Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 14(1): 188-98, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172270

ABSTRACT

PURPOSE: Our preclinical experiments indicated that Romidepsin (Depsipeptide FK228; DP) mediates growth arrest and apoptosis in cultured lung cancer cells. A phase II trial was done to examine clinical and molecular responses mediated by this histone deacetylase inhibitor in lung cancer patients. EXPERIMENTAL DESIGN: Nineteen patients with neoplasms refractory to standard therapy received 4-h DP infusions (17.8 mg/m(2)) on days 1 and 7 of a 21-day cycle. Each full course of therapy consisted of two identical 21-day cycles. Plasma DP levels were evaluated by liquid chromatography-mass spectrometry techniques. A variety of molecular end points were assessed in tumor biopsies via immunohistochemistry techniques. Long oligo arrays were used to examine gene expression profiles in laser-captured tumor cells before and after DP exposure, relative to lung cancer cells and adjacent normal bronchial epithelia from patients undergoing pulmonary resections. RESULTS: Nineteen patients were evaluable for toxicity assessment; 18 were evaluable for treatment response. Myelosuppression was dose limiting in one individual. No significant cardiac toxicities were observed. Maximum steady-state plasma DP concentrations ranged from 384 to 1,114 ng/mL. No objective responses were observed. Transient stabilization of disease was noted in nine patients. DP enhanced acetylation of histone H4, increased p21 expression in lung cancer cells, and seemed to shift global gene expression profiles in these cells toward those detected in normal bronchial epithelia. CONCLUSION: Although exhibiting minimal clinical efficacy at this dose and schedule, DP mediates biological effects that may warrant further evaluation of this histone deacetylase inhibitor in combination with novel-targeted agents in lung cancer patients.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Small Cell/drug therapy , Depsipeptides/therapeutic use , Lung Neoplasms/drug therapy , Acetylation/drug effects , Adult , Aged , Antibiotics, Antineoplastic/pharmacokinetics , Depsipeptides/pharmacokinetics , Female , Gene Expression/drug effects , Gene Expression Profiling , Histones/drug effects , Humans , Immunohistochemistry , Lasers , Male , Microdissection , Middle Aged , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
2.
Clin Cancer Res ; 12(19): 5777-85, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17020984

ABSTRACT

PURPOSE: The DNA methylation paradox, manifested as derepression of cancer-testis antigens, and silencing of tumor suppressors during malignant transformation, provides the rationale for the utilization of chromatin remodeling agents for cancer therapy. A phase I trial was done to examine pharmacokinetics, toxicities, and gene expression mediated by 5-aza-2'-deoxycytidine (DAC) in patients with thoracic malignancies. EXPERIMENTAL DESIGN: Thirty-five patients with cancers refractory to standard therapy received continuous 72-hour DAC infusions using a phase I dose-escalation schema. Each full course of therapy consisted of two identical 35-day cycles. Plasma DAC levels were evaluated by liquid chromatography-mass spectrometry techniques. Quantitative reverse transcription-PCR, methylation-specific PCR, and immunohistochemical techniques were used to evaluate NY-ESO-1, MAGE-3, and p16 expression in tumor biopsies. Long oligonucleotide arrays were used to evaluate gene expression profiles in laser-captured tumor cells before and after DAC exposure. RESULTS: Thirty-five patients were evaluable for toxicities; 25 were evaluable for treatment response. Myelosuppression constituted dose-limiting toxicity. The maximum tolerated dose of DAC was 60 to 75 mg/m(2) depending on the number of prior cytotoxic chemotherapy regimens. No objective responses were observed. Plasma DAC concentrations approximated thresholds for gene induction in cultured cancer cells. Target gene induction was observed in 36% of patients. Posttreatment antibodies to NY-ESO-1 were detected in three patients exhibiting NY-ESO-1 induction in their tumor tissues. Complex, heterogeneous gene expression profiles were observed in pretreatment and posttreatment tissues. CONCLUSION: Prolonged DAC infusions can modulate gene expression in primary thoracic malignancies. These findings support further evaluation of DNA-demethylating agents alone or in combination with other regimens targeting induced gene products for the treatment of these neoplasms.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/genetics , Pleural Neoplasms/genetics , Adult , Aged , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Azacitidine/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Female , Genes, p16/physiology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Maximum Tolerated Dose , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesothelioma/drug therapy , Mesothelioma/genetics , Mesothelioma/metabolism , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pleural Neoplasms/drug therapy , Pleural Neoplasms/metabolism , Transcriptional Activation
3.
Cancer Res ; 65(17): 7763-74, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140944

ABSTRACT

Regulatory sequences recognized by the unique pair of paralogous factors, CTCF and BORIS, have been implicated in epigenetic regulation of imprinting and X chromosome inactivation. Lung cancers exhibit genome-wide demethylation associated with derepression of a specific class of genes encoding cancer-testis (CT) antigens such as NY-ESO-1. CT genes are normally expressed in BORIS-positive male germ cells deficient in CTCF and meCpG contents, but are strictly silenced in somatic cells. The present study was undertaken to ascertain if aberrant activation of BORIS contributes to derepression of NY-ESO-1 during pulmonary carcinogenesis. Preliminary experiments indicated that NY-ESO-1 expression coincided with derepression of BORIS in cultured lung cancer cells. Quantitative reverse transcription-PCR analysis revealed robust, coincident induction of BORIS and NY-ESO-1 expression in lung cancer cells, but not normal human bronchial epithelial cells following 5-aza-2'-deoxycytidine (5-azadC), Depsipeptide FK228 (DP), or sequential 5-azadC/DP exposure under clinically relevant conditions. Bisulfite sequencing, methylation-specific PCR, and chromatin immunoprecipitation (ChIP) experiments showed that induction of BORIS coincided with direct modulation of chromatin structure within a CpG island in the 5'-flanking noncoding region of this gene. Cotransfection experiments using promoter-reporter constructs confirmed that BORIS modulates NY-ESO-1 expression in lung cancer cells. Gel shift and ChIP experiments revealed a novel CTCF/BORIS-binding site in the NY-ESO-1 promoter, which unlike such sites in the H19-imprinting control region and X chromosome, is insensitive to CpG methylation in vitro. In vivo occupancy of this site by CTCF was associated with silencing of the NY-ESO-1 promoter, whereas switching from CTCF to BORIS occupancy coincided with derepression of NY-ESO-1. Collectively, these data indicate that reciprocal binding of CTCF and BORIS to the NY-ESO-1 promoter mediates epigenetic regulation of this CT gene in lung cancer cells, and suggest that induction of BORIS may be a novel strategy to augment immunogenicity of pulmonary carcinomas.


Subject(s)
Antigens, Neoplasm/genetics , DNA-Binding Proteins/metabolism , Lung Neoplasms/genetics , Membrane Proteins/genetics , Repressor Proteins/metabolism , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/metabolism , Base Sequence , CCCTC-Binding Factor , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Histones/metabolism , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Sulfites/pharmacology
4.
Cancer Res ; 65(16): 7223-30, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103073

ABSTRACT

We have previously shown that the leader peptide (p14) of the Env-precursor of mouse mammary tumor virus is translocated into the nucleoli of murine T cell lymphomas that harbor this virus. Using a polyclonal antibody against recombinant p14, we show here that p14 is also localized to the nucleoli of murine mammary carcinomas and some human breast cancer samples. Affinity purification studies define a number of proteins, mostly nucleolar, that bind p14. Taken together, these findings point towards a more general involvement of p14 in lymphomagenesis and mammary carcinogenesis.


Subject(s)
Breast Neoplasms/virology , Cell Nucleolus/virology , Lymphoma, T-Cell/virology , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies, Viral/biosynthesis , Breast Neoplasms/metabolism , Cell Nucleolus/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry , Lymphoma, T-Cell/metabolism , Mammary Neoplasms, Experimental/virology , Mice , Molecular Sequence Data , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/immunology
5.
Oncogene ; 24(14): 2386-97, 2005 Mar 31.
Article in English | MEDLINE | ID: mdl-15735751

ABSTRACT

cDNA arrays were used to examine gene induction in CALU-6 and H460 lung cancer cells mediated by sequential 5-aza 2'-deoxycytidine (DAC)/depsipeptide FK228 (DP) exposure in order to identify translational end points for clinical trials evaluating these agents. In both cell lines, sequential DAC/DP treatment induced expression of tissue factor pathway inhibitor-2 (TFPI-2), an inhibitor of Factor VII: tissue factor signal transduction known to diminish the malignant phenotype of cancer cells. TFPI-2 expression was diminished or absent in 16 of 32 cell lines established from thoracic malignancies. Sequential DAC/DP treatment induced TFPI-2 in cancer cells deficient for TFPI-2 expression in the basal state. Promoter methylation coincided with loss of TFPI-2 expression in a number of cancer lines. TFPI-2 promoter methylation was observed in one of five pulmonary adenocarcinomas, and seven of seven esophageal adenocarcinomas, but not corresponding normal tissues. DP enhanced acetylation of TFPI-2-associated histones in CALU-6 cells. DP or PDBU, alone, induced TFPI-2 expression in cancer cells deficient for TFPI-2 expression in the absence of promoter methylation. In these cells, DP-mediated TFPI-2 induction was abrogated by calphostin. Induction of TFPI-2 by distinct, yet cooperative mechanisms involving chromatin remodeling and PKC signaling strengthens the preclinical rationale for sequential administration of DNA demethylating agents and HDAC inhibitors in cancer patients. Furthermore, induction of TFPI-2 may be a useful surrogate marker of treatment response in individuals receiving sequential DAC/DP infusions.


Subject(s)
Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Depsipeptides/pharmacology , Esophageal Neoplasms/metabolism , Glycoproteins/genetics , Lung Neoplasms/metabolism , Base Sequence , Cell Line, Tumor , DNA Primers , Decitabine , Esophageal Neoplasms/pathology , Humans , Lung Neoplasms/pathology
6.
Invest Ophthalmol Vis Sci ; 46(2): 415-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15671263

ABSTRACT

PURPOSE: Primary intraocular lymphoma (PIOL) is a subtype of central nervous system lymphoma. Although this lymphoma is rare, its incidence has tripled in the past 15 years. Currently, the only available model is a murine metastatic malignant lymphoma that occurs after intraperitoneal inoculation of Rev-2-T-6 lymphoma cells into newborn syngeneic mice. The current study was conducted to develop a new experimental model for PIOL. METHODS: Rev-2-T-6 cells (0.5 x 10(5) or 1.0 x 10(5)) were inoculated into the vitreous of adult BALB/c mice. Mice were monitored clinically every other day and under fundoscopic examination weekly. They were euthanatized on weeks 3, 5, 6, 7, or 8, after inoculation. All eyes were processed for histology. Immunohistochemistry was performed with an antibody (p14) specific for Rev-2-T-6 cells. Cytokine mRNA expression (IL-2, -4, -6, -10, and IFN-gamma and CC chemokine receptor-1 [CCR1]) was assayed in the lymphoma cells by microdissection and RT-PCR. IL-10 and -6 levels in the vitreous were measured by ELISA. RESULTS: Within 2 to 4 weeks, tumor cells from the vitreous migrate through the retina and gather between the RPE cell and retina. Rarely (>2 months after inoculation), Rev-2-T-6 cells may break through the RPE and infiltrate the choroid and sclera. Tumor localization was confirmed by immunohistochemistry. The intraocular lymphoma cells produce high levels of IL-10, IFN-gamma, and CCR1 transcripts. A high level of IL-10 was detected in the vitreous inoculated with Rev-2-T-6 cells. CONCLUSIONS: The data suggest that RPE cells constitute a barrier to the spread of intraocular lymphoma. Intravitreal injection of Rev-2-T-6 cells is a novel model of PIOL in immune-competent hosts that will aid in understanding the molecular mechanisms of the disease.


Subject(s)
Disease Models, Animal , Eye Neoplasms/pathology , Lymphoma, T-Cell/pathology , Retinal Neoplasms/pathology , Vitreous Body/pathology , Animals , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Eye Neoplasms/metabolism , Lymphoma, T-Cell/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Pigment Epithelium of Eye/pathology , RNA, Messenger/metabolism , Receptors, CCR1 , Receptors, Chemokine/metabolism , Retinal Neoplasms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Vitreous Body/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...