Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Lung Cancer ; 15(3): 188-196.e2, 2014 May.
Article in English | MEDLINE | ID: mdl-24560012

ABSTRACT

BACKGROUND: This phase II study examined the efficacy of mapatumumab in combination with paclitaxel and carboplatin in patients with non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: Patients with stage IIIB or stage IV advanced primary NSCLC were randomly assigned (1:1:1) to receive up to 6 courses of standard-dose paclitaxel and carboplatin or a combination of paclitaxel, carboplatin, and mapatumumab (10 mg/kg or 30 mg/kg). Primary efficacy end points were overall response rate and median progression-free survival (PFS). Secondary efficacy end points included disease control rate, overall survival (OS), time to response, and duration of response. Exploratory studies included evaluation of historical biopsy materials for TRAIL-R1 expression by immunohistochemical analysis and serum levels of M30, a marker of apoptosis, before and after the first 2 doses of mapatumumab. Safety parameters, including adverse events (AEs), laboratory tests, and immunogenicity, were assessed. RESULTS: The majority of patients had stage IV disease (79%) and an Eastern Cooperative Oncology Group (ECOG) performance status of 0 (58%); baseline characteristics were similar across treatment arms. No improvements in response or disease control rates, PFS, or OS were gained from the addition of mapatumumab. Adverse events in the mapatumumab arms were generally consistent with toxicities seen in the carboplatin and paclitaxel control arm. Levels of M30 were highly variable, and consistent patterns were not seen across treatment arms. CONCLUSION: This study showed no clinical benefit from adding mapatumumab to carboplatin and paclitaxel in unselected patients with NSCLC. The combination was generally well tolerated. The possibility of subgroups sensitive to mapatumumab is discussed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/drug effects , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Paclitaxel/administration & dosage , Survival Rate , Treatment Outcome
2.
Cancer Biol Ther ; 12(4): 335-48, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21785270

ABSTRACT

Tumor growth is often associated with insufficient apoptosis. The Tumor Necrosis Factor (TNF)-Related Apoptosis-Inducing Ligand (TRAIL) and its proapoptotic receptors death receptor 4 (DR4) and DR5 agonistic monoclonal antibodies are being developed as targeted therapeutics because they kill cancer cells while sparing normal cells. A challenge to targeted therapeutics is the selection of patients who are most likely to benefit from targeted drugs because of the heterogeneity of cancer. Molecular imaging may be useful in targeted drug development by assessing the target expression and drug-target interaction, and for predicting therapeutic response. We hypothesized that the cell surface expression level of DR4/5 may predict the proapoptotic targeted therapeutic response if the signaling pathway downstream is intact. The goal of this proof-of-concept study was to develop a molecular imaging strategy to predict proapoptotic anti-cancer therapy response at an early stage of treatment. TRAIL and the DR5 agonistic monoclonal antibody HGS-ETR2 (Lexatumumab, TRM-2) were labeled with a near-infrared dye and these were used to image the TRAIL receptors on cultured TRAIL sensitive and TRAIL resistant human tumor cells as well as tumor xenografts. Imaging of cells and tumor-bearing animals was conducted with near infrared fluorescence imagers and apoptosis in cells was assessed by western blots of PARP-cleavage and flow cytometry of sub-G1 content. Apoptosis in tumors was evaluated by imaging near-infrared dye-labeled Annexin V and tumor tissue activated caspase-3 staining. Both in vitro and in vivo studies showed that imaging of death inducing ligand-receptor interaction was consistent with the apoptosis readout. Thus TRAIL sensitive tumors that express TRAIL receptors underwent cell death following treatment whereas tumors lacking TRAIL receptor expression were shown to be TRAIL resistant. In vivo molecular imaging of TRAIL receptor expression correlated with response to TRAIL therapy and an apoptotic response in vivo.


Subject(s)
Apoptosis/physiology , Colorectal Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Blotting, Western , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , HT29 Cells , Humans , Immunohistochemistry , Luminescent Measurements/methods , Mice , Mice, Nude , Protein Binding , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Cancer Biol Ther ; 12(4): 349-56, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21757995

ABSTRACT

Spectral imaging systems are capable of detecting and quantifying subtle differences in light quality. In this study we coupled spectral imaging with fluorescence and white light microscopy to develop new methods for quantifying autophagy and apoptosis. For autophagy, we employed multispectral imaging to examine spectral changes in the fluorescence of LC3-GFP, a chimeric protein commonly used to track autophagosome formation. We found that punctate autophagosome-associated LC3-GFP exhibited a spectral profile that was distinctly different from diffuse cytosolic LC3-GFP. We then exploited this shift in spectral quality to quantify the amount of autophagosome-associated signal in single cells. Hydroxychloroquine (CQ), an anti-malarial agent that increases autophagosomal number, significantly increased the punctate LC3-GFP spectral signature, providing proof-of-principle for this approach. For studying apoptosis, we employed the Prism and Reflector Imaging Spectroscopy System (PARISS) hyperspectral imaging system to identify a spectral signature for active caspase-8 immunostaining in ex vivo tumor samples. This system was then used to rapidly quantify apoptosis induced by lexatumumab, an agonistic TRAIL-R2/DR5 antibody, in histological sections from a preclinical mouse model. We further found that the PARISS could accurately distinguish apoptotic tumor regions in hematoxylin and eosin-stained sections, which allowed us to quantify death receptor-mediated apoptosis in the absence of an apoptotic marker. These spectral imaging systems provide unbiased, quantitative and fast means for studying autophagy and apoptosis and complement the existing methods in their respective fields.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Diagnostic Imaging/methods , Spectrum Analysis/methods , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hydroxychloroquine/pharmacology , Mice , Mice, Nude , Microscopy, Fluorescence/methods , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Transplantation , Phagosomes/drug effects , Phagosomes/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Transplantation, Heterologous
4.
Adv Exp Med Biol ; 615: 127-58, 2008.
Article in English | MEDLINE | ID: mdl-18437894

ABSTRACT

A human tumor cell's ability to avoid the normal regulatory mechanisms of cell growth, division, and death are the hallmarks of transformation and cancer. Numerous novel therapeutic agents currently in preclinical or clinical evaluation aim to revive the normal regulation or evade these regulatory defects and induce growth arrest and cell death. One of the cell death pathways that has garnered significant interest, as a potential target for therapeutic intervention, is the programmed cell death pathway regulated by the tumor necrosis factor-related apoptosis-inducing ligand receptors (TRAIL-RS). Receptor agonist molecules including forms of the native ligand and monoclonal antibodies are being developed and tested as therapeutics in the treatment of human cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Animals , Apoptosis/drug effects , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
5.
Endocrinology ; 143(9): 3641-50, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12193580

ABSTRACT

The transcription factor Stat3 is activated through tyrosine phosphorylation by many cytokines and is a fundamental mediator of their signals. In the mammary gland, Stat3 activity increases sharply shortly after weaning, and involution is delayed in mice, that contain a mutant Stat3 lacking 33 amino acids including the key tyrosine residue. We have now generated a more extensive mutation of Stat3 through the deletion of exons 15-21 in mammary epithelium. This resulted in the loss of 245 amino acids including the DNA binding and SH2 domains, and Stat3 protein was undetectable. Pregnancy-mediated mammary development and lactation were normal in these mice. Involution was delayed and, remarkably, Stat3-null mammary epithelium maintained its functional integrity and competence even 6 d after weaning, whereas control mammary tissue was rendered nonfunctional within 2 d. The lack of remodeling and functional stasis of the epithelium correlated with the disruption of proteinase activity. Our data demonstrate that mammary tissue can retain its functional competence in the absence of external lactogenic stimuli and demonstrate a delay in the initiation of the irreversible stage of involution.


Subject(s)
DNA-Binding Proteins/genetics , Gene Deletion , Lactation/physiology , Mammary Glands, Animal/physiology , Trans-Activators/genetics , Alleles , Animals , Apoptosis , Binding Sites , Blotting, Western , Clusterin , DNA/metabolism , DNA-Binding Proteins/physiology , Endopeptidases/metabolism , Epithelium/physiology , Female , Glycoproteins/analysis , In Situ Nick-End Labeling , Male , Mammary Glands, Animal/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Milk Proteins/analysis , Molecular Chaperones/analysis , Pregnancy , STAT3 Transcription Factor , Trans-Activators/physiology , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL
...