Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Br J Haematol ; 153(5): 599-611, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21492126

ABSTRACT

Bcl-2 proteins represent a rheostat that controls cellular viability. Obatoclax, a BH3-mimetic, has been designed to specifically target and counteract anti-apoptotic Bcl-2 proteins. We evaluated the biological effects of obatoclax on the anti-tumour activity of rituximab and chemotherapy agents. Obatoclax induced cell death of rituximab/chemotherapy-sensitive (RSCL), -resistant cell lines (RRCL) and primary tumour-cells derived from patients with B-cell lymphomas (N=39). Obatoclax also enhanced the activity of rituximab and had synergistic activity when combined with chemotherapy agents. The ability of Obatoclax to induce PARP cleavage varied between patient samples and was not observed in some RRCL. Inhibition of caspase activity did not affect obatoclax activity, suggesting the existence of caspase-independent death pathways. Autophagy was detected by LC3 conversion and/or electron microscopy in RRCL and in patient-derived tumour cells. Moreover, obatoclax activity was inhibited by Beclin-1 knockdown. In summary, obatoclax is an active Bcl-2 inhibitor that potentiates the activity of chemotherapy agents and, to a lesser degree, rituximab. Defining the molecular events triggered by obatoclax is necessary to further its clinical development and identify potential biomarkers that are predictive of response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Lymphoma, B-Cell/pathology , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Autophagy/drug effects , Caspases/physiology , Cell Death/drug effects , Drug Evaluation, Preclinical/methods , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Indoles , Lymphoma, B-Cell/metabolism , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Pyrroles/administration & dosage , Pyrroles/pharmacology , Rituximab , Tumor Cells, Cultured , Up-Regulation/drug effects , bcl-2 Homologous Antagonist-Killer Protein/analysis , bcl-2-Associated X Protein/analysis
3.
Clin Cancer Res ; 14(5): 1561-70, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316581

ABSTRACT

Acquirement of resistance to rituximab has been observed in lymphoma patients. To define mechanisms associated with rituximab resistance, we developed various rituximab-resistant cell lines (RRCL) and studied changes in CD20 expression/structure, lipid raft domain (LRD) reorganization, calcium mobilization, antibody-dependent cellular cytotoxicity, and complement-mediated cytotoxicity (CMC) between parental and RRCL. Significant changes in surface CD20 antigen expression were shown in RRCL. Decreased calcium mobilization and redistribution of CD20 into LRD were found in RRCL. Western blotting identified a unique 35 kDa protein band in RRCL, which was not seen in parental cells and was secondary to an increase in surface and cytoplasmic expression of IgM light chains. CD20 gene expression was decreased in RRCL. In vitro exposure to PS341 increased CD20 expression in RRCL and minimally improved the sensitivity to rituximab-associated CMC. Our data strongly suggest that the acquisition of rituximab resistance is associated with global gene and protein down-regulation of the CD20 antigen affecting LRD organization and downstream signaling. CD20 expression seems to be regulated at the pretranscriptional and posttranscriptional levels. Proteasome inhibition partially reversed rituximab resistance, suggesting the existence of additional mediators of rituximab resistance. Future research is geared to identify drugs and/or biological agents that are effective against RRCL.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD20/genetics , Antigens, CD20/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Lymphoma, B-Cell/drug therapy , Transcription, Genetic/drug effects , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Northern , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Calcium/metabolism , Down-Regulation , Flow Cytometry , Gene Expression Profiling , Humans , Immunoglobulins/metabolism , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Membrane Microdomains/drug effects , Oligonucleotide Array Sequence Analysis , Phenotype , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rituximab , Tumor Cells, Cultured
4.
Br J Haematol ; 140(1): 36-45, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17995965

ABSTRACT

The immunomodulatory drugs (IMiDs) lenalidomide and actimid (also known as CC-4047) are thalidomide analogues which are more potent than their parental compound. In combination with rituximab, we have previously demonstrated that IMiDs have synergistic in vivo anti-tumour activity in preclinical studies in a human lymphoma severe combined immunodeficiency mouse model. This report further explored the mechanisms by which IMiDs exert their anti-lymphoma effects. Following exposure of subcutaneous lymphoma tumours in murine models to IMiDs, there was a significant increase in the recruitment of natural killer (NK) cells to tumour sites. This increase in NK cells was mediated via stimulation of dendritic cells and modification of the cytokine microenvironment associated with an increase in monocyte chemotactic protein-1, tumour necrosis factor-alpha and interferon-gamma and probably augmented rituximab-associated antibody-dependent cellular cytotoxicity. IMiDs also had significant anti-angiogenic effects in our B-cell lymphoma models. Thus, by modulation of the immune system mediated via dendritic cells and NK cells, changing the cytokine milieu, as well as by their anti-angiogenic effects, IMiDs in combination with rituximab resulted in augmented in vivo anti-tumour effects against B-cell lymphoma. Our positive preclinical data adds additional support for the evaluation of IMiDs plus rituximab in patients with relapsed/refractory B-cell lymphoma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Cytotoxicity, Immunologic/drug effects , Immunosuppressive Agents/pharmacology , Killer Cells, Natural/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal, Murine-Derived , Cell Line, Tumor , Cytokines/biosynthesis , Dendritic Cells/metabolism , Drug Interactions , Immunohistochemistry , Lenalidomide , Mice , Mice, SCID , Microcirculation , Neovascularization, Pathologic/prevention & control , Rituximab , Thalidomide/analogs & derivatives , Thalidomide/pharmacology
5.
Leuk Lymphoma ; 48(12): 2424-36, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18067019

ABSTRACT

In an attempt to define mechanisms by which B-cell non-Hodgkin lymphoma (NHL) may escape rituximab immunotherapy, we developed several rituximab-resistant cell lines (RRCL) generated from the rituximab-sensitive cell lines (RSCL) Raji and RL. Rituximab resistance was associated with CD20 downregulation and upregulation of CD52 and the complement inhibitory proteins (CIPs) CD55 and CD59. No significant alemtuzumab-associated complement-mediated cell lysis (CMC) or antibody-dependent cellular cytotoxicity (ADCC) was demonstrated in RSCL. In contrast, in vitro exposure of RRCL to alemtuzumab resulted in a significant degree of CMC and ADCC. Of note, in vitro blocking of CD52 with anti-CD52 F(ab')(2) fractions in RRCL improved rituximab-associated CMC as compared to unblocked RRCL. Our current data provides a basis for further evaluation of alemtuzumab-based clinical trials for patients with rituximab-resistant NHL.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/therapeutic use , Complement System Proteins/immunology , Glycoproteins/antagonists & inhibitors , Lymphoma, Non-Hodgkin/drug therapy , Alemtuzumab , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antigens, CD/analysis , Antigens, CD20/analysis , Antigens, Neoplasm/analysis , Apoptosis , CD52 Antigen , Cell Line, Tumor , Cytotoxicity, Immunologic , Drug Resistance, Neoplasm , Glycoproteins/analysis , Histocompatibility Antigens Class II/analysis , Humans , Lymphoma, Non-Hodgkin/immunology , Rituximab
6.
Clin Cancer Res ; 13(15 Pt 1): 4556-64, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671142

ABSTRACT

PURPOSE: Mapatumumab and lexatumumab are fully humanized, high-affinity immunoglobulin G(1 lambda) monoclonal antibodies (mAb) that target/activate the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor 1 (TRAIL-R1) and receptor 2 (TRAIL-R2), respectively, triggering the extrinsic apoptotic pathway. Theoretically, synergistic antitumor activity should be observed by combining TRAIL-R mAbs with agents (e.g., rituximab) that activate the intrinsic apoptotic pathway. EXPERIMENTAL DESIGN: To this end, targeted antigen expression in a NHL-cell panel was evaluated by flow cytometry. NHL cells were exposed to mapatumumab or lexatumumab followed by rituximab, isotype, or RPMI. DNA synthesis was quantified by [(3)H]-thymidine incorporation assays. Induction of apoptosis was detected by flow-cytometric analysis. For antibody-dependent cellular cytotoxicity (ADCC) and complement-mediated cytotoxicity (CMC) studies, standardized (51)Cr-release assays were done. We inoculated severe combined immunodeficiency (SCID) mouse with Raji cells i.v. The animals then were treated with various combinations of rituximab, mapatumumab, lexatumumab, and isotype alone or in combination. RESULTS: In vitro exposure to mapatumumab resulted in significant apoptosis (30-50%) and decreased DNA synthesis in sensitive lymphoma cells. Mapatumumab/rituximab combination resulted in a significant inhibition of cell proliferation (90% reduction) when compared with mapatumumab (60% reduction) or rituximab (5% reduction). In vivo, the median survival time of animals treated with mapatumumab and rituximab was longer (not reached) than those treated with rituximab monotherapy [33 days (95% confidence interval, 29-37), log-rank test, P = 0.05]. CONCLUSIONS: Mapatumumab induces apoptosis, cell growth arrest, ADCC, and CMC. The combination of mapatumumab plus rituximab is more effective in controlling lymphoma growth in vivo than either antibody. Rituximab and mapatumumab warrant further evaluation against B-cell lymphoma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Lymphoma, B-Cell/therapy , Receptors, Tumor Necrosis Factor/immunology , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity , Apoptosis , Cell Line, Tumor , Drug Therapy, Combination , Flow Cytometry , Lymphoma, B-Cell/immunology , Mice , Mice, SCID , Receptors, TNF-Related Apoptosis-Inducing Ligand , Rituximab , Xenograft Model Antitumor Assays
7.
Ann Surg Oncol ; 10(3): 284-90, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12679314

ABSTRACT

BACKGROUND: beta-Catenin functions as an integral part of the E-cadherin/catenin adhesion complex to maintain epithelial cell integrity. beta-Catenin also functions as part of the Wnt signal transduction pathway to transmit growth-promoting signals to the nucleus via its interactions with Tcf/Lef transcription factors. Previous reports have demonstrated altered beta-catenin expression in numerous tumor types; however, reports regarding beta-catenin expression in pancreatic cancer have been conflicting. METHODS: beta-Catenin expression was examined in 10 pancreatic cancer cell lines by Western and Northern analysis and by immunofluorescence. Expression was also examined by immunohistochemistry in 57 primary pancreatic cancers and 7 foci of carcinoma-in-situ. RESULTS: Reduced expression of beta-catenin was observed in 4 of 10 pancreatic cancer cell lines. Reduced membranous expression was noted in 32 pancreatic cancers (56%) and correlated with loss of tumor differentiation. Nuclear beta-catenin expression was identified in two tumors (4%). beta-Catenin expression was present in all seven foci of carcinoma-in-situ; however, nuclear expression was predominant in four of the seven cases. CONCLUSIONS: Alterations in beta-catenin expression are common in pancreatic cancer; however, signaling and adhesion functions may be perturbed at different times during tumor progression. Therefore, dysregulation of beta-catenin may contribute to the development and progression of this disease through distinct mechanisms.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Cytoskeletal Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/pathology , Trans-Activators/biosynthesis , Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/pharmacology , Disease Progression , Humans , Pancreatic Neoplasms/genetics , Signal Transduction , Trans-Activators/analysis , Trans-Activators/pharmacology , Tumor Cells, Cultured , beta Catenin
8.
Surgery ; 132(2): 141-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12219004

ABSTRACT

BACKGROUND: beta-Catenin is a component of the E-cadherin/catenin adhesion complex that maintains epithelial cell integrity. We have previously observed decreased beta-catenin expression in both human pancreatic cancer cell lines and primary tumors. To determine the significance of this finding with respect to pancreatic carcinogenesis, this study evaluated the effects of restoring expression of beta-catenin with and without E-cadherin in pancreatic cancer cells. METHODS: MiaPaca-2 cells were stably transfected with full-length cDNAs for beta-catenin, E-cadherin, or a mutated E-cadherin lacking the beta-catenin-binding domain. Doubly transfected cell clones containing beta-catenin and either E-cadherin or deleted E-cadherin were also selected. Assays for cell adhesion, cell cycle profile, motility, and apoptosis were performed. RESULTS: Cell clones expressing beta-catenin alone or beta-catenin and deleted E-cadherin did not differ significantly from the parental cell lines in any of the assays performed. In contrast, MiaPaca-2 cell clones expressing both beta-catenin and E-cadherin showed tight adhesion, decreased cell growth, and a significantly increased apoptotic index as compared to the parental line or singly transfected clones. CONCLUSIONS: MiaPaca-2 cells undergo apoptosis at a significantly increased rate after restoration of the E-cadherin/beta-catenin adhesion complex. This increase in apoptosis is dependent on the ability of E-cadherin to bind beta-catenin. Loss of beta-catenin expression may therefore provide pancreatic cancer cells with a growth advantage that contributes to tumor progression.


Subject(s)
Apoptosis/physiology , Cadherins/genetics , Cytoskeletal Proteins/genetics , Pancreatic Neoplasms , Trans-Activators/genetics , Breast Neoplasms , Cell Adhesion/physiology , Cell Division/physiology , Cell Movement/physiology , Female , Gene Expression Regulation, Neoplastic , Humans , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...