85, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19760074

RESUMEN

The transmembrane receptor CD30 (TNFRSF8) and its ligand CD30L (CD153, TNFSF8) are members of the tumor necrosis factor (TNF) superfamily and display restricted expression in subpopulations of activated T-and B-cells in nonpathologic conditions. CD30 expression is upregulated in various hematological malignancies, including Reed-Sternberg cells in Hodgkin's disease (HD), anaplastic large cell lymphoma (ALCL) and subsets of Non-Hodgkin's lymphomas (NHLs). Increased CD30L expression was found on mast cells within HD tumors and preclinical and clinical studies with compounds targeting the CD30/ CD30L system in HD and ALCL demonstrated therapeutic benefit. Upregulation of CD30 and CD30L is also linked to leukocytes in patients with chronic inflammatory diseases, including lupus erythematosus, asthma, rheumatoid arthritis and atopic dermatitis (AD). Preclinical studies conducted with transgenic mice or biologic compounds suggested important regulatory functions of the CD30-CD30L system in various aspects of the immune system. Such key regulatory roles and their low expression in normal conditions combined with increased expression in malignant tissues provided a strong rationale to investigate CD30 and CD30L as therapeutic targets in hematologic malignancies, autoimmune and inflammatory diseases. In this report, we review the pharmacodynamic effects of specific therapeutic compounds targeting the CD30/CD30L system in preclinical- and clinical studies.


Asunto(s)
Enfermedades Autoinmunes/terapia , Ligando CD30/antagonistas & inhibidores , Inflamación/terapia , Antígeno Ki-1/antagonistas & inhibidores , Neoplasias/terapia , Animales , Enfermedades Autoinmunes/inmunología , Ligando CD30/metabolismo , Humanos , Inflamación/inmunología , Antígeno Ki-1/metabolismo , Ratones , Neoplasias/inmunología
5.
Br J Haematol ; 142(1): 69-73, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18477046

RESUMEN

The antibody-drug conjugate (ADC) cAC10-vcMMAE consists of the tubulin inhibitor monomethyl auristatin E (MMAE) conjugated to the chimeric anti-CD30 monoclonal antibody cAC10. This ADC potently interferes with the growth of CD30-positive haematological tumours, including Hodgkin lymphoma (HL) and anaplastic large-cell lymphoma. This study found improved antitumour activity in a preclinical model of HL when SGN-35 was combined with chemotherapeutic regimens such as ABVD (doxorubicin, bleomycin, vinblastine and dacarbazine) or gemcitabine. Improved efficacy was also observed in high tumour burden models, indicating that combining ADCs with chemotherapeutic agents may be advantageous for the treatment of patients with relapsed or refractory HL.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Enfermedad de Hodgkin/terapia , Inmunoconjugados/administración & dosificación , Antígeno Ki-1/administración & dosificación , Oligopéptidos/administración & dosificación , Moduladores de Tubulina/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Bleomicina/administración & dosificación , Dacarbazina/administración & dosificación , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Doxorrubicina/administración & dosificación , Ratones , Ratones SCID , Trasplante de Neoplasias , Vinblastina/administración & dosificación , Gemcitabina
6.
J Leukoc Biol ; 82(5): 1322-31, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17711975

RESUMEN

Circulating NK cells normally experience temperature gradients as they move about the body, but the onset of inflammation can expose them and their targets to febrile temperatures for several hours. We found that exposure of human peripheral blood NK cells and target cells to fever-range temperatures significantly enhances lysis of Colo205 target cells. A similar effect was not observed when NK cell lines or IL-2-activated peripheral blood NK cells were used as effectors, indicating that thermal sensitivity of effectors is maturation or activation state-dependent. Use of blocking antibodies revealed that this effect is also dependent on the function of the activating receptor NKG2D and its ligand MHC class I-related chain A (MICA). On NK cells, it was observed that thermal exposure does not affect the total level of NKG2D surface expression, but does result in its distinct clustering, identical to that which occurs following IL-2-induced activation. On tumor target cells, a similar, mild temperature elevation results in transcriptional up-regulation of MICA in a manner that correlates with increased sensitivity to cytolysis. Overall, these data reveal that NK cells possess thermally responsive regulatory elements, which facilitate their ability to capitalize on reciprocal, stress-induced changes simultaneously occurring on target cells during inflammation and fever.


Asunto(s)
Membrana Celular/metabolismo , Supervivencia Celular/inmunología , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad Clase I/genética , Calor , Células Asesinas Naturales/citología , Receptores Inmunológicos/metabolismo , Comunicación Celular , Supervivencia Celular/fisiología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Vigilancia Inmunológica , Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK , Neoplasias/metabolismo , Neoplasias/patología , Transporte de Proteínas , Receptores Inmunológicos/genética , Receptores de Células Asesinas Naturales , Elementos Reguladores de la Transcripción , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Eur J Dermatol ; 18(1): 41-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18086588

RESUMEN

Atopic dermatitis (AD) is a chronic inflammatory skin disease associated with cutaneous hyperreactivity to environmental stimuli, resulting in increased infiltration of inflammatory cells, IgE production and enhanced expression of costimulatory molecules, cytokines and chemokines. CD30, a TNF receptor superfamily member, is a costimulatory molecule expressed on activated T and B cells. A positive correlation between soluble CD30 (sCD30) levels in patient serum and AD disease severity has been described previously. However, the relative frequencies and identities of cells expressing CD30 in AD patients and the relationship between the frequency of CD30 positive cells and serum sCD30 levels with disease severity remained unknown. To address these questions, immunofluorescence analysis of AD skin lesions representing different disease stages, was conducted. In addition to the CD4+ T cells, CD1a+ Langerhans cells and CD8+ T cells were found to express CD30 in AD lesions and the cell numbers correlated with disease severity. FACS analysis of AD patient blood samples revealed expression of CD30 on memory T-cells and a correlation with disease severity was identified. Finally, serum analysis of soluble mediators revealed positive correlations between sCD30, IgE, MDC, TARC and PARC levels with disease severity. Combined, our data provide correlative evidence that CD30+ cells, including Langerhans cells and CD8+ T-cells, may contribute to AD disease severity and that therapeutic strategies targeting CD30+ cells may provide benefit to AD patients.


Asunto(s)
Antígenos CD1/análisis , Linfocitos T CD8-positivos/inmunología , Dermatitis Atópica/inmunología , Antígeno Ki-1/análisis , Células de Langerhans/inmunología , Antígenos CD4/análisis , Quimiocina CCL17/sangre , Quimiocinas CC/sangre , Dermatitis Atópica/patología , Humanos , Inmunohistoquímica , Interleucina-8/sangre , Antígeno Ki-1/sangre , Piel/inmunología , Piel/patología
8.
Eur J Dermatol ; 18(5): 527-33, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18693155

RESUMEN

Atopic dermatitis (AD) is a chronic inflammatory skin disease associated with cutaneous hyperreactivity to environmental stimuli, resulting in increased infiltration of inflammatory cells, IgE production and enhanced expression of various co-stimulatory molecules, Th2 cytokines and chemokines. Antigen presenting cells (APCs) are critical for AD disease pathogenesis and interaction between APCs and inflammatory T cells represents an important signal required for induction and maintenance of the inflammatory process. CD40 was shown to be upregulated on inflammatory cells in patients with atopic dermatitis; however the identity of these cells and their correlation with disease severity remained unknown. To address these questions, we determined CD40 expression in skin lesions and on peripheral blood cells from AD patients and identified a positive correlation between the numbers of CD40 positive cells and disease severity. Furthermore, we identified the nature of CD40 positive cells in skin lesions to include CD1a+ and CD11b+ APCs. Finally, a correlation between serum PARC and IgE levels and the numbers of CD40+ cells in AD skin lesions was found. Combined, these findings demonstrate that CD40 is upregulated on APCs, cell types previously shown to contribute to AD disease pathology, and suggest that therapeutic strategies targeting CD40 positive cells may provide benefit to AD patients.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos CD40/biosíntesis , Dermatitis Atópica/inmunología , Humanos , Índice de Severidad de la Enfermedad
9.
Cancer Res ; 66(4): 2328-37, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16489038

RESUMEN

Metastatic renal cell carcinoma (RCC) is an aggressive disease refractory to most existing therapeutic modalities. Identifying new markers for disease progression and drug targets for RCC will benefit this unmet medical need. We report a subset of clear cell and papillary cell RCC aberrantly expressing the lymphocyte activation marker CD70, a member of the tumor necrosis factor superfamily. Importantly, CD70 expression was found to be maintained at the metastatic sites of RCC. Anti-CD70 antibody-drug conjugates (ADC) consisting of auristatin phenylalanine phenylenediamine (AFP) or monomethyl auristatin phenylalanine (MMAF), two novel derivatives of the anti-tubulin agent auristatin, mediated potent antigen-dependent cytotoxicity in CD70-expressing RCC cells. Cytotoxic activity of these anti-CD70 ADCs was associated with their internalization and subcellular trafficking through the endosomal-lysosomal pathway, disruption of cellular microtubule network, and G2-M phase cell cycle arrest. The efficiency of drug delivery using anti-CD70 as vehicle was illustrated by the much enhanced cytotoxicity of antibody-conjugated MMAF compared with free MMAF. Hence, ADCs targeted to CD70 can selectively recognize RCC, internalize, and reach the appropriate subcellular compartment(s) for drug release and tumor cell killing. In vitro cytotoxicity of these ADCs was confirmed in xenograft models using RCC cell lines. Our findings provide evidence that CD70 is an attractive target for antibody-based therapeutics against metastatic RCC and suggest that anti-CD70 ADCs can provide a new treatment approach for advanced RCC patients who currently have no chemotherapeutic options.


Asunto(s)
Antígenos CD/biosíntesis , Antineoplásicos/administración & dosificación , Carcinoma de Células Renales/inmunología , Inmunoconjugados/inmunología , Inmunoconjugados/farmacología , Neoplasias Renales/inmunología , Proteínas de la Membrana/biosíntesis , Factores de Necrosis Tumoral/biosíntesis , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Apoptosis/efectos de los fármacos , Ligando CD27 , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Ciclo Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Activación de Linfocitos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Desnudos , Microtúbulos/efectos de los fármacos , Oligopéptidos/administración & dosificación , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Transl Med ; 5: 29, 2007 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-17592641

RESUMEN

BACKGROUND: The anti-tumor efficacy of human immune effector cells, such as cytolytic T lymphocytes (CTLs), has been difficult to study in lung cancer patients in the clinical setting. Improved experimental models for the study of lung tumor-immune cell interaction as well as for evaluating the efficacy of adoptive transfer of immune effector cells are needed. METHODS: To address questions related to the in vivo interaction of human lung tumor cells and immune effector cells, we obtained an HLA class I (+) lung tumor cell line from a fresh surgical specimen, and using the infiltrating immune cells, isolated and characterized tumor antigen-specific, CD8(+) CTLs. We then established a SCID mouse-human tumor xenograft model with the tumor cell line and used it to study the function of the autologous CTLs provided via adoptive transfer. RESULTS: The tumor antigen specific CTLs isolated from the tumor were found to have an activated memory phenotype and able to kill tumor cells in an antigen specific manner in vitro. Additionally, the tumor antigen-specific CTLs were fully capable of homing to and killing autologous tumors in vivo, and expressing IFN-gamma, each in an antigen-dependent manner. A single injection of these CTLs was able to provide significant but temporary control of the growth of autologous tumors in vivo without the need for IL-2. The timing of injection of CTLs played an essential role in the outcome of tumor growth control. Moreover, immunohistochemical analysis of surviving tumor cells following CTL treatment indicated that the surviving tumor cells expressed reduced MHC class I antigens on their surface. CONCLUSION: These studies confirm and extend previous studies and provide additional information regarding the characteristics of CTLs which can be found within a patient's tumor. Moreover, the in vivo model described here provides a unique window for observing events that may also occur in patients undergoing adoptive cellular immunotherapy as effector cells seek and destroy areas of tumor growth and for testing strategies to improve clinical effectiveness.


Asunto(s)
Traslado Adoptivo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Linfocitos T Citotóxicos/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antígenos de Neoplasias/inmunología , Proliferación Celular , Citocinas/metabolismo , Citotoxicidad Inmunológica , Epítopos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Memoria Inmunológica , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Ratones SCID , Especificidad de Órganos , Fenotipo , Factores de Tiempo
11.
Br J Pharmacol ; 148(8): 1116-23, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16847437

RESUMEN

1. Cell-surface expression of CD40 in B-cell malignancies and multiple solid tumors has raised interest in its potential use as a target for antibody-based cancer therapy. SGN-40, a humanized monoclonal anti-CD40 antibody, mediates antibody-dependent cytotoxicity and inhibits B-cell tumor growth in vitro, properties of interest for the treatment of cancers, and is currently in Phase I clinical trials for B-cell malignancies. In this study, we determined in vivo activity and pharmacokinetics properties of SGN-40. 2. Effect of SGN-40 in xenograft model of CD40-expressing B-cell lymphoma in severe-combined immune deficiency mice and its in vivo pharmacokinetics properties in normal mice, rats and cynomolgus monkeys were studied. 3. Treatment with SGN-40 significantly increased the survival of mice xenografted with human B-cell lymphoma cell line. SGN-40 exhibited nearly 100% bioavailability in mice and it cleared faster when given at a low dose. In monkeys, clearance of SGN-40 was also much faster at low dose, suggesting nonlinear pharmacokinetics in these species. In rats, however, SGN-40 clearance at all tested doses was similar, suggesting that pharmacokinetics were linear in this dose range in rats. Administration of SGN-40 to monkeys also produced marked, dose-dependent, and persistent depletion of peripheral CD20(+) B lymphocytes. 4. Data presented in this report suggest that SGN-40 is active in in vivo, and based upon interspecies scaling, SGN-40 clearance in humans is predicted to be similar to observed SGN-40 clearance in monkeys. These data suggest that SGN-40 has appropriate pharmacokinetic properties that support its clinical use.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD40/inmunología , Animales , Anticuerpos Monoclonales/farmacocinética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Macaca fascicularis , Masculino , Ratones , Ratones SCID , Especificidad de la Especie
12.
Clin Cancer Res ; 9(16 Pt 1): 5866-73, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14676108

RESUMEN

PURPOSE: Rituximab is a chimeric antibody (Ab) directed against the cluster designated (CD) 20 antigen found on normal and malignant B cells. Rituximab activity has been associated with complement-mediated cytotoxicity, Ab-dependent cellular cytotoxicity (ADCC), and induction of apoptosis. Recent studies performed in severe combined immunodeficiency (SCID) mouse models suggest that in vivo rituximab-associated ADCC is mediated via the FcgammaRIII receptor on effector cells. Despite low level expression of FcgammaRIII, neutrophils are also known to induce ADCC primarily via FcgammaRI receptor (CD64). The purpose of this work was to study the effect(s) of neutrophils on the in vivo antitumor activity of rituximab. EXPERIMENTAL DESIGN: To better characterize the biological activity of rituximab, we used a human non-Hodgkin's lymphoma animal model by injecting Raji cells i.v. into natural killer (NK) cell-depleted SCID mice. Disseminated disease involving liver, lung, and central nervous system developed, with subsequent death occurring approximately 3 weeks after tumor inoculation. Specifically, 6-8-week-old NK cell-depleted SCID mice were inoculated by tail vein injection with 1 x 10(6) Raji cells on day 0. The animals then were divided into three cohorts: (a) group A received placebo (PBS); (b) group B received rituximab administered via tail vein injection at 10 mg/kg on days 3, 5, 7, and 11; and (c) group C consisted of neutrophil-depleted SCID mice treated with rituximab at 10 mg/kg on the same schedule. Neutrophils were depleted by i.p. administration of 80 microg of rat antimouse Ly-6G (Gr-1) Ab (BD PharMingen, Inc.) on days -1, 4, 9, and 14. The end point of the study was survival. Differences in outcome between treatment groups were analyzed by Kaplan-Meier methodology. RESULTS: Neutrophil- and NK cell-depleted SCID mice (group C) did not respond to rituximab, and the mean survival time was not significantly different from that of control mice. NK cell-depleted SCID mice with intact neutrophil function (group B) responded to rituximab, and 66% remained alive and appeared healthy after a mean follow-up period of 246 days. Overall, NK cell-depleted SCID mice with intact neutrophil function treated with rituximab had statistically longer mean survival as compared with mice in neutrophil-depleted and control groups (161 days versus 28 days versus 22 days, P=0.003). CONCLUSIONS: In the absence of neutrophils, rituximab was less effective in controlling lymphoma cell growth or prolonging survival in our B-cell lymphoma SCID mouse model. Neutrophil-induced ADCC appears to contribute to the in vivo antitumor activity of rituximab. Strategies that improve the function of neutrophils, such as granulocyte-macrophage colony-stimulating factor or G-CSF priming, may increase the antitumor effects of rituximab. Additional in vivo animal studies are warranted.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Células Asesinas Naturales/inmunología , Linfoma no Hodgkin/inmunología , Linfoma no Hodgkin/terapia , Neutrófilos/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos CD20/inmunología , Proteínas del Sistema Complemento/inmunología , Humanos , Linfoma no Hodgkin/patología , Ratones , Ratones SCID , Ratas , Rituximab , Tasa de Supervivencia , Células Tumorales Cultivadas
13.
MAbs ; 2(1): 14-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20081379

RESUMEN

Approval of an anti-CD20 chimeric monoclonal antibody, rituximab, has revolutionized cancer treatment and also validated CD20 targeting for providing benefit and improvement of overall response rate in B cell malignancies. Although many patients have benefited from the treatment of rituximab, there are still significant numbers of patients who are refractory or develop resistance to the treatment. Here we discuss pre-clinically well-defined potential mechanisms of action for rituximab and review the ways next generation anti-CD20 monoclonal antibodies can potentially exploit them to further enhance the treatment of B cell malignancies. Although the relative importance of each of these mechanism remains to be established in the clinic, well-designed clinical trials will help to define the efficacy and understanding of which effector activity of modified next generation anti-CD20 mAb will be important in the treatment of B-cell malignancies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/efectos de los fármacos , Inmunoterapia , Linfoma de Células B/tratamiento farmacológico , Oncología Médica , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales de Origen Murino , Apoptosis/inmunología , Linfocitos B/inmunología , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos , Humanos , Linfoma de Células B/inmunología , Rituximab , Transducción de Señal/inmunología
14.
Blood ; 110(13): 4370-2, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17909075

RESUMEN

Increased expression of CD30 is associated with a variety of hematologic malignancies, including Hodgkin disease (HD) and anaplastic large cell lymphoma (ALCL). The anti-CD30 monoclonal antibody SGN-30 induces direct antitumor activity by promoting growth arrest and DNA fragmentation of CD30(+) tumor cells. In this study, we investigated the contributions of Fc-mediated effector cell functions to SGN-30 activity. We determined that antibody-dependent cellular phagocytosis, mediated by macrophages, to contribute significantly to antitumor activity in vitro. To delineate the identity of the host effector cells involved in mediating antitumor activity in vivo, we studied the effects of effector cell ablation in a disseminated model of HD (L540cy). Depletion of macrophages markedly reduced efficacy of SGN-30, demonstrating that macrophages contribute significantly to SGN-30 efficacy in this model. These findings may have implications for patient stratification or combination treatment strategies in clinical trials conducted with SGN-30 in HD and ALCL.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedad de Hodgkin/terapia , Macrófagos/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Línea Celular Tumoral , Humanos , Antígeno Ki-1/inmunología , Ratones , Ratones SCID , Trasplante de Neoplasias , Fagocitosis/efectos de los fármacos , Trasplante Heterólogo
15.
Blood ; 109(3): 1185-92, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17038522

RESUMEN

Antigens expressed on malignant cells in the absence of significant expression on normal tissues are highly desirable targets for therapeutic antibodies. CD70 is a TNF superfamily member whose normal expression is highly restricted but is aberrantly expressed in hematologic malignancies including non-Hodgkin lymphoma (NHL), Hodgkin disease, and multiple myeloma. In addition, solid tumors such as renal cell carcinoma, nasopharyngeal carcinoma, thymic carcinoma, meduloblastoma, and glioblastoma express high levels of this antigen. To functionally target CD70-expressing cancers, a murine anti-CD70 monoclonal antibody was engineered to contain human IgG1 constant domains. The engineered antibody retained the binding specificity of the murine parent monoclonal antibody and was shown to induce Fc-mediated effector functions including antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis in vitro. Further, administration of this antibody significantly prolonged survival of severe combined immunodeficient (SCID) mice bearing CD70+ disseminated human NHL xenografts. Survival of these mice was dependent upon the activity of resident effector cells including neutrophils, macrophages, and natural killer (NK) cells. These data suggest that an anti-CD70 antibody, when engineered to contain human IgG1 constant domains, possesses effector cell-mediated antitumor activity and has potential utility for anticancer therapy.


Asunto(s)
Anticuerpos/uso terapéutico , Antineoplásicos , Ligando CD27/inmunología , Linfoma no Hodgkin/terapia , Ingeniería de Proteínas/métodos , Animales , Anticuerpos/genética , Anticuerpos/inmunología , Especificidad de Anticuerpos , Citotoxicidad Celular Dependiente de Anticuerpos , Línea Celular Tumoral , Proteínas del Sistema Complemento , Humanos , Sistema Inmunológico/citología , Inmunoglobulina G , Ratones , Ratones SCID , Fagocitosis , Tasa de Supervivencia , Trasplante Heterólogo
16.
Bioconjug Chem ; 17(1): 114-24, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16417259

RESUMEN

We have previously shown that antibody-drug conjugates (ADCs) consisting of cAC10 (anti-CD30) linked to the antimitotic agent monomethylauristatin E (MMAE) lead to potent in vitro and in vivo activities against antigen positive tumor models. MMAF is a new antimitotic auristatin derivative with a charged C-terminal phenylalanine residue that attenuates its cytotoxic activity compared to its uncharged counterpart, MMAE, most likely due to impaired intracellular access. In vitro cytotoxicity studies indicated that mAb-maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl-MMAF (mAb-L1-MMAF) conjugates were >2200-fold more potent than free MMAF on a large panel of CD30 positive hematologic cell lines. As with cAC10-L1-MMAE, the corresponding MMAF ADC induced cures and regressions of established xenograft tumors at well tolerated doses. To further optimize the ADC, several new linkers were generated in which various components within the L1 linker were either altered or deleted. One of the most promising linkers contained a noncleavable maleimidocaproyl (L4) spacer between the drug and the mAb. cAC10-L4-MMAF was approximately as potent in vitro as cAC10-L1-MMAF against a large panel of cell lines and was equally potent in vivo. Importantly, cAC10-L4-MMAF was tolerated at >3 times the MTD of cAC10-L1-MMAF. LCMS studies indicated that drug released from cAC10-L4-MMAF was the cysteine-L4-MMAF adduct, which likely arises from mAb degradation within the lysosomes of target cells. This new linker technology appears to be ideally suited for drugs that are both relatively cell-impermeable and tolerant of substitution with amino acids. Thus, alterations of the linker have pronounced impacts on toxicity and lead to new ADCs with greatly improved therapeutic indices.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Concentración 50 Inhibidora , Linfoma de Células B Grandes Difuso/patología , Dosis Máxima Tolerada , Ratones , Ratones SCID , Oligopéptidos/administración & dosificación , Oligopéptidos/síntesis química , Resultado del Tratamiento , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Eur J Immunol ; 34(12): 3326-36, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15484188

RESUMEN

Programmed death-1 ligand 2 (PD-L2) is a ligand for programmed death-1 (PD-1), a receptor that plays an inhibitory role in T cell activation. Since previous studies have shown up-regulation of PD-L2 expression by Th2 cytokines, and asthma is driven by a Th2 response, we hypothesized that PD-L2 might be involved in regulation of the immune response in this disease. We have found that lungs from asthmatic mice had sustained up-regulation of PD-1 and PD-L2, with PD-L2 primarily on dendritic cells. Although addition of PD-L2-Fc in vitro led to decreased T cell proliferation and cytokine production, administration of PD-L2-Fc in vivo in a mouse asthma model resulted in elevated serum IgE levels, increased eosinophilic and lymphocytic infiltration into bronchoalveolar lavage fluid, higher number of cells in the draining lymph nodes, and production of IL-5 and IL-13 from these cells. Although PD-1 was expressed on regulatory T cells, PD-L2-Fc did not affect regulatory T cell activity in vitro. This study provides in vivo evidence of an exacerbated inflammatory response following PD-L2-Fc administration and indicates a potential role for this molecule in Th2-mediated diseases such as asthma.


Asunto(s)
Antígenos de Superficie/inmunología , Asma/inmunología , Pulmón/inmunología , Péptidos/inmunología , Células Th2/inmunología , Animales , Antígenos CD/inmunología , Proteínas Reguladoras de la Apoptosis , Antígeno B7-1/inmunología , Antígeno B7-2 , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Glicoproteínas de Membrana/inmunología , Ratones , Proteína 2 Ligando de Muerte Celular Programada 1 , Receptor de Muerte Celular Programada 1 , Células Th2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA