Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Mutat Res ; 688(1-2): 12-8, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20167225

RESUMEN

This study was designed to obtain a better insight into the relative contribution of homologous recombination (HR) and non-homologous end joining (NHEJ) to the repair of radiation-induced DNA double-strand breaks (DSBs) at first meiotic prophase. Early and late pachytene and early diplotene spermatocytes that had completed crossing over were sampled. We studied the kinetics of gamma-H2AX chromatin foci removal after irradiation of mice deficient for HR and mice deficient for NHEJ. Analyzing gamma-H2AX signals in unirradiated RAD54/RAD54B deficient spermatocytes indicated incomplete meiotic recombination repair due to the pronounced increase of gamma-H2AX foci in late prophase primary spermatocytes. In these mice, 8h after irradiation, early pachytene spermatocytes showed a reduction of the numbers of gamma-H2AX foci by 52% compared to 82% in the wild type, the difference being significant. However, after crossing over (in late pachytene and early diplotene), no effect of RAD54/RAD54B deficiency on the reduction of irradiation-induced foci was observed. In NHEJ deficient SCID mice, repair kinetics in early spermatocytes were similar to those in wild type mice. However, 1h after irradiation in late pachytene and early diplotene spermatocytes 1.7 times more foci were found than in wild type mice. This difference might be related to the absence of a DNA-PKcs dependent fast repair component in SCID mice. As subsequent repair is normal, HR likely is taking over. Taken together, the results obtained in RAD54/RAD54B deficient mice and in SCID mice indicate that DSB repair in early pachytene spermatocytes is mainly carried out through HR. In late spermatocytes (late pachytenes and early diplotenes) NHEJ is active. However, probably there is an interplay between these repair pathways and when in late spermatocytes the NHEJ pathway is compromised HR may take over.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Recombinación Genética , Espermatocitos/efectos de la radiación , Animales , ADN Helicasas , Histonas/genética , Masculino , Profase Meiótica I , Ratones , Ratones SCID , Proteínas Nucleares/genética
2.
Mutat Res ; 683(1-2): 84-90, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19887075

RESUMEN

The repair of DNA double strand breaks (DSBs) in male germ cells is slower and differently regulated compared to that in somatic cells. Round spermatids show DSB repair and are radioresistant to apoptosis induction. Mutation induction studies using ionizing irradiation, indicated a high frequency of chromosome aberrations (CA) in the next generation. Since they are in a G1 comparable stage of the cell cycle, haploid spermatids are expected to repair DSBs by the non-homologous end-joining pathway (NHEJ). However, immunohistochemical evidence indicates that not all components of the classical NHEJ pathway are available since the presence of DNA-PKcs cannot be shown. Here, we demonstrate that round spermatids, as well as most other types of male germ cells express both Parp1 and XRCC1. Therefore, we have determined whether the alternative Parp1/XRCC1 dependent NHEJ pathway is active in these nuclei and also have tested for classical NHEJ activity by a genetic method. To evaluate DSB repair in SCID mice, deficient for DNA-PKcs, and to study the involvement of the Parp1/XRCC1 dependent NHEJ pathway in round spermatids, the loss of gamma-H2AX foci after irradiation has been determined in nucleus spreads of round spermatids of SCID mice and in nucleus spreads and histological sections of Parp1-inhibited mice and their respective controls. Results show that around half of the breaks in randomly selected round spermatids are repaired between 1 and 8h after irradiation. The repair of 16% of the induced DSBs requires DNA-PKcs and 21% Parp1. Foci numbers in the Parp1-inhibited testes tend to be higher in spermatids of all epithelial stages reaching significance in stages I-III which indicates an active Parp1/XRCC1 pathway in round spermatids and a decreased repair capacity in later round spermatid stages. In Parp1-inhibited SCID mice only 14.5% of the breaks were repaired 8h after irradiation indicating additivity of the two NHEJ pathways in round spermatids.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Espermátides/metabolismo , Espermátides/efectos de la radiación , Animales , Western Blotting , Núcleo Celular/genética , Núcleo Celular/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Proteína Quinasa Activada por ADN/metabolismo , Técnica del Anticuerpo Fluorescente , Rayos gamma , Células Germinativas/efectos de la radiación , Histonas/metabolismo , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones SCID , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Espermatogénesis , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
3.
Int J Oncol ; 34(3): 813-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19212686

RESUMEN

Cyclopentenyl cytosine (CPEC), targetting the de novo biosynthesis of cytidine triphosphate (CTP), increases the cytotoxicity of gemcitabine (2',2'-difluoro-2'-deoxycytidine, dFdC) alone and in combination with irradiation in several human tumour cells in vitro. We investigated whether CPEC enhances the therapeutic ratio of gemcitabine and irradiation in human pancreatic BxPC-3 xenografts and in rat syngeneic L44 lung tumours. These models were selected because gemcitabine and radiation are used to treat both pancreatic and lung cancer patients and both models differ in growth capacity and in gemcitabine-induced radiosensitisation. A profound dose-dependent CTP-depletion was observed after a single injection of CPEC in both tumour tissue and in normal jejunum. In both models, CPEC alone induced a slight but significant tumour growth delay. The combination of CPEC with gemcitabine, at time intervals that showed CTP-depletion after CPEC, enhanced neither tumour growth delay nor toxicity as compared to gemcitabine alone. In addition, no beneficial effect of CPEC was observed in combination with gemcitabine and radiation. These results suggest that CPEC and gemcitabine alone as well as in combination with radiation target a similar cell population in both tumour models. In conclusion, future clinical development of CPEC as a modulator of gemcitabine combined with radiation is unlikely.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Citidina/análogos & derivados , Desoxicitidina/análogos & derivados , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Animales , Línea Celular Tumoral , Terapia Combinada , Citidina/farmacología , Citidina Trifosfato/biosíntesis , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad , Ratas , Ratas Endogámicas BN , Gemcitabina
4.
DNA Repair (Amst) ; 6(9): 1243-54, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17376750

RESUMEN

In male germ cells the repair of DNA double strand breaks (DSBs) differs from that described for somatic cell lines. Irradiation induced immunofluorescent foci (IRIF's) signifying a double strand DNA breaks, were followed in spermatogenic cells up to 16 h after the insult. Foci were characterised for Mdc1, 53BP1 and Rad51 that always were expressed in conjecture with gamma-H2AX. Subsequent spermatogenic cell types were found to have different repair proteins. In early germ cells up to the start of meiotic prophase, i.e. in spermatogonia and preleptotene spermatocytes, 53BP1 and Rad51 are available but no Mdc1 is expressed in these cells before and after irradiation. The latter might explain the radiosensitivity of spermatogonia. Spermatocytes from shortly after premeiotic S-phase till pachytene in epithelial stage IV/V express Mdc1 and Rad51 but no 53BP1 which has no role in recombination involved repair during the early meiotic prophase. Mdc1 is required during this period as in Mdc1 deficient mice all spermatocytes enter apoptosis in epithelial stage IV when they should start mid-pachytene phase of the meiotic prophase. From stage IV mid pachytene spermatocytes to round spermatids, Mdc1 and 53BP1 are expressed while Rad51 is no longer expressed in the haploid round spermatids. Quantifying foci numbers of gamma-H2AX, Mdc1 and 53BP1 at various time points after irradiation revealed a 70% reduction after 16 h in pachytene and diplotene spermatocytes and round spermatids. Although the DSB repair efficiency is higher then in spermatogonia where only a 40% reduction was found, it still does not compare to somatic cell lines where a 70% reduction occurs in 2 h. Taken together, DNA DSBs repair proteins differ for the various types of spermatogenic cells, no germ cell type possessing the complete set. This likely leads to a compromised efficiency relative to somatic cell lines. From the evolutionary point of view it may be an advantage when germ cells die from DNA damage rather than risk the acquisition of transmittable errors made during the repair process.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , ADN/efectos de la radiación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosfoproteínas/metabolismo , Espermatocitos/efectos de la radiación , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN , Relación Dosis-Respuesta en la Radiación , Técnica del Anticuerpo Fluorescente , Células Germinativas/metabolismo , Células Germinativas/efectos de la radiación , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Meiosis/fisiología , Ratones , Ratones Endogámicos , Ratones Noqueados , Recombinasa Rad51/metabolismo , Espermatogonias/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53 , Rayos X
5.
Int J Radiat Oncol Biol Phys ; 71(5): 1444-54, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18294781

RESUMEN

PURPOSE: To evaluate results of high-dose total-body irradiation (TBI) regimens for hematopoietic stem cell transplantation. METHODS AND MATERIALS: A total of 1,032 patients underwent TBI in one or two fractions before autologous or allogeneic hematologic stem cell transplantation for acute leukemia and non-Hodgkin's lymphoma. The TBI regimens were normalized by using the biological effective dose (BED) concept. The BED values were divided into three dose groups. Study end points were relapse incidence (RI), non-relapse mortality (NRM), relapse-free survival (RFS), and overall survival (OS). Multivariate analysis was performed, stratified by disease. RESULTS: In the highest TBI dose group, RI was significantly lower and NRM was higher vs. the lower dose groups. However, a significant influence on RFS and OS was not found. Relapses in the eye region were found only after shielding to very low doses. Age was of significant influence on OS, RFS, and NRM in favor of younger patients. The NRM of patients older than 40 years significantly increased, and OS decreased. There was no influence of age on RI. Men had better OS and RFS and lower NRM. Type of transplantation significantly influenced RI and NRM for patients with acute leukemia and non-Hodgkin's lymphoma. There was no influence on RFS and OS. CONCLUSIONS: Both RI and NRM were significantly influenced by the size of the BED of single-dose or two-fraction TBI regimens; OS and RFS were not. Age was of highly significant influence on NRM, but there was no influence of age on RI. Hyperfractionated TBI with a high BED might be useful, assuming NRM can be reduced.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia/terapia , Linfoma no Hodgkin/terapia , Irradiación Corporal Total , Enfermedad Aguda , Adolescente , Adulto , Factores de Edad , Análisis de Varianza , Supervivencia sin Enfermedad , Femenino , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Leucemia/mortalidad , Linfoma no Hodgkin/mortalidad , Masculino , Persona de Mediana Edad , Países Bajos , Protección Radiológica/métodos , Recurrencia , Efectividad Biológica Relativa , Factores Sexuales , Acondicionamiento Pretrasplante , Resultado del Tratamiento , Irradiación Corporal Total/mortalidad
6.
Anticancer Res ; 28(2A): 1129-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18507064

RESUMEN

BACKGROUND: Delay in the initiation of radiotherapy after surgery is associated with an increase in local regional recurrence. A possible mechanism might be that remaining tumor cells proliferate significantly faster as a result of induced angiogenic cytokines. The growth rate of tumors arising from the inoculation of L44 tumor cells in the wound bed after surgical removal of L44 tumors was determined. MATERIALS AND METHODS: L44 tumors growing in the flank of female BN rats were surgically removed. In the wound bed, 5x10(6) L44 cells, harvested from the in vitro cell line, were injected. L44 cells were also injected in the contralateral flank and in control rats with and without surgical intervention. Tumor volumes as a function of time after injection of cells were recorded. From the attained volume at day 7, the cell doubling time was calculated, assuming 10(9) cells per cm3. RESULTS: Tumors arising in the wound bed had the fastest growth rate as compared to the tumors in the contralateral flank or tumors in control rats with or without surgical intervention. CONCLUSION: The results clearly indicate accelerated tumor growth after surgical stress. This indicates that delay in the initiation of radiotherapy after surgery with tumor cells remaining, results in a larger tumor burden and hence a higher probability of local recurrence.


Asunto(s)
Recurrencia Local de Neoplasia , Neoplasias/cirugía , Estrés Fisiológico/complicaciones , Procedimientos Quirúrgicos Operativos/efectos adversos , Animales , Proliferación Celular , Femenino , Neoplasias/patología , Ratas , Carga Tumoral
7.
Int J Radiat Oncol Biol Phys ; 69(4): 1205-8, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17967309

RESUMEN

BACKGROUND: The aim of this study is to show the efficiency of keloidectomy and postoperative interstitial high-dose-rate (HDR) brachytherapy in the prevention of keloids. METHODS AND MATERIALS: Between 1998 and 2004, 35 patients with 54 keloids were treated postoperatively with HDR brachytherapy. The first HDR dose was applied within 6 hours after surgery, and two additional HDR doses were administered on the next day with a six-hour interval. The majority of patients received 6 Gy as the first dose postsurgery and two fractions of 4 Gy (38 keloids) on the next day. Seven keloids were treated postoperatively with three fractions of 6 Gy. The biologically effective dose (BED), derived from the linear quadratic concept, was applied to calculate the BED for the various radiation regimens. The keloid recurrence rates at specific BED values were compared with those derived for other fractionation schemes in the literature. RESULTS: Four recurrences/nonsatisfactory results out of nine treated keloids were observed after treatments with 1 x 4 Gy + 2 x 3 Gy. Only one recurrence out of 38 was found after 1 x 6 Gy + 2 x 4 Gy and none after 3 x 6 Gy. Better cosmetic results were found at the higher-dose schemes. CONCLUSION: The results of this study prove the effectiveness of HDR brachytherapy after keloidectomy provided that the total HDR dose is sufficient. Currently our scheme is 3 x 6 Gy.


Asunto(s)
Braquiterapia/métodos , Queloide/prevención & control , Complicaciones Posoperatorias/prevención & control , Adolescente , Adulto , Anciano , Femenino , Humanos , Radioisótopos de Iridio/uso terapéutico , Masculino , Persona de Mediana Edad , Dosificación Radioterapéutica , Recurrencia , Efectividad Biológica Relativa
8.
Int J Radiat Oncol Biol Phys ; 65(4): 1228-32, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16682132

RESUMEN

PURPOSE: Late complications related to total body irradiation (TBI) as part of the conditioning regimen for hematopoietic stem cell transplantation have been increasingly noted. We reviewed and compared the results of treatments with various TBI regimens and tried to derive a dose-effect relationship for the endpoint of late renal dysfunction. The aim was to find the tolerance dose for the kidney when TBI is performed. METHODS AND MATERIALS: A literature search was performed using PubMed for articles reporting late renal dysfunction. For intercomparison, the various TBI regimens were normalized using the linear-quadratic model, and biologically effective doses (BEDs) were calculated. RESULTS: Eleven reports were found describing the frequency of renal dysfunction after TBI. The frequency of renal dysfunction as a function of the BED was obtained. For BED>16 Gy an increase in the frequency of dysfunction was observed. CONCLUSIONS: The tolerance BED for kidney tissue undergoing TBI is about 16 Gy. This BED can be realized with highly fractionated TBI (e.g., 6x1.7 Gy or 9x1.2 Gy at dose rates>5 cGy/min). To prevent late renal dysfunction, the TBI regimens with BED values>16 Gy (almost all found in published reports) should be applied with appropriate shielding of the kidneys.


Asunto(s)
Enfermedades Renales/etiología , Riñón/efectos de la radiación , Irradiación Corporal Total/efectos adversos , Trasplante de Células Madre Hematopoyéticas , Humanos , Dosis Máxima Tolerada , Efectividad Biológica Relativa , Acondicionamiento Pretrasplante/efectos adversos
9.
Anticancer Res ; 26(1A): 139-45, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16475690

RESUMEN

BACKGROUND: Gemcitabine (dFdC) may cause radiosensitization by specific interference with homologous recombination-mediated DNA double-strand break repair. The radiosensitizing effect of dFdC might be less in normal healthy tissue and more restricted to undifferentiated tumor cells, making it a tumor-selective radiosensitizer. Whether dFdC acts as a radiosensitizer in undifferentiated and well-differentiated rat tumors and on rat foot skin was tested. MATERIALS AND METHODS: Undifferentiated L44 lung tumors in BN rats, MLL prostate tumors in Copenhagen rats, and well-differentiated L42 lung tumors in WAG/Rij rats were used. The tumors were treated with a single X-ray dose, combined or not with dFdC (30 mg/kg) administered 24 h earlier. Tumor volume growth delay was the end-point used. In addition, rat foot skin was treated with a single dose of 22.5 Gy, with or without dFdC. The degree of skin damage was determined according to a scoring system. RESULTS: For tumor growth delay, the dose-enhancement ratios were 1.37 and 1.23-1.36 for the L44 and MLL tumors, respectively. No radiosensitization was observed for the well-differentiated L42 tumor and foot skin. CONCLUSION: Radiosensitization by dFdC was observed in the undifferentiated tumors, but not in the well-differentiated tumor and skin. Our data support further trials to evaluate the usefulness of dFdC as a radiosensitizer in undifferentiated tumors.


Asunto(s)
Carcinoma/tratamiento farmacológico , Carcinoma/radioterapia , Desoxicitidina/análogos & derivados , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Antimetabolitos Antineoplásicos/farmacología , Terapia Combinada , Desoxicitidina/farmacología , Relación Dosis-Respuesta en la Radiación , Femenino , Masculino , Ratas , Ratas Endogámicas BN , Piel/efectos de la radiación , Gemcitabina
10.
Int J Radiat Oncol Biol Phys ; 62(1): 62-9, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15850903

RESUMEN

PURPOSE: We analyzed toxicity and the local control rates for advanced laryngeal cancer, treated with two accelerated fractionation schedules. The main difference between the schedules was the onset of the concomitant boost, in Week 3 or Week 4. Overall treatment time and total dose were equivalent. METHODS AND MATERIALS: In a prospective, nonrandomized study of T3, T4, and advanced T2 laryngeal cancer, concomitant boost schedules were used in 100 patients. Thirty patients received a schedule of twice daily 1.2 Gy in Weeks 1-3, followed by twice daily 1.7 Gy in Weeks 4 and 5; total dose was 70 Gy (the hyperfractionated accelerated schedule [HAS] regimen). Seventy patients were treated with 5 times 2 Gy in Weeks 1 and 2, followed by daily 1.8 Gy and 1.5 Gy (boost) in Weeks 3-5; total dose 69.5 Gy (the accelerated schedule only [ASO] regimen). Distribution of T stage was 47%, 40%, and 12% for T2, T3, and T4, respectively. In 24% of the patients, lymph nodes were positive. Pretreatment tracheotomy or stridor or both occurred in 8 patients. The distribution of prognostic factors was not significantly different between the two fractionation schedules. Acute and late toxicity was assessed. Results were estimated by the use of actuarial methods. For late toxicity and local control univariate and multivariate analyses were performed. Tumor control probability analysis was used to model cure rate differences. RESULTS: Overall acute mucositis score was equal for both schedules. Acute mucositis started and decreased significantly earlier in the HAS regimen. In all patients acute mucositis healed completely. The treatment was completed within 38 days in all patients. The regional control rate was 100% for clinical N0, and 75% for the clinical N+ patients. The 3-year local control rate was 59% and 78% for the HAS and ASO regimens, respectively (p = 0.05); the ultimate local control was 80% and 94%, respectively. In multivariate analysis, besides the fractionation schedule (relative risk [RR], 2.6 for HAS vs. ASO), pretreatment tracheotomy/stridor (RR 4.3, yes vs. no), and local tumor response 3-6 weeks after radiotherapy (RR 5.1, no vs. yes) were independent factors for local control. Tumor control probability analysis indicated that the onset of repopulation may be about 4-6 days earlier for the HAS regimen. The onset of repopulation in the HAS regimen is probably at the end of the second week or at the beginning of the third week. Severe late toxicity was observed in the HAS group and ASO group in, respectively, 11% and 16%. In multivariate analysis this toxicity related significantly to the field size and pretreatment tracheotomy/stridor. CONCLUSIONS: In our study the timing of the boost in accelerated radiotherapy for advanced laryngeal cancer was an independent factor for local control, favoring the use of a concomitant boost in Week 3. This finding may indicate that accelerated repopulation of tumor cells starts early in the treatment phase.


Asunto(s)
Neoplasias Laríngeas/radioterapia , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Proliferación Celular , Fraccionamiento de la Dosis de Radiación , Femenino , Humanos , Neoplasias Laríngeas/patología , Masculino , Persona de Mediana Edad , Mucosa Bucal/efectos de la radiación , Estadificación de Neoplasias , Estudios Prospectivos , Traumatismos por Radiación/etiología , Ruidos Respiratorios/etiología , Estomatitis/etiología , Factores de Tiempo
11.
Int J Radiat Oncol Biol Phys ; 57(4): 1116-21, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14575844

RESUMEN

PURPOSE: Recently, low alpha/beta values of 1.2 and 1.5 Gy for prostate tumors have been derived from clinical results of external beam radiotherapy and of permanent implants of (125)I and (103)Pd. In the analyses the contributions of tumor repopulation, and edema as a result of inserting radioactive seeds in the prostate, have been ignored. In this paper we reanalyzed the clinical data and introduced the contribution of repopulation and edema. METHODS AND MATERIALS: The linear quadratic-biologically effective dose model was used for reanalysis. In this model, the influence of repopulation and edema has been taken into account. The biologically effective dose was calculated as a function of alpha/beta for 2 brachytherapy regimens with (125)I and (103)Pd and 2 fractionated treatments, and for different half-times for repair of sublethal damage for the brachytherapy regimens. RESULTS: We have found a plausible alpha/beta value of 3.1 to 3.9 Gy, an alpha value of 0.1 to 0.15 Gy(-1), and a half-time of repair of about 0.5 h. CONCLUSIONS: It seems now that the alpha/beta value is low, 3.1-3.9 Gy, but not as low as the 1.2 and 1.5 Gy reported earlier.


Asunto(s)
Braquiterapia , Radioisótopos de Yodo/uso terapéutico , Paladio/uso terapéutico , Neoplasias de la Próstata/radioterapia , Radioisótopos/uso terapéutico , Reparación del ADN , Edema/complicaciones , Humanos , Modelos Lineales , Masculino , Enfermedades de la Próstata/complicaciones , Dosificación Radioterapéutica , Valores de Referencia , Efectividad Biológica Relativa
12.
Int J Radiat Oncol Biol Phys ; 53(2): 422-33, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12023147

RESUMEN

PURPOSE: To study the influence of radiobiologic and physical parameters and parameters related to edema on the biologically effective dose (BED) for permanent prostate implants and to determine the optimal timing of seed reconstruction for BED calculation. METHODS AND MATERIALS: On the basis of the linear-quadratic model, an expression for the BED was derived, including the edema parameters. A set of parameter values was defined, and these parameter values were varied one at a time to examine the effect on the BED and the theoretically effective treatment time (t(eff)). A ratio epsilon was defined to investigate the optimal timing of seed reconstruction. RESULTS: The maximal BED decreases when the extent of lethal damage is smaller, the potential tumor doubling time is smaller, the half-life time of the seeds is shorter, and the magnitude of prostate volume increase is larger. For 125I, the optimal timing of seed reconstruction is 25 days after implantation. Seed reconstruction 1 day after the implantation results in an underestimation of the BED of at most 43%, depending on the magnitude and half-life of edema. An overestimation of the BED of at most 22% is calculated when seed reconstruction took place at the effective treatment time. CONCLUSION: The maximal BED depends strongly on the value of alpha, the potential tumor doubling time, and the choice of isotope. If prostate volume increase due to edema is not taken into account, the BED will be underestimated shortly after the implantation and overestimated if the calculations are based on images taken several months after implantation. The optimal timing of BED evaluation for 125I seed implants and typical prostate edema values is 25 days after implantation.


Asunto(s)
Algoritmos , Braquiterapia/estadística & datos numéricos , Neoplasias de la Próstata/radioterapia , Efectividad Biológica Relativa , Braquiterapia/efectos adversos , Edema/etiología , Radioisótopos de Oro/uso terapéutico , Semivida , Humanos , Radioisótopos de Yodo/uso terapéutico , Modelos Lineales , Masculino , Paladio/uso terapéutico , Próstata/patología , Próstata/efectos de la radiación , Enfermedades de la Próstata/etiología , Neoplasias de la Próstata/patología , Radiactividad , Radioisótopos/uso terapéutico , Factores de Tiempo
13.
Int J Radiat Oncol Biol Phys ; 52(5): 1367-74, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11955751

RESUMEN

PURPOSE: To determine a dose-effect relationship for cataract induction, the tissue-specific parameter, alpha/beta, and the rate of repair of sublethal damage, mu value, in the linear-quadratic formula have to be known. To obtain these parameters for the human eye lens, a large series of patients treated with different doses and dose rates is required. The data of patients with acute leukemia treated with single-dose total body irradiation (STBI) and bone marrow transplantation (BMT) collected by the European Group for Blood and Marrow Transplantation were analyzed. METHODS AND MATERIALS: The data of 495 patients who underwent BMT for acute leukemia, who had STBI as part of their conditioning regimen, were analyzed using the linear-quadratic concept. The end point was the incidence of cataract formation after BMT. Of the analyzed patients, 175 were registered as having cataracts. Biologic effective doses (BEDs) for different sets of values for alpha/beta and mu were calculated for each patient. With Cox regression analysis, using the overall chi-square test as the parameter evaluating the goodness of fit, alpha/beta and mu values were found. Risk factors for cataract induction were the BED of the applied TBI regimen, allogeneic BMT, steroid therapy for >14 weeks, and heparin administration. To avoid the influence of steroid therapy and heparin on cataract induction, patients who received steroid or heparin treatment were excluded, leaving only the BED as a risk factor. Next, the most likely set of alpha/beta and mu values was obtained. With this set, the cataract-free survival rates were calculated for specific BED intervals, according to the Kaplan-Meier method. From these calculations, cataract incidences were obtained as function of the BED at 120 months after STBI. RESULTS: The use of BED instead of the TBI dose enabled the incidence of cataract formation to be predicted in a reasonably consistent way. With Cox regression analysis for all STBI data, a maximal chi-square value was obtained for alpha/beta = 1.75 Gy and mu = 0.75 h(-1). When Cox regression analysis was applied for patients who had no steroid treatment after BMT, a maximal chi-square value was obtained for alpha/beta = 1 Gy and mu = 0.6 h(-1). Cox regression analysis was repeated using the data of patients who had not received posttransplant steroid treatment and also no heparin administration; we found alpha/beta = 0.75 Gy and mu= 0.65 h(-1). An increased cataract incidence was observed after steroid treatment of >14 weeks and heparin administration. CONCLUSION: The alpha/beta value of 0.75 Gy and mu value of 0.65 h(-1) found for the eye lens are characteristic for late-responding tissues. The incidence of cataract formation can now be quantified, taking into account the values calculated for alpha/beta and mu, TBI dose, and dose rate. Also, the reduction in cataract incidence as a result of lens dose reduction by eye shielding can be estimated.


Asunto(s)
Catarata/etiología , Cristalino/efectos de la radiación , Irradiación Corporal Total , Enfermedad Aguda , Adolescente , Adulto , Algoritmos , Anticoagulantes/uso terapéutico , Trasplante de Médula Ósea , Niño , Preescolar , Relación Dosis-Respuesta en la Radiación , Femenino , Heparina/uso terapéutico , Humanos , Lactante , Leucemia/terapia , Masculino , Modelos de Riesgos Proporcionales , Protección Radiológica , Efectividad Biológica Relativa , Esteroides/uso terapéutico , Acondicionamiento Pretrasplante
14.
Int J Radiat Oncol Biol Phys ; 57(3): 787-93, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14529785

RESUMEN

PURPOSE: To investigate the responses of two experimental rat tumors to single and fractionated X-ray doses whether or not combined with Motexafin gadolinium (MGd), and the distribution of MGd in R3327-MATLyLu (MLL) tumors using MRI. METHODS: L44 lung tumor in BN rats and MLL prostate tumor in Copenhagen rats were grown subcutaneously. MGd at concentrations of 8.7 to 25.1 micro mol/kg was administered 2 h before or just before treatments with single and fractionated X-ray doses. Tumor volume growth delay was the endpoint used. The two-dimensional distribution of the MGd concentration in time was analyzed simultaneously in slices through the center of MLL tumors using MRI. Directly after the MRI experiments, tumor sections were stained for cytoplasm, nuclei, and microvessel endothelium. RESULTS: MGd at different concentrations administered a few minutes or 2 h before X-ray doses produced no radiation enhancement in the two tumor models. The MGd concentration as determined by MRI was maximal 5 min after injection and decreased slowly thereafter. In a representative section at the center of the MLL tumor, the microvessel density is nearly homogeneous and correlates with a nearly homogeneous MGd distribution. Hardly any MGd is taken up in underlying muscle tissue. CONCLUSION: No radiosensitization was observed for the different irradiation regimens. The distribution of MGd is nearly homogeneous in the MLL tumor and hardly any MGd is taken up in underlying muscles. Our negative results on radiosensitivity in our two tumor models raise questions about the efficacy of MGd as a general radiosensitizing agent.


Asunto(s)
Neoplasias Pulmonares/radioterapia , Metaloporfirinas/uso terapéutico , Neoplasias de la Próstata/radioterapia , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Peso Corporal/efectos de la radiación , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Neoplasias Pulmonares/tratamiento farmacológico , Imagen por Resonancia Magnética , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Ratas
15.
Int J Radiat Oncol Biol Phys ; 52(5): 1375-80, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11955752

RESUMEN

PURPOSE: To assess the degree of visual impairment as a result of cataract formation after total body irradiation (TBI) for bone marrow transplantation. METHODS AND MATERIALS: The data from 93 patients who received TBI in 1 or 2 fractions as a part of their conditioning regimen for bone marrow transplantation were analyzed with respect to the degree of visual impairment as a result of cataract formation. The probability to develop severe visual impairment (SVI) was determined for all patients, and the degree of visual impairment was assessed for 56 patients with stabilized cataract, using three categories: no, mild, or severe. RESULTS: For all 93 patients, the probability of developing a cataract causing SVI was 0.44. For allogeneic patients, it was 0.33 without and 0.71 with steroid treatment (p <0.001). All SVI-free probability curves reached a plateau distinct from the cataract-free curves. Apparently, cataracts developing late in the follow-up period rarely cause SVI. Of the patients with stabilized cataract, 32% had no visual impairment, 16% had mild, and 52% severe impairment. No or mild visual impairment was present in 61% of all patients with stable cataract and no steroid treatment compared with only 13% of the patients treated with steroids (p = 0.035). CONCLUSION: SVI occurs in only some of the patients (52%) with stable cataract after TBI for bone marrow transplantation in 1 or 2 fractions. Steroid treatment markedly increases the probability of developing visual problems as result of a cataract after TBI.


Asunto(s)
Trasplante de Médula Ósea , Catarata/etiología , Acondicionamiento Pretrasplante/efectos adversos , Trastornos de la Visión/etiología , Irradiación Corporal Total/efectos adversos , Enfermedad Aguda , Adolescente , Adulto , Femenino , Estudios de Seguimiento , Humanos , Leucemia/terapia , Masculino , Persona de Mediana Edad , Probabilidad , Esteroides/uso terapéutico
16.
Int J Radiat Oncol Biol Phys ; 59(4): 1138-47, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15234049

RESUMEN

PURPOSE: To affirm the effectiveness and complication rate of postoperative single-dose beta-irradiation (RT) with (90)Sr in the case of primary pterygium in a clinical trial. Pterygium is a benign disease of the supporting orbital tissue that can cause impairment of visual function. Depending on the technique used for surgery, recurrence is described in up to 70% of cases-a reason to combine the initial treatment with radiotherapy or chemotherapy. METHODS AND MATERIALS: This trial was designed as a prospective, randomized, multicenter, double-blind study. Surgery was performed in all cases according to the bare sclera technique. Ninety-one patients with 96 pterygia were postoperatively randomized to either beta-RT or sham RT. In the case of beta-RT, a (90)Sr eye applicator was used to deliver 2500 cGy to the sclera surface at a dose rate of between 200 and 250 cGy/min. Sham RT was given using the same type of applicator without the (90)Sr layer. After treatment, both an ophthalmologist and a radiation oncologist performed the follow-up examinations. The accumulated data were analyzed using a group sequential test. RESULTS: Between February 1998 and September 2002, 96 eyes with primary pterygium were operated on according to the trial protocol. Additional treatment was performed within 24 hours postoperatively. Ten patients were lost to follow-up, resulting in 86 patients who could be analyzed. In the 44 eyes randomized to receive beta-RT, 3 relapses occurred compared with 28 recurrences in the 42 eyes that received sham RT, for a crude control rate of 93.2% vs. 33.3%, respectively. At a mean follow-up of 18 months, major treatment complications had not been observed. CONCLUSION: Single-dose beta-RT after bare sclera surgery is a simple, effective, and safe treatment that reduces the risk of primary pterygium recurrence.


Asunto(s)
Pterigion/radioterapia , Radioisótopos de Estroncio/uso terapéutico , Adulto , Anciano , Terapia Combinada , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Pterigion/cirugía , Prevención Secundaria
17.
Strahlenther Onkol ; 180(12): 798-804, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15592700

RESUMEN

AIM: To determine whether radiation combined with Trans-4-AminoMethyl Cyclohexane carboxylic Acid (AMCA, or tranexamic acid, Cyklokapron) results in a better tumor response than radiation alone. MATERIALS AND METHODS: We evaluated the responses of the L44 lung tumor in BN rats and R3327-MATLyLu (MLL) prostate tumor in Copenhagen rats, to single and fractionated X-ray doses with and without AMCA (1.5 g/kg). Tumors were grown subcutaneously in the flank of the animal. AMCA was administered subcutaneously twice daily for at least 2 weeks. Response to treatment was evaluated according to excess growth delay and specific growth delay. RESULTS: L44 and MLL tumors treated with AMCA only experienced a non-significant growth delay. L44 tumors treated with 4 daily dose fractions of 2.5 Gy had a significant excess and specific growth delay when treated with AMCA, the enhancement ratio was 1.6-1.7. The enhancement ratio based on the calculated excess biologically effective dose of the linear-quadratic concept was 1.4-1.5. MLL tumors treated with a single dose of 20 Gy and AMCA had no significant excess growth delay. CONCLUSION: The enhancement ratio of 1.4-1.7 for the L44 tumor, but not for the MLL tumor, due to AMCA treatment, indicates that AMCA may potentiate the anti-tumor effect of ionizing radiation in distinct tumor types.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Ácido Tranexámico/administración & dosificación , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Antifibrinolíticos/administración & dosificación , Línea Celular Tumoral , Quimioterapia Adyuvante/métodos , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Neoplasias Pulmonares/patología , Masculino , Neoplasias de la Próstata/patología , Radiación Ionizante , Ratas , Resultado del Tratamiento
18.
Oncol Rep ; 9(4): 683-8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12066192

RESUMEN

The brain requires a stable internal environment, which is established by the integrity of the blood-brain barrier (BBB). The efficacy of chemotherapeutics in the treatment of brain malignancies is often hampered by the presence of the BBB. BBB disruption can be performed either by osmotic disruption, bradykinin or irradiation. Radiotherapy with doses of 20 to 30 Gy with fraction size of 2 Gy may be used to increase the permeability of the BBB. These radiation doses by themselves will not give rise to serious side effects or long-term complications. Disruption of the BBB by radiotherapy might have implications in the treatment of primary brain tumors, cerebral metastases, and prophylactic cranial irradiation in small cell lung cancer since irradiation will cause cell kill and may enhance the effect of chemotherapy. We present a review on the effects of irradiation on the BBB and subsequently discuss the potential value for therapeutic applications.


Asunto(s)
Barrera Hematoencefálica/efectos de la radiación , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Encéfalo/irrigación sanguínea , Permeabilidad Capilar/efectos de la radiación , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Terapia Combinada , Humanos , Factores de Tiempo
20.
Radiother Oncol ; 98(3): 292-7, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21316787

RESUMEN

PURPOSE: Stereotactic radiotherapy (SRT) of brain metastases is considered effective when long-term local control is obtained. However, dose-effect data are scarce. We, therefore, performed a systematic literature search to assess the evidence concerning the relation of SRT dose and local control probability. METHODS AND MATERIALS: A search was performed for papers describing patients treated with SRT for brain metastases, published from 1990 through 2009, in the electronic databases Medline (Pubmed) and Embase. We selected only papers reporting actuarial local control probability, in which a fixed dose had been prescribed and in which the size of the metastases was given. Series with SRT as a boost after whole brain irradiation (WBI) or with SRT after surgery were excluded. From the selected papers we extracted data on dose, local control rates and necrosis rates. Biological effective doses of the linear-quadratic-cubic model, using an α/ß of 12Gy (BED(12)), were calculated and a dose-response curve was constructed. RESULTS: Eleven papers fulfilled the selection criteria for further analysis. Six-month local control rates were higher than 80% in almost all the series irrespective of dose. Twelve-month local control rates, however, varied and were higher than 80%, higher than 60% and lower than 50% with single doses of ≥21Gy, ≥18Gy and ≤15Gy, respectively, and 70% or higher with fractionated SRT (FSRT). A BED(12) of at least 40Gy was associated with a twelve-month local control rate of 70% or more. CONCLUSION: Local control after single fraction SRT is highly dependent upon dose and is high (>80%) after 21Gy or more, but low (<50%) after 15Gy or less. We conclude that SRT for brain metastases should preferably be applied with a BED(12) of at least 40Gy corresponding with a single fraction of 20Gy, two fractions of 11.6Gy or three fractions of 8.5Gy.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Relación Dosis-Respuesta en la Radiación , Recurrencia Local de Neoplasia/radioterapia , Humanos , Metástasis de la Neoplasia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA