Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Immunology ; 141(1): 123-31, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24116901

RESUMEN

Alemtuzumab is a humanized monoclonal antibody specific for the CD52 protein present at high levels on the surface of B and T lymphocytes. In clinical trials, alemtuzumab has shown a clinical benefit superior to that of interferon-ß in relapsing-remitting multiple sclerosis patients. Treatment with alemtuzumab leads to the depletion of circulating lymphocytes followed by a repopulation process characterized by alterations in the number, proportions and properties of lymphocyte subsets. Of particular interest, an increase in the percentage of T cells with a regulatory phenotype (Treg cells) has been observed in multiple sclerosis patients after alemtuzumab. Since Treg cells play an important role in the control of autoimmune responses, the effect of alemtuzumab on Treg cells was further studied in vitro. Alemtuzumab effectively mediated complement-dependent cytolysis of human T lymphocytes and the remaining population was enriched in T cells with a regulatory phenotype. The alemtuzumab-exposed T cells displayed functional regulatory characteristics including anergy to stimulation with allogeneic dendritic cells and ability to suppress the allogeneic response of autologous T cells. Consistent with the observed increase in Treg cell frequency, the CD25(hi) T-cell population was necessary for the suppressive activity of alemtuzumab-exposed T cells. The mechanism of this suppression was found to be dependent on both cell-cell contact and interleukin-2 consumption. These findings suggest that an alemtuzumab-mediated increase in the proportion of Treg cells may play a role in promoting the long-term efficacy of alemtuzumab in patients with multiple sclerosis.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Alemtuzumab , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Antígeno CD52 , Comunicación Celular/efectos de los fármacos , Comunicación Celular/inmunología , Supervivencia Celular , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Glicoproteínas/inmunología , Humanos , Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Masculino , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Linfocitos T Reguladores/patología
2.
Acta Neuropathol Commun ; 11(1): 200, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38111057

RESUMEN

TAR DNA-binding protein 43 (TDP-43) and Fused in Sarcoma/Translocated in Sarcoma (FUS) are ribonucleoproteins associated with pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Under physiological conditions, TDP-43 and FUS are predominantly localized in the nucleus, where they participate in transcriptional regulation, RNA splicing and metabolism. In disease, however, they are typically mislocalized to the cytoplasm where they form aggregated inclusions. A number of shared cellular pathways have been identified that contribute to TDP-43 and FUS toxicity in neurodegeneration. In the present study, we report a novel pathogenic mechanism shared by these two proteins. We found that pathological FUS co-aggregates with a ribosomal protein, the Receptor for Activated C-Kinase 1 (RACK1), in the cytoplasm of spinal cord motor neurons of ALS, as previously reported for pathological TDP-43. In HEK293T cells transiently transfected with TDP-43 or FUS mutant lacking a functional nuclear localization signal (NLS; TDP-43ΔNLS and FUSΔNLS), cytoplasmic TDP-43 and FUS induced co-aggregation with endogenous RACK1. These co-aggregates sequestered the translational machinery through interaction with the polyribosome, accompanied by a significant reduction of global protein translation. RACK1 knockdown decreased cytoplasmic aggregation of TDP-43ΔNLS or FUSΔNLS and alleviated associated global translational suppression. Surprisingly, RACK1 knockdown also led to partial nuclear localization of TDP-43ΔNLS and FUSΔNLS in some transfected cells, despite the absence of NLS. In vivo, RACK1 knockdown alleviated retinal neuronal degeneration in transgenic Drosophila melanogaster expressing hTDP-43WT or hTDP-43Q331K and improved motor function of hTDP-43WT flies, with no observed adverse effects on neuronal health in control knockdown flies. In conclusion, our results revealed a novel shared mechanism of pathogenesis for misfolded aggregates of TDP-43 and FUS mediated by interference with protein translation in a RACK1-dependent manner. We provide proof-of-concept evidence for targeting RACK1 as a potential therapeutic approach for TDP-43 or FUS proteinopathy associated with ALS and FTLD.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Degeneración Lobar Frontotemporal , Sarcoma , Animales , Humanos , Esclerosis Amiotrófica Lateral/patología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HEK293 , Neuronas Motoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/patología , Degeneración Lobar Frontotemporal/patología , Biosíntesis de Proteínas , Sarcoma/metabolismo , Sarcoma/patología , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/genética
3.
Biomedicines ; 10(9)2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36140270

RESUMEN

Misfolded toxic forms of alpha-synuclein (α-Syn) have been implicated in the pathogenesis of synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). The α-Syn oligomers and soluble fibrils have been shown to mediate neurotoxicity and cell-to-cell propagation of pathology. To generate antibodies capable of selectively targeting pathogenic forms of α-Syn, computational modeling was used to predict conformational epitopes likely to become exposed on oligomers and small soluble fibrils, but not on monomers or fully formed insoluble fibrils. Cyclic peptide scaffolds reproducing these conformational epitopes exhibited neurotoxicity and seeding activity, indicating their biological relevance. Immunization with the conformational epitopes gave rise to monoclonal antibodies (mAbs) with the desired binding profile showing selectivity for toxic α-Syn oligomers and soluble fibrils, with little or no reactivity with monomers, physiologic tetramers, or Lewy bodies. Recognition of naturally occurring soluble α-Syn aggregates in brain extracts from DLB and MSA patients was confirmed by surface plasmon resonance (SPR). In addition, the mAbs inhibited the seeding activity of sonicated pre-formed fibrils (PFFs) in a thioflavin-T fluorescence-based aggregation assay. In neuronal cultures, the mAbs protected primary rat neurons from toxic α-Syn oligomers, reduced the uptake of PFFs, and inhibited the induction of pathogenic phosphorylated aggregates of endogenous α-Syn. Protective antibodies selective for pathogenic species of α-Syn, as opposed to pan α-Syn reactivity, are expected to provide enhanced safety and therapeutic potency by preserving normal α-Syn function and minimizing the diversion of active antibody from the target by the more abundant non-toxic forms of α-Syn in the circulation and central nervous system.

4.
Immunology ; 128(2): 260-70, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19740383

RESUMEN

Alemtuzumab is a humanized monoclonal antibody against CD52, an antigen found on the surface of normal and malignant lymphocytes. It is approved for the treatment of B-cell chronic lymphocytic leukaemia and is undergoing Phase III clinical trials for the treatment of multiple sclerosis. The exact mechanism by which alemtuzumab mediates its biological effects in vivo is not clearly defined and mechanism of action studies have been hampered by the lack of cross-reactivity between human and mouse CD52. To address this issue, a transgenic mouse expressing human CD52 (hCD52) was created. Transgenic mice did not display any phenotypic abnormalities and were able to mount normal immune responses. The tissue distribution of hCD52 and the level of expression by various immune cell populations were comparable to those seen in humans. Treatment with alemtuzumab replicated the transient increase in serum cytokines and depletion of peripheral blood lymphocytes observed in humans. Lymphocyte depletion was not as profound in lymphoid organs, providing a possible explanation for the relatively low incidence of infection in alemtuzumab-treated patients. Interestingly, both lymphocyte depletion and cytokine induction by alemtuzumab were largely independent of complement and appeared to be mediated by neutrophils and natural killer cells because removal of these populations with antibodies to Gr-1 or asialo-GM-1, respectively, strongly inhibited the activity of alemtuzumab whereas removal of complement by treatment with cobra venom factor had no impact. The hCD52 transgenic mouse appears to be a useful model and has provided evidence for the previously uncharacterized involvement of neutrophils in the activity of alemtuzumab.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Antineoplásicos/inmunología , Glicoproteínas/metabolismo , Depleción Linfocítica/métodos , Modelos Animales , Adenoviridae/inmunología , Alemtuzumab , Animales , Anticuerpos Monoclonales Humanizados , Anticuerpos Antivirales/biosíntesis , Antígeno CD52 , Citocinas/biosíntesis , Citocinas/sangre , Relación Dosis-Respuesta Inmunológica , Humanos , Tejido Linfoide/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/inmunología , Bazo/inmunología , Distribución Tisular
5.
Sci Rep ; 9(1): 9870, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285517

RESUMEN

Advances in the understanding of Alzheimer's disease (AD) suggest that pathogenesis is not directly related to plaque burden, but rather to soluble toxic amyloid-beta oligomers (AßO). Therapeutic antibodies targeting Aß monomers and/or plaque have shown limited efficacy and dose-limiting adverse events in clinical trials. These findings suggest that antibodies capable of selectively neutralizing toxic AßO may achieve improved efficacy and safety. To this end, we generated monoclonal antibodies against a conformational Aß epitope predicted by computational modeling to be presented on toxic AßO but not monomers or fibrils. The resulting lead antibody, PMN310, showed the desired AßO-selective binding profile. In vitro, PMN310 inhibited AßO propagation and toxicity. In vivo, PMN310 prevented AßO-induced loss of memory formation and reduced synaptic loss and inflammation. A humanized version (huPMN310) compared favorably to other Aß-directed antibodies showing a lack of adverse event-associated binding to Aß deposits in AD brains, and greater selective binding to AßO-enriched AD brain fractions that contain synaptotoxic Aß species. Systemic administration of huPMN310 in mice resulted in brain exposure and kinetics comparable to those of other therapeutic human monoclonal antibodies. Greater selectivity for AßO and the potential to safely administer high doses of huPMN310 are expected to result in enhanced safety and therapeutic potency.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/inmunología , Niño , Cognición/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Placa Amiloide/inmunología , Adulto Joven
6.
J Immunol Methods ; 333(1-2): 51-60, 2008 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-18314132

RESUMEN

The efficacy and mechanism of action of therapeutic antibodies that target cancer cells have typically been evaluated using in vitro assays and long-term in vivo tumor models. To allow for a more efficient assessment of the function of candidate therapeutic antibodies, we have developed a flow cytometric-based method that rapidly and directly quantifies antibody-mediated killing in a short term in vivo assay. Target cells that express human CD52, including huCD52(+) splenocytes from huCD52 transgenic mice and Ramos cells, a CD52(+) human B cell lymphoma line, and CD52(-) reference cells were differentially labeled by using two fluorescent dyes to distinguish target and reference cell populations. Labeled cells were injected into mice with or without Campath-1H (Alemtuzumab) and then recovered for flow cytometric analysis 5 h later. We found that huCD52(+) transgenic splenocytes and Ramos cells were selectively depleted in Campath-treated animals but not in animals treated with a negative control antibody. Furthermore, it is likely that the cells were depleted in vivo by a complement-dependent mechanism since target cell depletion was significantly reversed after complement inactivation using cobra venom factor. This report demonstrates the feasibility and utility of a powerful method for the rapid evaluation in vivo of therapeutic antibody candidates for cancer.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Anticuerpos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Citometría de Flujo/métodos , Linfoma de Células B/inmunología , Alemtuzumab , Animales , Anticuerpos/inmunología , Anticuerpos Monoclonales Humanizados , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Antígeno CD52 , Línea Celular Tumoral , Colorantes Fluorescentes/química , Glicoproteínas/inmunología , Humanos , Inmunoterapia/métodos , Linfoma de Células B/terapia , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Organismos Libres de Patógenos Específicos
7.
Curr Gene Ther ; 5(6): 595-605, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16457649

RESUMEN

Over the past decade, adenovirus (Ad)-based vectors have been used extensively in the context of cancer gene therapy. Two basic strategies have been pursued for the use of Ad vectors in cancer gene therapy: 1) approaches aimed at direct tumor cell killing through delivery of replicating oncolytic viruses or non-replicating vectors encoding tumor suppressor genes, suicide genes or anti-angiogenic genes, and 2) immunotherapeutic approaches aimed at inducing host anti-tumor immune responses that can destroy tumor cells at both primary and metastatic locations. Both strategies offer the potential of selective tumor cell destruction without damage to normal tissues. Extensive pre-clinical and clinical studies have been conducted based on these strategies. Encouraging results have been obtained but robust clinical efficacy remains elusive. Several obstacles limiting the therapeutic activity of Ad vectors have been encountered, including efficiency of tumor cell transduction and inhibition of efficacy by anti-Ad host immune responses. However, expanding knowledge in the areas of Ad biology and tumor biology continues to lead to increasingly sophisticated approaches to address these issues. A review of various Ad-based cancer gene therapy approaches and recent progress in the area are presented herein.


Asunto(s)
Adenovirus Humanos/genética , Terapia Genética/métodos , Vectores Genéticos , Neoplasias/terapia , Adenovirus Humanos/inmunología , Animales , Terapia Combinada , Vectores Genéticos/inmunología , Vectores Genéticos/uso terapéutico , Humanos , Inmunoterapia/métodos , Modelos Biológicos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/radioterapia
8.
J Neuroimmunol ; 285: 4-12, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26198912

RESUMEN

Alemtuzumab, a monoclonal antibody directed against human CD52, is used in the treatment of MS. To characterize the impact of anti-CD52 administration, a monoclonal antibody to mouse CD52 (anti-muCD52) was generated and evaluated in EAE mouse models of MS. A single course of anti-muCD52 provided a therapeutic benefit accompanied by a reduction in the frequency of autoreactive T lymphocytes and production of pro-inflammatory cytokines. Examination of the CNS revealed a decrease in infiltrating lymphocytes, demyelination and axonal loss. Electrophysiological assessment showed preservation of axonal conductance in the spinal cord. These findings suggest that anti-CD52 therapy may help preserve CNS integrity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Glicoproteínas/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Axones/efectos de los fármacos , Axones/inmunología , Axones/patología , Antígeno CD52 , Enfermedades Desmielinizantes/patología , Encefalomielitis Autoinmune Experimental/patología , Glicoproteínas/antagonistas & inhibidores , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular
9.
J Clin Cell Immunol ; 4(4)2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24363961

RESUMEN

The pathogenesis of multiple sclerosis (MS) is thought to involve peripheral activation of immune cells against central nervous system (CNS) antigens and their migration across the blood-brain barrier, leading to CNS inflammation and neurodegeneration. Alemtuzumab, a humanized anti-CD52 monoclonal antibody that rapidly depletes CD52-expressing cells from the circulation, is being investigated as a new treatment option in relapsing-remitting MS (RRMS). Clinical and radiologic results indicate robust suppression of inflammation related to the depletion of T and B lymphocytes during each treatment course of alemtuzumab. Furthermore, several lines of evidence suggest that the long-term clinical effects of alemtuzumab are attributable to qualitative changes in repopulating lymphocyte subsets potentially leading to a rebalancing of the immune system. Here, we review the contribution of data from animal models, ex vivo human studies, and clinical trials to the understanding of the mechanisms underlying the therapeutic effect of alemtuzumab in patients with RRMS.

10.
J Neuroimmunol ; 261(1-2): 29-36, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23759318

RESUMEN

Alemtuzumab is a monoclonal antibody against the CD52 antigen present at high levels on the surface of lymphocytes. While treatment of multiple sclerosis patients with alemtuzumab results in marked depletion of lymphocytes from the circulation, it has not been associated with a high incidence of serious infections. In a human CD52 transgenic mouse, alemtuzumab treatment showed minimal impact on the number and function of innate immune cells. A transient decrease in primary adaptive immune responses was observed post-alemtuzumab but there was little effect on memory responses. These results potentially help explain the level of immunocompetence observed in alemtuzumab-treated MS patients.


Asunto(s)
Inmunidad Adaptativa/genética , Anticuerpos Monoclonales Humanizados/fisiología , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Glicoproteínas/inmunología , Alemtuzumab , Animales , Antígenos CD/genética , Antígenos de Neoplasias/genética , Linfocitos B/inmunología , Antígeno CD52 , Células Cultivadas , Glicoproteínas/genética , Humanos , Inmunidad Innata/genética , Ratones , Ratones Transgénicos , Linfocitos T/inmunología , Resultado del Tratamiento
11.
PLoS One ; 7(6): e39416, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761788

RESUMEN

Alemtuzumab is a monoclonal antibody that targets cell surface CD52 and is effective in depleting lymphocytes by cytolytic effects in vivo. Although the cytolytic effects of alemtuzumab are dependent on the density of CD52 antigen on cells, there is scant information regarding the expression levels of CD52 on different cell types. In this study, CD52 expression was assessed on phenotypically distinct subsets of lymphoid and myeloid cells in peripheral blood mononuclear cells (PBMCs) from normal donors. Results demonstrate that subsets of PBMCs express differing levels of CD52. Quantitative analysis showed that memory B cells and myeloid dendritic cells (mDCs) display the highest number while natural killer (NK) cells, plasmacytoid dendritic cells (pDCs) and basophils have the lowest number of CD52 molecules per cell amongst lymphoid and myeloid cell populations respectively. Results of complement dependent cytolysis (CDC) studies indicated that alemtuzumab mediated profound cytolytic effects on B and T cells with minimal effect on NK cells, basophils and pDCs, correlating with the density of CD52 on these cells. Interestingly, despite high CD52 levels, mDCs and monocytes were less susceptible to alemtuzumab-mediated CDC indicating that antigen density alone does not define susceptibility. Additional studies indicated that higher expression levels of complement inhibitory proteins (CIPs) on these cells partially contributes to their resistance to alemtuzumab mediated CDC. These results indicate that alemtuzumab is most effective in depleting cells of the adaptive immune system while leaving innate immune cells relatively intact.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Muerte Celular/efectos de los fármacos , Glicoproteínas/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Alemtuzumab , Antígenos CD/genética , Antígenos de Neoplasias/genética , Antígeno CD52 , Muerte Celular/inmunología , Glicoproteínas/genética , Humanos , Leucocitos Mononucleares/inmunología
12.
Curr Stem Cell Res Ther ; 6(4): 297-316, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21190531

RESUMEN

Mesenchymal stem cells (MSCs) were discovered as a rare population of non-hematopoietic stem cells that reside in the bone marrow and interact closely with hematopoietic stem cells to support their growth and differentiation. MSCs are multipotent cells that have the ability to differentiate into cells of the mesenchymal lineage including adipocytes, osteocytes and chondrocytes and they have been reported to home to areas of tissue injury and participate in tissue repair. More recently, MSCs have also been described to possess anti-inflammatory and immunomodulatory properties that can affect multiple arms of the immune system. MSCs have been shown to inhibit T and B cell proliferation, downregulate the lytic activity of cytotoxic T lymphocytes and NK cells, inhibit the maturation and antigen-presenting function of dendritic cells and modulate macrophage function through both contact-dependent and contact-independent mechanisms. The administration of MSCs in models of autoimmune disease such as collagen-induced arthritis, EAE and autoimmune diabetes has provided additional evidence for an immunoregulatory role of MSCs supporting their use in controlling autoimmunity. The administration of allogeneic MSCs as immunosuppressive agents represents a viable approach as they appear to be largely non-immunogenic and clinical trials with allogeneic MSCs are currently underway in graftversus- host disease, Crohn's disease and type I diabetes indications. The immunomodulatory properties, mechanism of action and potential clinical utility of MSCs are reviewed herein.


Asunto(s)
Inmunidad Celular , Células Madre Mesenquimatosas/inmunología , Trasplante de Células Madre , Animales , Células Presentadoras de Antígenos/inmunología , Enfermedades Autoinmunes/terapia , Diferenciación Celular , Ensayos Clínicos como Asunto , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunidad Humoral , Células Asesinas Naturales/inmunología , Células Madre Mesenquimatosas/citología , Ratones
13.
Leuk Lymphoma ; 51(7): 1293-304, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20377308

RESUMEN

Alemtuzumab is a recombinant humanized IgG1 monoclonal antibody directed against CD52, an antigen expressed on the surface of normal and malignant B and T lymphocytes. Alemtuzumab is approved for the treatment of B-cell chronic lymphocytic leukemia (B-CLL), but the exact mechanism by which the antibody depletes malignant lymphocytes in vivo is not clearly defined. To address this issue, the anti-tumor activity of alemtuzumab was studied in disseminated and subcutaneous xenograft tumor models. The density of CD52 target antigen on the surface of tumor cells appeared to correlate with the anti-tumor activity of alemtuzumab. Deglycosylation of alemtuzumab resulted in a loss of cytotoxicity in vitro and was found to abolish anti-tumor activity in vivo. Individual inactivation of effector mechanisms in tumor-bearing mice indicated that the protective activity of alemtuzumab in vivo was primarily dependent on ADCC mediated by neutrophils and to a lesser extent NK cells. Increasing the number of circulating neutrophils by treatment with G-CSF enhanced the anti-tumor activity of the antibody, thus providing further evidence for the involvement of neutrophils as effector cells in the activity of alemtuzumab.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Antineoplásicos/uso terapéutico , Células Asesinas Naturales/inmunología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Neutrófilos/inmunología , Alemtuzumab , Animales , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Antígeno CD52 , Células CHO , Cricetinae , Cricetulus , Femenino , Glicoproteínas/metabolismo , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Humanos , Técnicas para Inmunoenzimas , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/patología , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Ratones , Ratones SCID , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Clin Transl Sci ; 2(1): 75-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20443871

RESUMEN

Immunization with the electrofusion product of tumor cells and dendritic cells (DCs) is a promising approach to cancer immunotherapy. Production of electrofusion vaccines currently requires the acquisition of tumor material and must be tailored to each individual. Alternative vaccine configurations were explored in this study. Results indicated that fusion vaccines with fully syngeneic, semi-allogeneic or fully allogeneic components, were all effective in inducing specific, long-lasting antitumor immunity. This previously undescribed activity of a fully allogeneic fusion product introduces the possibility of using defined allogeneic tumor and DC lines to simplify vaccine manufacturing.


Asunto(s)
Fusión Celular/métodos , Células Dendríticas/inmunología , Electroporación/métodos , Inmunidad/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Reactividad Cruzada/inmunología , Citocinas/metabolismo , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Trasplante Homólogo
15.
Drugs Today (Barc) ; 40(11): 913-29, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15645004

RESUMEN

The use of tumor vaccines as a therapeutic modality for the treatment of cancer is a very exciting and promising area of research. Unlike chemotherapy, vaccination against tumor antigens has the potential to specifically target cancer cells without damage to normal tissues or debilitating side effects. Recent progress in immunology and tumor biology has allowed for the development of new vaccination strategies and approaches to enhance clinical efficacy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas de Choque Térmico/inmunología , Neoplasias , Antígenos de Neoplasias/clasificación , Antígenos de Neoplasias/efectos de los fármacos , Vacunas contra el Cáncer/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/prevención & control , Insuficiencia del Tratamiento
16.
Mol Ther ; 7(4): 498-505, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12727113

RESUMEN

Dendritic cells (DCs) are potent antigen-presenting cells capable of inducing primary T-cell responses. Several immunotherapy treatment strategies involve manipulation of DCs, both in vivo and ex vivo, to promote the immunogenic presentation of tumor-associated antigens. In this study, an electrofusion protocol was developed to induce fusion between tumor cells and allogeneic bone marrow-derived DCs. Preimmunization with irradiated electrofusion product was found to provide partial to complete protection from tumor challenge in the murine Renca renal cell carcinoma model and the B16 and M3 melanoma models. Vaccinated survivors developed specific immunological memory and were able to reject a subsequent rechallenge with the same tumor cells but not a syngeneic unrelated tumor line. Antitumor protection in the B16 model was accompanied by the development of a polyclonal cytotoxic T-lymphocyte response against defined melanoma-associated antigens. The therapeutic potential of this type of approach was suggested by the ability of a Renca-DC electrofusion product to induce tumor rejection in a substantial percentage of hosts (60%) bearing pre-established tumor cells. These results indicate that treatment with electrofused tumor cells and allogeneic DCs is capable of inducing a potent antitumor response and could conceivably be applied to a wide range of cancer indications for which tumor-associated antigens have not been identified.


Asunto(s)
Vacunas contra el Cáncer , Fusión Celular/métodos , Células Dendríticas/inmunología , Electroporación , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Animales , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/inmunología , Células Dendríticas/metabolismo , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas
17.
Mol Ther ; 6(4): 519-27, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12377194

RESUMEN

We have demonstrated recently that treatment of established peritoneal mesothelial tumors with complexes composed of cationic lipid and noncoding plasmid DNA (pNull) results in the inhibition of tumor growth accompanied by the induction of a tumor-specific cellular immune response. In this study, treatment of mice bearing intraperitoneal (i.p.) M3 melanoma tumors with i.p. injections of lipid/pNull complex was found to inhibit tumor growth and induce the development of a cytolytic response against several M3 melanoma-associated antigens. Depletion of CD8(+) T cells, as opposed to natural killer (NK) or CD4(+) T cells, essentially abrogated the therapeutic effect of lipid+pNull complex, thus supporting the involvement of cytotoxic CD8(+) T cells in the antitumor response. The antitumor effect of lipid/pNull complex was maximal following delivery into a tumor-bearing compartment. For example, i.p. delivery of complex was more effective than intravenous (i.v.) or subcutaneous (s.c.) treatment of i.p. M3 tumors. In addition, i.v. injection of complex displayed therapeutic activity against lung metastases caused by i.v. injection of tumor cells, and intratumoral injection of complex into solid s.c. tumors caused regression in most animals. Importantly, the immune response induced by local treatment of tumors with complex also offered systemic protection against tumor cells at distal sites, as illustrated by the eradication of both peritoneal tumors and lung metastases in mice treated with complex delivered i.p. Treatment with lipid/pNull complex, therefore, represents an attractive immune-based treatment modality that could potentially be applied to many tumor types.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica/genética , Terapia Genética , Melanoma Experimental/terapia , Plásmidos , Linfocitos T Citotóxicos/inmunología , Animales , Portadores de Fármacos , Liposomas , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Plásmidos/administración & dosificación
18.
J Immunother ; 27(4): 273-81, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15235388

RESUMEN

Provoking a specific cellular immune response against tumor-associated antigens is a promising therapeutic strategy to treat cancers with defined antigens such as melanoma. In recent clinical trials, however, immune responses against melanoma antigens have been elicited without consistent clinical responses, suggesting the need for approaches that potentiate the specific cellular immune response. Since B lymphocytes have been reported to exert a negative effect on the cellular arm of the immune response in certain model systems, the authors compared the protective immunity elicited by melanoma antigens in B cell-deficient microMT mice to that obtained in fully immunocompetent C57BL/6 mice. Immunization with melanoma-associated antigens was accomplished using recombinant adenovirus (Ad) vectors encoding human gp100 (Ad2/gp100) or murine TRP-2 (Ad2/mTRP-2). A single dose of Ad2/gp100 or Ad2/mTRP-2 inhibited the growth of established subcutaneous B16 melanoma tumors in B cell-deficient but not wild-type C57BL/6 mice. The enhanced tumor protection observed in B cell-deficient mice appeared to be associated with potentiation of the magnitude and longevity of the specific cellular immune response. Natural killer (NK) cells were also found to be essential to the protective immune response in microMT mice because NK cell depletion with anti-asialo-GM1 antibody resulted in both the loss of tumor growth suppression and attenuation of the specific cellular immune response. The authors conclude that the protective cell-mediated immunity provoked by Ad-based cancer vaccines is enhanced in the absence of B cells, suggesting that a therapeutic regimen that includes depletion of B lymphocytes may be beneficial to cancer vaccine therapy.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/patología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Melanoma/inmunología , Melanoma/terapia , Adenoviridae/genética , Animales , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Terapia Genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Inmunoterapia , Oxidorreductasas Intramoleculares/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Melanoma/genética , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Tasa de Supervivencia , Resultado del Tratamiento
19.
J Immunol ; 169(5): 2414-21, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12193709

RESUMEN

Self tolerance to MHC class I-restricted nonmutated self Ags is a significant hurdle to effective cancer immunotherapy. Compelling evidence is emerging that altered peptide ligands can be far more immunogenic than their corresponding native epitopes; however, there is no way to reliably predict which modifications will lead to enhanced native epitope-specific immune responses. We reasoned that this limitation could be overcome by devising an empirical screen in which the nearly complete combinatorial spectrum of peptides of optimal length can be rapidly assayed for reactivity with a MHC class I-restricted cytotoxic T cell clone. This method, solid-phase epitope recovery, quantitatively ranks all reactive peptides in the library and allows selection of altered peptide ligands having desirable immunogenic properties of interest. In contrast to rationally designed MHC anchor-modified peptides, peptides identified by the present method are highly substituted in predicted TCR contact residues and can reliably activate and expand effector cell populations in vitro which lyse target cells presenting the wild-type epitope. We demonstrate that solid-phase epitope recovery peptides corresponding to a poorly immunogenic epitope of the melanoma Ag, gp100, can reliably induce wild-type peptide-specific CTL using normal donor T cells in vitro. Furthermore, these peptides can complement one another to induce these responses in an overwhelming majority of normal individuals in vitro. These data provide a rationale for the design of superior vaccines comprising a mixture of structurally diverse yet functionally convergent peptides.


Asunto(s)
Pruebas Inmunológicas de Citotoxicidad/métodos , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Oligopéptidos/inmunología , Oligopéptidos/metabolismo , Presentación de Antígeno , Línea Celular , Técnicas de Cocultivo , Técnicas Químicas Combinatorias/métodos , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Humanos , Melanoma/inmunología , Microesferas , Oligopéptidos/análisis , Biblioteca de Péptidos , Poliestirenos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Células Tumorales Cultivadas , Antígeno gp100 del Melanoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA