Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cancers (Basel) ; 15(9)2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37174026

ABSTRACT

Focal radiation necrosis of the brain (fRNB) is a late adverse event that can occur following the treatment of benign or malignant brain lesions with stereotactic radiation therapy (SRT) or stereotactic radiosurgery (SRS). Recent studies have shown that the incidence of fRNB is higher in cancer patients who received immune checkpoint inhibitors. The use of bevacizumab (BEV), a monoclonal antibody that targets the vascular endothelial growth factor (VEGF), is an effective treatment for fRNB when given at a dose of 5-7.5 mg/kg every two weeks. In this single-center retrospective case series, we investigated the effectiveness of a low-dose regimen of BEV (400 mg loading dose followed by 100 mg every 4 weeks) in patients diagnosed with fRNB. A total of 13 patients were included in the study; twelve of them experienced improvement in their existing clinical symptoms, and all patients had a decrease in the volume of edema on MRI scans. No clinically significant treatment-related adverse effects were observed. Our preliminary findings suggest that this fixed low-dose regimen of BEV can be a well-tolerated and cost-effective alternative treatment option for patients diagnosed with fRNB, and it is deserving of further investigation.

2.
Oncotarget ; 8(22): 35728-35742, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28415723

ABSTRACT

Auranofin (AF) is an anti-arthritic drug considered for combined chemotherapy due to its ability to impair the redox homeostasis in tumor cells. In this study, we asked whether AF may in addition radiosensitize tumor cells by targeting thioredoxin reductase (TrxR), a critical enzyme in the antioxidant defense system operating through the reductive protein thioredoxin. Our principal findings in murine 4T1 and EMT6 tumor cells are that AF at 3-10 µM is a potent radiosensitizer in vitro, and that at least two mechanisms are involved in TrxR-mediated radiosensitization. The first one is linked to an oxidative stress, as scavenging of reactive oxygen species (ROS) by N-acetyl cysteine counteracted radiosensitization. We also observed a decrease in mitochondrial oxygen consumption with spared oxygen acting as a radiosensitizer under hypoxic conditions. Overall, radiosensitization was accompanied by ROS overproduction, mitochondrial dysfunction, DNA damage and apoptosis, a common mechanism underlying both cytotoxic and antitumor effects of AF. In tumor-bearing mice, a simultaneous disruption of the thioredoxin and glutathione systems by the combination of AF and buthionine sulfoximine was shown to significantly improve tumor radioresponse. In conclusion, our findings illuminate TrxR in cancer cells as an exploitable radiobiological target and warrant further validation of AF in combination with radiotherapy.


Subject(s)
Auranofin/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Disease Models, Animal , Glutathione/metabolism , Hypoxia/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Thioredoxin-Disulfide Reductase/metabolism
3.
Radiother Oncol ; 119(2): 291-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26874542

ABSTRACT

BACKGROUND AND PURPOSE: High arginase-1 (Arg) expression by myeloid-derived suppressor cells (MDSC) is known to inhibit antitumor T-cell responses through depletion of l-arginine. We have previously shown that nitric oxide (NO), an immune mediator produced from l-arginine, is a potent radiosensitizer of hypoxic tumor cells. This study therefore examines whether Arg(+) overexpressing MDSC may confer radioresistance through depleting the substrate for NO synthesis. MATERIAL AND METHODS: MDSC and Arg expression were studied in preclinical mouse CT26 and 4T1 tumor models and further validated in rectal cancer patients in comparison with healthy donors. The radioprotective effect of MDSC was analyzed in hypoxic tumor cells with regard to l-arginine depletion. RESULTS: In both mouse tumors and cancer patients, MDSC expansion was associated with Arg activation causing accelerated l-arginine consumption. l-Arginine depletion in turn profoundly suppressed the capacity of classically activated macrophages to synthesize NO resulting in impaired tumor cell radiosensitivity. In advanced cT3-4 rectal cancer, circulating neutrophils revealed Arg overexpression approaching that in MDSC, therefore mounting a protumor compartment wherein Arg(+) neutrophils increased from 17% to over 90%. CONCLUSIONS: Protumor Arg(+) MDSC reveal a unique ability to radioprotect tumor cells through l-arginine depletion, a common mechanism behind both T-cell and macrophage inhibition.


Subject(s)
Arginase/physiology , Arginine/metabolism , Myeloid-Derived Suppressor Cells/physiology , Rectal Neoplasms/radiotherapy , Animals , HCT116 Cells , Humans , Macrophages/physiology , Mice , Neutrophils/physiology , Nitric Oxide/biosynthesis , Rectal Neoplasms/metabolism , T-Lymphocytes/immunology
4.
Int J Radiat Oncol Biol Phys ; 85(3): 820-7, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-22975619

ABSTRACT

PURPOSE: To determine whether host hepatocytes may reverse hypoxic radioresistance through nitric oxide (NO)-induced oxygen sparing, in a model relevant to colorectal cancer (CRC) liver metastases. METHODS AND MATERIALS: Hepatocytes and a panel of CRC cells were incubated in a tissue-mimetic coculture system with diffusion-limited oxygenation, and oxygen levels were monitored by an oxygen-sensing fluorescence probe. To activate endogenous NO production, cocultures were exposed to a cytokine mixture, and the expression of inducible nitric oxide synthase was analyzed by reverse transcription-polymerase chain reaction, Western blotting, and NO/nitrite production. The mitochondrial targets of NO were examined by enzymatic activity. To assess hypoxic radioresponse, cocultures were irradiated and reseeded for colonies. RESULTS: Resting hepatocytes consumed 10-40 times more oxygen than mouse CT26 and human DLD-1, HT29, HCT116, and SW480 CRC cells, and thus seemed to be the major effectors of hypoxic conditioning. As a result, hepatocytes caused uniform radioprotection of tumor cells at a 1:1 ratio. Conversely, NO-producing hepatocytes radiosensitized all CRC cell lines more than 1.5-fold, similar to the effect of selective mitochondrial inhibitors. The radiosensitizing effect was associated with a respiratory self-arrest of hepatocytes at the level of aconitase and complex II, which resulted in profound reoxygenation of tumor cells through oxygen sparing. Nitric oxide-producing hepatocytes were at least 10 times more active than NO-producing macrophages to reverse hypoxia-induced radioresistance. CONCLUSIONS: Hepatocytes were the major determinants of the hypoxic microenvironment and radioresponse of CRC cells in our model of metabolic hypoxia. We provide evidence that reoxygenation and radiosensitization of hypoxic CRC cells can be achieved through oxygen sparing induced by endogenous NO production in host hepatocytes.


Subject(s)
Cell Respiration/physiology , Colorectal Neoplasms , Hepatocytes/metabolism , Liver Neoplasms, Experimental/radiotherapy , Mitochondria, Liver/physiology , Nitric Oxide/biosynthesis , Oxygen Consumption/physiology , Radiation Tolerance/physiology , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Coculture Techniques , Enzyme Induction , HCT116 Cells , HT29 Cells , Hepatocytes/radiation effects , Humans , Liver Neoplasms, Experimental/secondary , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/metabolism , Tumor Microenvironment/physiology
5.
J Clin Oncol ; 31(1): 65-72, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23213105

ABSTRACT

PURPOSE: This phase III trial compared adjuvant whole-brain radiotherapy (WBRT) with observation after either surgery or radiosurgery of a limited number of brain metastases in patients with stable solid tumors. Here, we report the health-related quality-of-life (HRQOL) results. PATIENTS AND METHODS: HRQOL was a secondary end point in the trial. HRQOL was assessed at baseline, at 8 weeks, and then every 3 months for 3 years with the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire C30 and Brain Cancer Module. The following six primary HRQOL scales were considered: global health status; physical, cognitive, role, and emotional functioning; and fatigue. Statistical significance required P ≤ .05, and clinical relevance required a ≥ 10-point difference. RESULTS: Compliance was 88.3% at baseline and dropped to 45.0% at 1 year; thus, only the first year was analyzed. Overall, patients in the observation only arm reported better HRQOL scores than did patients who received WBRT. The differences were statistically significant and clinically relevant mostly during the early follow-up period (for global health status at 9 months, physical functioning at 8 weeks, cognitive functioning at 12 months, and fatigue at 8 weeks). Exploratory analysis of all other HRQOL scales suggested worse scores for the WBRT group, but none was clinically relevant. CONCLUSION: This study shows that adjuvant WBRT after surgery or radiosurgery of a limited number of brain metastases from solid tumors may negatively impact some aspects of HRQOL, even if these effects are transitory. Consequently, observation with close monitoring with magnetic resonance imaging (as done in the EORTC trial) is not detrimental for HRQOL.


Subject(s)
Brain Neoplasms/radiotherapy , Cranial Irradiation , Neoplasms/complications , Postoperative Complications , Quality of Life , Radiosurgery/adverse effects , Brain Neoplasms/secondary , Europe , Follow-Up Studies , Health Status , Humans , International Agencies , Neoplasm Staging , Neoplasms/pathology , Neoplasms/surgery , Patient Compliance , Prognosis , Radiotherapy, Adjuvant , Survival Rate
6.
J Clin Oncol ; 29(2): 134-41, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-21041710

ABSTRACT

PURPOSE: This European Organisation for Research and Treatment of Cancer phase III trial assesses whether adjuvant whole-brain radiotherapy (WBRT) increases the duration of functional independence after surgery or radiosurgery of brain metastases. PATIENTS AND METHODS: Patients with one to three brain metastases of solid tumors (small-cell lung cancer excluded) with stable systemic disease or asymptomatic primary tumors and WHO performance status (PS) of 0 to 2 were treated with complete surgery or radiosurgery and randomly assigned to adjuvant WBRT (30 Gy in 10 fractions) or observation (OBS). The primary end point was time to WHO PS deterioration to more than 2. RESULTS: Of 359 patients, 199 underwent radiosurgery, and 160 underwent surgery. In the radiosurgery group, 100 patients were allocated to OBS, and 99 were allocated to WBRT. After surgery, 79 patients were allocated to OBS, and 81 were allocated to adjuvant WBRT. The median time to WHO PS more than 2 was 10.0 months (95% CI, 8.1 to 11.7 months) after OBS and 9.5 months (95% CI, 7.8 to 11.9 months) after WBRT (P = .71). Overall survival was similar in the WBRT and OBS arms (median, 10.9 v 10.7 months, respectively; P = .89). WBRT reduced the 2-year relapse rate both at initial sites (surgery: 59% to 27%, P < .001; radiosurgery: 31% to 19%, P = .040) and at new sites (surgery: 42% to 23%, P = .008; radiosurgery: 48% to 33%, P = .023). Salvage therapies were used more frequently after OBS than after WBRT. Intracranial progression caused death in 78 (44%) of 179 patients in the OBS arm and in 50 (28%) of 180 patients in the WBRT arm. CONCLUSION: After radiosurgery or surgery of a limited number of brain metastases, adjuvant WBRT reduces intracranial relapses and neurologic deaths but fails to improve the duration of functional independence and overall survival.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Disease Progression , Disease-Free Survival , Female , Humans , Male , Middle Aged , Radiosurgery/adverse effects , Radiotherapy/adverse effects , Radiotherapy, Adjuvant , Salvage Therapy , Survival Rate
7.
Int J Radiat Oncol Biol Phys ; 76(5): 1520-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20338478

ABSTRACT

PURPOSE: Nitric oxide (NO), synthesized by the inducible nitric oxide synthase (iNOS), is known to inhibit metabolic oxygen consumption because of interference with mitochondrial respiratory activity. This study examined whether activation of iNOS (a) directly in tumor cells or (b) in bystander macrophages may improve radioresponse through sparing of oxygen. METHODS AND MATERIALS: EMT-6 tumor cells and RAW 264.7 macrophages were exposed to bacterial lipopolysaccharide plus interferon-gamma, and examined for iNOS expression by reverse transcription polymerase chain reaction, Western blotting and enzymatic activity. Tumor cells alone, or combined with macrophages were subjected to metabolic hypoxia and analyzed for radiosensitivity by clonogenic assay, and for oxygen consumption by electron paramagnetic resonance and a Clark-type electrode. RESULTS: Both tumor cells and macrophages displayed a coherent picture of iNOS induction at transcriptional/translational levels and NO/nitrite production, whereas macrophages showed also co-induction of the inducible heme oxygenase-1, which is associated with carbon monoxide (CO) and bilirubin production. Activation of iNOS in tumor cells resulted in a profound oxygen sparing and a 2.3-fold radiosensitization. Bystander NO-producing, but not CO-producing, macrophages were able to block oxygen consumption by 1.9-fold and to radiosensitize tumor cells by 2.2-fold. Both effects could be neutralized by aminoguanidine, a metabolic iNOS inhibitor. An improved radioresponse was clearly observed at macrophages to tumor cells ratios ranging between 1:16 to 1:1. CONCLUSIONS: Our study is the first, as far as we are aware, to provide evidence that iNOS may induce radiosensitization through oxygen sparing, and illuminates NO-producing macrophages as a novel determinant of tumor cell radioresponse within the hypoxic tumor microenvironment.


Subject(s)
Macrophage Activation , Macrophages/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Oxygen Consumption/physiology , Radiation Tolerance/physiology , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Respiration , Electron Spin Resonance Spectroscopy , Enzyme Activation , Guanidines/pharmacology , Heme Oxygenase-1/biosynthesis , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors
8.
Int J Radiat Oncol Biol Phys ; 71(3): 647-51, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18514774

ABSTRACT

Activated T lymphocytes are known to kill tumor cells by triggering cytolytic mechanisms; however, their ability to enhance radiation responses remains unclear. This study examined the radiosensitizing potential of mouse CD8+ T cells, obtained by T-cell-tailored expansion and immunomagnetic purification. Activated CD8+ T cells displayed an interferon (IFN)-gamma+ phenotype and enhanced by 1.8-fold the radiosensitivity of EMT-6 tumor cells in 1% oxygen, which modeled tumor-relevant hypoxia. Radiosensitization was counteracted by neutralizing IFN-gamma or by blocking the inducible isoform of nitric oxide synthase, thus delineating the immune-tumor cell interaction through the IFN-gamma secretion pathway. Reverse transcriptase-polymerase chain reaction, enzyme-linked immunosorbent assay, and fluorescence-activated cell sorter data in agreement detected downregulation of the IFN-gamma gene by hypoxia, which caused IFN-gamma deficiency next to radioresistance. Therefore, immune and radiation responses are likely to be allied in the hypoxic tumor microenvironment, and CD8+ T cells may bridge immunostimulatory and radiosensitizing strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Immunity, Innate/immunology , Interferon-gamma/immunology , Mammary Neoplasms, Animal/immunology , Animals , Cell Hypoxia/immunology , Cell Hypoxia/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Immunity, Innate/radiation effects , Mice , Mice, Inbred BALB C , Radiation Dosage
9.
Int J Radiat Oncol Biol Phys ; 66(5): 1473-80, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17056198

ABSTRACT

PURPOSE: To explore whether antitumor immunoadjuvant OM-174 can stimulate immune cells to produce interferon-gamma (IFN-gamma) and thereby radiosensitize tumor cells. METHODS AND MATERIALS: Splenocytes from BALB/c mice were stimulated by OM-174 at plasma-achievable concentrations (0.03-3 mug/mL), and afterward analyzed for the expression and secretion of IFN-gamma by reverse transcriptase-polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. Stimulated splenocytes were used as a source of IFN-gamma to radiosensitize hypoxic EMT-6 tumor cells through the cytokine-inducible isoform of nitric oxide synthase (iNOS). RESULTS: OM-174 activated the production of IFN-gamma at high levels that reached 70 ng/mL in normoxia (21% oxygen) and 27 ng/mL in tumor-relevant hypoxia (1% oxygen). This caused up to 2.1-fold radiosensitization of EMT-6 tumor cells, which was associated with the iNOS-mediated production of the radiosensitizing molecule nitric oxide, as confirmed by accumulation of its oxidative metabolite nitrite, Western blot analysis, and reverse transcriptase-polymerase chain reaction. Both iNOS activation and radiosensitization were counteracted by neutralizing antibodies against IFN-gamma. The same mechanism of radiosensitization through the IFN-gamma secretion pathway was identified for IL-12 + IL-18, which are known to mediate IFN-gamma responses. Hypoxia displayed a dual effect on the immune-tumor cell interaction, by downregulating the expression of the IFN-gamma gene while upregulating iNOS at transcriptional level. CONCLUSION: Immunoadjuvant OM-174 is an efficient radiosensitizer of tumor cells through activation of the IFN-gamma secretion pathway in immune cells. This finding indicates a rationale for combining immunostimulatory and radiosensitizing strategies and extends the potential therapeutic applications of OM-174.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interferon-gamma/metabolism , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Radiation Tolerance , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Immunity, Cellular , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , Interleukin-12/metabolism , Interleukin-18/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/immunology , Spleen/cytology , Up-Regulation
10.
Int J Radiat Oncol Biol Phys ; 60(2): 598-606, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15380597

ABSTRACT

PURPOSE: This study examines whether activated macrophages may radiosensitize tumor cells through the release of proinflammatory mediators. METHODS AND MATERIALS: RAW 264.7 macrophages were activated by lipid A, and the conditioned medium (CM) was analyzed for the secretion of cytokines and the production of nitric oxide (NO) through inducible nitric oxide synthase (iNOS). EMT-6 tumor cells were exposed to CM and analyzed for hypoxic cell radiosensitivity. The role of nuclear factor (NF)-kappaB in the transcriptional activation of iNOS was examined by luciferase reporter gene assay. RESULTS: Clinical immunomodulator lipid A, at a plasma-relevant concentration of 3 microg/mL, stimulated RAW 264.7 macrophages to release NO, tumor necrosis factor (TNF)-alpha, and other cytokines. This in turn activated iNOS-mediated NO production in EMT-6 tumor cells and drastically enhanced their radiosensitivity. Radiosensitization was abrogated by the iNOS inhibitor aminoguanidine but not by a neutralizing anti-TNF-alpha antibody. The mechanism of iNOS induction was linked to NF-kappaB but not to JAK/STAT signaling. Interferon-gamma further increased the NO production by macrophages to a level that caused radiosensitization of EMT-6 cells through the bystanding effect of diffused NO. CONCLUSIONS: We demonstrate for the first time that activated macrophages may radiosensitize tumor cells through the induction of NO synthesis, which occurs in both tumor and immune cells.


Subject(s)
Lipid A/pharmacology , Macrophages/drug effects , Mammary Neoplasms, Animal/radiotherapy , Nitric Oxide/biosynthesis , Radiation Tolerance , Animals , Cell Hypoxia , Cell Line, Tumor , Culture Media, Conditioned/chemistry , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Guanidines/pharmacology , Interferon-gamma/pharmacology , Interleukins/analysis , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitrites/analysis , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/biosynthesis
11.
Neurosurgery ; 53(6): 1429-31; discussion 1431-2, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14633311

ABSTRACT

OBJECTIVE AND IMPORTANCE: Epidermoid cysts are benign lesions that account for 0.7% of all intraspinal tumors. Standard treatment is complete resection. The recurrence rate after surgery cannot be estimated from the scarce literature, but it has been acknowledged that, in some patients, curative surgery may be impossible and temporary relief of symptoms is the only aim. CLINICAL PRESENTATION: In 1996, a 59-year-old woman presented with a 30-year history of a spinal epidermoid cyst, for which she had previously undergone eight operations. Spastic paraplegia of the lower limbs had been present from initial diagnosis, and the neurological level had moved upward to C6. At the time of presentation, paresthesia in her arms and hands had developed. A magnetic resonance imaging scan revealed typical sequelae of previous surgery and a fibrotic region starting at the level of C5-C6 with formation of a cyst that extended to the conus terminalis. The spinal cord could not be visualized caudally at C7. INTERVENTION: Because both patient and surgeons did not want further surgical interventions that might comprise upper limb function, radiotherapy was performed. A dose of 46 Gy was delivered to the cervicothoracic spine (C4-T1) by opposing anteroposterior high-energy photon beams. The treatment was well tolerated, and after treatment, the patient's clinical symptoms improved slightly, without evolution on magnetic resonance imaging. CONCLUSION: Irradiation was effective in stabilizing the patient's disease. Although radiotherapy is not established in the treatment of epidermoid cysts, it should be considered as an alternative to palliative surgery and in patients who cannot undergo operations.


Subject(s)
Epidermal Cyst/radiotherapy , Spinal Diseases/radiotherapy , Female , Humans , Middle Aged
12.
Int J Radiat Oncol Biol Phys ; 57(3): 779-86, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14529784

ABSTRACT

PURPOSE: Lipid A has shown promising immunostimulatory effects in both experimental tumor models and advanced stage cancer patients. This study examines whether lipid A may directly modulate the radioresponse of tumor cells by activating inducible nitric oxide synthase (iNOS) or cyclooxygenase-2 (COX-2) through nuclear factor-kappaB (NF-kappaB) signaling. METHODS AND MATERIALS: Hypoxic EMT-6 tumor cells were exposed to lipid A and analyzed for the level of COX-2 and iNOS by Western blotting and enzymatic assays. The hypoxic radioresponse of EMT-6 cells was estimated by clonogenic survival. The activation of NF-kappaB was examined by immunostaining of its p65 subunit and by luciferase reporter gene assay. RESULTS: Lipid A dose-dependently increased the expression and activity of iNOS with a maximal effect at plasma achievable concentrations of 3-30 micro g/mL. The COX-2 mediated production of prostaglandin E2 was constitutively high and further upregulated by lipid A. The radiosensitivity of hypoxic EMT-6 cells was increased up to 2.5 times and counteracted by the iNOS inhibitor aminoguanidine but not by the COX-2 inhibitor NS-398. The mechanism of radiosensitization was linked to NF-kappaB signaling, because its inhibition by phenylarsine oxide impaired both iNOS activation and radioresponse. CONCLUSION: Lipid A is an efficient hypoxic cell radiosensitizer at plasma relevant concentrations, which provides a rationale to combine lipid A with radiotherapy in further studies.


Subject(s)
Isoenzymes/metabolism , Lipid A/pharmacology , NF-kappa B/physiology , Nitric Oxide Synthase/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Radiation-Sensitizing Agents/pharmacology , Signal Transduction/physiology , Cell Hypoxia/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Guanidines , Humans , Membrane Proteins , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology
13.
Int J Radiat Oncol Biol Phys ; 54(3): 948-52, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12377349

ABSTRACT

PURPOSE: To evaluate accuracy and time requirements of a stereoscopic X-ray-based positioning system in patients receiving conformal radiotherapy to the prostate. METHODS AND MATERIALS: Setup errors of the isocenter with regard to the bony pelvis were measured by means of orthogonal verification films and compared to conventional positioning (using skin drawings and lasers) and infrared marker (IR) based positioning in each of 261 treatments. In each direction, the random error represents the standard deviation and the systematic error the absolute value of the mean position. Time measurements were done in 75 treatments. RESULTS: Random errors with the X-ray positioning system in the anteroposterior (AP), lateral, and longitudinal direction were (average +/- 1 standard deviation) 2 +/- 0.6 mm, 1.7 +/- 0.6 mm, and 2.4 +/- 0.7 mm. The corresponding values of conventional as well as IR positioning were significantly higher (p < 0.01). Systematic errors for X-ray positioning were 1.1 +/- 1.2 mm AP, 0.6 +/- 0.5 mm laterally, and 1.5 +/- 1.6 mm longitudinally. Conventional and IR marker-based positioning showed significantly larger systematic errors AP and laterally, but longitudinally, the difference was not significant. Depending on the axis looked at, errors of >or=5 mm occurred in 2%-14% of treatments after X-ray positioning, 13%-29% using IR markers, and 28%-53% with conventional positioning. Total linac time for one treatment session was 14 min 51 s +/- 4 min 18 s, half of which was used for the X-ray-assisted positioning procedure. CONCLUSION: X-ray-assisted patient positioning significantly improves setup accuracy, at the cost of an increased treatment time.


Subject(s)
Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Radiotherapy, Conformal/methods , Humans , Male , Radiography , Time Factors
14.
Int J Radiat Oncol Biol Phys ; 52(3): 694-8, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11849791

ABSTRACT

PURPOSE: To evaluate an infrared (IR) marker-based positioning system in patients receiving conformal radiotherapy for prostate cancer. METHODS AND MATERIALS: During 553 treatments, the ability of the IR system to automatically position the isocenter was recorded. Setup errors were measured by means of orthogonal verification films and compared to conventional positioning (using skin drawings and lasers) in 184 treatments. RESULTS: The standard deviation of anteroposterior (AP) and lateral setup errors was significantly reduced with IR marker positioning compared to conventional: 2 vs. 4.8 mm AP (p < 0.01) and 1.6 vs. 3.5 mm laterally (p < 0.01). Longitudinally, the difference was not significant (3.5 vs. 3.0 mm). Systematic errors were on the average smaller AP and laterally for the IR method: 4.1 vs. 7.8 mm AP (p = 0.01) and 3.1 vs. 5.6 mm lateral (p = 0.07). Longitudinally, the IR system resulted in somewhat larger systematic errors: 5.0 vs. 3.4 mm for conventional positioning (p = 0.03). The use of an off-line correction protocol, based on the average deviation measured over the first four fractions, allowed virtual elimination of systematic errors. Inability of the IR system to correctly locate the markers, leading to an executional failure, occurred in 21% of 553 fractions. CONCLUSION: IR marker-assisted patient positioning significantly improves setup accuracy along the AP and lateral axes. Executional failures need to be reduced.


Subject(s)
Infrared Rays , Prostatic Neoplasms/radiotherapy , Radiotherapy, Conformal/methods , Humans , Immobilization , Male , Radiotherapy Planning, Computer-Assisted/methods , Radiotherapy, Conformal/statistics & numerical data , Supine Position
SELECTION OF CITATIONS
SEARCH DETAIL
...