Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Eur Heart J Cardiovasc Imaging ; 17(11): 1230-1238, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27550664

ABSTRACT

Magnetic resonance imaging (MRI) is increasingly being used in cardiology to detect heart disease and guide therapy. It is mooted to be a safer alternative to imaging techniques, such as computed tomography (CT) or coronary angiographic imaging. However, there has recently been an increased interest in the potential long-term health risks of MRI, especially in the light of the controversy resulting from a small number of research studies reporting an increase in DNA damage following exposure, with calls to limit its use and avoid unnecessary examination, according to the precautionary principle. Overall the published data are somewhat limited and inconsistent; the ability of MRI to produce DNA lesions has yet to be robustly demonstrated and future experiments should be carefully designed to optimize sensitivity and benchmarked to validate and assess reproducibility. The majority of the current studies have focussed on the initial induction of DNA damage, and this has led to comparisons between the reported induction of γH2AX and implied double-strand break (DSB) yields produced following MRI with induction by imaging techniques using ionizing radiation. However, γH2AX is not only a marker of classical double-ended DSB, but also a marker of stalled replication forks and in certain circumstances stalled DNA transcription. Additionally, ionizing radiation is efficient at producing complex DNA damage, unique to ionizing radiation, with an associated reduction in repairability. Even if the fields associated with MRI are capable of producing DNA damage, the lesions produced will in general be simple, similar to those produced by endogenous processes. It is therefore inappropriate to try and infer cancer risk by simply comparing the yields of γH2AX foci or DNA lesions potentially produced by MRI to those produced by a given exposure of ionizing radiation, which will generally be more biologically effective and have a greater probability of leading to long-term health effects. As a result, it is important to concentrate on more relevant downstream end points (e.g. chromosome aberration production), along with potential mechanisms by which MRI may lead to DNA lesions. This could potentially involve a perturbation in homeostasis of oxidative stress, modifying the background rate of endogenous DNA damage induction. In summary, what the field needs at the moment is more research and less fear mongering.


Subject(s)
DNA Damage/radiation effects , Magnetic Resonance Imaging, Cine/adverse effects , Radiation Tolerance/genetics , Radiation, Ionizing , Cardiovascular Diseases/diagnostic imaging , Evaluation Studies as Topic , Female , Humans , Magnetic Resonance Imaging, Cine/methods , Male , Needs Assessment , Qualitative Research , Radiation Dosage , Risk Assessment
2.
Br J Cancer ; 107(2): 291-9, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22713662

ABSTRACT

BACKGROUND: Most solid tumours contain regions of sub-optimal oxygen concentration (hypoxia). Hypoxic cancer cells are more resistant to radiotherapy and represent the most aggressive fraction of a tumour. It is therefore essential that strategies continue to be developed to target hypoxic cancer cells. Inhibition of the DNA damage response (DDR) might be an effective way of sensitising hypoxic tumour cells to radiotherapy. METHODS: Here, we describe the cellular effects of pharmacological inhibition of the apical DDR kinase ATR (Ataxia Telangiectasia and Rad 3 related) with a highly selective inhibitor, VE-821, in hypoxic conditions and its potential as a radiosensitiser. RESULTS: VE-821 was shown to inhibit ATR-mediated signalling in response to replication arrest induced by severe hypoxia. In these same conditions, VE-821 induced DNA damage and consequently increased Ataxia Telangiectasia Mutated-mediated phosphorylation of H2AX and KAP1. Consistently, ATR inhibition sensitised tumour cell lines to a range of oxygen tensions. Most importantly, VE-821 increased radiation-induced loss of viability in hypoxic conditions. Using this inhibitor we have also demonstrated for the first time a link between ATR and the key regulator of the hypoxic response, HIF-1. HIF-1 stabilisation and transcriptional activity were both decreased in response to ATR inhibition. CONCLUSION: These findings suggest that ATR inhibition represents a novel strategy to target tumour cells in conditions relevant to pathophysiology and enhance the efficacy of radiotherapy.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Hypoxia/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Radiation Tolerance/drug effects , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage , DNA Replication/drug effects , HCT116 Cells , HeLa Cells , Histones/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyrazines/pharmacology , Radiation Tolerance/genetics , Radiotherapy/methods , Repressor Proteins/genetics , Signal Transduction/drug effects , Sulfones/pharmacology , Tripartite Motif-Containing Protein 28
5.
Oncogene ; 29(3): 463-8, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19838217

ABSTRACT

A significant proportion of human cancers overexpress DNA polymerase beta (Pol beta), the major DNA polymerase involved in base excision repair. The underlying mechanism and biological consequences of overexpression of this protein are unknown. We examined whether Pol beta, expressed at levels found in tumor cells, is involved in the repair of DNA damage induced by oxaliplatin treatment and whether the expression status of this protein alters the sensitivity of cells to oxaliplatin. DNA damage induced by oxaliplatin treatment of HCT116 and HT29 colon cancer cells was observed to be associated with the stabilization of Pol beta protein on chromatin. In comparison with HCT116 colon cancer cells, isogenic oxaliplatin-resistant (HCT-OR) cells were found to have higher constitutive levels of Pol beta protein, faster in vitro repair of a DNA substrate containing a single nucleotide gap and faster repair of 1,2-GG oxaliplatin adduct levels in cells. In HCT-OR cells, small interfering RNA knockdown of Pol beta delayed the repair of oxaliplatin-induced DNA damage. In a different model system, Pol beta-deficient fibroblasts were less able to repair 1,2-GG oxaliplatin adducts and were hypersensitive to oxaliplatin treatment compared with isogenic Pol beta-expressing cells. Consistent with previous studies, Pol beta-deficient mouse fibroblasts were not hypersensitive to cisplatin treatment. These data provide the first link between oxaliplatin sensitivity and DNA repair involving Pol beta. They demonstrate that Pol beta modulates the sensitivity of cells to oxaliplatin treatment.


Subject(s)
DNA Polymerase beta/metabolism , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage , DNA Polymerase beta/deficiency , DNA Polymerase beta/genetics , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Knockout , Oxaliplatin , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
6.
Oncogene ; 26(48): 6851-62, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17471232

ABSTRACT

Caveolin-1 (Cav-1) is an integral transmembrane protein and a critical component in interactions of integrin receptors with cytoskeleton-associated and signaling molecules. Since integrin-mediated cell adhesion generates signals conferring radiation resistance, we examined the effects of small interfering RNA-mediated knockdown of Cav-1 alone or in combination with beta1-integrin or focal adhesion kinase (FAK) on radiation survival and proliferation of pancreatic carcinoma cell lines. Irradiation induced Cav-1 expression in PATU8902, MiaPaCa2 and Panc1 cell lines. The cell lines showed significant radiosensitization after knockdown of Cav-1, beta1-integrin or FAK and cholesterol depletion by beta-cyclodextrin relative to nonspecific controls. Under knockdown conditions, proliferation of non-irradiated and irradiated cells was significantly attenuated relative to controls. These findings correlated with changes in expression or phosphorylation of Akt, glycogen synthase kinase 3beta, Paxillin, Src, c-Jun N-terminal kinase and mitogen-activated protein kinase. Analysis of DNA microarray data revealed a Cav-1 overexpression in a subset of pancreatic ductal adenocarcinoma samples. The data presented show, for the first time, that disruption of interactions of Cav-1 with beta1-integrin or FAK affects radiation survival and proliferation of pancreatic carcinoma cells and suggest that Cav-1 is critical to these processes. These results indicate that strategies targeting Cav-1 may be useful as an approach to improve conventional therapies, including radiotherapy, for pancreatic cancer.


Subject(s)
Caveolin 1/metabolism , Pancreatic Neoplasms/radiotherapy , Radiation Tolerance/physiology , Blotting, Western , Cell Adhesion , Cell Cycle/physiology , Cell Cycle/radiation effects , Cell Proliferation/radiation effects , Colony-Forming Units Assay , Fluorescent Antibody Technique , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Profiling , Humans , Integrin beta1/chemistry , Integrin beta1/genetics , Integrin beta1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Pancreatic Neoplasms/metabolism , Paxillin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction , X-Rays
7.
Int J Radiat Biol ; 79(7): 569-76, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14530152

ABSTRACT

PURPOSE: The inhibition of activated Ras combined with radiotherapy was identified as a potential method for radiosensitization. MATERIALS AND METHODS: Immunoblotting was used to control for prenylation inhibition of the respective Ras isoforms and for changes in activity of downstream proteins. Clonogenic assays with human and rodent tumour cell lines and transfected cell lines served for the testing of radiosensitivity. Xenograft tumours were treated with farnesyl transferase inhibitors and radiation and assayed for ex vivo plating efficiency, regrowth of tumours and EF5 staining for detection of hypoxia. Concurrent treatment with L-778,123 and radiotherapy was performed in non-small cell lung cancer (NSCLC) and head and neck cancer (HNC) patients. RESULTS: Blocking the prenylation of Ras proteins in cell lines with Ras activated by mutations or receptor signalling resulted in radiation sensitization in in vitro and in vivo. The PI3 kinase downstream pathway was identified as a contributor to Ras-mediated radiation resistance. Additionally, increased oxygenation of xenograft tumours was observed after FTI treatment. Combined treatment in a phase I study was safe and effective in NSCLC and HNC. CONCLUSIONS: Tumour cells with activated Ras were sensitized to radiation. Unravelling the underlying mechanisms promises to lead to even more specific drugs with higher potency and safety.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Farnesyltranstransferase , Genes, ras/physiology , Humans , Imidazoles/pharmacology , Protein Processing, Post-Translational , Radiation Tolerance
8.
Semin Oncol ; 28(5 Suppl 16): 86-93, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11706400

ABSTRACT

The targeting of molecular abnormalities in neoplasms may provide an opportunity to improve the selectivity of cancer therapy. Ras mutations are a common genetic event in human cancers. Other genetic changes in tumors can signal through ras-dependent pathways as well. The targeting of ras through the inhibition of Ras protein farnesylation is one new cancer treatment strategy under clinical evaluation. Several farnesyltransferase inhibitors (FTIs) have been evaluated in phase I trials. The toxicity and maximally tolerated doses of several FTIs have been determined, and clinical trials are underway to evaluate FTIs in combination with conventional cytotoxic chemotherapy agents. Also underway are attempts to develop assays to measure the biological effects of the FTI in patients. Inhibition of farnesylation of a number of surrogate markers are currently being investigated. These efforts may provide insight into the mechanism of action of these compounds and lead to improved patient selection for clinical trials.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , ras Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , Clinical Trials as Topic , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Quinolones/pharmacology , Quinolones/therapeutic use
9.
Cancer Chemother Pharmacol ; 48(3): 177-87, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11592338

ABSTRACT

OBJECTIVES: Pharmacokinetic studies were performed on the first 28 patients enrolled in a phase I trial to determine the ability of EF5 [2-(2-nitro-1-H-imidazolI-yl)-N-(2,2,3,3,3-pentafluoropropyl) acetamide] to detect hypoxia in human tumors in the absence of patient toxicity. METHODS: EF5 was made in purified form and formulated for intravenous injection by the National Cancer Institute. After obtaining consent from the patients, EF5 was administered and blood samples were drawn at various times over approximately 48 h. For most patients it was possible to collect total urine at approximately 8-h intervals. EF5 in plasma and urine was analyzed by high-performance liquid chromatography. RESULTS: EF5's plasma concentration followed a simple exponential decay following infusion. The plasma half-life was 11.7 +/- 2.6 h (+/- SD) and was not affected by drug dose (9 to 28 mg/kg), fractional urine recovery, patient weight or gender. Absolute plasma values suggested even biodistribution of the drug throughout the soft tissue with a volume of distribution equal to 0.56 l/ kg. Despite the relatively high lipid partition coefficient (logP = 0.6), EF5 was excreted primarily (up to 70%) via kidney clearance. No drug metabolites (e.g. retaining the 2-nitroimidazole chromophore) were detected in either plasma or urine. No toxicity was found at drug doses adequate to detect tumor hypoxia. CONCLUSIONS: Currently held paradigms of 2-nitroimidazole metabolism (e.g. clearance rate and toxicity as affected by octanol/ water partition coefficient) are discussed. The results reported herein suggest that EF5 is biologically stable with predictable pharmacokinetics. EF5's consistent half-life and clearance properties will allow quantitative analysis of EF5 binding relative to tissue oxygen levels.


Subject(s)
Etanidazole/pharmacokinetics , Hydrocarbons, Fluorinated/pharmacokinetics , Hypoxia/diagnosis , Neoplasms/metabolism , Radiation-Sensitizing Agents/pharmacokinetics , Area Under Curve , Body Weight , Chromatography, High Pressure Liquid , Etanidazole/analogs & derivatives , Half-Life , Humans , Maximum Tolerated Dose , Metabolic Clearance Rate , Predictive Value of Tests , Sex Factors , Tissue Distribution
10.
Semin Radiat Oncol ; 11(4): 328-37, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11677657

ABSTRACT

Local recurrence after a definitive course of radiation therapy remains a significant clinical problem and represents a common pattern of failure for many solid tumors. The sensitivity of tumor cells to the cytotoxicity of ionizing radiation is thought to be one of the major determinants of local control for tumors in patients treated with radiation therapy. There is substantial experimental evidence to demonstrate that increased radiation resistance is associated with the expression of activated oncogenes, including Ras. Mutated forms of Ras are found in 30% of human cancers including a substantial proportion of pancreatic and colon adenocarcinomas. Mutated Ras produces proteins that remain locked in a constitutively active state, thereby relaying uncontrolled signals. Ras proteins are guanosine triphosphate-binding proteins that play a pivotal role in the control of many cellular processes, including growth and differentiation. Preclinical studies have shown that expression of mutant Ras increases cellular radioresistance. Ras function is dependent on its localization to the plasma membrane. This is achieved by posttranslational modifications, including the addition of a farnesyl isoprenoid moiety in a reaction catalyzed by the enzyme protein farnesyltransferase (FTase). This enzyme has become an important target for the design of new agents that target Ras. FTase inhibitors (FTIs) block the farnesylation of Ras and reverse Ras-mediated radioresistance in human cell lines. FTIs have been well tolerated in animal studies and appear not to cause generalized cytotoxicity. There are ongoing clinical trials to determine the optimal therapeutic schedules and dose for FTIs. A phase 1 trial of the FTI L778-123 and radiotherapy has recently been completed.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Neoplasm Recurrence, Local/radiotherapy , Neoplasms/genetics , Neoplasms/radiotherapy , ras Proteins/physiology , Alkyl and Aryl Transferases/metabolism , Humans , Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/physiology , Radiation Tolerance/genetics , Signal Transduction , ras Proteins/metabolism
11.
Oncogene ; 20(27): 3486-96, 2001 Jun 14.
Article in English | MEDLINE | ID: mdl-11429695

ABSTRACT

All eukaryotic cells manifest cell cycle delay after exposure to DNA damaging agents. It has been proposed that such cell cycle checkpoints may allow DNA repair but direct evidence of such activity during the radiation-induced G2 delay has been lacking. We report here that cells arrested in G2 by radiation (2-3 Gy) and etoposide incorporate bromodeoxyuridine (BrdU) at discrete foci in the nucleus. We detected G2 cells with CENP-F, a nuclear protein maximally expressed in G2. Caffeine and okadaic acid, both established radiosensitizers, inhibit the incorporation of BrdU in G2 cells. Radioresistant HT29 and OVCAR cells demonstrate BrdU foci formation more frequently during the G2 delay when compared to the more radiosensitive A2780 cell line. The repair foci formed during G2 may be followed through mitosis and observed in daughter cells in G1. Taken together, these observations are consistent with the detection of DNA repair activity during the radiation-induced G2 delay after relatively low doses of radiation.


Subject(s)
Cell Cycle/genetics , DNA Damage , DNA Repair , DNA, Neoplasm/genetics , Gamma Rays , Bromodeoxyuridine , Cell Cycle/drug effects , Cell Cycle/radiation effects , Centromere/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , Etoposide/toxicity , Female , Flow Cytometry , G2 Phase , HeLa Cells , Humans , Kinetics , Microfilament Proteins , Ovarian Neoplasms , Radiation Tolerance , Tumor Cells, Cultured
12.
Prostate ; 48(1): 16-28, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11391683

ABSTRACT

BACKGROUND: The Bowman-Birk inhibitor is a soybean-derived protease inhibitor that has anti-inflammatory and anticarcinogenic activities. METHODS: A Phase I trial of Bowman-Birk inhibitor concentrate (BBIC) in 19 male subjects with benign prostatic hyperplasia (BPH) and lower urinary tract symptoms (LUTS) has been performed. RESULTS: The results of the trial indicated that there was no dose-limiting toxicity of BBIC. There was a statistically significant decrease in serum PSA levels in all BBIC-treated patients. Some BBIC-treated patients exhibited a relatively large reduction in serum PSA levels, ranging up to a 43% reduction. There was also a statistically significant decrease in serum triglyceride levels and a decrease in prostate volume in the treated patients. The scores recorded in response to a urinary symptom questionnaire indicated improved urinary activities in the BBIC-treated patients; however, the control subjects exhibited similar improvements in urinary activities during the course of the trial. CONCLUSIONS: The data obtained in this trial, particularly the data suggesting that BBIC treatment may lead to reduced serum PSA levels and reduced prostate volumes, suggest that a Phase II clinical trial of BBIC for the therapy of BPH is warranted.


Subject(s)
Prostatic Hyperplasia , Prostatic Hyperplasia/drug therapy , Trypsin Inhibitor, Bowman-Birk Soybean/therapeutic use , Aged , Aged, 80 and over , Biopsy, Needle , Cholesterol/blood , Dose-Response Relationship, Drug , Humans , Male , Middle Aged , Prostate/drug effects , Prostate/pathology , Prostate-Specific Antigen/blood , Prostate-Specific Antigen/drug effects , Prostatic Hyperplasia/physiopathology , Quality of Life , Treatment Outcome , Triglycerides/blood , Trypsin Inhibitor, Bowman-Birk Soybean/administration & dosage , Trypsin Inhibitor, Bowman-Birk Soybean/urine , Urinary Retention/drug therapy
14.
Cancer Res ; 61(10): 4278-82, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358856

ABSTRACT

The critical pathways determining the resistance of tumor cells to ionizing radiation are poorly defined. Because the ras oncogene, a gene activated in many human cancers treated with radiotherapy, can induce increased radioresistance, we have asked which Ras effector pathways are significant in conferring a survival advantage to tumor cells. The phosphoinositide-3-kinase (PI3K) inhibitor LY294002 radiosensitized cells bearing mutant ras oncogenes, but the survival of cells with wild-type ras was not affected. Inhibition of the PI3K downstream target p70S6K by rapamycin, the Raf-MEK-MAPK pathway with PD98059, or the Ras-MEK kinase-p38 pathway with SB203580 had no effect on radiation survival in cells with oncogenic ras. Expression of active PI3K in cells with wild-type ras resulted in increased radiation resistance that could be inhibited by LY294002. These experiments have indicated the importance of PI3K in mediating enhanced radioresistance and have implicated PI3K as a potential target for specific radiosensitization of tumors.


Subject(s)
MAP Kinase Signaling System/physiology , Radiation Tolerance/physiology , ras Proteins/physiology , Animals , Chromones/pharmacology , Enzyme Induction , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Genes, ras , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Rats , Tumor Cells, Cultured , ras Proteins/antagonists & inhibitors , ras Proteins/genetics
15.
Neuro Oncol ; 3(1): 11-21, 2001 01.
Article in English | MEDLINE | ID: mdl-11305412

ABSTRACT

DNA damage produces delayed mitosis (G2/M delay) in proliferating cells, and shortening the delay sensitizes human malignant glioma and medulloblastoma cells to cytotoxic chemotherapy. Although activation of the cyclin-dependent kinase CDC2 mediates G2/M transition in all tumor cells studied to date, regulation of CDC2 varies between tumor types. Persistent hyperphosphorylation of kinase and reduced cyclin expression have been implicated as mediators of treatment-induced G2 delay in different tumor models. To evaluate regulation of G2/M transition in human brain tumors, we studied the expression and/or activity of CDC2 kinase and cyclins A and B1 in U-251 MG and DAOY medulloblastoma cells after their treatment with camptothecin (CPT). Synchronized cells were treated during S phase, then harvested at predetermined intervals for evaluation of cell cycle kinetics, kinase activity mRNA, and protein expression. CPT produced G2 delay associated with decreased CDC2 kinase activity and cyclin B1 expression. Kinase activity was associated with CDC2 bound to cyclin B1, not cyclin A, in both cell lines. Cyclin A mRNA and protein expression were reduced after CPT treatment; however, decreased protein expression was short lived and moderate in the glioma and primitive neuroectodermal tumor/medulloblastoma cells, respectively. We conclude that G2 delay is a common response of brain tumor cells to chemotherapy with topoisomerase I inhibitors and that a mechanism of this delay may be reduced expression of cyclin B1.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/genetics , Camptothecin/pharmacology , Cyclin B/biosynthesis , Enzyme Inhibitors/pharmacology , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Medulloblastoma/genetics , Neoplasm Proteins/biosynthesis , Topoisomerase I Inhibitors , Brain Neoplasms/pathology , CDC2 Protein Kinase/metabolism , Cyclin A/biosynthesis , Cyclin A/genetics , Cyclin B/genetics , Cyclin B1 , Glioma/pathology , Humans , Medulloblastoma/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured/drug effects
17.
Cancer Res ; 61(5): 2289-93, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280800

ABSTRACT

Many tumors contain extensive regions of hypoxia. Because hypoxic cells are markedly more resistant to killing by radiation, repeated attempts have been made to improve the oxygenation of tumors to enhance radiotherapy. We have studied the oxygenation of tumor xenografts in nude mice after treatment with the farnesyltransferase inhibitor L744,832. Hypoxia was assessed by measuring the binding of the hypoxic cell marker pentafluorinated 2-nitroimidazole. We show that xenografts from two tumor cell lines with mutations in H-ras had markedly improved oxygenation after farnesyltransferase treatment. In contrast, xenografts from two tumors without ras mutations had equivalent hypoxia regardless of treatment. The effect on tumor oxygenation could be detected at 3 days and remained after 7 days of treatment. These results indicate that treatment with farnesyltransferase inhibitors can alter the oxygenation of certain tumors and suggest that such treatment might be useful in the radiosensitization of these tumors.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Methionine/analogs & derivatives , Methionine/pharmacology , Neoplasms/metabolism , Oxygen/metabolism , ras Proteins/biosynthesis , Animals , Cell Hypoxia/drug effects , Farnesyltranstransferase , Gene Expression , Genes, ras/genetics , HT29 Cells/drug effects , HT29 Cells/enzymology , HT29 Cells/metabolism , Humans , Mice , Mice, Nude , Mutation , Neoplasms/drug therapy , Neoplasms/enzymology , Xenograft Model Antitumor Assays , ras Proteins/genetics
18.
Int J Radiat Oncol Biol Phys ; 49(2): 587-96, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11173159

ABSTRACT

PURPOSE: The presence of hypoxia, measured by needle electrodes, has been shown to be associated with poor patient outcome in several human tumor types, including soft tissue sarcomas. The present report emphasizes the evaluation of hypoxia in soft tissue sarcomas based upon the binding of the 2-nitroimidazole drug EF5 (2-[2-nitro-1H-imidazol-1-yl]-N-(2,2,3,3,3-pentafluoropropyl) acetamide). EF5 has previously been shown to be predictive of radiation response in animal tumors and in in vitro studies. We have also previously reported studies of EF5 binding in human squamous cell tumors. Using fluorescent immunohistochemical techniques, we provide data on the presence and distribution of EF5 binding, as a surrogate for hypoxia, in human spindle cell tumors. METHODS AND MATERIALS: Patients with spindle cell tumors who were scheduled for tumor surgery were asked to participate in the Phase I trial of EF5. Approximately 48 h preoperatively, EF5 was administered i.v. at doses between 9 and 21 mg/kg. Binding in frozen sections of biopsied tissues was determined using monoclonal antibodies labeled with the green-excited, orange-emitting fluorescent dye, Cy3. Calibration studies were performed in vitro by incubating fresh tumor tissue cubes obtained from each patient with EF3 (an analog of EF5) under hypoxic conditions ("reference binding"). The goal of these calibration studies was to quantify the maximal binding levels possible in individual patient's tissues. The relationship between binding (in situ based on EF5 binding) and reference binding (in vitro based on EF3 binding) was determined. RESULTS: Eight patients were studied; 3 of these patients had gastrointestinal stromal tumors (GIST). The incubation of tumor tissue cubes in EF3 under hypoxic conditions demonstrated that all tumors bound drug to a similar extent. Reference binding showed a 3.2-fold variation in median fluorescence (113-356) on an absolute fluorescence scale, calibrated by a Cy3 dye standard. In situ binding in the brightest tumor section varied by a factor of 25.4 between the lowest and highest binding tumor (7.5-190.2). Heterogeneity of highest binding was greater between tumors than within individual tumors. A correspondence between EF5 binding and Eppendorf needle electrode studies was seen in the 5 patients with non-GISTs. CONCLUSION: Inter- and intratumoral heterogeneity of EF5 binding in spindle cell tumors has been documented. Patterns of binding consistent with diffusion limited hypoxia are present in human spindle cell neoplasms.


Subject(s)
Cell Hypoxia , Etanidazole/analogs & derivatives , Etanidazole/metabolism , Gastrointestinal Neoplasms/metabolism , Hydrocarbons, Fluorinated/metabolism , Indicators and Reagents/metabolism , Radiation-Sensitizing Agents/metabolism , Sarcoma/metabolism , Adult , Aged , Extremities , Female , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/physiopathology , Humans , Immunohistochemistry , Male , Middle Aged , Sarcoma/pathology , Sarcoma/physiopathology
19.
Cancer Res ; 60(23): 6597-600, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11118040

ABSTRACT

Transformation with ras oncogenes results in increased radiation sur vival in many but not all cells. In addition, prenyltransferase inhibitors which inhibit ras proteins by blocking posttranslational modification radiosensitize cells with oncogenic ras. These findings suggest that oncogenic ras contributes to intrinsic radiation resistance. However, because introduction of ras oncogenes does not increase radiation survival in all cells and because prenyltransferase inhibitors target molecules other than ras, these studies left the conclusion that ras increases the intrinsic radi ation resistance of tumor cells in doubt. Here we show that genetic inactivation of K- or N-ras oncogenes in human tumor cells (DLD-1 and HT1080, respectively) leads to increased radiosensitivity. Reintroduction of the activated N-ras gene into the HT1080 line, having lost its mutant allele, resulted in increased radiation resistance. This study lends further support to the hypothesis that expression of activated ras can contribute to intrinsic radiation resistance in human tumor cells and extends this finding to the K- and N- members of the ras family. These findings support the development of strategies that target ras for inactivation in the treatment of cancer.


Subject(s)
Genes, ras/physiology , Radiation Tolerance/genetics , Tumor Cells, Cultured/radiation effects , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Alleles , Apoptosis/radiation effects , Cell Division/genetics , Cell Survival/radiation effects , Clone Cells , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Fibrosarcoma/radiotherapy , Gene Expression Regulation, Neoplastic , Humans , Tumor Cells, Cultured/physiology
20.
Int J Radiat Oncol Biol Phys ; 48(1): 89-94, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10924976

ABSTRACT

PURPOSE: To identify factors that may predict for severe radiation pneumonitis or pneumonopathy (RP), we reviewed a set of simple, commonly available characteristics. METHODS AND MATERIALS: Medical records of 148 lung cancer patients with good performance status (ECOG 0-1) treated definitively with chemoradiation from 6/92-6/98 at the University of Pennsylvania were reviewed. Actuarial survival and the crude rate of severe radiation pneumonitis were determined as a function of several variables. Potential predictive factors examined included age, gender, histology, stage, pulmonary function, performance status (0 vs. 1), weight loss, tumor location, radiation dose, initial radiation field size, chemotherapy regimen, and timing of chemotherapy. Univariate analysis (log-rank test) was performed for each variable. Multivariate analysis was performed using linear regression. RESULTS: Median survival for the entire cohort was 14.7 months. Four patients were inevaluable for pneumonitis due to early death from progressive disease. Of the remaining 144 evaluable patients, 12 (8.3%) experienced severe RP. The most significant factor predicting for severe RP was performance status (p < 0.003). The risk of severe RP was 16% for PS-1 patients vs. 2% for PS-0 patients. Women were significantly more likely to develop severe RP than men (p = 0.01). Among 67 patients for whom pre-radiation therapy pulmonary function data were available, forced expiratory volume of the lung in 1 second (FEV(1)) was also significant (p = 0. 03). No patient suffering severe RP had a pretreatment FEV(1) > 2.0 liters. The median radiation dose was 59.2 Gy and median initial radiation field size was 228 cm(2). Neither radiotherapy factor predicted for RP. Other factors studied, including chemotherapy drugs, and schedule, also were not significant predictors of severe RP. CONCLUSIONS: Pretreatment performance status, gender, and FEV(1) are significant predictors of severe radiation pneumonopathy, at least when using conventional radiation fields and doses. Complex radiation dose-volume algorithms that attempt to predict lung complication probabilities should probably incorporate these simply obtained clinical parameters.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Small Cell/radiotherapy , Lung Neoplasms/radiotherapy , Radiation Pneumonitis/etiology , Adult , Aged , Aged, 80 and over , Analysis of Variance , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Small Cell/drug therapy , Combined Modality Therapy , Female , Forced Expiratory Volume , Humans , Karnofsky Performance Status , Lung Neoplasms/drug therapy , Male , Middle Aged , Radiotherapy Dosage , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...