Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 467
Filter
1.
Cancer Res Treat ; 53(2): 471-479, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33091966

ABSTRACT

PURPOSE: Hypoxia is widely known as one of the mechanisms of chemoresistance and as an environmental condition which triggers invasion and metastasis of cancer. Evofosfamide is a hypoxia-activated prodrug of the cytotoxin bromo-isophosphoramide mustard conjugated with 2-nitroimidazole. Biliary tract cancer (BTC) is known to contain large hypoxic area. This study evaluated the efficacy and safety of evofosfamide as a second-line treatment of advanced BTC. MATERIALS AND METHODS: Patients received evofosfamide at a dose of 340 mg/m2 on days 1, 8, and 15 of every 28-day cycle. Primary end-point was progression-free survival (PFS) rate at 4-months (4m-PFSR). Secondary end-points included overall survival (OS), PFS, disease control rate (DCR), metabolic response by 18F-fluorodeoxyglucose positron emission tomography (PET), hypoxic parameters evaluated by 18F-fluoromisonidazole (FMISO) PET and toxicity. RESULTS: Twenty patients were treated with evofosfamide, with 16 response-evaluable patients. There was no objective response; stable disease was observed in nine patients, with a DCR of 56.25%. 4m-PFSR was 40.6%. Median PFS was 3.60 months (95% confidence interval [CI], 1.68 to 5.52). Median OS was 6.37 months (95% CI, 3.94 to 8.79). Reduction of tumor metabolic activity was observed in eight of 15 patients (53.3%). High baseline hypoxic parameters were associated with poor PFS. Change of hypoxic parameters between pretreatment and post-treatment reflected hypoxic-activated drug response. There was no treatment-related death. CONCLUSION: Evofosfamide as second-line treatment of advanced BTC showed acceptable safety and comparable efficacy to other agents. Changes in volumetric parameters measured with FMISO PET, showing the degree of tumor hypoxia, reflected the response to evofosfamide based on the mode of action.


Subject(s)
Biliary Tract Neoplasms/drug therapy , Cell Hypoxia/drug effects , Misonidazole/analogs & derivatives , Nitroimidazoles/therapeutic use , Phosphoramide Mustards/therapeutic use , Positron-Emission Tomography/methods , Biliary Tract Neoplasms/pathology , Female , Humans , Male , Middle Aged , Misonidazole/pharmacology , Misonidazole/therapeutic use , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Prospective Studies
2.
Br J Radiol ; 92(1093): 20170915, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29303355

ABSTRACT

Nitroimidazoles have been extensively explored as hypoxic cell radiosensitizers but have had limited clinical success, with efficacy restricted by toxicity. However, they have proven clinically useful as probes for tumour hypoxia. Both applications, and probably much of the dose-limiting toxicities, reflect the dominant chemical property of electron affinity or ease of reduction, associated with the nitro substituent in an aromatic structure. This single dominant property affords unusual, indeed extraordinary flexibility in drug or probe design, suggesting further development is possible in spite of earlier limitations, in particular building on the benefit of hindsight and an appreciation of errors made in earlier studies. The most notable errors were: the delay in viewing cellular thiol depletion as a likely common artefact in testing in vitro; slow recognition of pH-driven concentration gradients when compounds were weak acids and bases; and a failure to explore the possible involvement of pH and ascorbate in influencing hypoxia probe binding. The experience points to the need to involve a wider range of expertise than that historically involved in many laboratories when studying the effects of chemicals on radiation response or using diagnostic probes.


Subject(s)
Cell Hypoxia/drug effects , Misonidazole/pharmacology , Oxygen Consumption/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Hypoxia/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Drug , Humans , Sensitivity and Specificity , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects
3.
Sci Rep ; 8(1): 17832, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30546057

ABSTRACT

This study aimed to explore the application of two radiotracers (18F-fluorodeoxyglucose (FDG) and 18F-fluoromisonidazole (FMISO)) in monitoring hepatic metastases of human colorectal cancer (CRC). Mouse models of CRC hepatic metastases were established by implantation of the human CRC cell lines LoVo and HT29 by intrasplenic injection. Wound healing and Transwell assays were performed to examine cell migration and invasion abilities. Radiotracer-based cellular uptake in vitro and micro-positron emission tomography imaging of liver metastases in vivo were performed. The incidence of liver metastases in LoVo-xenografted mice was significantly higher than that in HT29-xenografted ones. The SUVmax/mean values of 18F-FMISO, but not 18F-FDG, in LoVo xenografts were significantly greater than in HT29 xenografts. In vitro, LoVo cells exhibited stronger metastatic potential and higher radiotracer uptake than HT29 cells. Mechanistically, the expression of HIF-1α and GLUT-1 in LoVo cells and LoVo tumor tissues was remarkably higher than in HT29 cells and tissues. Linear regression analysis demonstrated correlations between cellular 18F-FDG/18F-FMISO uptake and HIF-1α/GLUT-1 expression in vitro, as well as between 18F-FMISO SUVmax and GLUT-1 expression in vivo. 18F-FMISO uptake may serve as a potential biomarker for the detection of liver metastases in CRC, whereas its clinical use warrants validation.


Subject(s)
Colorectal Neoplasms , Fluorodeoxyglucose F18/pharmacology , Liver Neoplasms , Misonidazole/analogs & derivatives , Positron-Emission Tomography/methods , Animals , Cell Line, Tumor , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Heterografts , Humans , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Misonidazole/pharmacology , Neoplasm Metastasis , Neoplasm Transplantation
4.
Curr Radiopharm ; 9(3): 235-243, 2016.
Article in English | MEDLINE | ID: mdl-27562785

ABSTRACT

BACKGROUND AND OBJECTIVE: The hypoxia PET tracer, 1-[18F]fluoro-3-(2-nitro-1Himidazol- 1-yl)-propan-2-ol ([18F]FMISO) is the first radiotracer developed for hypoxia PET imaging and has shown promising for cancer diagnosis and prognosis. However, access to [18F]FMISO radiotracer is limited due to the needed cyclotron and radiochemistry expertise. The study aimed to develop the automated production method on the [18F]FMISO radiotracer with the novel fully automated platform of the BG75 system and validate its usage on animal tumor models. METHOD: [18F]FMISO was produced with the dose synthesis cartridge automatically on the BG75 system. Validation of [18F]FMISO hypoxia imaging functionality was conducted on two tumor mouse models (FaDu/U87 tumor). The distribution of [18F]FMISO within tumor was further validated by the standard hypoxia marker EF5. RESULTS: The average radiochemical purity was (99±1) % and the average pH was 5.5±0.2 with other quality attributes passing standard criteria (n=12). Overall biodistribution for [18F]FMISO in both tumor models was consistent with reported studies where bladder and large intestines presented highest activity at 90 min post injection. High spatial correlation was found between [18F]FMISO autoradiography and EF5 hypoxia staining, indicating high hypoxia specificity of [18MF]FMISO. CONCLUSION: This study shows that qualified [18F]FMISO can be efficiently produced on the BG75 system in an automated "dose-on-demand" mode using single dose disposable cards. The possibilities of having a low-cost, automated system manufacturing ([18F]Fluoride production + synthesis + QC) different radiotracers will greatly enhance the potential for PET technology to reach new geographical areas and underserved patient populations.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , Glioblastoma/diagnostic imaging , Misonidazole/analogs & derivatives , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology , Animals , Autoradiography , Cell Hypoxia , Cell Line, Tumor , Chromatography, High Pressure Liquid , Humans , Mice , Misonidazole/chemical synthesis , Misonidazole/pharmacology , Radiographic Image Interpretation, Computer-Assisted , Tissue Distribution
5.
Sci Rep ; 6: 31551, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27546160

ABSTRACT

Photodynamic therapy (PDT) is an efficacious treatment for some types of cancers. However, PDT-induced tumor hypoxia as a result of oxygen consumption and vascular damage can reduce the efficacy of this therapy. Measuring and monitoring intrinsic and PDT-induced tumor hypoxia in vivo during PDT is of high interest for prognostic and treatment evaluation. In the present study, static and dynamic (18)F-FMISO PET were performed with mice bearing either U87MG or MDA-MB-435 tumor xenografts immediately before and after PDT at different time points. Significant difference in tumor hypoxia in response to PDT over time was found between the U87MG and MDA-MB-435 tumors in both static and dynamic PET. Dynamic PET with pharmacokinetics modeling further monitored the kinetics of (18)F-FMISO retention to hypoxic sites after treatment. The Ki and k3 parametric analysis provided information on tumor hypoxia by distinction of the specific tracer retention in hypoxic sites from its non-specific distribution in tumor. Dynamic (18)F-FMISO PET with pharmacokinetics modeling, complementary to static PET analysis, provides a potential imaging tool for more detailed and more accurate quantification of tumor hypoxia during PDT.


Subject(s)
Cell Tracking/methods , Misonidazole/analogs & derivatives , Neoplasms, Experimental , Positron-Emission Tomography/methods , Animals , Cell Hypoxia , Cell Line, Tumor , Heterografts , Humans , Mice , Mice, Nude , Misonidazole/pharmacokinetics , Misonidazole/pharmacology , Neoplasm Transplantation , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism
6.
Clin Cancer Res ; 22(20): 5079-5086, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27185374

ABSTRACT

PURPOSE: Structural and functional alterations in tumor vasculature are thought to contribute to tumor hypoxia which is a primary driver of malignancy through its negative impact on the efficacy of radiation, immune surveillance, apoptosis, genomic stability, and accelerated angiogenesis. We performed a prospective, multicenter study to test the hypothesis that abnormal tumor vasculature and hypoxia, as measured with MRI and PET, will negatively impact survival in patients with newly diagnosed glioblastoma. EXPERIMENTAL DESIGN: Prior to the start of chemoradiation, patients with glioblastoma underwent MRI scans that included dynamic contrast enhanced and dynamic susceptibility contrast perfusion sequences to quantitate tumor cerebral blood volume/flow (CBV/CBF) and vascular permeability (ktrans) as well as 18F-Fluoromisonidazole (18F-FMISO) PET to quantitate tumor hypoxia. ROC analysis and Cox regression models were used to determine the association of imaging variables with progression-free and overall survival. RESULTS: Fifty patients were enrolled of which 42 had evaluable imaging data. Higher pretreatment 18F-FMISO SUVpeak (P = 0.048), mean ktrans (P = 0.024), and median ktrans (P = 0.045) were significantly associated with shorter overall survival. Higher pretreatment median ktrans (P = 0.021), normalized RCBV (P = 0.0096), and nCBF (P = 0.038) were significantly associated with shorter progression-free survival. SUVpeak [AUC = 0.75; 95% confidence interval (CI), 0.59-0.91], nRCBV (AUC = 0.72; 95% CI, 0.56-0.89), and nCBF (AUC = 0.72; 95% CI, 0.56-0.89) were predictive of survival at 1 year. CONCLUSIONS: Increased tumor perfusion, vascular volume, vascular permeability, and hypoxia are negative prognostic markers in newly diagnosed patients with gioblastoma, and these important physiologic markers can be measured safely and reliably using MRI and 18F-FMISO PET. Clin Cancer Res; 22(20); 5079-86. ©2016 AACR.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/mortality , Glioblastoma/blood supply , Glioblastoma/mortality , Magnetic Resonance Imaging , Neovascularization, Pathologic/pathology , Positron-Emission Tomography , Tumor Hypoxia/physiology , Adult , Aged , Biomarkers/analysis , Brain Neoplasms/pathology , Disease-Free Survival , Female , Glioblastoma/pathology , Humans , Male , Middle Aged , Misonidazole/analogs & derivatives , Misonidazole/pharmacology , Prospective Studies , Radiopharmaceuticals/pharmacology
7.
Chem Pharm Bull (Tokyo) ; 62(3): 301-3, 2014.
Article in English | MEDLINE | ID: mdl-24583786

ABSTRACT

The synthesis of a small series of 2-nitroimidazoles in which the ß-amino alcohol side chain was amidated with a range of alkylating/acylating functionality is described. Synthetic methodologies were developed that generally provided for selective N-acyl versus N,O-bisacyl products. In vitro, target analogs showed minimal radiosensitization activity, with only a few exhibiting a sensitizer enhancement ratio (SER) >2.0 and C(1.6) values comparable to reference agents RB-6145 and RSU-1069. In an assay to determine potential to alkylate biomolecules, representative analogs showed <1% of the alkylating activity of RSU-1069. In vivo, one analog showed an enhancement ratio of 1.6 relative to vehicle control when tested in B6C3F1 mice with an implanted KHT sarcoma. The data reinforce prior findings that there is a correlation between alkylation potential and in vivo activity.


Subject(s)
Acylation/drug effects , Alkylating Agents/chemical synthesis , Alkylating Agents/pharmacology , Drug Design , Nitroimidazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Drug Screening Assays, Antitumor , Mice , Misonidazole/analogs & derivatives , Misonidazole/pharmacology , Nitroimidazoles/chemical synthesis , Nitroimidazoles/chemistry , Radiation-Sensitizing Agents/chemical synthesis , Structure-Activity Relationship
8.
Arch Pharm (Weinheim) ; 347(3): 153-60, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24375829

ABSTRACT

Racemic misonidazole, a radiosensitizer formally used in radiation therapy of cancer and to date still applied, was once reported to exhibit strong inhibitory effects on mouse glutathione peroxidases (GPX). This appeared to qualify misonidazole as a lead structure for the development of novel GPX inhibitors to cause oxidative stress in chemotherapy-resistant tumors. A unique feature of misonidazole as an inhibitor of GPX is the absence of a thiol functionality. Therefore, it was expected to selectively target inhibition devoid of promiscuous interactions with cations and sulfhydryl groups. We synthesized the isomers of misonidazole and analyzed the ability of chiroptical high-performance liquid chromatography (HPLC) to identify the particular enantiomers. Due to the chiral pool synthesis, the assignment of the correct configuration could be verified. Finally, we evaluated both isomers for their inhibitory activities on bovine erythrocyte GPx-1, which is 87% homologous to the human enzyme. Despite the previously reported inhibition of racemic misonidazole on the less homologous mouse GPx-1, we did not find any significant inhibitory activity on the bovine enzyme for either isomer. Though misonidazole appears unlikely to be an inhibitor of human GPx-1 activity, we still spotlight misonidazole as a promising fragment-like lead structure in general.


Subject(s)
Circular Dichroism , Drug Discovery/methods , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glutathione Peroxidase/antagonists & inhibitors , Misonidazole/chemical synthesis , Misonidazole/pharmacology , Oxidants/chemical synthesis , Oxidants/pharmacology , Amino Acid Sequence , Animals , Cattle , Chromatography, High Pressure Liquid , Glutathione Peroxidase/metabolism , Isomerism , Molecular Sequence Data , Glutathione Peroxidase GPX1
9.
Acta Biochim Pol ; 60(3): 427-33, 2013.
Article in English | MEDLINE | ID: mdl-23888297

ABSTRACT

A series of new benzimidazole derivatives were synthesized and tested in vitro for possible anticancer activity. Their effect of proliferation into selected tumor cell lines at normoxia and hypoxia conditions was determined by WST-1 test. Additionally, apoptosis test (caspase 3/7 assay) was used to check the mode caused by the agents of cell death. Four of the examined compounds (7, 8, 13, 11) showed a very good antiproliferative effect and three of them were specific for hypoxia conditions (8, 14, 11). Compound 8 was the most cytotoxic against human lung adenocarcinoma A549 cells at hypoxic conditions. Hypoxia/ normoxia cytotoxic coefficient of compound 14 (4.75) is close to hypoxia/normoxia cytotoxic coefficient of tirapazamine (5.59) - a reference compound in our experiments and this parameter locates it between mitomycin C and 2-nitroimidazole (misonidazole). Screening test of caspase-dependent apoptosis proved that exposure to A549 cells of compounds 7-8 and 13-14 for 48 h promote apoptotic cell death. These results supplement our earlier study of the activity of new potentialy cytotoxic heterocyclic compounds against selected tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Hypoxia , Cell Line, Tumor , Gene Expression , Humans , Misonidazole/pharmacology , Mitomycin/pharmacology , Structure-Activity Relationship
10.
Int J Radiat Oncol Biol Phys ; 79(4): 1188-95, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21183291

ABSTRACT

PURPOSE: Tumor hypoxia has been observed in many human cancers and is associated with treatment failure in radiation therapy. The purpose of this study is to quantify the effect of different radiation fractionation schemes on tumor cell killing, assuming a realistic distribution of tumor oxygenation. METHODS AND MATERIALS: A probability density function for the partial pressure of oxygen in a tumor cell population is quantified as a function of radial distance from the capillary wall. Corresponding hypoxia reduction factors for cell killing are determined. The surviving fraction of a tumor consisting of maximally resistant cells, cells at intermediate levels of hypoxia, and normoxic cells is calculated as a function of dose per fraction for an equivalent tumor biological effective dose under normoxic conditions. RESULTS: Increasing hypoxia as a function of distance from blood vessels results in a decrease in tumor cell killing for a typical radiotherapy fractionation scheme by a factor of 10(5) over a distance of 130 µm. For head-and-neck cancer and prostate cancer, the fraction of tumor clonogens killed over a full treatment course decreases by up to a factor of ∼10(3) as the dose per fraction is increased from 2 to 24 Gy and from 2 to 18 Gy, respectively. CONCLUSIONS: Hypofractionation of a radiotherapy regimen can result in a significant decrease in tumor cell killing compared to standard fractionation as a result of tumor hypoxia. There is a potential for large errors when calculating alternate fractionations using formalisms that do not account for tumor hypoxia.


Subject(s)
Cell Hypoxia/physiology , Cell Survival/radiation effects , Dose Fractionation, Radiation , Oxygen Consumption/physiology , Radiation Tolerance/physiology , Algorithms , Capillaries/anatomy & histology , Capillaries/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Humans , Linear Models , Male , Misonidazole/pharmacology , Partial Pressure , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Treatment Failure
11.
Nucl Med Rev Cent East Eur ; 14(2): 90-5, 2011.
Article in English | MEDLINE | ID: mdl-22219149

ABSTRACT

During the carcinogenesis process, tumour cells often have a more rapid proliferation potential than cells that participate in blood capillary formation by neoangiogenesis. As a consequence of the poorly organized vasculature of various solid tumours, a limited oxygen delivery is observed. This hypoxic mechanism frequently occurs in solid cancers and can lead to therapeutic resistance. The present selected literature review is focused on the comparison of two positron emitting radiopharmaceuticals agents, which are currently leaders in tumour hypoxia imaging by PET. {18F}-fluoromisonidazole (=FMISO) is most commonly used as an investigational PET agent with an investigational new drug exemption from the FDA, while {64Cu}-diacetyl-bis(N4-methylthiosemicarbazone) (64Cu-ATSM) has been presented as an alternative radiopharmaceutical not yet readily available. The comparison of these two radiopharmaceutical agents is particularly focused on isotope properties, radiopharmaceutical labelling process, pharmacological mechanisms, dosimetry data in patients, and clinical results in terms of image contrast. PET imaging has demonstrated a good efficacy in tumour hypoxia imaging with both FMISO and Cu-ATSM, but FMISO has presented too slow an in vivo accumulation and a weak image contrast of the hypoxia area. Despite a less favourable dosimetry, 64Cu-ATSM appears superior in terms of imaging performance, calling for industrial and clinical development of this innovative radiopharmaceutical.


Subject(s)
Copper Radioisotopes/pharmacology , Misonidazole/analogs & derivatives , Neoplasms/diagnostic imaging , Organometallic Compounds/pharmacology , Positron-Emission Tomography/methods , Radiation-Sensitizing Agents/pharmacology , Thiosemicarbazones/pharmacology , Cell Hypoxia , Coordination Complexes , Humans , Misonidazole/pharmacology
12.
Proc Natl Acad Sci U S A ; 106(33): 14004-9, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19666490

ABSTRACT

Tumor hypoxia and the hypoxia-inducible factors (HIFs) play a central role in the development of cancer. To study the relationship between tumor growth, tumor hypoxia, the stabilization of HIF-1alpha, and HIF transcriptional activity, we have established an in vivo imaging tool that allows longitudinal and noninvasive monitoring of these processes in a mouse C51 allograft tumor model. We used positron emission tomography (PET) with the hypoxia-sensitive tracer [(18)F]-fluoromisonidazole (FMISO) to measure tumor hypoxia over 14 days. Stabilization of HIF-1alpha and HIF transcriptional activity were assessed by bioluminescence imaging using the reporter constructs HIF-1alpha-luciferase and hypoxia response element-luciferase, respectively, stably expressed in C51 cells. Interestingly, we did not observe any major change in the level of tumor hypoxia throughout the observation period whereas HIF-1alpha levels and HIF activity showed drastic temporal variations. When comparing the readouts as a function of time we found a good correlation between HIF-1alpha levels and HIF activity. In contrast, there was no significant correlation between the [(18)F]-FMISO PET and HIF readouts. The tool developed in this work allows for the longitudinal study of tumor hypoxia and HIF-1alpha in cancer in an individual animal and will be of value when monitoring the efficacy of therapeutical interventions targeting the HIF pathway.


Subject(s)
Hypoxia , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases/metabolism , Mice , Misonidazole/analogs & derivatives , Misonidazole/pharmacology , Models, Biological , Neoplasm Transplantation , Positron-Emission Tomography/methods , Response Elements , Time Factors , Transcription, Genetic
13.
Brain Res ; 1288: 135-42, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19595680

ABSTRACT

The hypoxia tracer fluorine-18 fluoromisonidazole ([18F]FMISO) and its tritiated counterpart ([(3)H]FMISO) have been used as markers of potentially salvageable brain (ischemic penumbra) after stroke. In experimental models, the dynamics and half-life of [3H]FMISO allow concurrent histology after 24 h. Our aim was to further validate these techniques, by determining the optimum tracer exposure interval to delineate ischemic penumbra, and the effects of prolonged exposure on tracer retention in permanent ischemia. Middle cerebral artery occlusion (MCAO) of varying durations was created in rats using the thread occlusion model. Autoradiography using objective thresholding to define tracer-retention volume was performed to determine the time course of tracer retention in hypoxic tissues and the duration of ongoing retention after bolus administration. An ischemic duration of < or =90 min resulted in a tracer-retention volume underestimating 'tissue at risk' (histological infarction 24 h after permanent occlusion) by >1/2. Two hour ischemia resulted in a volume equal to 'tissue at risk'. Twenty-four hour permanent ischemia resulted in tracer-retaining tissue volumes greater than final infarction. However, the use of more stringent thresholding of autoradiographic signal produced a volume of FMISO retention closely approximating infarct volume. The findings indicate that the timing of imaging is crucial, with an optimal imaging time of 2 h using the current threshold. Earlier imaging is limited by tracer dynamics with this particular agent, however autoradiography with a longer ischemic interval (permanent occlusion) is feasible with modified thresholds. These findings support a role for hypoxia tracers in providing new insight into the ischemic penumbra.


Subject(s)
Brain/diagnostic imaging , Hypoxia-Ischemia, Brain/diagnostic imaging , Infarction, Middle Cerebral Artery/diagnostic imaging , Misonidazole/analogs & derivatives , Animals , Autoradiography , Brain/drug effects , Brain/pathology , Cerebrovascular Circulation , Hypoxia-Ischemia, Brain/pathology , Image Interpretation, Computer-Assisted , Infarction, Middle Cerebral Artery/pathology , Misonidazole/pharmacology , Radiation-Sensitizing Agents/pharmacology , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Time Factors
15.
Anticancer Res ; 26(2A): 1261-70, 2006.
Article in English | MEDLINE | ID: mdl-16619533

ABSTRACT

BACKGROUND: Intratumor quiescent (Q) cells and p53-mutated tumor cells are more difficult to control than intratumor proliferating (P) cells and p53 wild-type tumor cells, respectively. The usefulness of 3 hypoxic cell radio-sensitizers was compared in terms of a radio-sensitizing effect under aerobic and hypoxic conditions and a repair-inhibiting effect following irradiation on both Q and total (P + Q) cell populations in solid tumors. The dependency of these effects on the p53 status of tumor cells was also examined using tumor cell lines with identical genetic backgrounds except for their p53 status. MATERIALS AND METHODS: Human head and neck squamous cell carcinoma cells transfected with mutant TP53 (SAS/mp53) or with neo vector as a control (SAS/neo) were inoculated subcutaneously into both the hind legs of Balb/cA nude mice. The nude mice bearing the tumors and C3H/He mice bearing SCC VII tumors received 5-bromo-2'-deoxyuridine (BrdU) continuously to label all the P cells in the tumors. Tumor-bearing mice received gamma-ray irradiation while alive or following tumor clamping after being administered no drug, nimorazole, SR-2514 or misonidazole, or received no drug, nimorazole, SR-2514 or misonidazole straight after gamma-ray irradiation. For the group irradiated after receiving the drug, the tumors were excised immediately following irradiation, while for the group irradiated before receiving the drug, the tumors were excised 24 h after irradiation. The excised tumors were minced and trypsinized. The tumor cell suspensions thus obtained were incubated with cytochalasin-B (a cytokinesis blocker), and the micronucleus (MN) frequency in the cells without BrdU labelling (= quiescent (Q) cells) was determined using immunofluorescence staining for BrdU. Meanwhile, the MN frequency in the total tumor cell population was determined from the tumors that had not been pretreated with BrdU. The clonogenic cell survival was also determined in the mice given no BrdU. RESULTS: Both the radio-sensitizing effects under aerobic and hypoxic conditions and the repair-inhibiting effects following gamma-ray irradiation increased in the following order: nimorazole < SR-2514 < misonidazole in both total and Q cells in these 3 tumors. Both effects were more marked in the Q cells and p53-mutated tumors than in the total cells and p53-wild tumors, respectively. CONCLUSION: In terms of controlling radio-resistant Q tumor cells and p53-mutated tumor cells, the combination of radio-sensitizers and conventional radiotherapy is promising both for radio-sensitization and for repair-inhibition, but further study of the toxicity to normal tissues is needed.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Genes, p53/physiology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Cell Hypoxia/physiology , Combined Modality Therapy , DNA Repair/genetics , Dose-Response Relationship, Radiation , Female , Gamma Rays , Head and Neck Neoplasms/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Misonidazole/pharmacology , Nimorazole/pharmacology , Xenograft Model Antitumor Assays
16.
BMC Cancer ; 5: 152, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16321146

ABSTRACT

BACKGROUND: Hypoxia compromises local control in patients with head-and-neck cancer (HNC). In order to determine the value of [18F]-fluoromisonidazole (Fmiso) with regard to tumor hypoxia, a patient study with dynamic Fmiso PET was performed. For a better understanding of tracer uptake and distribution, a kinetic model was developed to analyze dynamic Fmiso PET data. METHODS: For 15 HNC patients, dynamic Fmiso PET examinations were performed prior to radiotherapy (RT) treatment. The data was analyzed using a two compartment model, which allows the determination of characteristic hypoxia and perfusion values. For different parameters, such as patient age, tumor size and standardized uptake value, the correlation to treatment outcome was tested using the Wilcoxon-Mann-Whitney U-test. Statistical tests were also performed for hypoxia and perfusion parameters determined by the kinetic model and for two different metrics based on these parameters. RESULTS: The kinetic Fmiso analysis extracts local hypoxia and perfusion characteristics of a tumor tissue. These parameters are independent quantities. In this study, different types of characteristic hypoxia-perfusion patterns in tumors could be identified. The clinical verification of the results, obtained on the basis of the kinetic analysis, showed a high correlation of hypoxia-perfusion patterns and RT treatment outcome (p = 0.001) for this initial patient group. CONCLUSION: The presented study established, that Fmiso PET scans may benefit from dynamic acquisition and analysis by a kinetic model. The pattern of distribution of perfusion and hypoxia in the tissue is correlated to local control in HNC.


Subject(s)
Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Misonidazole/analogs & derivatives , Positron-Emission Tomography/methods , Radiation-Sensitizing Agents/pharmacology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Humans , Hypoxia , Image Processing, Computer-Assisted , Kinetics , Male , Middle Aged , Misonidazole/pharmacology , Models, Statistical , Software , Treatment Outcome
17.
Nucl Med Commun ; 24(5): 597-606, 2003 May.
Article in English | MEDLINE | ID: mdl-12717079

ABSTRACT

99mTc-glucarate is an investigational radiopharmaceutical which has been shown to accumulate in acute cerebral and myocardial injuries and in some tumours. In the present work, a survey of possible factors affecting the cellular accumulation of 99mTc-glucarate was carried out in cell lines and strains in vitro and in murine tumours in vivo. Accumulation was enhanced under hypoxic conditions in 12 of the 16 human and murine cell lines and strains studied, and inhibited in the presence of nitroimidazoles. At temperatures lower than 37 degrees C, accumulation was reduced, but a hypoxic/aerobic differential was maintained. Aerobic accumulation of 99mTc-glucarate was enhanced by cyanide. In transplanted tumours in mice, 99mTc-glucarate showed high tumour/muscle and tumour/blood ratios at early times after injection. Pharmacological enhancement of the extent of hypoxia by the administration of hydralazine or nitro-L-arginine resulted in significantly increased accumulation of 99mTc-glucarate in the tumour. The in vitro and in vivo properties of 99mTc-glucarate suggest that it may be useful for tumour imaging in the clinic, although the exact mechanism(s) by which it localizes in tumours remains unknown.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Fibrosarcoma/metabolism , Glucaric Acid/analogs & derivatives , Glucaric Acid/pharmacokinetics , Hypoxia/metabolism , Organotechnetium Compounds/pharmacokinetics , Animals , Azides/pharmacology , CHO Cells , Carcinoma, Squamous Cell/diagnostic imaging , Cricetinae , Cricetulus , Fibrosarcoma/diagnostic imaging , Fructose/metabolism , Hypoxia/diagnostic imaging , Male , Metabolic Clearance Rate , Metronidazole/pharmacology , Mice , Mice, Inbred C3H , Misonidazole/pharmacology , Oxygen/metabolism , Potassium Cyanide/pharmacology , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
19.
Radiat Res ; 158(3): 302-10, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12175307

ABSTRACT

The nitroimidazole-linked phenanthridines 2-NLP-3 (5-[3-(2-nitro-1-imidazoyl)-propyl]-phenanthridinium bromide) and 2-NLP-4 (5-[3-(2-nitro-1-imidazoyl)-butyl]-phenanthridinium bromide) are composed of the radiosensitizer, 2-nitroimidazole, attached to the DNA intercalator phenanthridine by a 3- and 4-carbon linker, respectively. Previous in vitro assays showed both compounds to be 10-100 times more efficient as hypoxic cell radiosensitizers (based on external drug concentrations) than the untargeted 2-nitroimidazole radiosensitizer, misonidazole (Cowan et al., Radiat. Res. 127, 81-89, 1991). Here we have used a (32)P postlabeling assay and 5'-end-labeled oligonucleotide assay to compare the radiation-induced DNA damage generated in the presence of 2-NLP-3, 2-NLP-4, phenanthridine and misonidazole. After irradiation of the DNA under anoxic conditions, we observed a significantly greater level of 3'-phosphoglycolate DNA damage in the presence of 2-NLP-3 or 2-NLP-4 compared to irradiation of the DNA in the presence of misonidazole. This may account at least in part for the greater cellular radiosensitization shown by the nitroimidazole-linked phenanthridines over misonidazole. Of the two nitroimidazole-linked phenanthridines, the better in vitro radiosensitizer, 2-NLP-4, generated more 3'-phosphoglycolate in DNA than did 2-NLP-3. At all concentrations, phenanthridine had little effect on the levels of DNA damage, suggesting that the enhanced radiosensitization displayed by 2-NLP-3 and 2-NLP-4 is due to the localization of the 2-nitroimidazole to the DNA by the phenanthridine substituent and not to radiosensitization by the phenanthridine moiety itself.


Subject(s)
DNA Damage , DNA/radiation effects , Intercalating Agents/pharmacology , Nitroimidazoles/pharmacology , Phenanthridines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , CHO Cells/radiation effects , Cattle , Cell Hypoxia , Cricetinae , Cricetulus , DNA/drug effects , DNA Repair , Dose-Response Relationship, Drug , Gamma Rays , Misonidazole/pharmacology , Oxygen/pharmacology , Structure-Activity Relationship
20.
Acta Oncol ; 39(2): 199-205, 2000.
Article in English | MEDLINE | ID: mdl-10859011

ABSTRACT

Two nitroimidazole compounds, misonidazole (MISO) and nimorazole (NIMO), were evaluated for their potential to modify uptake of [5,6-3H] 2-fluoro-2-deoxy-D-glucose (3H-FDG) in the human squamous carcinoma cell line UT-SCC-5 exposed to increasing levels of hypoxia. UT-SCC-5 cells were incubated with 0-10 mM of MISO or NIMO under normal or reduced oxygen concentrations of 20%, 1.5%, or 0% with 5% CO2 for 6 h, after which 74 KBq of 3H-FDG was added in media for 1 h. In the presence of normal concentrations of O2, both sensitizers increased 3H-FDG uptake by up to 178% (MISO) or 84% (NIMO) when compared with untreated cells. In anoxia, MISO decreased 3H-FDG uptake to 35% of that of control whereas NIMO-treated cells showed a respective decrease in tracer uptake to 62%. Clonogenic assays clearly indicated that MISO was toxic and NIMO moderately toxic for hypoxic cells, whereas both sensitizers exerted only a very modest effect on survival of fully oxygenated cells. Our findings indicate that nitroimidazole treatment consistently increases 3H-FDG uptake into UT-SCC-5 cells under normal oxygen concentrations. In hypoxia, the observed decrease in tracer uptake is dependent on both the level of ambient oxygen and drug concentration and may reflect both direct toxicity and inhibition of glycolysis. The observations may be useful for further applications of 18F-FDG positron emission tomography (PET) to monitor effects of hypoxic cell radiosensitizers on tumor metabolism in vivo.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Misonidazole/pharmacology , Nimorazole/pharmacology , Radiation-Sensitizing Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Death , Cell Hypoxia/physiology , Fluorodeoxyglucose F18/pharmacokinetics , Humans , Tomography, Emission-Computed , Tritium , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...