Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Clin Cancer Res ; 21(24): 5630-8, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26265694

ABSTRACT

PURPOSE: Predominant causes of head and neck cancer recurrence after radiotherapy are rapid repopulation, hypoxia, fraction of cancer stem cells, and intrinsic radioresistance. Currently, intrinsic radioresistance can only be assessed by ex vivo colony assays. Besides being time-consuming, colony assays do not identify causes of intrinsic resistance. We aimed to identify a biomarker for intrinsic radioresistance to be used before start of treatment and to reveal biologic processes that could be targeted to overcome intrinsic resistance. EXPERIMENTAL DESIGN: We analyzed both microRNA and mRNA expression in a large panel of head and neck squamous cell carcinoma (HNSCC) cell lines. Expression was measured on both irradiated and unirradiated samples. Results were validated using modified cell lines and a series of patients with laryngeal cancer. RESULTS: miRs, mRNAs, and gene sets that correlated with resistance could be identified from expression data of unirradiated cells. The presence of epithelial-to-mesenchymal transition (EMT) and low expression of miRs involved in the inhibition of EMT were important radioresistance determinants. This finding was validated in two independent cell line pairs, in which the induction of EMT reduced radiosensitivity. Moreover, low expression of the most important miR (miR-203) was shown to correlate with local disease recurrence after radiotherapy in a series of patients with laryngeal cancer. CONCLUSIONS: These findings indicate that EMT and low expression of EMT-inhibiting miRs, especially miR-203, measured in pretreatment material, causes intrinsic radioresistance of HNSCC, which could enable identification and treatment modification of radioresistant tumors. Clin Cancer Res; 21(24); 5630-8. ©2015 AACR.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , MicroRNAs/genetics , Radiation Tolerance/genetics , Cell Line, Tumor , Dose-Response Relationship, Radiation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/radiation effects , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/radiotherapy , Humans , Neoplasm Staging , Prognosis , Proportional Hazards Models , Radiation Dosage , Time Factors , Transcriptome
2.
Radiother Oncol ; 108(3): 529-34, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23849170

ABSTRACT

BACKGROUND AND PURPOSE: Tumor hypoxia is associated with therapy resistance and malignancy. Previously we demonstrated that activation of autophagy and the unfolded protein response (UPR) promote hypoxia tolerance. Here we explored the importance of ULK1 in hypoxia tolerance, autophagy induction and its prognostic value for recurrence after treatment. MATERIAL AND METHODS: Hypoxic regulation of ULK1 mRNA and protein was assessed in vitro and in primary human head and neck squamous cell carcinoma (HNSCC) xenografts. Its importance in autophagy induction, mitochondrial homeostasis and tolerance to chronic and acute hypoxia was evaluated in ULK1 knockdown cells. The prognostic value of ULK1 mRNA expression was assessed in 82 HNSCC patients. RESULTS: ULK1 enrichment was observed in hypoxic tumor regions. High enrichment was associated with a high hypoxic fraction. In line with these findings, high ULK1 expression in HNSCC patients appeared associated with poor local control. Exposure of cells to hypoxia induced ULK1 mRNA in a UPR and HIF1α dependent manner. ULK1 knockdown decreased autophagy activation, increased mitochondrial mass and ROS exposure and sensitized cells to acute and chronic hypoxia. CONCLUSIONS: We demonstrate that ULK1 is a hypoxia regulated gene and is associated with hypoxia tolerance and a worse clinical outcome.


Subject(s)
Autophagy , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Autophagy-Related Protein-1 Homolog , Carcinoma, Squamous Cell/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Head and Neck Neoplasms/pathology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Squamous Cell Carcinoma of Head and Neck , Unfolded Protein Response
3.
Nat Rev Clin Oncol ; 10(1): 27-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23165123

ABSTRACT

With the emergence of individualized medicine and the increasing amount and complexity of available medical data, a growing need exists for the development of clinical decision-support systems based on prediction models of treatment outcome. In radiation oncology, these models combine both predictive and prognostic data factors from clinical, imaging, molecular and other sources to achieve the highest accuracy to predict tumour response and follow-up event rates. In this Review, we provide an overview of the factors that are correlated with outcome-including survival, recurrence patterns and toxicity-in radiation oncology and discuss the methodology behind the development of prediction models, which is a multistage process. Even after initial development and clinical introduction, a truly useful predictive model will be continuously re-evaluated on different patient datasets from different regions to ensure its population-specific strength. In the future, validated decision-support systems will be fully integrated in the clinic, with data and knowledge being shared in a standardized, instant and global manner.


Subject(s)
Decision Support Systems, Clinical , Models, Theoretical , Neoplasms/radiotherapy , Precision Medicine , Radiation Oncology , Humans , Neoplasms/mortality , Treatment Outcome
4.
Semin Radiat Oncol ; 22(2): 108-18, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22385918

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Radiotherapy is a mainstay of treatment, either alone for early stage tumors or combined with chemotherapy for late stage tumors. An overall 5-year survival rate of around 50% for HNSCC demonstrates that treatment is often unsuccessful. Prediction of outcome is, therefore, aimed at sparing patients from ineffective and toxic treatments on the one hand, and indicating more successful treatment modalities on the other. Both functional and genetic assays have been developed to predict intrinsic radiosensitivity, hypoxia, and repopulation rate. Few, however, have shown consistent correlations with outcome across multiple studies. Messenger RNA and microRNA profiling show promise for predicting hypoxia, whereas epidermal growth factor receptor expression combined with other measures of tumor differentiation grade shows promise for predicting repopulation rate. Intrinsic radiosensitivity assays have not proven useful to date, although development of repair protein foci assays indicates promise from preclinical studies. Assays for cancer stem cell content have shown promise in several clinical studies. In addition, 2 assays showing robustness as predictors for outcome in HNSCC are human papilloma virus status and epidermal growth factor receptor expression. Neither these nor stem cell assays, however, can as yet reliably indicate alternative and better treatments for poor prognosis patients. It would be of great value to have assays that predict the benefit for an individual from combining new molecularly targeted agents with radiotherapy to increase response, in particular those that exploit tumor mutations to provide tumor specificity. Predictive assays are being developed for detecting defects in repair pathways for single- and double-strand DNA breaks, which should allow selection of drugs targeting the appropriate backup pathway, thus exploiting the concept of synthetic lethality. This is one of the most promising areas for prediction, both currently and in the future.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/radiotherapy , Neoplasm Recurrence, Local , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Humans , Predictive Value of Tests , Prognosis , Risk Factors
5.
Int J Radiat Oncol Biol Phys ; 81(5): 1515-23, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21944462

ABSTRACT

PURPOSE: The nucleoside 5-aminoimidazole-4-carboxamide riboside (AICAR) is a low-energy mimetic and adenosine monophosphate (AMP)-activated protein kinase (AMPK) agonist that can affect the phenotype of malignant cells by diminishing their anabolism. It does this by being converted to 5-aminoimidazole-4-carboxamide ribotide (ZMP), an AMP analog. We combined this promising antineoplastic agent with ionizing radiation in an attempt to increase its efficacy. METHODS AND MATERIALS: The effect of AICAR on cell proliferation, cell viability, apoptosis, reactive oxygen species production, radiosensitivity, and AMPK activation was determined in the human prostate cancer cell line PC3. To elucidate the radiosensitizing mechanism, clonogenic survival assays in the presence of a drug agonist or antagonist or with small interfering RNA targeting AMPK were done, as well as measurements of ZMP production and double strand break repair. Moreover, immunoblot analysis of the radiation response signaling pathways after AICAR treatment was performed. RESULTS: The incubation of human PC3 prostate cancer cells with AICAR-activated AMPK inhibited cell proliferation, decreased viability, increased apoptosis, and generated reactive oxygen species in a dose- and time-dependent manner. None of these endpoints gave more than additive effects when radiation was added. Radiosensitization was observed but only after 72 hours of treatment with 250 µM AICAR, suggesting that it was independent of AMPK activation. This finding was confirmed by small interfering RNA knockdown of AMPK. The mechanism of radiosensitization was associated with imbalanced deoxynucleotide pools owing to ZMP accumulation after AICAR administration that interfered with DNA repair. CONCLUSIONS: Our findings on the favorable interaction between low doses of AICAR and ionizing radiation in PC3 cells could open new perspectives for the clinical use of this or similar compounds. However, additional research is still required to establish the ZMP pathway as being of general applicability.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/metabolism , Aminoimidazole Carboxamide/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation , Gene Knockdown Techniques , Humans , Male , Metabolism , Prostatic Neoplasms/metabolism , RNA, Small Interfering , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Ribonucleotides/metabolism , Time Factors , Tumor Stem Cell Assay
6.
Nat Rev Cancer ; 11(4): 239-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21430696

ABSTRACT

Radiotherapy is used to treat approximately 50% of all cancer patients, with varying success. The dose of ionizing radiation that can be given to the tumour is determined by the sensitivity of the surrounding normal tissues. Strategies to improve radiotherapy therefore aim to increase the effect on the tumour or to decrease the effects on normal tissues. These aims must be achieved without sensitizing the normal tissues in the first approach and without protecting the tumour in the second approach. Two factors have made such approaches feasible: namely, an improved understanding of the molecular response of cells and tissues to ionizing radiation and a new appreciation of the exploitable genetic alterations in tumours. These have led to the development of treatments combining pharmacological interventions with ionizing radiation that more specifically target either tumour or normal tissue, leading to improvements in efficacy.


Subject(s)
Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation Oncology , Radiotherapy Planning, Computer-Assisted , Humans
7.
Cancer Res ; 70(21): 8706-14, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20978197

ABSTRACT

Ionizing radiation (IR) is an effective anticancer treatment, although failures still occur. To improve radiotherapy, tumor-targeted strategies are needed to increase radiosensitivity of tumor cells, without influencing normal tissue radiosensitivity. Base excision repair (BER) and single-strand break repair (SSBR) contribute to the determination of sensitivity to IR. A crucial protein in BER/SSBR is DNA polymerase ß (polß). Aberrant polß expression is commonly found in human tumors and leads to inhibition of BER. Here, we show that truncated polß variant (polß-Δ)-expressing cells depend on homologous recombination (HR) for survival after IR, indicating that a considerable fraction of polß-Δ-induced lesions are subject to repair by HR. Increased sensitization was found not to result from involvement in DNA-dependent protein kinase-dependent nonhomologous end joining, the other major double-strand break repair pathway. Caffeine and the ATM inhibitor Ku55933 cause polß-Δ-dependent radiosensitization. Consistent with the observed HR dependence and the known HR-modulating activity of ATM, polß-Δ-expressing cells showed increased radiosensitization after BRCA2 knockdown that is absent under ATM-inhibited conditions. Our data suggest that treatment with HR modulators is a promising therapeutic strategy for exploiting defects in the BER/SSBR pathway in human tumors.


Subject(s)
Chromones/pharmacology , DNA Polymerase beta/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/radiotherapy , Morpholines/pharmacology , Radiation Tolerance/drug effects , Recombination, Genetic/drug effects , Ataxia Telangiectasia Mutated Proteins , BRCA2 Protein/antagonists & inhibitors , BRCA2 Protein/metabolism , Blotting, Western , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , DNA Damage/drug effects , DNA Polymerase beta/genetics , DNA Repair/radiation effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Humans , Lung Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Radiation, Ionizing , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Stem Cell Assay , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism
8.
Clin Cancer Res ; 16(21): 5329-38, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20837694

ABSTRACT

PURPOSE: To find molecular markers from expression profiling data to predict recurrence of laryngeal cancer after radiotherapy. EXPERIMENTAL DESIGN: We generated gene expression data on pre-treatment biopsies from 52 larynx cancer patients. Patients developing a local recurrence were matched for T-stage, subsite, treatment, gender and age with non-recurrence patients. Candidate genes were then tested by immunohistochemistry on tumor material from a second series of 76 patients. Both series comprised early stage cancer treated with radiotherapy alone. Finally, gene expression data of eight larynx cancer cell lines with known radiosensitivity were analyzed. RESULTS: Nineteen patients with a local recurrence were matched with 33 controls. Gene sets for hypoxia, proliferation and intrinsic radiosensitivity did not correlate with recurrence, whereas expression of the putative stem cell marker CD44 did. In a supervised analysis, probes for all three splice variants of CD44 on the array appeared in the top 10 most significantly correlated with local recurrence. Immunohistochemical analysis of CD44 expression on the independent validation series confirmed CD44's predictive potential. In 8 larynx cancer cell lines, CD44 gene expression did not correlate with intrinsic radiosensitivity although it did correlate significantly with plating efficiency, consistent with a relationship with stem cell content. CONCLUSIONS: CD44 was the only biological factor tested which significantly correlated with response to radiotherapy in early stage larynx cancer patients, both at the mRNA and protein levels. Further studies are needed to confirm this and to assess how general these findings are for other head and neck tumor stages and sites.


Subject(s)
Carcinoma/diagnosis , Carcinoma/radiotherapy , Hyaluronan Receptors/genetics , Laryngeal Neoplasms/diagnosis , Laryngeal Neoplasms/radiotherapy , Neoplasm Recurrence, Local/diagnosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma/genetics , Carcinoma/pathology , Case-Control Studies , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Male , Microarray Analysis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Prognosis , Tumor Cells, Cultured , Validation Studies as Topic
9.
Semin Radiat Oncol ; 20(4): 241-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20832016

ABSTRACT

Cellular DNA repair determines survival after ionizing radiation. Human tumors commonly exhibit aberrant DNA repair since they drive mutagenesis and chromosomal instability. Recent reports have shown alterations in the base excision repair (BER) and single strand break repair (SSBR) pathways in human tumors. Here we review these reports with respect to radiation sensitivity and the attempts to target such tumor-specific BER/SSBR aberrations. These aberrations can alter cellular resistance to therapeutic agents, including radiation. Some strategies therefore aim to counteract the radioresistance mediated by such aberrant DNA repair. Other strategies aim to exploit the dependence of the tumor, but not the normal cells, on backup repair mechanisms after radiation, therefore increasing the therapeutic window. Such tumor-targeted radiosensitization holds promise for increasing the efficacy of radiotherapy.


Subject(s)
DNA Breaks, Single-Stranded/radiation effects , DNA Repair/radiation effects , Neoplasms/radiotherapy , DNA Breaks, Double-Stranded/radiation effects , DNA Damage , DNA Replication , DNA, Neoplasm/radiation effects , Dose-Response Relationship, Radiation , Humans , Radiation Oncology/methods , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology
10.
Radiother Oncol ; 95(3): 365-70, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20346528

ABSTRACT

PURPOSE: The purpose of this study was to combine gene expression profiles and clinical factors to provide a better prediction model of local control after chemoradiotherapy for advanced head and neck cancer. MATERIAL AND METHODS: Gene expression data were available for a series of 92 advanced stage head and neck cancer patients treated with primary chemoradiotherapy. The effect of the Chung high-risk and Slebos HPV expression profiles on local control was analyzed in a model with age at diagnosis, gender, tumor site, tumor volume, T-stage and N-stage and HPV profile status. RESULTS: Among 75 patients included in the study, the only factors significantly predicting local control were tumor site (oral cavity vs. Pharynx, hazard ratio 4.2 [95% CI 1.4-12.5]), Chung gene expression status (high vs. Low risk profile, hazard ratio 4.4 [95% CI 1.5-13.3]) and HPV profile (negative vs. Positive profile, hazard ratio 6.2 [95% CI 1.7-22.5]). CONCLUSIONS: Chung high-risk expression profile and a negative HPV expression profile were significantly associated with increased risk of local recurrence after chemoradiotherapy in advanced pharynx and oral cavity tumors, independent of clinical factors.


Subject(s)
Gene Expression Profiling , Head and Neck Neoplasms/therapy , Head and Neck Neoplasms/virology , Papillomaviridae/isolation & purification , Adult , Aged , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/etiology , Papillomaviridae/genetics , Risk Factors
11.
Int J Radiat Biol ; 86(1): 71-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20070218

ABSTRACT

PURPOSE: To review the present state of the art of the major current research areas in radiobiology in the form of a conference report. BACKGROUND: To celebrate their 50th anniversary, the Dutch Radiobiology Society recently held a meeting entitled "50 Years of Radiation Science in The Netherlands: From Molecular Research to Medical Application". Speakers were attracted from the USA and various European countries, covering topics including hypoxia, genomics and proteomics, DNA repair, DNA damage and signalling, chromosomal instability, stem cells, and normal tissue responses. Given the occasion, a history of Dutch radiobiology was also presented. CONCLUSION: Understanding the molecular pathways influencing the radiation response of cells, tumours and normal tissues has progressed dramatically over the last decades. Papers presented at this meeting showed that this understanding is leading to new and more effective ways to treat cancer with radiation.


Subject(s)
Radiobiology , Animals , Cell Hypoxia/physiology , Chromosomal Instability , DNA Damage , DNA Repair , Genomics , Humans , Proteomics , Stem Cell Transplantation
12.
Int J Radiat Biol ; 85(10): 825-36, 2009.
Article in English | MEDLINE | ID: mdl-19863199

ABSTRACT

PURPOSE: To review the many changes which have occurred in the past decades in the field of predicting outcome after radiotherapy from biological characteristics of the tumour or normal tissue. This review will also describe the present state of the art and emerging trends for the future. CONCLUSIONS: From using explanted cells, glass electrodes, exogenous proliferation and hypoxia tracers, and others, there has been a move towards monitoring expression and mutation of genes. Initially this was possible for just one or a few genes, but methods are now available which allow genome-wide monitoring at either the DNA or RNA level. The potential advantage of this evolution is not only to predict but also to understand potential causes of failure, allowing more rational and effective interventions. Comparative genomic hybridisation, mRNA expression profiling, microRNA profiling and promoter methylation profiling have all shown promise in finding signatures correlating with outcome, including after treatment involving radiotherapy. Expected trends for the future are: more signatures relevant to radiotherapy will be discovered; signatures will be refined and reduced to their essentials, such that genome-wide screening will give way to tailored signatures, quantifiable by routine non-array technology; more focus will be on assays predicting which pathway-specific radiosensitising drugs will be effective (exploiting tumour weaknesses); more signatures will be subjected to validation in randomised trials; and proteomics, DNA sequencing and imaging methods will play progressively increasing roles.


Subject(s)
Neoplasms/radiotherapy , Radiotherapy/trends , Gene Expression Profiling , Genome-Wide Association Study , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Prognosis , Radiation-Sensitizing Agents/therapeutic use , Treatment Outcome
13.
Int J Radiat Oncol Biol Phys ; 75(2): 489-96, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19735873

ABSTRACT

PURPOSE: Development of a radiosensitivity predictive assay is a central goal of radiation oncology. We reasoned a gene expression model could be developed to predict intrinsic radiosensitivity and treatment response in patients. METHODS AND MATERIALS: Radiosensitivity (determined by survival fraction at 2 Gy) was modeled as a function of gene expression, tissue of origin, ras status (mut/wt), and p53 status (mut/wt) in 48 human cancer cell lines. Ten genes were identified and used to build a rank-based linear regression algorithm to predict an intrinsic radiosensitivity index (RSI, high index = radioresistance). This model was applied to three independent cohorts treated with concurrent chemoradiation: head-and-neck cancer (HNC, n = 92); rectal cancer (n = 14); and esophageal cancer (n = 12). RESULTS: Predicted RSI was significantly different in responders (R) vs. nonresponders (NR) in the rectal (RSI R vs. NR 0.32 vs. 0.46, p = 0.03), esophageal (RSI R vs. NR 0.37 vs. 0.50, p = 0.05) and combined rectal/esophageal (RSI R vs. NR 0.34 vs. 0.48, p = 0.001511) cohorts. Using a threshold RSI of 0.46, the model has a sensitivity of 80%, specificity of 82%, and positive predictive value of 86%. Finally, we evaluated the model as a prognostic marker in HNC. There was an improved 2-year locoregional control (LRC) in the predicted radiosensitive group (2-year LRC 86% vs. 61%, p = 0.05). CONCLUSIONS: We validate a robust multigene expression model of intrinsic tumor radiosensitivity in three independent cohorts totaling 118 patients. To our knowledge, this is the first time that a systems biology-based radiosensitivity model is validated in multiple independent clinical datasets.


Subject(s)
Esophageal Neoplasms , Gene Expression/genetics , Head and Neck Neoplasms , Models, Genetic , Radiation Tolerance/genetics , Rectal Neoplasms , Aged , Aged, 80 and over , Algorithms , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/radiotherapy , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Humans , Linear Models , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Prognosis , Prospective Studies , Rectal Neoplasms/drug therapy , Rectal Neoplasms/genetics , Rectal Neoplasms/radiotherapy , Topotecan/administration & dosage
14.
Int J Cancer ; 124(11): 2643-50, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19253368

ABSTRACT

Not all patients with squamous cell carcinomas of the head and neck (HNSCC) benefit from concurrent cisplatin-based chemoradiation, but reliable predictive markers for outcome after chemoradiation are scarce. We have investigated potential prognostic biomarkers for outcome in a large group of patients. Ninety-one tumor biopsies taken from consecutive HNSCC patients were evaluated for protein expression on a tissue microarray. Using immunohistochemistry, 18 biomarkers, involved in various cellular pathways were investigated. Univariable and multivariable proportional hazard analyses were performed to investigate associations between each individual marker and outcome. In addition, the global test was used to test all variables simultaneously and selected combinations of markers for an overall association with local control. Univariable analysis showed statistically significant increased relative risks of RB, P16 and MRP2 for local control and MDR1 and HIF-1alpha for overall survival. MRP2, MDR1 and P16 levels were positively associated with outcome whereas RB and HIF-1alpha had a negative relationship. Using Goeman's global testing no combination of markers was identified that was associated with local control. Grouping the markers according to their function revealed an association between a combination of 3 markers (P16, P21 and P27) and outcome (p = 0.05) was found. In the multivariable analysis, MRP2 and RB remained significant independent predictive markers for local control. This study describes the prognostic value of biomarkers for the outcome in patients uniformly treated with concurrent chemoradiation. MRP2 and RB were found to be associated with outcome in patients treated with concurrent chemoradiation.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/therapy , Cisplatin/therapeutic use , Head and Neck Neoplasms/therapy , Adult , Aged , Carcinoma, Squamous Cell/mortality , Combined Modality Therapy , Female , Head and Neck Neoplasms/mortality , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Immunohistochemistry , Male , Middle Aged , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/analysis , Multivariate Analysis , Prognosis , Proto-Oncogene Proteins c-bcl-2/analysis , Retinoblastoma Protein/analysis , Treatment Outcome
15.
Laryngoscope ; 119(2): 316-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19160427

ABSTRACT

OBJECTIVES/HYPOTHESIS: To find biomarkers associated with response to radiotherapy in laryngeal cancer that can be used together with clinical parameters to improve outcome prediction. METHODS: In this study, 26 patients irradiated for laryngeal carcinomas with a local recurrence within two years (cases) and 33 patients without recurrence (controls) were included. All pretreatment biopsies were arrayed onto a tissue array. Immunohistochemistry was performed for 13 biomarkers that were selected from the literature as potential predictors for radioresponse in head and neck (HN) cancer: Bcl-2, Bcl-xL, p16, p21, p27, p53, cyclin D1, HIF-1alpha, CA9, COX-2, EGFR, ki-67, and pRB. RESULTS: Univariate logistic regression models showed borderline statistically significant increased relative risks, with positivity for CA9, COX-2, and p53. Goeman's global testing revealed an overall association between outcome and the 13 markers together with clinical variables. The most important markers were CA9 and COX-2. CONCLUSIONS: In laryngeal carcinoma, hypoxia and COX-2 overexpression provide a stronger contribution to an increased risk of local recurrence after radiotherapy compared with the well-known candidate markers p53, Bcl-2, and cyclin D1. However, no robust expression profile for the prediction of radioresistance was found.


Subject(s)
Biomarkers, Tumor/analysis , Laryngeal Neoplasms/radiotherapy , Aged , Antigens, Neoplasm/analysis , Carbonic Anhydrase IX , Carbonic Anhydrases/analysis , Case-Control Studies , Cyclooxygenase 2/analysis , Female , Humans , Immunoenzyme Techniques , Logistic Models , Male , Neoplasm Recurrence, Local , Predictive Value of Tests
16.
DNA Repair (Amst) ; 8(3): 336-46, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19059500

ABSTRACT

Several types of DNA lesion are induced after ionizing irradiation (IR) of which double strand breaks (DSBs) are expected to be the most lethal, although single strand breaks (SSBs) and DNA base damages are quantitatively in the majority. Proteins of the base excision repair (BER) pathway repair these numerous lesions. DNA polymerase beta has been identified as a crucial enzyme in BER and SSB repair (SSBR). We showed previously that inhibition of BER/SSBR by expressing a dominant negative DNA polymerase beta (polbetaDN) resulted in radiosensitization. We hypothesized increased kill to result from DSBs arising from unrepaired SSBs and BER intermediates. We find here higher numbers of IR-induced chromosome aberrations in polbetaDN expressing cells, confirming increased DSB formation. These aberrations did not result from changes in DSB induction or repair of the majority of lesions. SSB conversion to DSBs has been shown to occur during replication. We observed an increased induction of chromatid aberrations in polbetaDN expressing cells after IR, suggesting such a replication-dependence of secondary DSB formation. We also observed a pronounced increase of chromosomal deletions, the most likely cause of the increased kill. After H(2)O(2) treatment, polbetaDN expression only resulted in increased chromatid (not chromosome) aberrations. Together with the lack of sensitization to H(2)O(2), these data further suggest that the additional secondarily induced lethal DSBs resulted from repair attempts at complex clustered damage sites, unique to IR. Surprisingly, the polbetaDN induced increase in residual gammaH2AX foci number was unexpectedly low compared with the radiosensitization or induction of aberrations. Our data thus demonstrate the formation of secondary DSBs that are reflected by increased kill but not by residual gammaH2AX foci, indicating an escape from gammaH2AX-mediated DSB repair. In addition, we show that in the polbetaDN expressing cells secondary DSBs arise in a radiation-specific and partly replication-dependent manner.


Subject(s)
Cell Death/radiation effects , DNA Polymerase beta/physiology , Radiation, Ionizing , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/radiation effects , Chromatids/radiation effects , DNA/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Breaks, Single-Stranded/radiation effects , DNA Repair/radiation effects , DNA Replication , Dose-Response Relationship, Radiation , Histones , Humans , Oxidative Stress/genetics
18.
Radiother Oncol ; 86(3): 391-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18237797

ABSTRACT

PURPOSE: The purpose of the present study was to determine the role of DNA polymerase beta in repair and response after ionizing radiation in different phases of the cell cycle. METHODS AND MATERIALS: Synchronized cells deficient and proficient in DNA polymerase beta were irradiated in different phases of the cell cycle as determined by BrdU/flow cytometry. Cell kill and DNA repair were assessed by colony formation and alkaline comet assays, respectively. RESULTS: We first demonstrated delayed repair of ionizing radiation induced DNA damage in confluent polymerase beta deficient cells. Cell synchronization experiments revealed a cell cycle phase dependence by demonstrating radiation hypersensitivity of polymerase beta-deficient cells in G1, but not in the S-phase. Complementing polymerase beta-deficient cells with polymerase beta reverted the hypersensitivity in G1. Ionizing radiation damage repair was found to be delayed in beta-deficient cells when irradiated in G1, but not in S. CONCLUSIONS: The data show a differential role of DNA polymerase beta driven base excision and single strand break repair throughout the cell cycle after ionizing radiation damage.


Subject(s)
Cell Cycle/radiation effects , DNA Breaks, Single-Stranded/radiation effects , DNA Polymerase beta/metabolism , DNA Repair/radiation effects , Radiation, Ionizing , Animals , Cell Line , Mice , Radiation Injuries, Experimental , Radiation Tolerance
19.
Gynecol Oncol ; 108(2): 317-25, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18037478

ABSTRACT

PURPOSE: To determine whether maintaining HGB levels > or = 12.0 g/dL with recombinant human erythropoietin (R-HUEPO) compared to "standard" treatment (transfusion for HGB < or = 10.0 g/dL) improves progression-free survival (PFS), overall survival (OS) and local control (LC) in women receiving concurrent weekly cisplatin and radiation (CT/RT) for carcinoma of the cervix. In addition, to determine whether platinum-DNA adducts were associated with clinical characteristics or outcome. METHODS: Patients with stage IIB-IVA cervical cancer and HGB < 14.0 g/dL were randomly assigned to CT/RT+/-R-HUEPO (40,000 units s.c. weekly). R-HUEPO was stopped if HGB > 14.0 g/dL. Endpoints were PFS, OS and LC. Platinum-DNA adducts were quantified using immunocytochemistry assay in buccal cells. RESULTS: Between 08/01 and 09/03, 109 of 114 patients accrued were eligible. Fifty-two received CT/RT and 57 CT/RT+R-HUEPO. The study closed prematurely, with less than 25% of the planned accrual, due to potential concerns for thromboembolic event (TE) with R-HUEPO. Median follow-up was 37 months (range 9.8-50.4 months). PFS and OS at 3 years should be 65% and 75% for CT/RT and 58% and 61% for CT/RT+R-HUEPO, respectively. TE occurred in 4/52 receiving CT/RT and 11/57 with CT/RT+R-HUEPO, not all considered treatment related. No deaths occurred from TE. High-platinum adducts were associated with inferior PFS and LC. CONCLUSION: TE is common in cervical cancer patients receiving CT/RT. Difference in TE rate between the two treatments was not statistically significant. The impact of maintaining HGB level > 12.0 g/dL on PFS, OS and LC remains undetermined.


Subject(s)
Anemia/drug therapy , Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Erythropoietin/therapeutic use , Hemoglobins/metabolism , Uterine Cervical Neoplasms/blood , Uterine Cervical Neoplasms/therapy , Adult , Aged , Anemia/blood , Combined Modality Therapy , DNA Adducts , Erythropoietin/adverse effects , Female , Humans , Middle Aged , Neoplasm Staging , Recombinant Proteins , Thromboembolism/chemically induced , Uterine Cervical Neoplasms/pathology
20.
Radiat Res ; 168(6): 683-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18088185

ABSTRACT

Ionizing radiation induces a diverse spectrum of DNA lesions, including strand breaks and oxidized bases. In mammalian cells, ionizing radiation-induced lesions are targets of non-homologous end joining, homologous recombination, and base excision repair. In vitro assays show a potential involvement of DNA polymerase lambda in non-homologous end joining and base excision repair. In this study, we investigated whether DNA polymerase lambda played a significant role in determining ionizing radiation sensitivity. Despite increased sensitivity to hydrogen peroxide, lambda-deficient mouse embryonic fibroblasts displayed equal survival after exposure to ionizing radiation compared to their wild-type counterparts. In addition, we found increased sensitivity to the topoisomerase inhibitors camptothecin and etoposide in the absence of polymerase lambda. These results do not reveal a major role for DNA polymerase lambda in determining radiosensitivity in vivo.


Subject(s)
DNA Polymerase beta/deficiency , DNA Polymerase beta/metabolism , Radiation Tolerance , Animals , Camptothecin/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/drug effects , DNA Polymerase beta/genetics , Etoposide/pharmacology , Genome/genetics , Genotype , Hydrogen Peroxide/pharmacology , Mice , Radiation, Ionizing
SELECTION OF CITATIONS
SEARCH DETAIL
...