74 results on '"Kao GD"'
Search Results
2. Multiblock Discriminant Analysis of Integrative 18 F-FDG-PET/CT Radiomics for Predicting Circulating Tumor Cells in Early-Stage Non-small Cell Lung Cancer Treated With Stereotactic Body Radiation Therapy.
- Author
-
Lee SH, Kao GD, Feigenberg SJ, Dorsey JF, Frick MA, Jean-Baptiste S, Uche CZ, Cengel KA, Levin WP, Berman AT, Aggarwal C, Fan Y, and Xiao Y
- Subjects
- Aged, Aged, 80 and over, Area Under Curve, Biomarkers, Tumor, Carcinoma, Non-Small-Cell Lung pathology, Discriminant Analysis, Female, Fluorodeoxyglucose F18, Humans, Lung Neoplasms pathology, Male, Middle Aged, Prospective Studies, Radiopharmaceuticals, Statistics, Nonparametric, Tumor Burden, Carcinoma, Non-Small-Cell Lung blood, Carcinoma, Non-Small-Cell Lung radiotherapy, Lung Neoplasms blood, Lung Neoplasms radiotherapy, Neoplastic Cells, Circulating, Positron Emission Tomography Computed Tomography
- Abstract
Purpose: The main objective of the present study was to integrate
18 F-FDG-PET/CT radiomics with multiblock discriminant analysis for predicting circulating tumor cells (CTCs) in early-stage non-small cell lung cancer (ES-NSCLC) treated with stereotactic body radiation therapy (SBRT)., Methods: Fifty-six patients with stage I NSCLC treated with SBRT underwent18 F-FDG-PET/CT imaging pre-SBRT and post-SBRT (median, 5 months; range, 3-10 months). CTCs were assessed via a telomerase-based assay before and within 3 months after SBRT and dichotomized at 5 and 1.3 CTCs/mL. Pre-SBRT, post-SBRT, and delta PET/CT radiomics features (n = 1548 × 3/1562 × 3) were extracted from gross tumor volume. Seven feature blocks were constructed including clinical parameters (n = 12). Multiblock data integration was performed using block sparse partial least squares-discriminant analysis (sPLS-DA) referred to as Data Integration Analysis for Biomarker Discovery Using Latent Components (DIABLO) for identifying key signatures by maximizing common information between different feature blocks while discriminating CTC levels. Optimal input blocks were identified using a pairwise combination method. DIABLO performance for predicting pre-SBRT and post-SBRT CTCs was evaluated using combined AUC (area under the curve, averaged across different blocks) analysis with 20 × 5-fold cross-validation (CV) and compared with that of concatenation-based sPLS-DA that consisted of combining all features into 1 block. CV prediction scores between 1 class versus the other were compared using the Wilcoxon rank sum test., Results: For predicting pre-SBRT CTCs, DIABLO achieved the best performance with combined pre-SBRT PET radiomics and clinical feature blocks, showing CV AUC of 0.875 (P = .009). For predicting post-SBRT CTCs, DIABLO achieved the best performance with combined post-SBRT CT and delta CT radiomics feature blocks, showing CV AUCs of 0.883 (P = .001). In contrast, all single-block sPLS-DA models could not attain CV AUCs higher than 0.7., Conclusions: Multiblock integration with discriminant analysis of18 F-FDG-PET/CT radiomics has the potential for predicting pre-SBRT and post-SBRT CTCs. Radiomics and CTC analysis may complement and together help guide the subsequent management of patients with ES-NSCLC., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
3. Integration of Risk Survival Measures Estimated From Pre- and Posttreatment Computed Tomography Scans Improves Stratification of Patients With Early-Stage Non-small Cell Lung Cancer Treated With Stereotactic Body Radiation Therapy.
- Author
-
Jiao Z, Li H, Xiao Y, Aggarwal C, Galperin-Aizenberg M, Pryma D, Simone CB 2nd, Feigenberg SJ, Kao GD, and Fan Y
- Subjects
- Aged, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung radiotherapy, Cohort Studies, Dose Fractionation, Radiation, Female, Forecasting methods, Humans, Kaplan-Meier Estimate, Lung Neoplasms pathology, Lung Neoplasms radiotherapy, Male, Models, Theoretical, Postoperative Care, Preoperative Care, Prognosis, Radiosurgery mortality, Treatment Outcome, Carcinoma, Non-Small-Cell Lung diagnostic imaging, Carcinoma, Non-Small-Cell Lung mortality, Lung Neoplasms diagnostic imaging, Lung Neoplasms mortality, Radiosurgery methods, Tomography, X-Ray Computed
- Abstract
Purpose: To predict overall survival of patients receiving stereotactic body radiation therapy (SBRT) for early-stage non-small cell lung cancer (ES-NSCLC), we developed a radiomic model that integrates risk of death estimates and changes based on pre- and posttreatment computed tomography (CT) scans. We hypothesize this innovation will improve our ability to stratify patients into various oncologic outcomes with greater accuracy., Methods and Materials: Two cohorts of patients with ES-NSCLC uniformly treated with SBRT (a median dose of 50 Gy in 4-5 fractions) were studied. Prediction models were built on a discovery cohort of 100 patients with treatment planning CT scans, and then were applied to a separate validation cohort of 60 patients with pre- and posttreatment CT scans for evaluating their performance., Results: Prediction models achieved a c-index up to 0.734 in predicting survival outcomes of the validation cohort. The integration of the pretreatment risk of survival measures (risk-high vs risk-low) and changes (risk-increase vs risk-decrease) in risk of survival measures between the pretreatment and posttreatment scans further stratified the patients into 4 subgroups (risk: high, increase; risk: high, decrease; risk: low, increase; risk: low, decrease) with significant difference (χ
2 = 18.549, P = .0003, log-rank test). There was also a significant difference between the risk-increase and risk-decrease groups (χ2 = 6.80, P = .0091, log-rank test). In addition, a significant difference (χ2 = 7.493, P = .0062, log-rank test) was observed between the risk-high and risk-low groups obtained based on the pretreatment risk of survival measures., Conclusion: The integration of risk of survival measures estimated from pre- and posttreatment CT scans can help differentiate patients with good expected survival from those who will do more poorly following SBRT. The analysis of these radiomics-based longitudinal risk measures may help identify patients with early-stage NSCLC who will benefit from adjuvant treatment after lung SBRT, such as immunotherapy., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
4. Personal and Prognostic: Tissue and Liquid Biomarkers of Radiotherapeutic Response in Non-Small Cell Lung Cancer.
- Author
-
Jean-Baptiste SR, Feigenberg SJ, Dorsey JF, and Kao GD
- Subjects
- Biomarkers, Tumor genetics, Humans, Mutation, Prognosis, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung radiotherapy, Lung Neoplasms genetics, Lung Neoplasms pathology, Lung Neoplasms radiotherapy
- Abstract
Recent treatment advances have improved outcomes for patients with non-small cell lung cancer (NSCLC), often utilizing tumor molecular characterization to identify targetable mutations. This is further enhanced by advancements in "liquid biopsies", using peripheral blood for noninvasive, serial sampling of tumor biology. While tumor genomic alterations have established therapeutic implications in metastatic NSCLC, research is also ongoing to develop applications for tissue and liquid biomarkers in earlier stage disease, such as patients treated with radiation for early stage or locoregional NSCLC., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
5. Circulating Tumor Cells Are Associated with Recurrent Disease in Patients with Early-Stage Non-Small Cell Lung Cancer Treated with Stereotactic Body Radiotherapy.
- Author
-
Frick MA, Feigenberg SJ, Jean-Baptiste SR, Aguarin LA, Mendes A, Chinniah C, Swisher-McClure S, Berman A, Levin W, Cengel KA, Hahn SM, Dorsey JF, Simone CB 2nd, and Kao GD
- Subjects
- Aged, Aged, 80 and over, Carcinoma, Non-Small-Cell Lung blood, Carcinoma, Non-Small-Cell Lung surgery, Disease Progression, Female, Humans, Lung Neoplasms blood, Lung Neoplasms surgery, Male, Neoplasm Recurrence, Local blood, Neoplasm Recurrence, Local surgery, Neoplasm Staging, Retrospective Studies, Survival Rate, Telomerase blood, Treatment Outcome, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung radiotherapy, Lung Neoplasms pathology, Lung Neoplasms radiotherapy, Neoplasm Recurrence, Local pathology, Neoplastic Cells, Circulating pathology, Radiosurgery methods
- Abstract
Purpose: Although stereotactic body radiotherapy (SBRT) is effective in early-stage non-small cell lung cancer (NSCLC), approximately 10%-15% of patients will fail regionally and 20%-25% distantly. We evaluate a novel circulating tumor cell (CTC) assay as a prognostic marker for increased risk of recurrence following SBRT., Experimental Design: Ninety-two subjects (median age, 71 years) with T1a (64%), T1b (23%), or T2a (13%) stage I NSCLC treated with SBRT were prospectively enrolled. CTCs were enumerated by utilizing a GFP-expressing adenoviral probe that detects elevated telomerase activity in cancer cells. Samples were obtained before, during, and serially up to 24 months after treatment. SBRT was delivered to a median dose of 50 Gy (range, 40-60 Gy), mostly commonly in four to five fractions (92%)., Results: Thirty-eight of 92 subjects (41%) had a positive CTC test prior to SBRT. A cutoff of ≥5 CTCs/mL before treatment defined favorable ( n = 78) and unfavorable ( n = 14) prognostic groups. Increased risk of nodal ( P = 0.04) and distant ( P = 0.03) failure was observed in the unfavorable group. Within 3 months following SBRT, CTCs continued to be detected in 10 of 35 (29%) subjects. Persistent detection of CTCs was associated with increased risk of distant failure ( P = 0.04) and trended toward increased regional ( P = 0.08) and local failure ( P = 0.16)., Conclusions: Higher pretreatment CTCs and persistence of CTCs posttreatment is significantly associated with increased risk of recurrence outside the targeted treatment site. This suggests that CTC analysis may potentially identify patients at higher risk for regional or distant recurrences and who may benefit from either systemic therapy and/or timely locoregional salvage treatment., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
6. Early Detection of Recurrence in Patients With Locally Advanced Non-Small-Cell Lung Cancer via Circulating Tumor Cell Analysis.
- Author
-
Chinniah C, Aguarin L, Cheng P, Decesaris C, Cutillo A, Berman AT, Frick M, Doucette A, Cengel KA, Levin W, Hahn S, Dorsey JF, Simone CB 2nd, and Kao GD
- Subjects
- Adult, Aged, Aged, 80 and over, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung therapy, Chemoradiotherapy, Feasibility Studies, Female, Humans, Lung Neoplasms pathology, Lung Neoplasms therapy, Male, Middle Aged, Neoplasm Recurrence, Local, Neoplasm Staging, Carcinoma, Non-Small-Cell Lung diagnosis, Cell Count methods, Lung Neoplasms diagnosis, Neoplastic Cells, Circulating pathology
- Abstract
Background: Assays to identify circulating tumor cells (CTCs) might allow for noninvasive and sequential monitoring of lung cancer. We investigated whether serial CTC analysis could complement conventional imaging for detecting recurrences after treatment in patients with locally advanced non-small-cell lung cancer (LA-NSCLC)., Patients and Methods: Patients with LA-NSCLC (stage II-III) who definitively received concurrent chemoradiation were prospectively enrolled, with CTCs from peripheral blood samples identified using an adenoviral probe that detects elevated telomerase activity present in nearly all lung cancer cells. A "detectable" CTC level was defined as 1.3 green flourescent protein-positive cells per milliliter of collected blood. Samples were obtained before, during (at weeks 2, 4, and 6), and after treatment (post-radiation therapy [RT]; at months 1, 3, 6, 12, 18, and 24)., Results: Forty-eight patients were enrolled. At a median follow-up of 10.9 months, 22 (46%) patients had disease recurrence at a median time of 7.6 months post-RT (range, 1.3-32.0 months). Of the 20 of 22 patients for whom post-RT samples were obtained, 15 (75%) had an increase in CTC counts post-RT. In 10 of these 15 patients, CTCs were undetectable on initial post-RT draw but were then detected again before radiographic detection of recurrence, with a median lead time of 6.2 months and mean lead time of 6.1 months (range, 0.1-12.0 months) between CTC count increase and radiographic evidence of recurrence. One patient with an early recurrence (4.7 months) had persistently elevated detectable CTC levels during and after treatment., Conclusion: These results indicate that longitudinal CTC monitoring in patients with LA-NSCLC treated with chemoradiation is feasible, and that detectable CTC levels in many patients meaningfully precede radiologic evidence of disease recurrence., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
7. Circulating Glioma Cells Exhibit Stem Cell-like Properties.
- Author
-
Liu T, Xu H, Huang M, Ma W, Saxena D, Lustig RA, Alonso-Basanta M, Zhang Z, O'Rourke DM, Zhang L, Gong Y, Kao GD, Dorsey JF, and Fan Y
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis, Biomarkers, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Line, Tumor, Cell Survival, Drug Resistance, Neoplasm, Female, Humans, Immunophenotyping, Male, Mice, Neoplastic Cells, Circulating drug effects, Neoplastic Cells, Circulating pathology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Phenotype, Stress, Physiological, Wnt Proteins metabolism, Glioma metabolism, Glioma pathology, Neoplastic Cells, Circulating metabolism, Neoplastic Stem Cells metabolism
- Abstract
: Circulating tumor cells (CTC) are known to be present in the blood of patients with glioblastoma (GBM). Here we report that GBM-derived CTC possess a cancer stem cell (CSC)-like phenotype and contribute to local tumorigenesis and recurrence by the process of self-seeding. Genetic probes showed that mouse GBM-derived CTC exhibited Sox2/ETn transcriptional activation and expressed glioma CSC markers, consistent with robust expression of stemness-associated genes including SOX2, OCT4, and NANOG in human GBM patient-derived samples containing CTC. A transgenic mouse model demonstrated that CTC returned to the primary tumor and generated new tumors with enhanced tumorigenic capacity. These CTCs were resistant to radiotherapy and chemotherapy and to circulation stress-induced cell apoptosis. Single-cell RNA-seq analysis revealed that Wnt activation induced stemness and chemoresistance in CTC. Collectively, these findings identify GBM-derived CTC as CSC-like cells and suggest that targeting Wnt may offer therapeutic opportunities for eliminating these treatment-refractory cells in GBM. SIGNIFICANCE: These findings identify CTCs as an alternative source for in situ tumor invasion and recurrence through local micrometastasis, warranting eradication of systemic "out-of-tumor" CTCs as a promising new therapeutic opportunity for GBM., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
8. Circulating Tumor Cell Assessment in Presumed Early Stage Non-Small Cell Lung Cancer Patients Treated with Stereotactic Body Radiation Therapy: A Prospective Pilot Study.
- Author
-
Frick MA, Kao GD, Aguarin L, Chinniah C, Swisher-McClure S, Berman AT, Levin WP, Cengel KA, DeCesaris C, Hahn SM, Dorsey JF, and Simone CB 2nd
- Subjects
- Aged, Aged, 80 and over, Disease Progression, Dose Fractionation, Radiation, Female, Fluorescent Dyes chemistry, Follow-Up Studies, Green Fluorescent Proteins chemistry, Humans, Male, Middle Aged, Neoplasm Recurrence, Local, Pilot Projects, Prospective Studies, Recurrence, Telomerase blood, Carcinoma, Non-Small-Cell Lung blood, Carcinoma, Non-Small-Cell Lung radiotherapy, Lung Neoplasms blood, Lung Neoplasms radiotherapy, Neoplastic Cells, Circulating, Radiosurgery
- Abstract
Purpose: In patients treated with stereotactic body radiation therapy (SBRT) for presumed early stage non-small cell lung cancer (NSCLC), detection and monitoring of circulating tumor cells (CTCs) may be useful for assessing treatment response safely and noninvasively. No published reports of CTC trends in this patient population exist to date., Methods and Materials: Patients with clinically diagnosed stage I NSCLC treated with SBRT were eligible for this institutional review board-approved prospective clinical trial. Peripheral blood samples were assayed for CTCs via a green fluorescent protein-expressing adenoviral probe. CTC positivity was defined as 1.3 green fluorescent protein-positive cells/mL of collected blood. Samples were obtained before (pre-radiation therapy [RT]), during, and after SBRT (post-RT; months 1, 3, 6, 12, 18, and 24). SBRT was delivered in ≤5 fractions (median dose of 50 Gy in 12.5 Gy fractions) to a biological equivalent dose of ≥100 Gy in all cases., Results: Forty-eight consecutive patients (T1a [73%], T1b [21%], and T2a [6%]) were enrolled. Median follow-up was 14.2 months. Twenty patients (42%) had a positive CTC level pre-RT, with a median CTC count of 4.2 CTCs per mL (interquartile range [IQR], 2.2-18.7). Of these 20 patients, 17 had evaluable post-RT CTC evaluations showing reduced CTC counts at 1 month (median, 0.2; IQR, 0.1-0.8) and 3 months (median, 0.6; IQR, 0-1.1). Three of these 17 patients experienced disease progression at a median of 19.9 months; all 3 experienced ≥1 positive post-RT CTC test predating clinical progression by a median of 16 months (range, 2-17 months). In contrast, among patients presenting with CTC-detectable disease and for whom all post-RT CTC tests were negative, none experienced recurrence or progression., Conclusions: CTC monitoring after SBRT for presumed early stage NSCLC may give lead-time notice of disease recurrence or progression. Conversely, negative CTC counts after treatment may provide reassurance of disease control. CTC analysis is thus potentially useful in enhancing clinical diagnosis and follow-up in this population., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
9. Temporal DNA-PK activation drives genomic instability and therapy resistance in glioma stem cells.
- Author
-
Wang Y, Xu H, Liu T, Huang M, Butter PP, Li C, Zhang L, Kao GD, Gong Y, Maity A, Koumenis C, and Fan Y
- Subjects
- Acid Anhydride Hydrolases, Animals, Cell Line, Tumor, DNA Damage radiation effects, DNA Repair, DNA Repair Enzymes metabolism, DNA-Activated Protein Kinase genetics, DNA-Binding Proteins metabolism, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic radiation effects, Glioma genetics, Humans, Male, Mice, Neoplastic Stem Cells metabolism, Nuclear Proteins genetics, RNA, Small Interfering metabolism, Xenograft Model Antitumor Assays, DNA-Activated Protein Kinase metabolism, Genomic Instability radiation effects, Glioma radiotherapy, Neoplastic Stem Cells radiation effects, Nuclear Proteins metabolism, Radiation Tolerance genetics
- Abstract
Cancer stem cells (CSCs) - known to be resistant to genotoxic radiation and chemotherapy - are fundamental to therapy failure and cancer relapse. Here, we reveal that glioma CSCs are hypersensitive to radiation, but a temporal DNA repair mechanism converts the intrinsic sensitivity to genomic instability and treatment resistance. Transcriptome analysis identifies DNA-dependent protein kinase (DNA-PK) as a predominant DNA repair enzyme in CSCs. Notably, DNA-PK activity is suppressed after irradiation when ROS induce the dissociation of DNA-PKcs with Ku70/80, resulting in delayed DNA repair and radiosensitivity; subsequently, after ROS clearance, the accumulated DNA damage and robust activation of DNA-PK induce genomic instability, facilitated by Rad50-mediated cell-cycle arrest, leading to enhanced malignancy, CSC overgrowth, and radioresistance. Finally, we show a requisite in vivo role for DNA-PK in CSC-mediated radioresistance and glioma progression. These findings identify a time-sensitive mechanism controlling CSC resistance to DNA-damaging treatments and suggest DNA-PK/Rad50 as promising targets for CSC eradication.
- Published
- 2018
- Full Text
- View/download PDF
10. Circulating Tumor Cells, DNA, and mRNA: Potential for Clinical Utility in Patients With Melanoma.
- Author
-
Xu MJ, Dorsey JF, Amaravadi R, Karakousis G, Simone CB 2nd, Xu X, Xu W, Carpenter EL, Schuchter L, and Kao GD
- Subjects
- Humans, Melanoma genetics, Melanoma pathology, Neoplastic Cells, Circulating pathology, Prognosis, Biomarkers, Tumor blood, DNA, Neoplasm blood, Melanoma blood, RNA, Messenger blood
- Abstract
Unlabelled: : Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and messenger RNA (mRNA), collectively termed circulating tumor products (CTPs), represent areas of immense interest from scientists' and clinicians' perspectives. In melanoma, CTP analysis may have clinical utility in many areas, from screening and diagnosis to clinical decision-making aids, as surveillance biomarkers or sources of real-time genetic or molecular characterization. In addition, CTP analysis can be useful in the discovery of new biomarkers, patterns of treatment resistance, and mechanisms of metastasis development. Here, we compare and contrast CTCs, ctDNA, and mRNA, review the extent of translational evidence to date, and discuss how future studies involving both scientists and clinicians can help to further develop this tool for the benefit of melanoma patients., Implications for Practice: Scientific advancement has enabled the rapid development of tools to analyze circulating tumor cells, tumor DNA, and messenger RNA, collectively termed circulating tumor products (CTPs). A variety of techniques have emerged to detect and characterize melanoma CTPs; however, only a fraction has been applied to human subjects. This review summarizes the available human data that investigate clinical utility of CTP in cancer screening, melanoma diagnosis, prognosis, prediction, and genetic or molecular characterization. It provides a rationale for how CTPs may be useful for future research and discusses how clinicians can be involved in developing this exciting new technology., (©AlphaMed Press.)
- Published
- 2016
- Full Text
- View/download PDF
11. A novel approach for the detection and genetic analysis of live melanoma circulating tumor cells.
- Author
-
Xu MJ, Cooke M, Steinmetz D, Karakousis G, Saxena D, Bartlett E, Xu X, Hahn SM, Dorsey JF, and Kao GD
- Subjects
- Adenoviridae genetics, Adult, Aged, Area Under Curve, Cell Line, Tumor, Female, Fluorescent Antibody Technique, Genetic Vectors genetics, Genetic Vectors metabolism, Humans, Linear Models, Male, Melanoma metabolism, Middle Aged, Mutation, Neoplasm Metastasis, Neoplastic Cells, Circulating pathology, Pilot Projects, Proto-Oncogene Proteins B-raf genetics, ROC Curve, Telomerase metabolism, Melanoma pathology, Neoplastic Cells, Circulating metabolism
- Abstract
Background: Circulating tumor cell (CTC) detection and genetic analysis may complement currently available disease assessments in patients with melanoma to improve risk stratification and monitoring. We therefore sought to establish the feasibility of a telomerase-based assay for detecting and isolating live melanoma CTCs., Methods: The telomerase-based CTC assay utilizes an adenoviral vector that, in the presence of elevated human telomerase activity, drives the amplification of green fluorescent protein. Tumor cells are then identified via an image processing system. The protocol was tested on melanoma cells in culture or spiked into control blood, and on samples from patients with metastatic melanoma. Genetic analysis of the isolated melanoma CTCs was then performed for BRAF mutation status., Results: The adenoviral vector was effective for all melanoma cell lines tested with sensitivity of 88.7% (95%CI 85.6-90.4%) and specificity of 99.9% (95%CI 99.8-99.9%). In a pilot trial of patients with metastatic disease, CTCs were identified in 9 of 10 patients, with a mean of 6.0 CTCs/mL. At a cutoff of 1.1 CTCs/mL, the telomerase-based assay exhibits test performance of 90.0% sensitivity and 91.7% specificity. BRAF mutation analysis of melanoma cells isolated from culture or spiked control blood, or from pilot patient samples was found to match the known BRAF mutation status of the cell lines and primary tumors., Conclusions: To our knowledge, this is the first report of a telomerase-based assay effective for detecting and isolating live melanoma CTCs. These promising findings support further studies, including towards integrating into the management of patients with melanoma receiving multimodality therapy.
- Published
- 2015
- Full Text
- View/download PDF
12. Tracking viable circulating tumor cells (CTCs) in the peripheral blood of non-small cell lung cancer (NSCLC) patients undergoing definitive radiation therapy: pilot study results.
- Author
-
Dorsey JF, Kao GD, MacArthur KM, Ju M, Steinmetz D, Wileyto EP, Simone CB 2nd, and Hahn SM
- Subjects
- Aged, Biomarkers, Tumor blood, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung blood, Carcinoma, Non-Small-Cell Lung radiotherapy, Cell Line, Tumor, Female, Humans, Lung Neoplasms blood, Lung Neoplasms radiotherapy, Male, Middle Aged, Pilot Projects, Telomerase blood, Treatment Outcome, Carcinoma, Non-Small-Cell Lung diagnosis, Lung Neoplasms diagnosis, Neoplastic Cells, Circulating metabolism, Telomerase metabolism
- Abstract
Background: Assays identifying circulating tumor cells (CTCs) allow noninvasive and sequential monitoring of the status of primary or metastatic tumors, potentially yielding clinically useful information. However, to the authors' knowledge, the effect of radiation therapy (RT) on CTCs in patients with non-small cell lung cancer (NSCLC) has not been previously explored., Methods: This report describes results from a pilot study of 30 patients with NSCLC who received RT. Peripheral blood samples obtained from these patients were assayed for CTCs using an assay that identified live cells using an adenoviral probe that detected the elevated telomerase activity present in almost all cancer cells, but not in normal cells, and the validity of the assay was confirmed with secondary tumor-specific markers. Patients were assayed before initiation of RT (pre-RT), during the RT course, and/or after the completion of RT (post-RT)., Results: The assay successfully detected CTCs in the majority of patients, including 65% of patients before the start of RT, and in patients with both epidermal growth factor receptor wild-type and mutation-positive tumors. The median CTC counts in patients before RT was 9.1 CTCs per mL (range, undetectable to 571 CTCs per mL) and was significantly higher than the average post-RT count of 0.6 CTCs per mL (range, undetectable to 1.8 CTCs per mL; P<.001). Sequential CTC counts were available in a subset of patients and demonstrated decreases after RT, except for 1 patient who subsequently developed distant failure., Conclusions: The current pilot data suggest that CTC counts appear to reflect response to RT in patients with localized NSCLC. On the basis of these promising results, the authors have launched a more comprehensive and detailed clinical trial., (© 2014 American Cancer Society.)
- Published
- 2015
- Full Text
- View/download PDF
13. Application of a telomerase-based circulating tumor cell (CTC) assay in bladder cancer patients receiving postoperative radiation therapy: a case study.
- Author
-
Ju M, Kao GD, Steinmetz D, Chandrasekaran S, Keefe SM, Guzzo TJ, Christodouleas JP, Hahn SM, and Dorsey JF
- Subjects
- Aged, Carcinoma, Transitional Cell enzymology, Carcinoma, Transitional Cell therapy, Enzyme Assays, Fatal Outcome, Female, Humans, Male, Middle Aged, Radiotherapy, Adjuvant, Treatment Outcome, Urinary Bladder Neoplasms enzymology, Urinary Bladder Neoplasms therapy, Biomarkers, Tumor metabolism, Carcinoma, Transitional Cell diagnosis, Neoplastic Cells, Circulating metabolism, Telomerase metabolism, Urinary Bladder Neoplasms diagnosis
- Abstract
Background: Muscle invasive bladder carcinoma is an often lethal disease that requires aggressive treatment. Improved assays would contribute to better risk prediction and clinical management of this disease. A telomerase-based assay to detect circulating tumor cells (CTCs) may usefully fulfill this role., Methods: Two patients (C1 and C2) were enrolled onto an IRB-approved bladder biomarker study before initiating post-operative radiation therapy (RT) for muscle invasive bladder carcinoma. Blood samples were taken at predefined intervals: before, during, and after RT and then retrospectively correlated with imaging studies and disease course., Results: C1 began RT for positive resection margins on surgical pathology, at which time CTCs were undetectable and pelvic imaging demonstrated no evidence of disease. However, following the completion of treatment, the patient's CTC count was found to have increased to 202 CTCs/mL, and MRI demonstrated new abdominal and pelvic masses consistent with progressive disease. C1 ultimately died of disease with distant and local failure. Conversely, C2 was found to have 632 CTCs/mL before the initiation of RT for positive surgical margins, although imaging demonstrated no visible masses. At the conclusion of RT, repeat imaging showed changes that were indeterminate for either tumor recurrence or post-radiation effects. However, the patient's CTC count had dropped to 184 CTCs/mL. Furthermore, a second follow-up assay performed 6 months later revealed no detectable CTCs and repeat imaging showed complete resolution of worrisome imaging changes, thus excluding tumor progression., Conclusions: To our knowledge this is the first report of a telomerase-based assay to identify CTCs in bladder cancer patients. Further studies are required to fully determine the ultimate clinical utility of this assay. However, the two patient vignettes described here illustrate how serial CTC assays may track the disease course and inform the management of bladder cancer patients undergoing adjuvant RT and potentially chemotherapy.
- Published
- 2014
- Full Text
- View/download PDF
14. Detection of brain tumor cells in the peripheral blood by a telomerase promoter-based assay.
- Author
-
Macarthur KM, Kao GD, Chandrasekaran S, Alonso-Basanta M, Chapman C, Lustig RA, Wileyto EP, Hahn SM, and Dorsey JF
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Animals, Biomarkers, Tumor blood, Biomarkers, Tumor metabolism, Brain Neoplasms enzymology, Female, Glioma metabolism, Heterografts, Humans, Male, Mice, Promoter Regions, Genetic, Telomerase analysis, Telomerase genetics, Xenograft Model Antitumor Assays, Brain Neoplasms blood, Brain Neoplasms pathology, Glioma blood, Glioma pathology, Neoplastic Cells, Circulating metabolism, Neoplastic Cells, Circulating pathology, Telomerase metabolism
- Abstract
Blood tests to detect circulating tumor cells (CTC) offer great potential to monitor disease status, gauge prognosis, and guide treatment decisions for patients with cancer. For patients with brain tumors, such as aggressive glioblastoma multiforme, CTC assays are needed that do not rely on expression of cancer cell surface biomarkers like epithelial cell adhesion molecules that brain tumors tend to lack. Here, we describe a strategy to detect CTC based on telomerase activity, which is elevated in nearly all tumor cells but not normal cells. This strategy uses an adenoviral detection system that is shown to successfully detect CTC in patients with brain tumors. Clinical data suggest that this assay might assist interpretation of treatment response in patients receiving radiotherapy, for example, to differentiate pseudoprogression from true tumor progression. These results support further development of this assay as a generalized method to detect CTC in patients with cancer., (©2014 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
15. Theranostic gold nanoparticles modified for durable systemic circulation effectively and safely enhance the radiation therapy of human sarcoma cells and tumors.
- Author
-
Joh DY, Kao GD, Murty S, Stangl M, Sun L, Al Zaki A, Xu X, Hahn SM, Tsourkas A, and Dorsey JF
- Abstract
Radiation therapy (RT) is an integral component of the treatment of many sarcomas and relies on accurate targeting of tumor tissue. Despite conventional treatment planning and RT, local failure rates of 10% to 28% at 5 years have been reported for locally advanced, unresectable sarcomas, due in part to limitations in the cumulative RT dose that may be safely delivered. We describe studies of the potential usefulness of gold nanoparticles modified for durable systemic circulation (through polyethylene glycosylation; hereinafter "P-GNPs") as adjuvants for RT of sarcomas. In studies of two human sarcoma-derived cell lines, P-GNP in conjunction with RT caused increased unrepaired DNA damage, reflected by approximately 1.61-fold increase in γ-H2AX (histone phosphorylated on Ser(139)) foci density compared with RT alone. The combined RT and P-GNP also led to significantly reduced clonogenic survival of tumor cells, compared to RT alone, with dose-enhancement ratios of 1.08 to 1.16. In mice engrafted with human sarcoma tumor cells, the P-GNP selectively accumulated in the tumor and enabled durable imaging, potentially aiding radiosensitization as well as treatment planning. Mice pretreated with P-GNP before targeted RT of their tumors exhibited significantly improved tumor regression and overall survival, with long-term survival in one third of mice in this treatment group compared to none with RT only. Interestingly, prior RT of sarcoma tumors increased subsequent extravasation and in-tumor deposition of P-GNP. These results together suggest P-GNP may be integrated into the RT of sarcomas, potentially improving target imaging and radiosensitization of tumor while minimizing dose to normal tissues.
- Published
- 2013
- Full Text
- View/download PDF
16. Gold nanoparticles in radiation research: potential applications for imaging and radiosensitization.
- Author
-
Dorsey JF, Sun L, Joh DY, Witztum A, Kao GD, Alonso-Basanta M, Avery S, Hahn SM, Al Zaki A, and Tsourkas A
- Abstract
The potential of gold nanoparticles (GNPs) in therapeutic and diagnostic cancer applications is becoming increasingly recognized. These biologically compatible particles can be easily synthesized, tuned to different sizes, and functionalized by conjugation to various biologically useful materials. Efficient and specific delivery to tumor tissue can then be accomplished either by passive accumulation in leaky tumor vessels and tissue, or by directly targeting tumor-specific biomarkers. Tumor-localized GNPs can serve as both adjuvants for enhancing the efficacy of radiation therapy and also as contrast agents for various imaging modalities. In this review, we will discuss recent advancements and future potential in the application of GNP as both a radiosensitizer and an imaging contrast agent. Due to their versatility and biocompatibility, gold nanoparticles may represent a novel theranostic adjuvant for radiation applications in cancer management.
- Published
- 2013
- Full Text
- View/download PDF
17. Selective targeting of brain tumors with gold nanoparticle-induced radiosensitization.
- Author
-
Joh DY, Sun L, Stangl M, Al Zaki A, Murty S, Santoiemma PP, Davis JJ, Baumann BC, Alonso-Basanta M, Bhang D, Kao GD, Tsourkas A, and Dorsey JF
- Subjects
- Animals, Brain drug effects, Brain pathology, Brain radiation effects, Brain Neoplasms pathology, Cell Line, Tumor, Female, Glioblastoma pathology, Mice, Mice, Nude, Nanoparticles ultrastructure, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Glioblastoma drug therapy, Glioblastoma radiotherapy, Gold therapeutic use, Nanoparticles therapeutic use
- Abstract
Successful treatment of brain tumors such as glioblastoma multiforme (GBM) is limited in large part by the cumulative dose of Radiation Therapy (RT) that can be safely given and the blood-brain barrier (BBB), which limits the delivery of systemic anticancer agents into tumor tissue. Consequently, the overall prognosis remains grim. Herein, we report our pilot studies in cell culture experiments and in an animal model of GBM in which RT is complemented by PEGylated-gold nanoparticles (GNPs). GNPs significantly increased cellular DNA damage inflicted by ionizing radiation in human GBM-derived cell lines and resulted in reduced clonogenic survival (with dose-enhancement ratio of ~1.3). Intriguingly, combined GNP and RT also resulted in markedly increased DNA damage to brain blood vessels. Follow-up in vitro experiments confirmed that the combination of GNP and RT resulted in considerably increased DNA damage in brain-derived endothelial cells. Finally, the combination of GNP and RT increased survival of mice with orthotopic GBM tumors. Prior treatment of mice with brain tumors resulted in increased extravasation and in-tumor deposition of GNP, suggesting that RT-induced BBB disruption can be leveraged to improve the tumor-tissue targeting of GNP and thus further optimize the radiosensitization of brain tumors by GNP. These exciting results together suggest that GNP may be usefully integrated into the RT treatment of brain tumors, with potential benefits resulting from increased tumor cell radiosensitization to preferential targeting of tumor-associated vasculature.
- Published
- 2013
- Full Text
- View/download PDF
18. A 10-year analysis of American Society For Radiation Oncology Junior Faculty Career Development Awards.
- Author
-
Kimple RJ and Kao GD
- Subjects
- Career Mobility, Efficiency, Faculty, Medical supply & distribution, Humans, National Institutes of Health (U.S.) economics, Publishing statistics & numerical data, Research economics, Research statistics & numerical data, United States, Awards and Prizes, Financing, Organized economics, Radiation Oncology economics, Radiation Oncology education, Radiation Oncology statistics & numerical data, Societies, Medical economics, Societies, Medical statistics & numerical data, Staff Development economics, Staff Development statistics & numerical data
- Abstract
Purpose: Between 2000 and 2010, the American Society for Radiation Oncology (ASTRO) awarded 22 Junior Faculty Career Development Awards (JFA) totaling $4.4 million. This study aimed to evaluate the impact of these awards on the grantees' career development, including current position, publications, and subsequent independent grant funding., Methods: Each awardee was requested via email and telephone to provide an updated curriculum vitae, a National Institutes of Health (NIH) biosketch, and information regarding current position of employment. Twenty-one of the 22 JFA recipients complied. Reported grant funding was extracted from each candidate's CV, and the amounts of NIH grants obtained were confirmed via NIH REPORTER. Reported publications were confirmed via PubMed., Results: All survey respondents (21 of 21) have remained in academic positions. Subsequent aggregate grant funding totaled more than $25 million (range, $0-$4.1 million), 5.9 times the initial investment. NIH grant funding totaled almost $15 million, 3 times the initial investment. Awardees have published an average of 34.6 publications (range, 0-123) for an overall rate of 4.5 papers/year (range, 1-11)., Conclusions: ASTRO JFAs over the past decade have been strongly associated with grantees remaining in academic positions, success in attracting private and NIH grants, and publication productivity. In an era of dwindling federal research funding, the support provided by the ASTRO JFA may be especially helpful to support the research careers of promising junior faculty members., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
19. Enhancing the efficacy of drug-loaded nanocarriers against brain tumors by targeted radiation therapy.
- Author
-
Baumann BC, Kao GD, Mahmud A, Harada T, Swift J, Chapman C, Xu X, Discher DE, and Dorsey JF
- Subjects
- Animals, Antineoplastic Agents, Phytogenic administration & dosage, Blood-Brain Barrier drug effects, Blood-Brain Barrier pathology, Blood-Brain Barrier radiation effects, Brain Neoplasms pathology, Cell Line, Tumor, Chemoradiotherapy, Drug Carriers chemistry, Glioblastoma pathology, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Luciferases genetics, Luciferases metabolism, Mice, Mice, Nude, Micelles, Microscopy, Fluorescence, Nanostructures chemistry, Paclitaxel chemistry, Polymers chemistry, Treatment Outcome, Xenograft Model Antitumor Assays, Brain Neoplasms therapy, Drug Delivery Systems methods, Glioblastoma therapy, Paclitaxel administration & dosage
- Abstract
Glioblastoma multiforme (GBM) is a common, usually lethal disease with a median survival of only ~15 months. It has proven resistant in clinical trials to chemotherapeutic agents such as paclitaxel that are highly effective in vitro, presumably because of impaired drug delivery across the tumor's blood-brain barrier (BBB). In an effort to increase paclitaxel delivery across the tumor BBB, we linked the drug to a novel filomicelle nanocarrier made with biodegradable poly(ethylene-glycol)-block-poly(ε-caprolactone-r-D,L-lactide) and used precisely collimated radiation therapy (RT) to disrupt the tumor BBB's permeability in an orthotopic mouse model of GBM. Using a non-invasive bioluminescent imaging technique to assess tumor burden and response to therapy in our model, we demonstrated that the drug-loaded nanocarrier (DLN) alone was ineffective against stereotactically implanted intracranial tumors yet was highly effective against GBM cells in culture and in tumors implanted into the flanks of mice. When targeted cranial RT was used to modulate the tumor BBB, the paclitaxel-loaded nanocarriers became effective against the intracranial tumors. Focused cranial RT improved DLN delivery into the intracranial tumors, significantly improving therapeutic outcomes. Tumor growth was delayed or halted, and survival was extended by >50% (p less than 0.05) compared to the results obtained with either RT or the DLN alone. Combinations of RT and chemotherapeutic agents linked to nanocarriers would appear to be an area for future investigations that could enhance outcomes in the treatment of human GBM.
- Published
- 2013
- Full Text
- View/download PDF
20. Stereotactic intracranial implantation and in vivo bioluminescent imaging of tumor xenografts in a mouse model system of glioblastoma multiforme.
- Author
-
Baumann BC, Dorsey JF, Benci JL, Joh DY, and Kao GD
- Subjects
- Animals, Brain Neoplasms chemistry, Cell Line, Tumor, Glioblastoma chemistry, Humans, Mice, Mice, Nude, Brain Neoplasms pathology, Glioblastoma pathology, Luminescent Measurements methods, Neoplasm Transplantation methods, Stereotaxic Techniques, Transplantation, Heterologous methods
- Abstract
Glioblastoma multiforme (GBM) is a high-grade primary brain cancer with a median survival of only 14.6 months in humans despite standard tri-modality treatment consisting of surgical resection, post-operative radiation therapy and temozolomide chemotherapy. New therapeutic approaches are clearly needed to improve patient survival and quality of life. The development of more effective treatment strategies would be aided by animal models of GBM that recapitulate human disease yet allow serial imaging to monitor tumor growth and treatment response. In this paper, we describe our technique for the precise stereotactic implantation of bio-imageable GBM cancer cells into the brains of nude mice resulting in tumor xenografts that recapitulate key clinical features of GBM. This method yields tumors that are reproducible and are located in precise anatomic locations while allowing in vivo bioluminescent imaging to serially monitor intracranial xenograft growth and response to treatments. This method is also well-tolerated by the animals with low perioperative morbidity and mortality.
- Published
- 2012
- Full Text
- View/download PDF
21. An integrated method for reproducible and accurate image-guided stereotactic cranial irradiation of brain tumors using the small animal radiation research platform.
- Author
-
Baumann BC, Benci JL, Santoiemma PP, Chandrasekaran S, Hollander AB, Kao GD, and Dorsey JF
- Abstract
Preclinical studies of cranial radiation therapy (RT) using animal brain tumor models have been hampered by technical limitations in the delivery of clinically relevant RT. We established a bioimageable mouse model of glioblastoma multiforme (GBM) and an image-guided radiation delivery system that facilitated precise tumor localization and treatment and which closely resembled clinical RT. Our novel radiation system makes use of magnetic resonance imaging (MRI) and bioluminescent imaging (BLI) to define tumor volumes, computed tomographic (CT) imaging for accurate treatment planning, a novel mouse immobilization system, and precise treatments delivered with the Small Animal Radiation Research Platform. We demonstrated that, in vivo, BLI correlated well with MRI for defining tumor volumes. Our novel restraint system enhanced setup reproducibility and precision, was atraumatic, and minimized artifacts on CT imaging used for treatment planning. We confirmed precise radiation delivery through immunofluorescent analysis of the phosphorylation of histone H2AX in irradiated brains and brain tumors. Assays with an intravenous near-infrared fluorescent probe confirmed that radiation of orthografts increased disruption of the tumor blood-brain barrier (BBB). This integrated model system, which facilitated delivery of precise, reproducible, stereotactic cranial RT in mice and confirmed RT's resultant histologic and BBB changes, may aid future brain tumor research.
- Published
- 2012
- Full Text
- View/download PDF
22. A role for the histone deacetylase HDAC4 in the life-cycle of HIV-1-based vectors.
- Author
-
Smith JA, Yeung J, Kao GD, and Daniel R
- Subjects
- DNA, Viral metabolism, Genetic Vectors, HIV-1 genetics, HeLa Cells, Histone Deacetylase 2 metabolism, Histone Deacetylase 6, Humans, Protein Binding, Transduction, Genetic, HIV Integrase metabolism, HIV-1 physiology, Histone Deacetylases metabolism, Host-Pathogen Interactions, Repressor Proteins metabolism, Virus Integration
- Abstract
HIV-1 integration is mediated by the HIV-1 integrase protein, which joins 3'-ends of viral DNA to host cell DNA. To complete the integration process, HIV-1 DNA has to be joined to host cell DNA also at the 5'-ends. This process is called post-integration repair (PIR). Integration and PIR involve a number of cellular co-factors. These proteins exhibit different degrees of involvement in integration and/or PIR. Some are required for efficient integration or PIR. On the other hand, some reduce the efficiency of integration. Finally, some are involved in integration site selection. We have studied the role of the histone deacetylase HDAC4 in these processes. HDAC4 was demonstrated to play a role in both cellular double-strand DNA break repair and transcriptional regulation. We observed that HDAC4 associates with viral DNA in an integrase-dependent manner. Moreover, infection with HIV-1-based vectors induces foci of the HDAC4 protein. The related histone deacetylases, HDAC2 and HDAC6, failed to associate with viral DNA after infection. These data suggest that HDAC4 accumulates at integration sites. Finally, overexpression studies with HDAC4 mutants suggest that HDAC4 may be required for efficient transduction by HIV-1-based vectors in cells that are deficient in other DNA repair proteins. We conclude that HDAC4 is likely involved in PIR.
- Published
- 2010
- Full Text
- View/download PDF
23. Modulation of the anti-cancer efficacy of microtubule-targeting agents by cellular growth conditions.
- Author
-
Dorsey JF, Dowling ML, Kim M, Voong R, Solin LJ, and Kao GD
- Subjects
- Cell Count, Cell Culture Techniques, Cell Survival drug effects, Drug Resistance, Neoplasm drug effects, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Humans, Mitosis drug effects, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Microtubules drug effects, Microtubules metabolism, Neoplasms metabolism, Tubulin Modulators pharmacology
- Abstract
Mitotic spindle-disrupting agents target and disrupt microtubule dynamics. These agents include clinically important chemotherapies, including taxanes (paclitaxel (Taxol), docetaxel (Taxotere)) and vinca alkaloids (vincristine (Oncovin), vinblastine). Taxanes are a standard component of treatment for many malignancies, often in conjunction with other cytotoxic agents. However, the optimal sequencing of these treatments and whether efficacy may be influenced by in vitro cellular growth conditions remain incompletely investigated. Yet such preclinical investigations may guide clinical decision making. We therefore studied the effect of cell density on rapid killing by paclitaxel and vincristine. Breast, ovarian and prostate cancer cells were sensitive to rapid killing by either agent when grown at low density, but were markedly resistant when grown at high density, i.e. nearly confluent. The resistance of densely growing cells to rapid killing by these drugs translated to increased clonogenic survival. Pretreatment of densely growing cancer cells with cisplatin followed by paclitaxel, partially reversed the treatment resistance. Gene ontology associations from microarray analyses of cells grown at low and high density, suggested roles for membrane signal transduction and adhesion, but potentially also DNA damage repair and metabolism. Taken together, the treatment resistance at higher cell density may be associated with a lower proportion of active cycling in cells growing at high density as well as transduction of survival signals induced by increased cell-cell adhesion. Collectively these findings suggest mechanisms by which growth conditions may contribute to resistance to rapid killing by microtubule-disrupting drugs.
- Published
- 2010
- Full Text
- View/download PDF
24. Lack of compliance with national vaccination guidelines in oncology patients receiving radiation therapy.
- Author
-
Yee SS, Dutta PR, Solin LJ, Vapiwala N, and Kao GD
- Subjects
- Adult, Aged, Aged, 80 and over, Attitude of Health Personnel, Female, Guidelines as Topic, Health Surveys, Humans, Influenza Vaccines administration & dosage, Male, Middle Aged, Pneumococcal Vaccines administration & dosage, Young Adult, Influenza, Human prevention & control, Neoplasms prevention & control, Neoplasms radiotherapy, Patient Compliance, Pneumonia, Pneumococcal prevention & control, Vaccination standards
- Abstract
Cancer patients are at increased risk for potentially life-threatening infections. Patient safety goals recently issued by the Joint Commission on the Accreditation of Healthcare Organizations (JCAHO) and current Centers for Disease Control and Prevention (CDC) guidelines recommend vaccinations for all cancer patients over the age of 65 (for Pneumococcus) and 50 years of age (annually, for Influenza). The authors investigated vaccination practices in patients over a season of risk at a university-based outpatient cancer treatment clinic. Of 204 patients recruited, 196 (93%) completed the survey. Overall, 30% of patients reported never receiving the Influenza vaccine (33% of patients >50 years old), and 56% reported never receiving the Pneumococcal vaccine (30% of patients >65 years old). Only 7% of patients reported being asked or informed about vaccination by their oncologists. Substantial proportions of patients undergoing cancer treatment have not received vaccinations as recommended by national guidelines. The reasons cited for lack of compliance seem correctable, and doing so would potentially prevent mortality and morbidity, thereby improving the care of cancer patients. Recommended vaccinations may now include that for the Influenza A virus (H1N1).
- Published
- 2010
25. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and paclitaxel have cooperative in vivo effects against glioblastoma multiforme cells.
- Author
-
Dorsey JF, Mintz A, Tian X, Dowling ML, Plastaras JP, Dicker DT, Kao GD, and El-Deiry WS
- Subjects
- Animals, Blotting, Western, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Brain Neoplasms pathology, Caspases metabolism, Cell Cycle drug effects, Cell Line, Tumor, Cell Survival drug effects, Drug Synergism, Fluorodeoxyglucose F18, Glioblastoma metabolism, Glioblastoma pathology, Humans, Luminescent Measurements, Mice, Mice, Nude, Paclitaxel administration & dosage, Positron-Emission Tomography methods, TNF-Related Apoptosis-Inducing Ligand administration & dosage, Treatment Outcome, Tumor Burden drug effects, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, X-Ray Microtomography methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Glioblastoma drug therapy, Paclitaxel pharmacology, TNF-Related Apoptosis-Inducing Ligand pharmacology, Xenograft Model Antitumor Assays
- Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in conjunction with microtubule-targeting agents may be a promising novel anticancer treatment strategy. In vitro studies have suggested that relatively low concentrations of TRAIL enhance the lethality of paclitaxel (Taxol) against human cancer cells. The increased efficacy may be due to the triggering of caspase activation, resulting in mitotic checkpoint abrogation and catastrophe. We show here that wild-type p53 protects cells from caspase-dependent death induced by this therapeutic combination in vitro. We have now also developed an imaging-based model system to test the in vivo efficacy of combined TRAIL and Taxol, in which tumor growth and treatment response can be monitored noninvasively and in real-time. We further utilize bioluminescence, F18-fluorodeoxyglucose-positron emission tomography, and microscale computed tomography imaging to confirm the effects of combined treatment on tumors. These studies together provide the first in vivo confirmation that combined TRAIL plus paclitaxel results in better tumor control compared with either TRAIL or paclitaxel alone, and with no discernable increased normal tissue toxicity in the mouse. Interestingly, the in vivo antitumor response elicited by combined treatment was not affected by the p53 status of the tumor cells. These preclinical observations together suggest the therapeutic potential of combining TRAIL plus paclitaxel in cancer treatment, and support further preclinical and future clinical testing.
- Published
- 2009
- Full Text
- View/download PDF
26. The HDAC inhibitor panobinostat (LBH589) inhibits mesothelioma and lung cancer cells in vitro and in vivo with particular efficacy for small cell lung cancer.
- Author
-
Crisanti MC, Wallace AF, Kapoor V, Vandermeers F, Dowling ML, Pereira LP, Coleman K, Campling BG, Fridlender ZG, Kao GD, and Albelda SM
- Subjects
- Carcinoma, Small Cell enzymology, Carcinoma, Small Cell pathology, Cell Line, Tumor, Humans, Indoles, Lung Neoplasms enzymology, Lung Neoplasms pathology, Mesothelioma pathology, Panobinostat, Carcinoma, Small Cell drug therapy, Histone Deacetylase Inhibitors pharmacology, Hydroxamic Acids pharmacology, Lung Neoplasms drug therapy, Mesothelioma drug therapy
- Abstract
Lung cancer is the leading cause of cancer deaths in the United States. Current therapies are inadequate. Histone deacetylase inhibitors (HDACi) are a recently developed class of anticancer agents that cause increased acetylation of core histones and nonhistone proteins leading to modulation of gene expression and protein activity involved in cancer cell growth and survival pathways. We examined the efficacy of the HDACi panobinostat (LBH589) in a wide range of lung cancers and mesotheliomas. Panobinostat was cytotoxic in almost all 37 cancer cell lines tested. IC(50) and LD(50) values were in the low nmol/L range (4-470 nmol/L; median, 20 nmol/L). Small cell lung cancer (SCLC) cell lines were among the most sensitive lines, with LD(50) values consistently <25 nmol/L. In lung cancer and mesothelioma animal models, panobinostat significantly decreased tumor growth by an average of 62% when compared with vehicle control. Panobinostat was equally effective in immunocompetent and severe combined immunodeficiency mice, indicating that the inhibition of tumor growth by panobinostat was not due to direct immunologic effects. Panobinostat was, however, particularly effective in SCLC xenografts, and the addition of the chemotherapy agent etoposide augmented antitumor effects. Protein analysis of treated tumor biopsies revealed elevated amounts of cell cycle regulators such as p21 and proapoptosis factors, such as caspase 3 and 7 and cleaved poly[ADP-ribose] polymerase, coupled with decreased levels of antiapoptotic factors such as Bcl-2 and Bcl-X(L). These studies together suggest that panobinostat may be a useful adjunct in the treatment of thoracic malignancies, especially SCLC.
- Published
- 2009
- Full Text
- View/download PDF
27. HDAC4 promotes growth of colon cancer cells via repression of p21.
- Author
-
Wilson AJ, Byun DS, Nasser S, Murray LB, Ayyanar K, Arango D, Figueroa M, Melnick A, Kao GD, Augenlicht LH, and Mariadason JM
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Cell Proliferation, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Down-Regulation, Humans, Male, Mice, Mice, SCID, Models, Biological, Neoplasm Transplantation, RNA, Small Interfering metabolism, Cyclin-Dependent Kinase Inhibitor p21 antagonists & inhibitors, Gene Expression Regulation, Neoplastic, Histone Deacetylases physiology, Repressor Proteins physiology
- Abstract
The class II Histone deacetylase (HDAC), HDAC4, is expressed in a tissue-specific manner, and it represses differentiation of specific cell types. We demonstrate here that HDAC4 is expressed in the proliferative zone in small intestine and colon and that its expression is down-regulated during intestinal differentiation in vivo and in vitro. Subcellular localization studies demonstrated HDAC4 expression was predominantly nuclear in proliferating HCT116 cells and relocalized to the cytoplasm after cell cycle arrest. Down-regulating HDAC4 expression by small interfering RNA (siRNA) in HCT116 cells induced growth inhibition and apoptosis in vitro, reduced xenograft tumor growth, and increased p21 transcription. Conversely, overexpression of HDAC4 repressed p21 promoter activity. p21 was likely a direct target of HDAC4, because HDAC4 down-regulation increased p21 mRNA when protein synthesis was inhibited by cycloheximide. The importance of p21 repression in HDAC4-mediated growth promotion was demonstrated by the failure of HDAC4 down-regulation to induce growth arrest in HCT116 p21-null cells. HDAC4 down-regulation failed to induce p21 when Sp1 was functionally inhibited by mithramycin or siRNA-mediated down-regulation. HDAC4 expression overlapped with that of Sp1, and a physical interaction was demonstrated by coimmunoprecipitation. Chromatin immunoprecipitation (ChIP) and sequential ChIP analyses demonstrated Sp1-dependent binding of HDAC4 to the proximal p21 promoter, likely directed through the HDAC4-HDAC3-N-CoR/SMRT corepressor complex. Consistent with increased transcription, HDAC4 or SMRT down-regulation resulted in increased histone H3 acetylation at the proximal p21 promoter locus. These studies identify HDAC4 as a novel regulator of colon cell proliferation through repression of p21.
- Published
- 2008
- Full Text
- View/download PDF
28. TRAIL inactivates the mitotic checkpoint and potentiates death induced by microtubule-targeting agents in human cancer cells.
- Author
-
Kim M, Liao J, Dowling ML, Voong KR, Parker SE, Wang S, El-Deiry WS, and Kao GD
- Subjects
- Caspase 3 metabolism, Drug Evaluation, Preclinical, Drug Synergism, Gene Expression Regulation, Neoplastic drug effects, HCT116 Cells, Humans, Mitosis genetics, Models, Biological, Neoplasms genetics, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Treatment Outcome, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Microtubules drug effects, Mitosis drug effects, Neoplasms drug therapy, Nocodazole administration & dosage, TNF-Related Apoptosis-Inducing Ligand administration & dosage, TNF-Related Apoptosis-Inducing Ligand pharmacology
- Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted interest as an anticancer treatment, when used in conjunction with standard chemotherapy. We investigated the mechanistic basis for combining low-dose TRAIL with microtubule-targeting agents that invoke the mitotic checkpoint. Treatment of T98G and HCT116 cells with nocodazole alone resulted in a robust mitotic block with initially little cell death; low levels of cell death were also seen with TRAIL alone at 10 ng/mL final concentration. In contrast, the addition of low-dose TRAIL to nocodazole was associated with maximally increased caspase-3, caspase-8, and caspase-9 activation, which efficiently abrogated the mitotic delay and markedly increased cell death. In contrast, the abrogation of mitotic checkpoint and increased cell death were blocked by inhibitors of caspase-8 and caspase-9 or pan-caspase inhibitor. The addition of TRAIL to either nocodazole or paclitaxel (Taxol) reduced levels of the mitotic checkpoint proteins BubR1 and Bub1. BubR1 mutated for the caspase cleavage sites, but not wild-type BubR1, was resistant to cleavage induced by TRAIL added to nocodazole, and partially blocked the checkpoint abrogation. These results suggest that adding a relatively low concentration of TRAIL to antimicrotubule agents markedly increases complete caspase activation. This in turn accentuates degradation of spindle checkpoint proteins such as BubR1 and Bub1, contributes to abrogation of the mitotic checkpoint, and induces cancer cell death. These results suggest that TRAIL may increase the anticancer efficacy of microtubule-targeting drugs.
- Published
- 2008
- Full Text
- View/download PDF
29. Temozolomide-mediated radiosensitization of human glioma cells in a zebrafish embryonic system.
- Author
-
Geiger GA, Fu W, and Kao GD
- Subjects
- Animals, Animals, Genetically Modified, Blood Vessels physiology, Cell Proliferation drug effects, Cell Proliferation radiation effects, Cell Survival drug effects, Cell Survival radiation effects, Dacarbazine pharmacology, Dacarbazine therapeutic use, Embryo, Nonmammalian, Embryonic Development drug effects, Feasibility Studies, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Luminescent Proteins metabolism, Radiation-Sensitizing Agents pharmacology, Temozolomide, Tumor Cells, Cultured, Red Fluorescent Protein, Dacarbazine analogs & derivatives, Glioma radiotherapy, Xenograft Model Antitumor Assays methods, Zebrafish embryology
- Abstract
The zebrafish (Danio rerio) is a popular vertebrate model for biomedical research. The rapid development, transparency, and experimental accessibility of the embryo offer opportunities for assessing the developmental effects of anticancer treatment strategies. We therefore systematically investigated parameters for growing U251 human glioma cells expressing red fluorescent protein (U251-RFP) in zebrafish embryos. Factors optimized include injection volume, number of cells injected, anatomic site of injection, age of the embryo at the time of injection, and postinjection incubation temperature. After injection into the embryos, the U251-RFP cells proliferated and the resultant tumors, and even individual cells, could be visualized in real-time via fluorescence microscopy without the need for sacrifice. These tumors recruited host zebrafish vasculature, suggesting cancer cell-host tissue interactions. Having optimized parameters for introducing and growing these human cells in the zebrafish embryos, we exposed both embryos and transplanted cancer cells to ionizing radiation and temozolomide, either alone or in combination. The human tumors in each embryo were substantially diminished following exposure to ionizing radiation and the decrease was further enhanced by pretreatment with temozolomide. In contrast, temozolomide had no discernible effects on embryonic development. These results together support the relative safety of temozolomide during embryonic development, as well as its anticancer efficacy when combined with radiation. These results suggest the value of the zebrafish model for in vivo testing of the efficacy and safety of anticancer strategies, especially on the very young.
- Published
- 2008
- Full Text
- View/download PDF
30. Identification and biological evaluation of a novel and potent small molecule radiation sensitizer via an unbiased screen of a chemical library.
- Author
-
Lally BE, Geiger GA, Kridel S, Arcury-Quandt AE, Robbins ME, Kock ND, Wheeler K, Peddi P, Georgakilas A, Kao GD, and Koumenis C
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma radiotherapy, Animals, Colorectal Neoplasms drug therapy, Colorectal Neoplasms radiotherapy, Combinatorial Chemistry Techniques methods, Combined Modality Therapy, DNA Breaks, Double-Stranded, DNA Repair drug effects, Embryo, Nonmammalian drug effects, Embryo, Nonmammalian radiation effects, Female, Glioma drug therapy, Glioma radiotherapy, HT29 Cells, Humans, Lung Neoplasms drug therapy, Lung Neoplasms radiotherapy, Mice, Mice, Nude, Xenograft Model Antitumor Assays, Zebrafish embryology, Neoplasms drug therapy, Neoplasms radiotherapy, Propiophenones pharmacology, Radiation-Sensitizing Agents pharmacology
- Abstract
For patients with solid tumors, the tolerance of surrounding tissues often limits the dose of radiation that can be delivered. Thus, agents that preferentially increase the cytotoxic effects of radiation toward tumor cells would significantly alter the therapeutic ratio and improve patient survival. Using a high-throughput, unbiased screening approach, we have identified 4'-bromo-3'-nitropropiophenone (NS-123) as a radiosensitizer of human glioma cells in vitro and in vivo. NS-123 radiosensitized U251 glioma cells in a dose-dependent and time-dependent manner, with dose enhancement ratios ranging from 1.3 to 2.0. HT-29 colorectal carcinoma and A549 lung adenocarcinoma cells were also radiosensitized by NS-123 in vitro, whereas NS-123 did not increase the radiation sensitivity of normal human astrocytes or developmental abnormalities or lethality of irradiated Zebrafish embryos. In a novel xenograft model of U251 cells implanted into Zebrafish embryos, NS-123 enhanced the tumor growth-inhibitory effects of ionizing radiation (IR) with no apparent effect on embryo development. Similar results were obtained using a mouse tumor xenograft model in which NS-123 sensitized U251 tumors to IR while exhibiting no overt toxicity. In vitro pretreatment with NS-123 resulted in accumulation of unrepaired IR-induced DNA strand breaks and prolonged phosphorylation of the surrogate markers of DNA damage H2AX, ataxia telangiectasia mutated protein, DNA-dependent protein kinase, and CHK2 after IR, suggesting that NS-123 inhibits a critical step in the DNA repair pathway. These results show the potential of this cell-based, high-throughput screening method to identify novel radiosensitizers and suggest that NS-123 and similar nitrophenol compounds may be effective in antiglioma modalities.
- Published
- 2007
- Full Text
- View/download PDF
31. Inhibition of phosphatidylinositol-3-OH kinase/Akt signaling impairs DNA repair in glioblastoma cells following ionizing radiation.
- Author
-
Kao GD, Jiang Z, Fernandes AM, Gupta AK, and Maity A
- Subjects
- Cell Cycle, Cell Line, Tumor, Chromones pharmacology, DNA Damage, DNA Repair, Dose-Response Relationship, Drug, Doxycycline pharmacology, Histones metabolism, Humans, Morpholines pharmacology, Phosphoric Monoester Hydrolases metabolism, Radiation, Ionizing, Signal Transduction, Glioblastoma metabolism, Phosphatidylinositol 3-Kinases chemistry, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Radiation therapy is a mainstay in the treatment of glioblastomas, but these tumors are often associated with radioresistance. Activation of the phosphatidylinositol-3-OH kinase (PI3K)/Akt pathway, which occurs frequently in glioblastomas due to inactivation of the tumor suppressor phosphatase and tensin homologue (PTEN), correlates with radioresistance. To directly test the link between Akt activation and radioresistance, we utilized PTEN-deficient U251 glioblastoma cells engineered to inducibly restore PTEN upon exposure to doxycycline. These cells showed high basal levels of Akt activation (i.e. high levels of phospho-Akt), but induction of PTEN led to substantially decreased phospho-Akt and was associated with radiosensitization. To investigate whether the PTEN-induced radiosensitization was attributable to impaired sensing versus repair of DNA damage, we assessed levels of gamma-H2AX after ionizing radiation in U251 cells induced for PTEN. Initial post-radiation levels of gamma-H2AX foci were not decreased in PTEN-induced cells; however, the resolution of these foci was significantly delayed. In contrast to these results, induction of phosphatase-dead PTEN showed no appreciable effect. Finally, exposure of cells to the PI3K inhibitor LY294002 did not decrease the occurrence of gamma-H2AX foci after irradiation but did markedly delay their resolution. These results together support a direct link between Akt activation, repair of DNA damage, and radioresistance in glioblastoma. Targeting the PI3K/Akt pathway may modulate DNA repair to improve the efficacy of radiation therapy.
- Published
- 2007
- Full Text
- View/download PDF
32. "No Turning Bax" in the combined battle against prostate cancer:.
- Author
-
Rajendran RR and Kao GD
- Subjects
- Animals, Clinical Trials as Topic, Humans, Male, Proto-Oncogene Proteins c-bcl-2 metabolism, Apoptosis physiology, Biomarkers, Tumor analysis, Prostatic Neoplasms metabolism, Signal Transduction physiology, bcl-2-Associated X Protein metabolism
- Published
- 2007
- Full Text
- View/download PDF
33. Tripin/hSgo2 recruits MCAK to the inner centromere to correct defective kinetochore attachments.
- Author
-
Huang H, Feng J, Famulski J, Rattner JB, Liu ST, Kao GD, Muschel R, Chan GK, and Yen TJ
- Subjects
- Aurora Kinase B, Aurora Kinases, HeLa Cells, Humans, Kinetochores ultrastructure, Phosphoprotein Phosphatases metabolism, Protein Binding physiology, Protein Kinases metabolism, Protein Serine-Threonine Kinases metabolism, Protein Transport physiology, Anaphase physiology, Cell Cycle Proteins metabolism, Kinesins metabolism, Kinetochores metabolism
- Abstract
hSgo2 (previously annotated as Tripin) was recently reported to be a new inner centromere protein that is essential for centromere cohesion (Kitajima et al., 2006). In this study, we show that hSgo2 exhibits a dynamic distribution pattern, and that its localization depends on the BUB1 and Aurora B kinases. hSgo2 is concentrated at the inner centromere of unattached kinetochores, but extends toward the kinetochores that are under tension. This localization pattern is reminiscent of MCAK, which is a microtubule depolymerase that is believed to be a key component of the error correction mechanism at kinetochores. Indeed, we found that hSgo2 is essential for MCAK to localize to the centromere. Delocalization of MCAK accounts for why cells depleted of hSgo2 exhibit kinetochore attachment defects that go uncorrected, despite a transient delay in the onset of anaphase. Consequently, these cells exhibit a high frequency of lagging chromosomes when they enter anaphase. We confirmed that hSgo2 is associated with PP2A, and we propose that it contributes to the spatial regulation of MCAK activity within inner centromere and kinetochore.
- Published
- 2007
- Full Text
- View/download PDF
34. Oncogenic K-Ras signals through epidermal growth factor receptor and wild-type H-Ras to promote radiation survival in pancreatic and colorectal carcinoma cells.
- Author
-
Cengel KA, Voong KR, Chandrasekaran S, Maggiorella L, Brunner TB, Stanbridge E, Kao GD, McKenna WG, and Bernhard EJ
- Subjects
- Cell Line, Tumor, Cell Survival genetics, Cell Survival radiation effects, Colorectal Neoplasms genetics, Colorectal Neoplasms radiotherapy, Gene Expression Regulation, Neoplastic radiation effects, Humans, Pancreatic Neoplasms genetics, Pancreatic Neoplasms radiotherapy, Colorectal Neoplasms pathology, ErbB Receptors physiology, ErbB Receptors radiation effects, Genes, ras physiology, Genes, ras radiation effects, Pancreatic Neoplasms pathology, Signal Transduction genetics, Signal Transduction radiation effects
- Abstract
Pancreatic and colorectal carcinomas frequently express oncogenic/mutant K-Ras that contributes to both tumorigenesis and clinically observed resistance to radiation treatment. We have previously shown that farnesyltransferase inhibitors (FTI) radiosensitize many pancreatic and colorectal cancer cell lines that express oncogenic K-ras at doses that inhibit the prenylation and activation of H-Ras but not K-Ras. In the present study, we have examined the mechanism of FTI-mediated radiosensitization in cell lines that express oncogenic K-Ras and found that wild-type H-Ras is a contributor to radiation survival in tumor cells that express oncogenic K-Ras. In these experiments, inhibiting the expression of oncogenic K-Ras, wild-type H-Ras, or epidermal growth factor receptor (EGFR) led to similar levels of radiosensitization as treatment with the FTI tipifarnib. Treatment with the EGFR inhibitor gefitinib led to similar levels of radiosensitization, and the combinations of tipifarnib or gefitinib plus inhibition of K-Ras, H-Ras, or EGFR expression did not provide additional radiosensitization compared with tipifarnib or gefitinib alone. Finally, supplementing culture medium with the EGFR ligand transforming growth factor alpha was able to reverse the radiosensitizing effect of inhibiting K-ras expression. Taken together, these findings suggest that EGFR-activated H-Ras signaling is initiated by oncogenic K-Ras to promote radiation survival in pancreatic and colorectal cancers.
- Published
- 2007
- Full Text
- View/download PDF
35. Aloe(-emodin) for cancer? More than just a comforting salve.
- Author
-
Dorsey JF and Kao GD
- Subjects
- Anthraquinones, Cell Line, Tumor, Cell Proliferation drug effects, Emodin isolation & purification, Humans, Emodin pharmacology, Rheum chemistry, Stomach Neoplasms metabolism
- Published
- 2007
- Full Text
- View/download PDF
36. A novel histone deacetylase pathway regulates mitosis by modulating Aurora B kinase activity.
- Author
-
Li Y, Kao GD, Garcia BA, Shabanowitz J, Hunt DF, Qin J, Phelan C, and Lazar MA
- Subjects
- A Kinase Anchor Proteins, Acetylation, Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing metabolism, Aurora Kinase B, Aurora Kinases, Base Sequence, Cell Division, Cell Line, Enzyme Inhibitors pharmacology, G2 Phase, HeLa Cells, Histone Deacetylase Inhibitors, Histone Deacetylases chemistry, Histone Deacetylases genetics, Histones chemistry, Histones metabolism, Humans, Hydroxamic Acids pharmacology, Models, Biological, Multiprotein Complexes, RNA genetics, RNA Interference, Histone Deacetylases metabolism, Mitosis physiology, Protein Serine-Threonine Kinases metabolism
- Abstract
Histone deacetylase (HDAC) inhibitors perturb the cell cycle and have great potential as anti-cancer agents, but their mechanism of action is not well established. HDACs classically function as repressors of gene expression, tethered to sequence-specific transcription factors. Here we report that HDAC3 is a critical, transcription-independent regulator of mitosis. HDAC3 forms a complex with A-Kinase-Anchoring Proteins AKAP95 and HA95, which are targeted to mitotic chromosomes. Deacetylation of H3 in mitosis requires AKAP95/HA95 and HDAC3 and provides a hypoacetylated H3 tail that is the preferred substrate for Aurora B kinase. Phosphorylation of H3S10 by Aurora B leads to dissociation of HP1 proteins from methylated H3K9 residues on mitotic heterochromatin. This transcription-independent pathway, involving interdependent changes in histone modification and protein association, is required for normal progression through mitosis and is an unexpected target of HDAC inhibitors, a class of drugs currently in clinical trials for treating cancer.
- Published
- 2006
- Full Text
- View/download PDF
37. Radiosensitizing effects of the prenyltransferase inhibitor AZD3409 against RAS mutated cell lines.
- Author
-
Cengel KA, Deutsch E, Stephens TC, Voong KR, Kao GD, and Bernhard EJ
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Mice, Mice, Nude, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Xenograft Model Antitumor Assays, ras Proteins biosynthesis, ras Proteins genetics, Dimethylallyltranstransferase antagonists & inhibitors, Genes, ras, Mutation, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms radiotherapy, Radiation-Sensitizing Agents pharmacology
- Abstract
Mutations at the H-, N- and K-ras loci are among the most frequent genetic alterations in human cancers. In this study, we have investigated the effect of AZD3409, a novel, peptidomimetic prenyltransferase inhibitor (PTI), on the radiosensitivity of cells with mutated ras alleles. AZD3409, developed by AstraZeneca, inhibits both farnesyl- and geranylgeranyl transferase in cell free systems. AZD3409 inhibits the growth of a variety of human cancer cell lines, including cells that express mutant alleles of either K- or H- ras and was well tolerated when administered orally to healthy volunteers in a phase I clinical trial. We have previously shown that PTI can radiosensitize human and rodent cancer cell lines that express activated RAS. Here we assessed the ability of AZD3409 to radiosensitize human cancer cell lines in vivo and in vitro and the activation state of RAS proteins in treated cells. Once daily oral administration of AZD3409 to nude mice bearing PSN-1 and MiaPaCa-2 human pancreatic cancer xenografts expressing mutant K-ras was well tolerated and resulted in a supra-additive reduction in clonogenic cell survival after irradiation. Similarly, AZD3409 reduced clonogenic survival in cells that express either mutant K- or H- ras in vitro. We next examined the effect of AZD3409 on the processing and activation of K- and H-RAS. AZD3409-mediated radiosensitization, both in vivo and in vitro, correlates with a decrease in H-RAS processing without detectable effect on K-RAS processing. RAS activation assays show that the decreased H-RAS processing is accompanied by decreased H-RAS activation in cell lines with mutations in either K- or H-ras. However, no decrease in K-RAS activation was detected. Thus, radiosensitization of human cancer cells that express mutated K-RAS occurred under conditions where AZD3409 inihibits the activation of farneyslated H-RAS, but did not inhibit K-RAS activation.
- Published
- 2006
- Full Text
- View/download PDF
38. Zebrafish as a "biosensor"? Effects of ionizing radiation and amifostine on embryonic viability and development.
- Author
-
Geiger GA, Parker SE, Beothy AP, Tucker JA, Mullins MC, and Kao GD
- Subjects
- Animals, Embryo, Nonmammalian radiation effects, Embryonic Development drug effects, Radiation, Ionizing, Radiation-Protective Agents pharmacology, Amifostine pharmacology, Cell Survival radiation effects, Embryo, Nonmammalian physiology, Embryonic Development radiation effects, Zebrafish embryology
- Abstract
The zebrafish (Danio rerio) has emerged as a popular vertebrate model system for cancer and treatment-related research. Benefits include ease of care, rapid development, optical clarity of embryos, which allows visualization of major organ systems, and opportunities for genetic manipulation. However, specific parameters of radiation sensitivity have not been systematically documented. We investigated the effects of radiation and a radiomodifier on zebrafish viability and embryonic development. Embryos were exposed to gamma-radiation (5, 10, or 20 Gy) at sequential times postfertilization and serially assessed for viability and morphologic abnormalities. As expected, lethality and morphologic perturbations were more pronounced earlier in embryogenesis and with higher radiation doses and were partially reversed by amifostine. The effects of radiation and concurrent treatment with amifostine on the developmental organization of the eye and brain were striking. Radiation resulted in hypocellularity and disorganization of the cellular layers of the retina, effects partially reversed by amifostine, as well as lens opacification. Radiation strikingly reduced the volume of brain, but the volume loss was substantially blocked by amifostine. Increased terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling signal was noted in both the irradiated eye and brain, but reduced by amifostine. Finally, irradiating embryos resulted in caspase activation detectable in 96-well microplates, which was proportional to the number of embryos and radiation dose; the degree of activation was markedly reduced by amifostine. These results together suggest the power and versatility of the zebrafish in assessing the effects of radiation and radiomodifiers on organ and tissue development.
- Published
- 2006
- Full Text
- View/download PDF
39. Akt1 activation can augment hypoxia-inducible factor-1alpha expression by increasing protein translation through a mammalian target of rapamycin-independent pathway.
- Author
-
Pore N, Jiang Z, Shu HK, Bernhard E, Kao GD, and Maity A
- Subjects
- Cell Line, Tumor, Chromones pharmacology, Enzyme Activation, Enzyme Inhibitors pharmacology, Glioblastoma enzymology, Glioblastoma genetics, Glioblastoma metabolism, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Male, Morpholines pharmacology, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Protein Biosynthesis drug effects, Proto-Oncogene Proteins c-akt antagonists & inhibitors, RNA, Small Interfering genetics, TOR Serine-Threonine Kinases, Transfection, Vascular Endothelial Growth Factor A immunology, Vascular Endothelial Growth Factor A metabolism, Hypoxia-Inducible Factor 1, alpha Subunit biosynthesis, Protein Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism
- Abstract
The phosphoinositide 3-kinase (PI3K)/Akt pathway is commonly activated in cancer; therefore, we investigated its role in hypoxia-inducible factor-1alpha (HIF-1alpha) regulation. Inhibition of PI3K in U87MG glioblastoma cells, which have activated PI3K/Akt activity secondary to phosphatase and tensin homologue deleted on chromosome 10 (PTEN) mutation, with LY294002 blunted the induction of HIF-1alpha protein and its targets vascular endothelial growth factor and glut1 mRNA in response to hypoxia. Introduction of wild-type PTEN into these cells also blunted HIF-1alpha induction in response to hypoxia and decreased HIF-1alpha accumulation in the presence of the proteasomal inhibitor MG132. Akt small interfering RNA (siRNA) also decreased HIF-1alpha induction under hypoxia and its accumulation in normoxia in the presence of dimethyloxallyl glycine, a prolyl hydroxylase inhibitor that prevents HIF-1alpha degradation. Metabolic labeling studies showed that Akt siRNA decreased HIF-1alpha translation in normoxia in the presence of dimethyloxallyl glycine and in hypoxia. Inhibition of mammalian target of rapamycin (mTOR) with rapamycin (10-100 nmol/L) had no significant effect on HIF-1alpha induction in a variety of cell lines, a finding that was confirmed using mTOR siRNA. Furthermore, neither mTOR siRNA nor rapamycin decreased HIF-1alpha translation as determined by metabolic labeling studies. Therefore, our results indicate that Akt can augment HIF-1alpha expression by increasing its translation under both normoxic and hypoxic conditions; however, the pathway we are investigating seems to be rapamycin insensitive and mTOR independent. These observations, which were made on cells grown in standard tissue culture medium (10% serum), were confirmed in PC3 prostate carcinoma cells. We did find that rapamycin could decrease HIF-1alpha expression when cells were cultured in low serum, but this seems to represent a different pathway.
- Published
- 2006
- Full Text
- View/download PDF
40. EGFR tyrosine kinase inhibitors decrease VEGF expression by both hypoxia-inducible factor (HIF)-1-independent and HIF-1-dependent mechanisms.
- Author
-
Pore N, Jiang Z, Gupta A, Cerniglia G, Kao GD, and Maity A
- Subjects
- Binding Sites, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell enzymology, Cell Line, Tumor, Down-Regulation drug effects, Epidermal Growth Factor antagonists & inhibitors, Erlotinib Hydrochloride, Gefitinib, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms enzymology, Humans, Promoter Regions, Genetic, Quinazolines pharmacology, Sp1 Transcription Factor metabolism, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor A genetics, Carcinoma, Squamous Cell metabolism, ErbB Receptors antagonists & inhibitors, Head and Neck Neoplasms metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Protein Kinase Inhibitors pharmacology, Vascular Endothelial Growth Factor A biosynthesis
- Abstract
Epidermal growth factor receptor (EGFR) inhibitors can decrease vascular endothelial growth factor (VEGF) expression and tumor angiogenesis. In the current study, we investigate the molecular pathways by which this occurs using two drugs that have been used in the clinic, gefitinib (Iressa) and erlotinib (Tarceva). The decrease in VEGF expression by gefitinib in SQ20B squamous cell carcinoma cells was opposed by adenoviral expression of Akt in these cells. The hypoxia-inducible factor-1 (HIF-1) binding site located at approximately -1 kbp in the VEGF promoter was not required for down-regulation of promoter activity by gefitinib under normoxia. Furthermore, the drug decreased activity of a reporter containing the -88/+54 region. In a gel shift assay, gefitinib led to decreased retardation of a labeled DNA oligonucleotide probe corresponding to the -88/-66 region of the VEGF promoter, which contains Sp1 binding sites. These effects of gefitinib on VEGF promoter activity and DNA binding were both reversed by Akt expression. Phosphorylation of Sp1 was decreased in the presence of gefitinib. Gefitinib also decreases VEGF expression by decreasing HIF-1alpha expression. This occurs due to decreased protein translation without any change in the level of HIF-1alpha mRNA. Together, these results suggest that gefitinib decreases VEGF expression both by decreasing Sp1 binding to the proximal core VEGF promoter and by down-regulating HIF-1alpha expression. Similar results were obtained with erlotinib in SQ20B and gefitinib in HSC3 squamous carcinoma cells. These results indicate that there are at least two separate mechanisms by which EGFR inhibitors decrease VEGF expression.
- Published
- 2006
- Full Text
- View/download PDF
41. Gamma radiation increases endonuclease-dependent L1 retrotransposition in a cultured cell assay.
- Author
-
Farkash EA, Kao GD, Horman SR, and Prak ET
- Subjects
- Aminoglycosides pharmacology, Animals, Cells, Cultured, Cricetinae, DNA Damage, Endodeoxyribonucleases genetics, Enediynes, Genes, Reporter, Genomics, Histones analysis, Humans, Mutagens pharmacology, Mutation, Endodeoxyribonucleases metabolism, Gamma Rays, Long Interspersed Nucleotide Elements
- Abstract
Long Interspersed Elements (LINE-1s, L1s) are the most active mobile elements in the human genome and account for a significant fraction of its mass. The propagation of L1 in the human genome requires disruption and repair of DNA at the site of integration. As Barbara McClintock first hypothesized, genotoxic stress may contribute to the mobilization of transposable elements, and conversely, element mobility may contribute to genotoxic stress. We tested the ability of genotoxic agents to increase L1 retrotransposition in a cultured cell assay. We observed that cells exposed to gamma radiation exhibited increased levels of L1 retrotransposition. The L1 retrotransposition frequency was proportional to the number of phosphorylated H2AX foci, an indicator of genotoxic stress. To explore the role of the L1 endonuclease in this context, endonuclease-deficient tagged L1 constructs were produced and tested for their activity in irradiated cells. The activity of the endonuclease-deficient L1 was very low in irradiated cells, suggesting that most L1 insertions in irradiated cells still use the L1 endonuclease. Consistent with this interpretation, DNA sequences that flank L1 insertions in irradiated cells harbored target site duplications. These results suggest that increased L1 retrotransposition in irradiated cells is endonuclease dependent. The mobilization of L1 in irradiated cells potentially contributes to genomic instability and could be a driving force for secondary mutations in patients undergoing radiation therapy.
- Published
- 2006
- Full Text
- View/download PDF
42. Regulation of histone deacetylase 4 expression by the SP family of transcription factors.
- Author
-
Liu F, Pore N, Kim M, Voong KR, Dowling M, Maity A, and Kao GD
- Subjects
- Animals, Base Sequence, Binding Sites, Cells, Cultured, Consensus Sequence physiology, Drosophila cytology, HeLa Cells, Histone Deacetylases drug effects, Histone Deacetylases metabolism, Humans, Male, Molecular Sequence Data, Plicamycin pharmacology, Promoter Regions, Genetic, Repressor Proteins drug effects, Repressor Proteins metabolism, Sp1 Transcription Factor genetics, Sp3 Transcription Factor genetics, Tissue Distribution, Gene Expression Regulation, Enzymologic drug effects, Histone Deacetylases genetics, Repressor Proteins genetics, Sp1 Transcription Factor physiology, Sp3 Transcription Factor physiology
- Abstract
Histone deacetylases mediate critical cellular functions but relatively little is known about mechanisms controlling their expression, including expression of HDAC4, a class II HDAC implicated in the modulation of cellular differentiation and viability. Endogenous HDAC4 mRNA, protein levels and promoter activity were all readily repressed by mithramycin, suggesting regulation by GC-rich DNA sequences. We validated consensus binding sites for Sp1/Sp3 transcription factors in the HDAC4 promoter through truncation studies and targeted mutagenesis. Specific and functional binding by Sp1/Sp3 at these sites was confirmed with chromatin immunoprecipitation (ChIP) and electromobility shift assays (EMSA). Cotransfection of either Sp1 or Sp3 with a reporter driven by the HDAC4 promoter led to high activities in SL2 insect cells (which lack endogenous Sp1/Sp3). In human cells, restored expression of Sp1 and Sp3 up-regulated HDAC4 protein levels, whereas levels were decreased by RNA-interference-mediated knockdown of either protein. Finally, variable levels of Sp1 were in concordance with that of HDAC4 in a number of human tissues and cancer cell lines. These studies together characterize for the first time the activity of the HDAC4 promoter, through which Sp1 and Sp3 modulates expression of HDAC4 and which may contribute to tissue or cell-line-specific expression of HDAC4.
- Published
- 2006
- Full Text
- View/download PDF
43. Histone deacetylase inhibitor-mediated radiosensitization of human cancer cells: class differences and the potential influence of p53.
- Author
-
Kim IA, Shin JH, Kim IH, Kim JH, Kim JS, Wu HG, Chie EK, Ha SW, Park CI, and Kao GD
- Subjects
- Amides pharmacology, Benzothiazoles, Biphenyl Compounds pharmacology, Cell Cycle drug effects, Cell Cycle radiation effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation radiation effects, Dose-Response Relationship, Drug, Dose-Response Relationship, Radiation, Fatty Acids, Unsaturated pharmacology, Gene Expression Regulation drug effects, HeLa Cells, Histone Deacetylases radiation effects, Humans, Hydroxamic Acids antagonists & inhibitors, Hydroxamic Acids pharmacology, Naphthalenes pharmacology, Pyrones pharmacology, Thiazoles pharmacology, Toluene analogs & derivatives, Toluene pharmacology, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 radiation effects, Enzyme Inhibitors pharmacology, Histone Deacetylase Inhibitors, Histone Deacetylases classification, Radiation-Sensitizing Agents pharmacology, Tumor Suppressor Protein p53 drug effects
- Abstract
Histone deacetylase inhibitors (HDI) are emerging as potentially useful components of the anticancer armamentarium and as useful tools to dissect mechanistic pathways. HDIs that globally inhibit histone deacetylases (HDAC) have radiosensitizing effects, but the relative contribution of specific HDAC classes remains unclear. Newly characterized HDIs are now available that preferentially inhibit specific HDAC classes, including SK7041 (inhibits class I HDACs) and splitomicin (inhibits class III HDACs). We investigated in human cancer cells the relative radiosensitizations that result from blocking specific HDAC classes. We found that trichostatin A (TSA; inhibitor of both class I and II HDACs) was the most effective radiosensitizer, followed by the class I inhibitor SK7041, whereas splitomicin (inhibitor of class III) had least effect. Interestingly, radiosensitization by TSA in cell lines expressing p53 was more pronounced than in isogenic lines lacking p53. Radiosensitization of cells expressing p53 by TSA was reduced by pifithrin-alpha, a small-molecule inhibitor of p53. In contrast, the radiosensitization by TSA of cells expressing low levels of p53 was enhanced by transfection of wild-type p53-expressing vector or pretreatment with leptomycin B, an inhibitor of nuclear export that increased intracellular levels of p53. These effects on radiosensitization were respectively muted or not seen in cells treated with SK7041 or splitomicin. To our knowledge, this may be among the first systematic investigations of the comparative anticancer effects of inhibiting specific classes of HDACs, with results suggesting differences in the degrees of radiosensitization, which in some cell lines may be influenced by p53 expression.
- Published
- 2006
- Full Text
- View/download PDF
44. Caspase-mediated specific cleavage of BubR1 is a determinant of mitotic progression.
- Author
-
Kim M, Murphy K, Liu F, Parker SE, Dowling ML, Baff W, and Kao GD
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, Caspase 3, Caspase Inhibitors, Cell Cycle drug effects, Cell Cycle Proteins, Chickens, Conserved Sequence, Doxycycline pharmacology, Enzyme Activation, HeLa Cells, Humans, Mice, Microtubules drug effects, Microtubules metabolism, Molecular Sequence Data, Mutagenesis, Site-Directed, Nocodazole pharmacology, Paclitaxel pharmacology, Protein Kinases biosynthesis, Protein Kinases genetics, Protein Serine-Threonine Kinases, Sequence Homology, Amino Acid, Spindle Apparatus drug effects, Xenopus laevis, Caspases metabolism, Cell Cycle physiology, Protein Kinases metabolism, Spindle Apparatus metabolism
- Abstract
The fidelity of chromosomal duplication is monitored by cell cycle checkpoints operational during mitosis. One such cell cycle delay is invoked by microtubule-targeting agents such as nocodazole or paclitaxel (Taxol) and is mediated by mitotic checkpoint proteins that include BubR1. Relatively little is known about the regulation of expression and stability of BubR1 (or other checkpoint proteins) and how these factors dictate the durability of the cell cycle delay. We report here that treatment of HeLa cells with spindle-disrupting agents resulted in caspase activation and precipitated the cleavage of BubR1. This mechanism ultimately leads to reduced levels of full-length protein, which are accompanied by abrogation of the mitotic block; the checkpoint abrogation is substantially accelerated by inhibition of de novo protein synthesis. In contrast, inhibition of caspase activity blocked BubR1 degradation and prolonged mitosis. To confirm a direct link between caspase activity and BubR1 protein expression, we identified by site-directed mutagenesis the specific caspase cleavage sites cleaved after exposure to paclitaxel. Surprisingly, BubR1 has two sites of cleavage: primarily at Asp607/Asp610 and secondarily at Asp576/Asp579. BubR1 mutated at both locations (BubR1Delta579Delta610) was resistant to paclitaxel-induced degradation. Expression of BubR1Delta579Delta610 augmented the mitotic delay induced by spindle disruption in transfected cells as well as in clones engineered to inducibly express the mutant protein upon exposure to doxycycline and ultimately led to increased aneuploidy. Underscoring the importance of these caspase cleavage sites, both tetrapeptide motifs are identified in the amino acid sequences of human, mouse, chicken, and Xenopus BubR1. These results are potentially the first to link the control of the stability of a key mitotic checkpoint protein to caspase activation, a regulatory pathway that may be involved in killing defective cells and that has been evolutionarily conserved.
- Published
- 2005
- Full Text
- View/download PDF
45. Protein kinase C delta stimulates apoptosis by initiating G1 phase cell cycle progression and S phase arrest.
- Author
-
Santiago-Walker AE, Fikaris AJ, Kao GD, Brown EJ, Kazanietz MG, and Meinkoth JL
- Subjects
- Adenoviridae genetics, Animals, Blotting, Western, Bromodeoxyuridine pharmacology, Cell Cycle, Cell Death, Cell Proliferation, Checkpoint Kinase 1, Chromones pharmacology, DNA metabolism, DNA Damage, Enzyme Inhibitors pharmacology, Epithelial Cells metabolism, Flow Cytometry, G1 Phase, Models, Biological, Morpholines pharmacology, Phosphorylation, Protein Kinase C metabolism, Protein Kinase C-delta, Protein Kinases metabolism, Rats, Rats, Wistar, S Phase, Thyroid Gland cytology, Tumor Suppressor Protein p53 metabolism, Up-Regulation, Apoptosis, Protein Kinase C physiology
- Abstract
Overexpression of protein kinase C delta (PKCdelta) stimulates apoptosis in a wide variety of cell types through a mechanism that is incompletely understood. PKCdelta-deficient cells are impaired in their response to DNA damage-induced apoptosis, suggesting that PKCdelta is required to mount an appropriate apoptotic response under conditions of stress. The mechanism through which it does so remains elusive. In addition to effects on cell survival, PKCdelta elicits pleiotropic effects on cellular proliferation. We now provide the first evidence that the ability of PKCdelta to stimulate apoptosis is intimately linked to its ability to stimulate G(1) phase cell cycle progression. Using an adenoviral-based expression system to express PKCalpha,-delta, and -epsilon in epithelial cells, we demonstrate that a modest increase in PKCdelta activity selectively stimulates quiescent cells to initiate G(1) phase cell cycle progression. Rather than completing the cell cycle, PKCdelta-infected cells arrest in S phase, an event that triggers caspase-dependent apoptotic cell death. Apoptosis was preceded by the activation of cell cycle checkpoints, culminating in the phosphorylation of Chk-1 and p53. Strikingly, blockade of S phase entry using the phosphatidylinositol 3-kinase inhibitor LY294002 prevented checkpoint activation and apoptosis. In contrast, inhibitors of mitogen-activated protein kinase cascades failed to prevent apoptosis. These findings demonstrate that the biological effects of PKCdelta can be extended to include positive regulation of G(1) phase cell cycle progression. Importantly, they reveal the existence of a novel, cell cycle-dependent mechanism through which PKCdelta stimulates cell death.
- Published
- 2005
- Full Text
- View/download PDF
46. Newly identified roles for an old guardian: profound deficiency of the mitotic spindle checkpoint protein BubR1 leads to early aging and infertility.
- Author
-
Kim M and Kao GD
- Subjects
- Animals, Cell Cycle Proteins, Humans, Protein Serine-Threonine Kinases, Spindle Apparatus, Aging metabolism, Infertility metabolism, Protein Kinases deficiency
- Abstract
The mitotic spindle checkpoint ensures proper chromosome segregation, and thereby guards against the deleterious effects of aneuploidy. The protein components of the check-point machinery include evolutionarily conserved proteins such as BubR1. While the molecular and cellular biology of this checkpoint is becoming increasingly clarified, the ultimate consequences for overall health of deficiency of specific components such as BubR1 are much less clear--in part due to the embryonic lethality of complete knockouts. Through a clever combination of hypomorphic and knockout alleles, Baker and colleagues were able to engineer mice with graduated levels of BubR1 protein. In doing so, they established the threshold permitting survival to adulthood, but even more intriguingly, they discovered critical roles for BubR1 in preventing early aging and infertility.
- Published
- 2005
- Full Text
- View/download PDF
47. Mitotic spindle checkpoint inactivation by trichostatin a defines a mechanism for increasing cancer cell killing by microtubule-disrupting agents.
- Author
-
Dowling M, Voong KR, Kim M, Keutmann MK, Harris E, and Kao GD
- Subjects
- Antineoplastic Agents pharmacology, Centrosome drug effects, Drug Therapy, Combination, Flow Cytometry, Fluorescent Antibody Technique, HeLa Cells, Histone Deacetylase Inhibitors, Histones metabolism, Humans, Immunoblotting, Kinetochores drug effects, Kinetochores metabolism, Mitosis drug effects, Neoplasms metabolism, Neoplasms pathology, Nocodazole pharmacology, Paclitaxel pharmacology, Phosphorylation drug effects, Protein Serine-Threonine Kinases, Cell Cycle drug effects, Cell Cycle Proteins metabolism, Cell Survival drug effects, Enzyme Inhibitors pharmacology, Hydroxamic Acids pharmacology, Microtubules drug effects, Neoplasms drug therapy, Protein Kinases metabolism, Spindle Apparatus drug effects
- Abstract
Microtubule-disrupting agents such as the taxanes comprise some of the most clinically useful chemotherapeutic agents and invoke the spindle checkpoint in proliferating cells. A robust spindle checkpoint in turn may forestall mitotic catastrophe, potentially providing a mechanism that permits cancer cells to survive transient exposure to these drugs. Previous reports on G2-M cell cycle progression by histone deacetylase inhibitors suggested a potential role in modulating the therapeutic efficacy of microtubule-disrupting agents. As both classes of agents are generally administered in clinical trials as pulse treatments, we investigated in human cancer cells the effects of brief treatments with the histone deacetylase inhibitor trichostatin A (TSA) alone or with nocodazole or paclitaxel (Taxol) on cell cycle progression and the spindle checkpoint. Treatment of synchronized cells with 200 ng/ml of TSA alone for eight hours to completely block class I and II HDACs did not interfere with progression into mitosis with chromosomal condensation as confirmed by MPM-2 expression. TSA treatment at this concentration surprisingly did not interfere with formation of the mitotic spindle or centrosomal separation, but instead led to missegregation of chromosomes, suggesting effects on the spindle checkpoint. Consistent with this hypothesis, TSA abrogated the phosphorylation and kinetochore localization of the mitotic checkpoint protein BubR1 and the phosphorylation of histone H3 after paclitaxel and nocodazole treatment. These effects in turn led to rapid cell death and considerably reduced clonogenic survival. These results together suggest that by inactivating the spindle checkpoint, TSA can potentiate the lethal effects of microtubule-disrupting drugs, a strategy that might be usefully exploited for optimizing anticancer therapy.
- Published
- 2005
48. Mitotic checkpoint, aneuploidy and cancer.
- Author
-
Yen TJ and Kao GD
- Subjects
- Animals, Humans, Kinetochores, Models, Animal, Neoplasms metabolism, Aneuploidy, Genes, cdc, Mitosis genetics, Neoplasms genetics
- Published
- 2005
- Full Text
- View/download PDF
49. Caspase-mediated specific cleavage of human histone deacetylase 4.
- Author
-
Liu F, Dowling M, Yang XJ, and Kao GD
- Subjects
- Amino Acid Motifs, Animals, Apoptosis, Aspartic Acid chemistry, Binding Sites, Biological Transport, CHO Cells, Caspase 3, Cell Death, Cell Differentiation, Cell Line, Tumor, Cell Survival, Cricetinae, Cycloheximide pharmacology, Cysteine Endopeptidases metabolism, Dactinomycin pharmacology, Doxycycline pharmacology, Genetic Vectors, Glutamic Acid chemistry, HeLa Cells, Humans, Models, Biological, Multienzyme Complexes metabolism, Mutagenesis, Site-Directed, Proline chemistry, Proteasome Endopeptidase Complex, Protein Binding, Protein Structure, Tertiary, Protein Synthesis Inhibitors pharmacology, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Serine chemistry, Threonine chemistry, Time Factors, Ultraviolet Rays, Caspases metabolism, Histone Deacetylases metabolism, Repressor Proteins metabolism
- Abstract
Histone deacetylase 4 (HDAC4) is a class II HDAC implicated in controlling gene expression important for diverse cellular functions, but little is known about how its expression and stability are regulated. We report here that this deacetylase is unusually unstable, with a half-life of less than 8 h. Consistent with the instability of HDAC4 protein, its mRNA was also highly unstable (with a half-life of less than 4 h). The degradation of HDAC4 could be accelerated by exposure of cells to ultraviolet irradiation. HDAC4 degradation was not dependent on proteasome or CRM1-mediated export activity but instead was caspase-dependent and was detectable in diverse human cancer lines. Of two potential caspase consensus motifs in HDAC4, both lying within a region containing proline-, glutamic acid-, serine-, and threonine-rich (PEST) sequences, we identified, by site-directed mutagenesis, Asp-289 as the prime cleavage site. Notably, this residue is not conserved among other class IIa members, HDAC5, -7, and -9. Finally, the induced expression of caspase-cleavable HDAC4 led to markedly increased apoptosis. These results therefore unexpectedly link the regulation of HDAC4 protein stability to caspases, enzymes that are important for controlling cell death and differentiation.
- Published
- 2004
- Full Text
- View/download PDF
50. "Hood ornament" or "V-10 engine"? Myths and realities regarding physician-scientists in academic radiation oncology departments.
- Author
-
Kao GD and McKenna WG
- Subjects
- United States, Academic Medical Centers organization & administration, Biomedical Research organization & administration, Physicians, Radiation Oncology organization & administration, Research organization & administration, Science organization & administration
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.