Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Therm Biol ; 115: 103619, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37437370

RESUMEN

INTRODUCTION: Irreversible electroporation (IRE) is an ablation modality that applies short, high-voltage electric pulses to unresectable cancers. Although considered a non-thermal technique, temperatures do increase during IRE. This temperature rise sensitizes tumor cells for electroporation as well as inducing partial direct thermal ablation. AIM: To evaluate the extent to which mild and moderate hyperthermia enhance electroporation effects, and to establish and validate in a pilot study cell viability models (CVM) as function of both electroporation parameters and temperature in a relevant pancreatic cancer cell line. METHODS: Several IRE-protocols were applied at different well-controlled temperature levels (37 °C ≤ T ≤ 46 °C) to evaluate temperature dependent cell viability at enhanced temperatures in comparison to cell viability at T = 37 °C. A realistic sigmoid CVM function was used based on thermal damage probability with Arrhenius Equation and cumulative equivalent minutes at 43 °C (CEM43°C) as arguments, and fitted to the experimental data using "Non-linear-least-squares"-analysis. RESULTS: Mild (40 °C) and moderate (46 °C) hyperthermic temperatures boosted cell ablation with up to 30% and 95%, respectively, mainly around the IRE threshold Eth,50% electric-field strength that results in 50% cell viability. The CVM was successfully fitted to the experimental data. CONCLUSION: Both mild- and moderate hyperthermia significantly boost the electroporation effect at electric-field strengths neighboring Eth,50%. Inclusion of temperature in the newly developed CVM correctly predicted both temperature-dependent cell viability and thermal ablation for pancreatic cancer cells exposed to a relevant range of electric-field strengths/pulse parameters and mild moderate hyperthermic temperatures.


Asunto(s)
Hipertermia Inducida , Neoplasias Pancreáticas , Humanos , Proyectos Piloto , Electroporación/métodos , Temperatura , Neoplasias Pancreáticas/terapia
2.
Sci Rep ; 12(1): 7017, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35488115

RESUMEN

Hyperthermic intravesical chemotherapy (HIVEC)-whereby the bladder is heated to ± 43 °C during a chemotherapy instillation-can improve outcomes of non-muscle invasive bladder cancer (NMIBC) treatments. Experiments in animal models are required to explore new hyperthermia based treatments. Existing HIVEC devices are not suitable for rodents or large-scale animal trials. We present a HIVEC setup compatible with orthotopic rat models. An externally heated chemotherapeutic solution is circulated in the bladder through a double-lumen catheter with flow rates controlled using a peristaltic pump. Temperature sensors in the inflow channel, bladder and outflow channel allow temperature monitoring and adjustments in real-time. At a constant flow rate of 2.5 mL/min the system rapidly reaches the desired bladder temperature of 42-43 °C with minimal variability throughout a one-hour treatment in a rat bladder phantom, as well as in euthanised and live rats. Mean intraluminal bladder temperatures were 42.92 °C (SD = 0.15 °C), 42.45 °C (SD = 0.37 °C) and 42.52 °C (SD = 0.09 °C) in the bladder phantom, euthanised, and live rats respectively. Thermal camera measurements showed homogenous heat distributions over the bladder wall. The setup provides well-controlled thermal dose and the upscaling needed for performing large scale HIVEC experiments in rats.


Asunto(s)
Hipertermia Inducida , Neoplasias de la Vejiga Urinaria , Administración Intravesical , Animales , Femenino , Calor , Humanos , Masculino , Ratas , Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
3.
J Clin Transl Res ; 6(6): 225-235, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33564727

RESUMEN

BACKGROUND/AIM/OBJECTIVE: Late side effects of radiotherapy (RT) in the treatment for head and neck (HN) malignancies involve an inadequate healing response of the distressed tissue due to RT-induced hypovascularity. The aim of this study was to develop a pilot model in which vascular alterations associated with the onset of late irradiation (IR) injury could be measured in rabbit oral mucosa and mandibular bone. MATERIALS AND METHODS: Eight male New Zealand white rabbits were divided over four treatment groups. Group I-III received four fractions of RT (5.6 Gy, 6.5 Gy, and 8 Gy, respectively) and Group IV received 1 fraction of 30 Gy. Oral microcirculatory measurements were performed at baseline (before RT) and once a week during 11 consecutive weeks after RT assessing perfusion parameters, that is, total vessel density (TVD), perfused vessel density (PVD), proportion of perfused vessels (PPV), and microvascular flow index (MFI). Post-mortem histopathology specimens were analyzed. RESULTS: Five weeks after RT, TVD, and PVD in all groups showed a decrease of >10% compared to baseline, a significant difference was observed for Groups I, II, and IV (P<0.05). At T11, no lasting effect of decreased vessel density was observed. PPV and MFI remained unaltered at all-time points. Group IV showed a marked difference in scattered telangiectasia such as microangiopathies, histological necrosis, and loss of vasculature. CONCLUSION: No significant lasting effect in mucosal microcirculation density due to IR damage was detected. Observed changes in microcirculation vasculature and histology may align preliminary tissue transition towards clinical pathology in a very early state associated with late IR injury in the oral compartment. RELEVANCE FOR PATIENTS: Enhancing knowledge on the onset of late vascular IR injury in the HN region could help the development, monitoring, and timing of therapies that act on prevention, discontinuation, or repair of radiation pathology.

4.
Oncogene ; 35(46): 6026-6037, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27157610

RESUMEN

Colorectal cancer (CRC) is a heterogeneous disease posing a challenge for accurate classification and treatment of this malignancy. There is no common genetic molecular feature that would allow for the identification of patients at risk for developing recurrences and thus selecting patients who would benefit from more stringent therapies still poses a major clinical challenge. Recently, an international multicenter consortium (CRC Subtyping Consortium) was established aiming at the classification of CRC patients in biologically homogeneous CRC subtypes. Four consensus molecular subtypes (CMSs) were identified, of which the mesenchymal CMS4 presented with worse prognosis signifying the importance of identifying these patients. Despite the large number of samples analyzed and their clear association with unifying biological programs and clinical features, single-driver mutations could not be identified and patients are heterogeneous with regard to currently used clinical markers. We therefore set out to define the regulatory mechanisms underlying the distinct gene expression profiles using a network-based approach involving multiple molecular modalities such as gene expression, methylation levels and microRNA (miR) expression. The miR-200 family presented as the most powerful determinant of CMS4-specific gene expression, tuning the majority of genes differentially expressed in the poor prognosis subtype, including genes associated with the epithelial-mesenchymal transition program. Furthermore, our data show that two epigenetic marks, namely the methylation of the two miR-200 promoter regions, can identify tumors belonging to the mesenchymal subtype and is predictive of disease-free survival in CRC patients. Importantly, epigenetic silencing of the miR-200 family is also detected in epithelial CRC cell lines that belong to the mesenchymal CMS. We thus show that determining regulatory networks is a powerful strategy to define drivers of distinct cancer subtypes, which possess the ability to identify subtype affiliation and to shed light on biological behavior.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , MicroARNs/genética , Línea Celular Tumoral , Neoplasias Colorrectales/mortalidad , Biología Computacional/métodos , Metilación de ADN , Epigénesis Genética , Femenino , Humanos , Masculino , Familia de Multigenes , Fenotipo , Pronóstico , Regiones Promotoras Genéticas , Transcriptoma
5.
Oncogene ; 32(12): 1539-48, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22614016

RESUMEN

Glioblastoma multiforme (GBM) is a devastating disease with high mortality and poor prognosis. Cancer stem cells (CSCs) have recently been defined as a fraction of tumor cells highly resistant to therapy and subsequently considered to be responsible for tumor recurrence. These cells have been characterized in GBM and suggested to reside in and be supported by the tumor microvascular niche. Here we evaluated the response of tumor microvascular endothelial cells (tMVECs) to radio- and chemotherapy, and analyzed how this affects their interaction with CSCs. Our data demonstrate that tMVECs exhibit extreme resistance to both therapies, with the main response to irradiation being senescence. Importantly, senescent tMVECs can be detected in human GBM samples as well as in mice upon irradiation. Even though permanently arrested, they are still viable and able to support CSC growth with the same efficacy as non-senescent tMVECs. Intriguingly, GBM CSCs themselves are capable of differentiating into cells with similar features as tMVECs that subsequently undergo senescence when exposed to radiation. This indicates that endothelial-like cells are therapy resistant and, more importantly, support expansion of GBM cells.


Asunto(s)
Neoplasias Encefálicas/terapia , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Glioblastoma/terapia , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Femenino , Glioblastoma/irrigación sanguínea , Glioblastoma/patología , Humanos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/efectos de la radiación , Insuficiencia del Tratamiento
6.
Cell Death Differ ; 19(11): 1826-35, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22705846

RESUMEN

The tumor necrosis factor (TNF) family member APRIL (A proliferation inducing ligand) is a disease promoter in B-cell malignancies. APRIL has also been associated with a wide range of solid malignancies, including colorectal cancer (CRC). As evidence for a supportive role of APRIL in solid tumor formation was still lacking, we studied the involvement of APRIL in CRC. We observed that ectopic APRIL expression exacerbates the number and size of adenomas in Apc(Min) mice and in a mouse model for colitis-associated colon carcinogenesis. Furthermore, knockdown of APRIL in primary spheroid cultures of colon cancer cells and both mouse and human CRC cell lines reduced tumor clonogenicity and in vivo outgrowth. Taken together, our data therefore indicate that both tumor-derived APRIL and APRIL produced by non-tumor cells is supportive in colorectal tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Colorrectales/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Modelos Animales de Enfermedad , Humanos , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Células Tumorales Cultivadas , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
7.
Int J Hyperthermia ; 25(3): 220-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19212861

RESUMEN

The analysis of chromosomal aberrations by premature chromosome condensation (PCC) induced by Calyculin A (Cal) is feasible in tumor biopsies from patients and has the potential to predict sensitivity to radiotherapy. As hyperthermia (HT) improves radiotherapy outcome in certain tumor sites, it was investigated whether PCC induction is still possible after temperatures reached in the clinic. Human cervical carcinoma (CaSki) and lung carcinoma (SW-1573) cells were incubated with Cal to induce PCC immediately after 1 h treatment at temperatures ranging from 41 degrees C to 43 degrees C and after recovery for up to 24 h after treatment with 43 degrees C. Levels of phosphorylated Cdc2 (at the Tyr15 residue), histone H3 (at the Ser10 residue) and Cyclin B1 were investigated by immunoblotting. The amount of cells positive for phosphorylated histone H3 was determined by flow cytometry. Temperatures > or =42.5 degrees C inhibited the induction of PCC by Cal, while recovery of PCC-induction was observed at >20 h after treatment in both cell lines. The phosphorylation status of Cdc2 as well as of histone H3 in cells treated with Cal directly after HT at 43 degrees C was similar to that of cells treated with Cal alone or treated with Cal 24 h after HT at 43 degrees C. HT alone did not affect the levels of phosphorylated Cdc2, while phosphorylation levels of histone H3 were increased as compared with control status of these two proteins. Phosphorylated and total Cyclin B1 levels were not influenced by any of the treatments. Flow cytometric analysis confirmed that HT at 43 degrees C did not interfere with phosphorylation of histone H3. Our data indicate that HT transiently inhibits PCC induction by Cal in a temperature-dependent manner. Therefore, an interval of at least 24 h after HT should be applied before taking tumor biopsies for karyogram analysis of patients treated with temperatures above 42.5 degrees C.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , Aberraciones Cromosómicas/inducido químicamente , Hipertermia Inducida , Oxazoles/farmacología , Proteína Quinasa CDC2 , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Ciclina B/metabolismo , Ciclina B1 , Quinasas Ciclina-Dependientes , Femenino , Fiebre/metabolismo , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Toxinas Marinas , Oxazoles/antagonistas & inhibidores , Fosforilación , Neoplasias del Cuello Uterino/metabolismo
8.
Thyroid ; 16(4): 387-95, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16646686

RESUMEN

Radiation to the head-neck region may damage the thyroid gland, leading to hypothyroidism or thyroid carcinoma. Outcomes of radiation protection by lowering plasma thyroid-stimulating hormone (TSH) have thus far been ambiguous. Our aim was to evaluate the radioprotective effect of inhibiting the thyroid gland's activity during x-radiation. For this purpose, of 80 5-week old Wistar rats, 64 received cervical irradiation with 15 Gy (single dose). During irradiation, endocrine intervention was done, using thyroxine (T(4)), T(4) plus iodine, or iodine alone compared to placebo. During the endocrine interventions and follow-up, TSH and T(4) concentrations were measured periodically. Histologic examination of thyroid, pituitary gland, or the hypothalamus and any suspect lymph nodes, lungs, and liver was performed after 6 and 54 weeks. It was found that during the endocrine intervention, plasma levels of TSH were lower in rats given T(4) and higher in rats given iodine. After 6 and 54 weeks, no significant reduction in hypothyroidism or thyroid carcinoma was found between the different groups of rats given any endocrine intervention or no intervention. In conclusion, the administration of T(4), iodine or the combination during x-irradiation does not protect against radiation-induced thyroid damage.


Asunto(s)
Protección Radiológica/métodos , Glándula Tiroides/efectos de la radiación , Tirotropina/antagonistas & inhibidores , Animales , Combinación de Medicamentos , Estudios de Seguimiento , Masculino , Neoplasias Inducidas por Radiación/prevención & control , Ratas , Ratas Wistar , Yoduro de Sodio/uso terapéutico , Glándula Tiroides/patología , Glándula Tiroides/fisiología , Neoplasias de la Tiroides/prevención & control , Tirotropina/sangre , Tiroxina/sangre , Tiroxina/uso terapéutico
9.
J Neurooncol ; 74(2): 99-103, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16193379

RESUMEN

OBJECTIVE: Postoperative radiotherapy is standard treatment for patients with a glioblastoma multiforme (GBM). However, a GBM is radioresistant and almost always recurs, even after a high dose of radiation. A GBM is characterized by its extensive neo-angiogenesis, which can be attributed to the high levels of vascular endothelial growth factor (VEGF). The scope of this study is to investigate the VEGF secretion by GBM cells with different radiosensitivity after irradiation. METHODS: Three human GBM cell lines (U251, U251-NG2 and U87) were irradiated with single doses of 0, 5, 10 and 20 Gy of gamma-rays from a (137)Cs source. VEGF levels in medium were measured by ELISA at 24, 48 and 72 h after radiation. Cell survival was measured by the XTT assay 7 days after irradiation. RESULTS: Following single dose radiation, the VEGF levels showed a dose dependent increase in U251, U251-NG2 and U87 glioma cells. Both base-line and radiation-enhanced VEGF levels were about 10-fold higher in U87 compared to U251 and U251-NG2 cells. In addition, in the XTT assay, the U87 was more radioresistant than both U251 and U251-NG2 cell lines (dose modifying factor (DMF) = 1.6 and 1.7 resp). CONCLUSION: Irradiation enhanced VEGF secretion in all three tested glioma cell lines (up to eight times basal levels). It is tempting to associate the radiation-enhanced VEGF secretion with an increased angiogenic potential of the tumor, which may be a factor in radioresistance.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , Tolerancia a Radiación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias Encefálicas/metabolismo , Proliferación Celular/efectos de la radiación , Radioisótopos de Cesio , Relación Dosis-Respuesta en la Radiación , Glioblastoma/metabolismo , Humanos , Radiación Ionizante , Células Tumorales Cultivadas
10.
J Endocrinol Invest ; 28(3): 261-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15952412

RESUMEN

Because radiotherapy in the head and neck region is necessary in the treatment of childhood cancer, possibilities to prevent damage to the thyroid gland must be explored. We developed a model in which radiation-induced effects can be investigated in a way that these effects can be quantified, using thyroid dysmorphology and plasma TSH. Thirty-five Wistar rats, 5 weeks old, were X-irradiated on the cervical region, with a single dose varying from 0 to 20 Gy. After 6 weeks, TSH, T4 and T3 were determined, and thyroid glands were processed for histological examination by two independent pathologists. A histological classification scale was developed, using follicular size, colloid density and cell height of thyrocytes to measure hyperplasia and hypertrophy. By the sum of these scores, a cell-activity index was calculated, which was related to plasma TSH concentration. Numbers of PAS-positive droplets and epithelial desquamation were also counted. Inter-observer reliability was assessed. Good to very good reliability was found for scores of follicular size, colloid density and cell height. Significant increase of cell-activity index was found after 10, 15 and 20 Gy. The plasma TSH concentration was positively correlated to the cell-activity index, increasing with radiation-doses up to 15 Gy. The number of desquamated cells was significantly increased after radiation doses >10 Gy, with moderate reliability. In conclusion, this model using cell-activity index of thyrocytes together with plasma thyrotropin concentrations and desquamation of cells can be used for interpretation and future (pre-clinical) studies of prevention of radiation-induced thyroid damage.


Asunto(s)
Plexo Cervical/efectos de la radiación , Traumatismos Experimentales por Radiación/patología , Glándula Tiroides/citología , Glándula Tiroides/efectos de la radiación , Tirotropina/sangre , Animales , Masculino , Traumatismos Experimentales por Radiación/sangre , Ratas , Ratas Wistar , Pruebas de Función de la Tiroides , Glándula Tiroides/patología , Tiroxina/sangre , Triyodotironina/sangre
11.
J Radiat Res ; 45(3): 385-91, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15613783

RESUMEN

The role of DNA repair mechanisms in the cellular response to low dose rate (LDR) irradiation was studied with the aim to gain insight in the process of sublethal damage (SLD) repair. Chinese hamster cell lines mutated in either DNA single strand break (ssb) repair or DNA double strand break (dsb) repair by non homologous end joining (NHEJ) and homologous recombination (HR), or showing an AT-like phenotype, were irradiated in plateau-phase either at high dose rate (HDR, 3.3 Gy/min) or at pulsed low dose rate (p-LDR, average 1 Gy/h). Cell survival after irradiation was assessed using the clonogenic assay. A change in sensitivity when the dose rate was decreased was observed for all parental cell lines and the DNA ssb repair mutant. No difference in cell survival after p-LDR versus. HDR irradiation was observed for the two NHEJ mutants, the AT-like mutant and the HR mutant. Based on these results we conclude that single strand break repair does not play a role in the dose rate effect. The AT like protein, functional NHEJ and XRCC3 are required for the dose rate effect.


Asunto(s)
Células CHO/fisiología , Células CHO/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Mutación , Tolerancia a Radiación/genética , Animales , Células CHO/citología , Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Cricetinae , Cricetulus , ADN , Daño del ADN , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , ADN de Cadena Simple
12.
Oncol Rep ; 12(1): 187-92, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15201982

RESUMEN

Five mutant Chinese hamster cell lines deficient in DNA repair with the corresponding parental cell lines were used to determine their sensitivity to cisplatin, 5-fluorouracil and gemcitabine. The mutations in the cell lines led to defective single strand break repair (EM-C11), defective recombination mediated repair (irs1SF), defective double strand break repair (XR-V15B, a Ku-80 mutant and CR-C1, a DNA-PKcs mutant) and an AT-like mutation (VC-4). All mutant cell lines had an impaired doubling time during exponential growth and an increased sensitivity to X-irradiation. We may conclude that for cisplatin-induced cytotoxicity the homologous recombination-associated DNA repair plays an important role in the repair of the cisplatin induced lesions, confirming previous results. In 5-FU and gemcitabine induced toxicity to cells, repair processes involved with radiation-induced damage were not implicated. This is in striking contrast to the role of cisplatin in radiosensitization where inhibition of the NHEJ pathway is implicated, and to the role of gemcitabine in sensitization where specific interference with the HR pathway is implicated.


Asunto(s)
Cisplatino/toxicidad , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Desoxicitidina/análogos & derivados , Desoxicitidina/toxicidad , Fluorouracilo/toxicidad , Animales , Antimetabolitos Antineoplásicos/toxicidad , Células CHO , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Línea Celular , Cricetinae , ADN/genética , Relación Dosis-Respuesta a Droga , Rayos X , Gemcitabina
13.
Radiat Res ; 161(5): 511-6, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15161373

RESUMEN

The functionality of G(1)-phase arrest was investigated in relation to repair of potentially lethal damage (PLD) in human glioblastoma Gli-06 cells. Confluent cultures were irradiated and plated for clonogenic survival either immediately or 24 h after gamma irradiation. Bivariate flow cytometry was performed to assess the distribution over the cell cycle. Levels of TP53 and CDKN1A protein were assessed with Western blotting and levels of CDKN1A mRNA with RT-PCR. Confluence significantly reduced the number of proliferating cells. Marked PLD repair was found in the absence of an intact G(1) arrest. No accumulation of TP53 was observed, and the protein was smaller than the wild-type TP53 of RKO cells. No increased expression of CDKN1A at the mRNA or protein levels was found in Gli-06 cells. The TP53 of Gli-06 cells was unable to transactivate the CDKN1A gene. From this study, it is evident that PLD repair may be present without a functional TP53 or G(1) arrest.


Asunto(s)
Apoptosis/efectos de la radiación , Daño del ADN , Reparación del ADN/efectos de la radiación , ADN/efectos de la radiación , Glioblastoma/metabolismo , Glioblastoma/patología , Proteína p53 Supresora de Tumor/metabolismo , División Celular/efectos de la radiación , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Humanos , Dosis de Radiación
14.
Int J Hyperthermia ; 19(1): 45-57, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12519711

RESUMEN

The rat bladder was heated using a microwave applicator which was equipped with a system of circulating deionized water. The applicator was operated at 434 MHz and was placed at the ventral side with the rats in supine position. Temperatures in the bladder and adjacent were monitored using thermocouples with single or multiple sensors. One thermocouple located most centrally in the bladder served as reference. The rats were treated at intravesical reference temperature of 41, 42, 43, 44 and 45 degrees C for 1 h. The heating time to reach the reference temperature was approximately 5 min. Temperatures inside the bladder varied within 0.5 degrees C from the reference value, while the temperatures in the urethra were approximately 1.0 degrees C lower. At the left and the right side of the outer bladder wall, temperatures were approximately 0.5 degrees C lower than the reference value, while the temperature on the dorsal and ventral sides of the bladder wall were 1.0-1.5 degrees C lower. In the rectum, located in the treatment field, the temperature reached 39.1, 40.5, 42.4 and 42.5 degrees C after 1 h of hyperthermia at intravesical reference temperatures of 41, 42, 43, 44 and 45 degrees C, respectively. Body core temperature measured in the esophagus behind the pericardium never exceeded 40.0 degrees C. The capacity of the bladder was assessed after 1 h at 43, 44 and 45 degrees C at various intervals after heat treatment. In the sham treated control group and in the animals treated at 43 degrees C, no reduction in bladder capacity was observed. The treatment group where the bladder was kept at 44 degrees C for 1 h showed a clear reduction in bladder capacity at days 1 and 3 after hyperthermia. In the 45 degrees C treatment group, four out of seven rats died, this within a few days after treatment. The three surviving rats were tested for bladder capacity and all had a reduced capacity at days 3 and 7 post-treatment. Four weeks after 44 degrees C hyperthermia, all rats had recovered. After hyperthermia, depending on the heat-dose, an increase in blood urea nitrogen (BUN) was observed. After treatment at 42, 43 and 44 degrees C, peak values were observed after approximately 1 day (16 or 24 h) followed by recovery; after 42 degrees C, BUN levels were almost back to normal after 1 week; after 43 degrees C, the level was still twice as high as control levels; and after 44 degrees C, recovery of BUN levels to normal seemed slow, 1 week after treatment it was still five times as high as control. From these results, it is concluded that temperatures in the bladder below 44 degrees C are well tolerated. After 1 h at 44 degrees C, a transient decrease in bladder capacity was observed, as well as a high level of azotemia. After 1 h at 45 degrees C, a high mortality rate was observed. These observations agree with early clinical observations and may be used as guidelines for further clinical work.


Asunto(s)
Hipertermia Inducida/efectos adversos , Vejiga Urinaria/fisiopatología , Vejiga Urinaria/efectos de la radiación , Micción , Animales , Nitrógeno de la Urea Sanguínea , Temperatura Corporal , Cistitis/etiología , Femenino , Microondas/efectos adversos , Ratas , Ratas Endogámicas BN , Termografía
15.
J Radiat Res ; 42(2): 179-90, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11599884

RESUMEN

Sensitization by bromodeoxyuridine (BrdUrd) and hyperthermia (HT) on cell reproductive death induced by ionizing radiation was analyzed using the linear-quadratic [S(D)/S(0)=exp(-(alphaD + betaD2)]] model. Plateau-phase human lung tumor cells (SW-1573) and human colorectal carcinonoma cells (RKO) were treated with BrdUrd, radiation and HT. LQ-analysis was performed at iso-incubation dose and at iso-incorporation level of BrdUrd. and at iso-HT doses and iso-survival levels after HT. Clonogenic assays were performed 24 h after treatment to allow repair of potentially lethal damage (PLD). In SW cells BrdUrd. HT or the combination significantly increased the alpha-parameter (factor 2.0-5.7), without altering the beta-parameter. In RKO cells sensitization with BrdUrd increased both a (factor 1.4) and beta (factor 1.3) while HT only influenced beta (factor 2.1-4.0). The combination did not further increase the a and beta. The results indicate that BrdUrd has its main effect on the parameter alpha, dominant at clinically relevant radiation doses but that HT can affect both a and beta. The addition of BrdUrd and HT provides a method to enhance the efficacy of radiotherapy.


Asunto(s)
Bromodesoxiuridina/farmacología , Calor , Tolerancia a Radiación/fisiología , Fármacos Sensibilizantes a Radiaciones/farmacología , Muerte Celular , Humanos , Tolerancia a Radiación/efectos de los fármacos , Células Tumorales Cultivadas
16.
Oncol Rep ; 7(5): 937-40, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10948317

RESUMEN

Bromodeoxyuridine (BrdUrd)-induced radiosensitization of two different tumour cell lines was compared at equal levels of thymidine replacement. Human lung carcinoma cells (SW-1573) and human colorectal carcinoma cells (RKO) were grown for 48 h in the presence of respectively 1 microM BrdUrd and 4 microM of BrdUrd in order to obtain equal levels of BrdUrd into the DNA. In SW cells the level of thymidine replacement by BrdUrd was 6.7+/-0.5% and in RKO cells this was 7.1+/-0.8. Cell survival after irradiation with single doses up to 8 Gy, was determined with clonogenic assay. The magnitude of BrdUrd-induced radiosensitization was determined by analyzing radiation-dose survival curves with the linear-quadratic formula [S(D)/S(0)=exp-(alphaD+betaD2)]. In the SW cells BrdUrd radiosensitization led to a significant increase of the linear parameter, alpha, determining the initial slope of the survival curves, by a factor of about 2. In the RKO cells BrdUrd increased the value of alpha by a factor 1.4. This suggests that repair of potentially lethal damage (PLD) is inhibited. In both cell lines the quadratic term, beta, strongly influencing the high dose region of the survival curves, was not altered by sensitization by BrdUrd. The increase of alpha is of interest for clinical applications as BrdUrd sensitizes tumour cells after low doses of radiation.


Asunto(s)
Bromodesoxiuridina/farmacología , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Bromodesoxiuridina/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/radioterapia , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Clonales , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Neoplasias del Colon/radioterapia , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Humanos , Cinética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia , Fármacos Sensibilizantes a Radiaciones/metabolismo , Timidina/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación
17.
Int J Hyperthermia ; 15(6): 549-56, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10598951

RESUMEN

PURPOSE: To investigate the schedule-dependency of 2',2'difluorodeoxycytidine (dFdC, Gemcitabine) combined with hyperthermia (HT), in vitro as well as in vivo. MATERIALS AND METHODS: Rat R-1 rhabdomyosarcoma cells were treated with various concentrations of dFdC for 70 min, 4 h and 24 h. After various time intervals HT (60 min at 43 degrees C) was applied. Cell survival was determined by clonogenic assays. Female Wag/Rij rats bearing R-1 tumours on the hind limbs were treated with dFdC (20 mg/kg), with locally applied HT (60 min at 43 degrees C) or with a combined treatment using different time intervals (0, 24 and 48 h). Tumour growth delay (TGD) and normal tissue toxicity were assessed. RESULTS: With dFdC alone, significant cytotoxicity was observed after a 24 h-exposure. Except for the 24 h-exposure, HT reduced the cytotoxicity of dFdC in simultaneous applications. An enhanced cytotoxic effect was found when HT was applied 20 h after a 4 h-incubation with dFdC. In vivo, HT applied 48 h after dFdC-administration resulted in potentiation of the effect of dFdC with respect to TGD without an increase in toxicity. CONCLUSIONS: The efficacy of dFdC combined with HT is schedule-dependent both in vitro and in vivo. The addition of HT enhances the effectiveness of dFdC in the R-1 tumour model.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Desoxicitidina/análogos & derivados , Hipertermia Inducida , Rabdomiosarcoma/terapia , Animales , División Celular , Supervivencia Celular , Terapia Combinada , Desoxicitidina/efectos adversos , Desoxicitidina/uso terapéutico , Femenino , Hipertermia Inducida/efectos adversos , Ratas , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Gemcitabina
18.
Oncol Rep ; 5(3): 679-83, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9538175

RESUMEN

The levels of TNF, IL-1 and IL-6 in circulating blood of female WAG/Rij rats were assessed both after total-body irradiation (TBI) and localized irradiation of the right hind leg. The results show that enhanced levels of IL-1 in the circulation reflect a stress situation presumably resulting from handling and halothane anesthesia of the animal. Neither localized irradiation nor TBI resulted in further enhanced levels of IL-1. Both TBI and localized irradiation, lead to a small but significant increase in IL-6 levels in serum from circulating blood. After TBI this increase dissipated rapidly, 24 h after TBI increased levels are not found. After localized irradiation IL-6 levels remain elevated for a longer period. Still two weeks after irradiation, the longest time investigated, increased levels were observed. We did not observe increased TNF levels after localized irradiation or after TBI.


Asunto(s)
Miembro Posterior/efectos de la radiación , Interleucina-1/sangre , Interleucina-6/sangre , Ratas/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Irradiación Corporal Total , Animales , Línea Celular , Femenino , Miembro Posterior/metabolismo , Rayos X
19.
Oncol Rep ; 4(5): 1073-6, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-21590199

RESUMEN

To investigate radiosensitization by halogenated pyrimidines in human cells, SW-1573 lung tumor cells were grown in absence or presence of 4 mu M of iododeoxyuridine (IdUrd). Cell survival after irradiation with gamma-rays from a Cs-137 source with single doses up to 8 Gy, was determined by clonogenic assay, Radiosensitization by IdUrd was observed in both exponentially growing and plateau phase cells. Linear-quadratic analyses of the radiation survival curves demonstrated that radiosensitization was mainly expressed by an increase of the value of alpha (determining the initial slope of the survival curve) which can be interpreted as an enhancement of fixation of potentially lethal damage (PLD). The value of beta (determining the curvature of the survival curve), which might be attributed to enhanced interaction or to fixation of sublethal damage (SLD), was not influenced. In view of clinical applications of IdUrd it is of interest that radiosensitization has its main effect in the low dose region of the cell survival curve, described by the linear term alpha D.

20.
Int J Hyperthermia ; 12(6): 791-800, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8950159

RESUMEN

The levels of TNF, IL-1 and IL-6 in circulating blood female WAG/Ry rats were assessed in relation to treatment with localized hyperthermia of the right hind leg or with whole-body hyperthermia (WBH). After a localized treatment for 30 min at 43 or 44 degrees C no detectable increase in levels of IL-6 or TNF was obtained. Hyperthermia for 30 min at 45 degrees C led to an elevated level of IL-6 of 19.4 +/- 5.2 U/ml above the control level of 24 h after treatment. Levels of IL-1 were never higher than those in control animals that received only anaesthesia. Anaesthesia induced a peak level of approximately 131 U/ml IL-1 6 h after treatment. Serum levels of IL-1 and IL-6 are enhanced after WBH. IL-1 reaches a peak level already during WBH about 15 after reaching 41.5 degrees C. IL-6 levels were not enhanced during WBH but 1 h after WBH a clear peak was observed. Anaesthesia with sham WBH did not lead to enhanced IL-6 levels but enhanced IL-1 levels were clearly detected. We did not detect TNF in any sample after WBH. It is concluded from the present results that IL-6 is not induced by a 'standard' treatment of localized hyperthermia as used in oncotherapy (i.e. 60 min at 43 degrees C) to such a high level locally that this is reflected in increased levels in circulating blood. WBH at clinically relevant temperatures leads to enhanced levels of IL-1 and IL-6. The difference in IL-6 response after WBH or localized hyperthermia probably is related to the fact that in WBH also the bone marrow is treated. This may lead to stimulation of this important stem cell compartment of the peripheral blood. The sequence of appearance of IL-1 and IL-6 after hyperthermia is akin to the sequence in an inflammatory response. However, the experiments with sham treatment show that IL-1 may appear in the circulating blood not followed by IL-6. These results indicate that enhanced IL-1 levels may reflect a stress reaction of the animal related to the (sham) treatment. Enhanced levels of IL-1 after WBH correlate with enhanced levels of ACTH in the circulating blood.


Asunto(s)
Fiebre/sangre , Interleucina-1/sangre , Interleucina-6/sangre , Factor de Necrosis Tumoral alfa/análisis , Animales , Femenino , Hipertermia Inducida , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA