Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cancer Res ; 67(16): 7646-53, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699769

RESUMEN

Tumor hypoxia is commonly observed in primary solid malignancies but the hypoxic status of subclinical micrometastatic disease is largely unknown. The distribution of hypoxia in microscopic tumors was studied in animal models of disseminated peritoneal disease and intradermal (i.d.) growing tumors. Tumors derived from human colorectal adenocarcinoma cell lines HT29 and HCT-8 ranged in size from a few hundred microns to several millimeters in diameter. Hypoxia was detected by immunofluorescent visualization of pimonidazole and the hypoxia-regulated protein carbonic anhydrase 9. Tumor blood perfusion, cellular proliferation, and vascularity were visualized using Hoechst 33342, bromodeoxyuridine, and CD31 staining, respectively. In general, tumors of <1 mm diameter were intensely hypoxic, poorly perfused, and possessed little to no vasculature. Larger tumors (approximately 1-4 mm diameter) were well perfused with widespread vasculature and were not significantly hypoxic. Patterns of hypoxia in disseminated peritoneal tumors and i.d. tumors were similar. Levels of hypoxia in microscopic peritoneal tumors were reduced by carbogen breathing. Peritoneal and i.d. tumor models are suitable for studying hypoxia in microscopic tumors. If the patterns of tumor hypoxia in human patients are similar to those observed in these animal experiments, then the efficacy of systemic treatments of micrometastatic disease may be compromised by hypoxic resistance.


Asunto(s)
Neoplasias del Colon/metabolismo , Oxígeno/metabolismo , Neoplasias Peritoneales/metabolismo , Animales , Ascitis/metabolismo , Ascitis/patología , Dióxido de Carbono/farmacología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Neoplasias del Colon/patología , Femenino , Células HT29 , Humanos , Ratones , Ratones Desnudos , Microscopía Fluorescente , Trasplante de Neoplasias , Oxígeno/farmacología , Neoplasias Peritoneales/patología , Trasplante Heterólogo
2.
Int J Radiat Oncol Biol Phys ; 71(2): 533-41, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18374502

RESUMEN

PURPOSE: To evaluate the response of cells over-expressing dominant negative (DN) Ku70 to single and multiple small radiation doses. METHODS AND MATERIALS: Clones of fibroblasts over-expressing DNKu70, DNKu70-7, DNKu70-11, and parental Rat-1 cells were irradiated under oxic or hypoxic conditions with single or multiple doses. Cells were trypsinized 0 or 6 h after irradiation to determine surviving fraction (SF). RESULTS: Oxic DNKu70-7 or -11 cells trypsinized 6 h after irradiation were 1.52 or 1.25 and 1.28 or 1.15 times more sensitive than oxic Rat-1 at SF of 0.5 and 0.1, respectively. Hypoxic DNKu70-7 or -11 cells trypsinized 6 h after irradiation were 1.44 or 1.70 and 1.33 or 1.51 times more sensitive than hypoxic Rat-1 at SF of 0.5 and 0.1, respectively. To the multiple doses, oxic and hypoxic DNKu70-7 or -11 cells were 1.35 or 1.37 and 2.23 or 4.61 times more sensitive than oxic and hypoxic Rat-1, respectively, resulting in very small oxygen enhancement ratios. Namely, enhancement caused by DNKu70 under hypoxia after multiple doses was greater than that under oxic conditions and greater than that after single dose. CONCLUSIONS: Over-expression of DNKu70 enhances cells' response to radiation given as a single dose and as multiple small doses. The enhancement after multiple doses was stronger under hypoxic than under oxic conditions. These results encourage the use of DNKu70 fragment in a gene-radiotherapy.


Asunto(s)
Antígenos Nucleares/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibroblastos/efectos de la radiación , Tolerancia a Radiación/fisiología , Animales , Antígenos Nucleares/genética , Antígenos Nucleares/uso terapéutico , Hipoxia de la Célula , Línea Celular , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Daño del ADN , Reparación del ADN , Proteína Quinasa Activada por ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/uso terapéutico , Relación Dosis-Respuesta en la Radiación , Fibroblastos/metabolismo , Autoantígeno Ku , Modelos Lineales , Dosis de Radiación , Ratas , Factores de Tiempo
3.
Radiother Oncol ; 88(2): 269-76, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18538874

RESUMEN

PURPOSE: To study the changes in hypoxia resulting from mild temperature hyperthermia (MTH) in a subcutaneous xenograft model using dual-tracer immunohistochemical techniques. MATERIALS AND METHODS: HT29 tumors were locally heated at 41 degrees C. Changes in tumor hypoxia were investigated by pimonidazole and EF5. Pimonidazole was given 1h preheating, EF5 at various times during or after treatment, 1h later the animals were sacrificed. Blood vessels were identified by CD31 staining, and perfusion by Hoechst 33342 injected 1 min pre-sacrifice. RESULTS: The overall hypoxic fraction was significantly decreased by MTH during and immediately after heating. However, MTH induced both increases and decreases in tumor hypoxia in different parts of the tumor. Specifically, MTH decreased hypoxia in the regions with relatively well-perfused blood vessels, but increased hypoxia in regions that were poorly perfused. At 24-h post heating, newly formed hypoxic regions surrounded previously-hypoxic foci, which in turn surrounded pimonidazole-stained debris. Quantitative analysis did not evince changes in tumor oxygenation due to MTH at 24h post-treatment. CONCLUSION: In this xenograft model, the effect of MTH on tumor oxygenation was variable, both spatially and kinetically. Overall tumor oxygenation was improved during and after heating, but the effect was short-lived.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Colorrectales/metabolismo , Hipertermia Inducida , Inmunohistoquímica/métodos , Oxígeno/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Adenocarcinoma/irrigación sanguínea , Análisis de Varianza , Animales , Hipoxia de la Célula , Neoplasias Colorrectales/irrigación sanguínea , Etanidazol/análogos & derivados , Etanidazol/farmacología , Femenino , Hidrocarburos Fluorados/farmacología , Ratones , Nitroimidazoles/farmacología , Trasplante Heterólogo
4.
Radiat Res ; 169(1): 67-75, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18159950

RESUMEN

Wen, B., Urano, M., Humm, J. L., Seshan, V. E., Li, G. C. and Ling, C. C. Comparison of Helzel and OxyLite Systems in the Measurements of Tumor Partial Oxygen Pressure (pO(2)). Radiat. Res. 168, 67-75 (2008). It has been demonstrated in both experimental and human malignancies that hypoxic tumor cells are linked with aggressive disease phenotype. One of the methods to identify these cells is by direct physical measurement of tumor pO(2). This study compared pO(2) values measured with two systems, the Helzel Hypoximeter (successor of the polarographic Eppendorf electrode) and the Oxford-Optronix OxyLite (fiber-optic probe), in R3327-AT and R3327-AT/tkeGFP tumors. Partial oxygen pressure was measured in individual tumors with either system or in the same tumor with both systems. The similarities and discrepancies in pO(2) measurements between the two systems were also investigated when tumor-bearing animals were breathing pure oxygen. Our data showed a considerable heterogeneity in pO(2) values in each tumor using both the Helzel and OxyLite systems. Similar results were obtained with both systems for the mean and median pO(2) values, and the distributions of pO(2) values within the interval 0 < pO(2) < 40 mmHg (the range important for defining tumor hypoxia) were found to be statistically equivalent. However, the frequencies of high pO(2) values (>40 mmHg) and zero values measured by the two systems were statistically significantly different.


Asunto(s)
Neoplasias , Oxígeno/metabolismo , Animales , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patología , Presión Parcial
5.
Radiat Res ; 166(3): 512-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16953670

RESUMEN

The presence of oxygen-deficient tumor cells is a critical issue in cancer therapy. To identify tumor hypoxia, tissue partial oxygen pressure (pO2) can be measured directly. The OxyLite system allows determination of pO2 in tumors and permits continuous measurements of pO2 at a fixed point. In this study, this system was used to continuously measure pO2 in R3327-AT tumors in animals anesthetized with isoflurane. In addition, continuous pO2 measurement was performed in the muscle in non-tumor-bearing animals. In animals breathing isoflurane balanced by air, tumor pO2 at fixed positions decreased rapidly within 1-2 min of probe positioning but remained stable thereafter. In animals breathing isoflurane balanced by pure oxygen, tumor pO2 was higher and remained high. We also measured pO2 values at multiple positions in R3327-AT tumors of various sizes, with anesthetized animals breathing either air or pure oxygen. Our data showed that the frequency of pO2 measurements below 2.5 or 5.0 mmHg was significantly higher in animals breathing air than in animals breathing pure oxygen. Measurements in different-sized tumors showed that the mean pO2 value decreased as tumor volume increased, with the largest change occurring between tumor volumes of 100 and 200 mm3. Our data demonstrate that the OxyLite system, when used with isoflurane anesthesia, is a valuable tool in the study of tumor hypoxia.


Asunto(s)
Adenocarcinoma/metabolismo , Isoflurano/administración & dosificación , Oximetría/instrumentación , Oxígeno/análisis , Anestésicos por Inhalación/administración & dosificación , Animales , Diseño de Equipo , Análisis de Falla de Equipo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oximetría/métodos , Presión , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
6.
Cancer Chemother Pharmacol ; 58(6): 719-24, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16614851

RESUMEN

BACKGROUND: Hyperthermia enhances the cytotoxicity of some chemotherapeutic agents. Both clinical and laboratory studies suggest melphalan may be an important drug when hyperthermia is added to chemotherapy treatments. Factors that may modify the thermal enhancement of melphalan were studied to optimize its clinical use with hyperthermia. METHODS: The tumor studied was an early-generation isotransplant of a spontaneous C3Hf/Sed mouse fibrosarcoma, Fsa-II. All studies were performed under supervision of the Animal Care and Use Committee. Hyperthermia was administered by immersing the tumor-bearing foot into a constant temperature water bath. Four factors were studied: duration of hyperthermia, sequencing of hyperthermia and melphalan, intensity of hyperthermia, and tumor size. To study duration of hyperthermia tumors were treated at 41.5 degrees C for 30 or 90 min immediately after intraperitoneal administration of melphalan. For sequencing of hyperthermia and melphalan, animals received hyperthermia treatment of tumors for 30 min at 41.5 degrees C immediately after drug administration, both immediately and 3 h after administration of drug or only at 3 h after administration of drug. Intensity of hyperthermia was studied using heat treatment of tumors for 30 min at 41.5 or 43.5 degrees C immediately following drug administration. Effect of tumor size was studied by delaying experiments until three times the tumor volume (113 mm3) was observed. Treatment of tumors was for 30 min at 41.5 degrees C immediately following drug administration. Tumor response was studied by the mean tumor growth time. RESULTS: Hyperthermia in the absence of melphalan had a small but significant effect on tumor growth time at 43.5 degrees C but not at 41.5 degrees C. Hyperthermia at 41.5 degrees C immediately after melphalan administration doubled mean tumor growth time at 30 min and caused a threefold increase at 90 min (P=0.0002) when compared to tumors treated with melphalan alone at room temperature. Application of hyperthermia for one-half hour immediately following drug administration was the most effective in delaying tumor growth. No significant difference in mean tumor growth time was observed with an increase in temperature from 41.5 to 43.5 degrees C. For large tumors heat alone and melphalan alone caused a moderate increase in tumor growth delay. These effects in large tumors were greatly increased by a combination of chemotherapy and hyperthermia. CONCLUSIONS: From our data it would appear that the administration of intraperitoneal melphalan immediately prior to 90 min of heat at 41.5 degrees C may optimize anti-neoplastic activity. These data may be useful in formulating clinical protocols in which melphalan and heat are combined.


Asunto(s)
Fibrosarcoma/terapia , Hipertermia Inducida , Melfalán/uso terapéutico , Animales , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/uso terapéutico , Terapia Combinada , Fibrosarcoma/patología , Calor , Inyecciones Intraperitoneales , Melfalán/administración & dosificación , Ratones , Ratones Endogámicos C3H , Sarcoma Experimental/patología , Sarcoma Experimental/terapia , Temperatura , Factores de Tiempo , Resultado del Tratamiento
7.
Int J Oncol ; 26(4): 853-62, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15753978

RESUMEN

Manganese superoxide dismutase (MnSOD) is a mitochondrial enzyme that converts cytotoxic superoxide radicals into hydrogen peroxide. MnSOD activity is lower in tumor cells, and MnSOD overexpression reportedly ameliorates malignant phenotypes. We established stable MnSOD overexpressing cell lines from a human osteosarcoma cell line, SaOS2, and then investigated the effects of MnSOD overexpression on plating efficiency (PE) and the involvement of reactive oxygen species, including nitric oxide (NO) in those effects. The PE of SaOS2FM(L), a moderate MnSOD overexpression cell line, increased, while that of SaOS2FM(H), a high MnSOD overexpression cell line, decreased. Although we assessed PE using a colony-formation assay, time-lapse microscopic observation revealed that cells attached to the flasks had undergone neither apoptosis nor necrosis. Moreover, MnSOD overexpression did not affect cell doubling time. Therefore, MnSOD overexpression might correlate directly with cellular adhesion's effect on PE changes. When L-buthionine-[S,R]-sulfoximine (BSO) was administered to increase the intracellular concentration of hydrogen peroxide, the PEs of both cell lines decreased, and when hydrogen peroxide was eliminated by the administration of sodium pyruvate, only the PE of SaOS2FM(H) increased. The combination of BSO and NO (NOR4 or isosorbide 5-mononitrate) administration synergistically decreased PE in both cell lines. These findings suggest that changes in cellular adhesion properties correlate with the balance between increased hydrogen peroxide levels and decreased superoxide radical levels. This is the first report to indicate that PE and cellular adhesion properties change bidirectionally according to the levels of MnSOD overexpression: first increasing then decreasing as MnSOD activity increases. Our results indicate that PE changes might be decided by the balance between two cytotoxic compounds (decreased superoxide radical levels and increased hydrogen peroxide levels), and that NO loading and increased hydrogen peroxide synergistically reduce PE and cellular adhesion.


Asunto(s)
Neoplasias Óseas/enzimología , Adhesión Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/enzimología , Osteosarcoma/patología , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/farmacología , Neoplasias Óseas/patología , Ensayo de Unidades Formadoras de Colonias , Genes p53 , Humanos , Peróxido de Hidrógeno/farmacología , Óxido Nítrico/farmacología , Oxidantes/química , Fenotipo , Especies Reactivas de Oxígeno/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba
8.
Radiat Res ; 158(2): 167-73, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12105986

RESUMEN

Recently, a system that measures tissue oxygen tension using time-resolved luminescence-based optical sensors has become available commercially (Oxford Optronix, Oxford, England). Two experiments were conducted using this system. First, the oxygen tension distribution was measured in two tumor lines: a spontaneous mouse fibrosarcoma, FSa-II, and a human squamous cell carcinoma xenograft, FaDu. The area in which the pO(2) was equal to or lower than 2.5 mmHg was defined as the hypoxic lesion, and the hypoxic cell fraction was taken as the fraction of these measurements in a tumor. The measured hypoxic cell fractions were compared with those determined by the paired cell survival assay for tumors of various sizes. Second, the tumor tissue pO(2) was measured continuously after administration of two different anesthetics to evaluate the effect of these drugs on tissue pO(2). Results indicated a good agreement between the hypoxic cell fractions measured by this system and those determined by the paired cell survival curve assay for tumors smaller than approximately 500 mm(3). For tumors larger than approximately 500 mm(3), the hypoxic cell fractions measured by the oxygen probe system were higher than those measured by the paired cell survival assay. This may suggest that the hypoxic cell fraction measured by the oxygen probes included both hypoxic and necrotic areas in large tumors where necrotic lesions occupied a significant portion of the tumor. Continuous measurements of pO(2) after anesthesia (Nembutal, or ketamine plus xylazine) showed a consistent rise in the pO(2) during the first 20-30 min of measurement. Subsequently, the pO(2) values became constant or continued to rise slowly. For comparison, the tumor cell survivals were assayed after a dose of 20 Gy given in air at 5, 20 and 60 min after anesthesia. The result showed a decrease in cell survival only in tumors irradiated 20 min after an injection of Nembutal.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Fibrosarcoma/metabolismo , Oxígeno/análisis , Animales , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Hipoxia de la Célula/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Fibrosarcoma/patología , Fibrosarcoma/radioterapia , Humanos , Ketamina/farmacología , Ratones , Ratones Endogámicos C3H , Presión Parcial , Factores de Tiempo , Células Tumorales Cultivadas
9.
Int J Radiat Biol ; 88(7): 515-22, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22617044

RESUMEN

PURPOSE: To study the effect of DN (dominant-negative) Ku70 and reoxygenation on the hypoxia-induced cell-kill. MATERIALS AND METHODS: Cell lines were human colorectal carcinoma HCT8 and HT29 cells and their respective derivatives, v-HCT8 and v-HT29 infected with DNKu70-containing adenovirus. Cells were plated in glass tubes and made hypoxic by flushing N(2) gas containing 0, 0.1 or 0.5% O(2). Cell survival was determined by colony formation assay immediately after 0-96 h hypoxia. To reoxygenate medium were replaced fresh following 48 or 72 h in hypoxia and cells were incubated in aerobic environment for 2-24 h before survival assay. RESULTS: When incubated in hypoxia, cells lost reproductive capability ∼ exponentially as a function of time in hypoxia, and depending on the O(2) concentration. DNKu70 rendered cells more prone to hypoxia-induced cell-kill. Following reoxygenation cell survival increased rapidly but without detectable cell proliferation during first 24 hours. This evinced hypoxia-induced potentially lethal damage (PLD) that was repairable upon reoxygenation. DNKu70 did not significantly inhibit this repair. CONCLUSION: Hypoxia-induced cell lethality was facilitated by DNKu70, but substantially repaired upon reoxygenation. This may have negative impact on the effect of reoxygenation in cancer therapy.


Asunto(s)
Antígenos Nucleares/metabolismo , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/fisiopatología , Consumo de Oxígeno , Oxígeno/metabolismo , Antígenos Nucleares/genética , Apoptosis , Hipoxia de la Célula , Proliferación Celular , Supervivencia Celular , Proteínas de Unión al ADN/genética , Células HT29 , Humanos , Autoantígeno Ku , Regulación hacia Arriba
10.
Int J Radiat Oncol Biol Phys ; 77(3): 877-85, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20510198

RESUMEN

PURPOSE: To investigate the effect of recombinant replication-defective adenovirus containing dominant-negative Ku70 fragment on the response of tumor cells to multiple small radiation doses. Our ultimate goal is to demonstrate the feasibility of using this virus in gene-radiotherapy to enhance the radiation response of tumor cells. METHODS AND MATERIALS: Human colorectal HCT8 and HT29 carcinoma cells were plated in glass tubes, infected with virus (25 multiplicity of infection), and irradiated with a single dose or zero to five doses of 3 Gy each at 6-h intervals. Hypoxia was induced by flushing with 100% nitrogen gas. The cells were trypsinized 0 or 6 h after the final irradiation, and cell survival was determined by colony formation. The survival data were fitted to linear-quadratic model or exponential line. RESULTS: Virus infection enhanced the radiation response of the HCT8 and HT29 cells. The virus enhancement ratio for single-dose irradiation at a surviving fraction of 0.1 was approximately 1.3 for oxic and hypoxic HCT8 and 1.4 and 1.1 for oxic and hypoxic HT29, respectively. A similar virus enhancement ratio of 1.2-1.3 was observed for both oxic and hypoxic cells irradiated with multiple doses; however, these values were smaller than the values found for dominant-negative Ku70-transfected Rat-1 cells. This difference has been discussed. The oxygen enhancement ratio for HCT8 and HT29 receiving fractionated doses was 1.2 and 2.0, respectively, and virus infection altered them slightly. CONCLUSION: Infection of recombinant replication-defective adenovirus containing dominant-negative Ku70 fragment enhanced the response of human colorectal cancer cells to single and multiple radiation doses.


Asunto(s)
Adenoviridae/fisiología , Antígenos Nucleares/metabolismo , Neoplasias Colorrectales , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Virus Defectuosos/fisiología , Tolerancia a Radiación/fisiología , Adenoviridae/genética , Animales , Antígenos Nucleares/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral/efectos de la radiación , Línea Celular Tumoral/virología , Supervivencia Celular , Ensayo de Unidades Formadoras de Colonias/métodos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/radioterapia , Neoplasias Colorrectales/virología , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Virus Defectuosos/genética , Terapia Genética/métodos , Células HT29/efectos de la radiación , Células HT29/virología , Humanos , Autoantígeno Ku , Modelos Lineales , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Dosis de Radiación , Ratas , Transfección/métodos
11.
Radiother Oncol ; 92(3): 345-52, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19433338

RESUMEN

PURPOSE: To test the hypothesis that, with 5-fluorocytosine (5-FC) treatment, the co-expression of cytosine deaminase (CD) and uracil phosphoribosyltransferase (UPRT) can lead to greater radiosensitization and bystander effect than CD-expression alone. METHODS AND MATERIALS: R3327-AT cell lines stably expressing CD or CDUPRT were generated. The 5-FC and 5-FU cytotoxicity, and the radiosensitivity with/without 5-FC treatment, of these cells were evaluated under both aerobic and hypoxic conditions. The bystander effect was assessed by apoptosis staining and clonogenic survival. The pharmacokinetics of 5-FU and 5-FC metabolism was monitored in mice bearing CD- or CDUPRT-expressing tumors using 19F MR spectroscopy (MRS). RESULTS: CDUPRT-expressing cells were more sensitive to 5-FC and 5-FU than CD-expressing cells. CDUPRT-expression further enhanced the radiosensitizing effect of 5-FC, relative to that achieved by CD-expression alone. A 25-fold lower dose of 5-FC resulted in the same magnitude of radiosensitization in CDUPRT-expressing cells, relative to that in CD-expressing cells. The 5-FC cytotoxicity in co-cultures of parental cells mixed with 10-20% CDUPRT cells was similar to that in 100% CDUPRT cells. 19F MRS measurements showed that expression of CDUPRT leads to enhanced accumulation of fluorine nucleotide (FNuc), relative to that associated with CD-expression alone. CONCLUSION: Our study suggests that CDUPRT/5-FC strategy may be more effective than CD/5-FC, especially when used in combination with radiation.


Asunto(s)
Efecto Espectador/genética , Flucitosina/farmacología , Pentosiltransferasa/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/radioterapia , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Animales , Western Blotting , Línea Celular Tumoral/efectos de los fármacos , Técnicas de Cocultivo , Citosina Desaminasa/efectos de los fármacos , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Modelos Animales de Enfermedad , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Desnudos , Pentosiltransferasa/efectos de los fármacos , Pentosiltransferasa/metabolismo , Probabilidad , Neoplasias de la Próstata/patología , Valores de Referencia , Transfección
12.
J Surg Oncol ; 88(1): 14-20, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15384091

RESUMEN

BACKGROUND: Hyperthermia enhances the cytotoxicity of some chemotherapeutic agents and recent studies suggest that docetaxel may show improved response at elevated temperatures. Factors that may modify the thermal enhancement of docetaxel were studied to optimize its clinical use with hyperthermia. METHODS: The tumor studied was an early-generation isotransplant of a spontaneous C3Hf/Sed mouse fibrosarcoma, Fsa-II. All studies were approved by the Animal Care and Use Committee. Docetaxel was given as a single intraperitoneal injection. Hyperthermia was achieved by immersing the tumor-bearing foot into a constant temperature water bath. Four factors were studied: duration of hyperthermia, sequencing of hyperthermia with docetaxel, intensity of hyperthermia, and tumor size. To study duration of hyperthermia tumors were treated at 41.5 degrees C for 30 or 90 min immediately after intraperitoneal administration of docetaxel. For sequencing of hyperthermia and docetaxel, animals received hyperthermia treatment of tumors for 30 min at 41.5 degrees C immediately after drug administration, hyperthermia both immediately and 3 hr after docetaxel administration and hyperthermia given only at 3 hr after administration of docetaxel. Intensity of hyperthermia was studied using heat treatment of tumors for 30 min at 41.5 or 43.5 degrees C immediately following docetaxel administration. Effect of tumor size was studied by delaying experiments until three times the tumor volume (113 mm(3)) was observed. Treatment of tumors lasted for 30 min at 41.5 degrees C immediately following drug administration. Tumor response was studied using the mean tumor growth time. RESULTS: Hyperthermia in the absence of docetaxel had a small but significant effect on tumor growth time at 43.5 degrees C but not at 41.5 degrees C. Hyperthermia at 41.5 degrees C for 90 min immediately after docetaxel administration significantly increased mean tumor growth time (P = 0.0435) when compared to tumors treated with docetaxel at room temperature. Treatment for 30 min had no effect. Application of hyperthermia immediately and immediately plus 3 hr following docetaxel was effective in delaying tumor growth. Treatment at 3 hr only had no effect. No significant difference in mean tumor growth time was observed with docetaxel and one half hour of hyperthermia at 41.5 or 43.5 degrees C. For larger tumors, hyperthermia alone caused a significant delay in tumor growth time. Docetaxel at 41.5 degrees C for 30 min did not significantly increase mean tumor growth time compared to large tumors treated with docetaxel at room temperature. CONCLUSIONS: Docetaxel shows a moderate increase in anti-tumor activity with hyperthermia. At 41.5 degrees C the thermal enhancement of docetaxel is time dependent if hyperthermia is applied immediately following drug administration. With large tumors docetaxel alone or docetaxel plus hyperthemia showed the greatest delays in tumor growth time in the experiments.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Fibrosarcoma/terapia , Hipertermia Inducida , Taxoides/administración & dosificación , Animales , Modelos Animales de Enfermedad , Docetaxel , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C3H
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda