Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Pract Radiat Oncol ; 14(2): e97-e104, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37984711

RESUMEN

PURPOSE: Hypofractionated radiation therapy (HFRT) is a common treatment for thoracic tumors, typically delivered as 60 Gy in 15 fractions. We aimed to identify dosimetric risk factors associated with radiation pneumonitis in patients receiving HFRT at 4 Gy per fraction, focusing on lung V20, mean lung dose (MLD), and lung V5 as potential predictors of grade ≥2 pneumonitis. METHODS AND MATERIALS: All patients were treated with thoracic HFRT to 60 Gy in 15 fractions or 72 Gy in 18 fractions at a single health care system from 2013 to 2020. Tumors near critical structures (trachea, proximal tracheobronchial tree, esophagus, spinal cord, or heart) were considered central (within 2 cm), and those closer were classified as ultracentral (within 1 cm). The primary endpoint was grade ≥2 pneumonitis. Logistic regression analyses, adjusting for target size and dosimetric variables, were used to establish a dose threshold associated with <20% risk of grade ≥2 pneumonitis. RESULTS: During a median 24.3-month follow-up, 18 patients (16.8%) developed grade ≥2 radiation pneumonitis, with no significant difference between the 2 dose regimens (17.3% vs 16.3%, P = .88). Four patients (3.7%) experienced grade ≥3 pneumonitis, including 2 grade 5 cases. Patients with grade ≥2 pneumonitis had significantly higher lung V20 (mean 23.4% vs 14.5%, P < .001), MLD (mean 13.0 Gy vs 9.5 Gy, P < .001), and lung V5 (mean 49.6% vs 40.6%, P = .01). Dose thresholds for a 20% risk of grade ≥2 pneumonitis were lung V20 <17.7%, MLD <10.6 Gy, and V5 <41.3%. Multivariable analysis revealed a significant association between lung V20 and grade ≥2 pneumonitis (adjusted odds ratio, 1.48, P = .03). CONCLUSIONS: To minimize the risk of grade ≥2 radiation pneumonitis when delivering 4 Gy per fraction at either 60 Gy or 72 Gy, it is advisable to maintain lung V20<17.7%. MLD <10.6 Gy and V5<41.3% can also be considered as lower-priority constraints. However, additional validation is necessary before incorporating these constraints into clinical practice or trial planning guidelines.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neumonía , Neumonitis por Radiación , Humanos , Neumonitis por Radiación/epidemiología , Neumonitis por Radiación/etiología , Neoplasias Pulmonares/patología , Pulmón/patología , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Neumonía/complicaciones , Estudios Retrospectivos , Dosificación Radioterapéutica
2.
Adv Radiat Oncol ; 6(3): 100648, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34195487

RESUMEN

PURPOSE: To determine the incidence and predictors of gastric bleeding after chemoradiation for esophageal or gastroesophageal junction cancer. METHODS AND MATERIALS: We reviewed patients receiving chemoradiation to at least 41.4 Gy for localized esophageal cancer whose fields included the stomach and who did not undergo surgical resection. The primary endpoint was grade ≥3 gastric hemorrhage (GB3+). Comprehensive stomach dose-volume parameters were collected, and stomach dose-volume histograms were generated for analysis. RESULTS: A total of 145 patients met our inclusion criteria. Median prescribed dose was 50.4 Gy (range, 41.4-56 Gy). Median stomach Dmax was 53.0 Gy (1.0-62.7 Gy), and median stomach V40, V45, and V50 Gy were 112 cm3 (0-667 cm3), 84 cm3 (0-632 cm3), and 50 cm3 (0-565 cm3), respectively. Two patients (1.4%) developed radiation-induced GB3+. The only dosimetric factor that was significantly different for these patients was a higher stomach Dmax (58.1 and 58.3 Gy) than the cohort median (53 Gy). One of these patients also had cirrhosis, and the other had a history of nonsteroidal anti-inflammatory drug use. Five other patients had GB3+ events associated with documented tumor progression. A Cox proportional hazards model based on stomach Dmax with respect to the development of GB3+ was found to be statistically significant. Time-to-event curves and dose-volume atlases were generated, demonstrating an increased risk of GB3+ only when stomach Dmax was >58 Gy (P < .05). CONCLUSIONS: We observed a low rate of GB3+ events in patients who received chemoradiation to a median dose of 50.4 Gy to volumes that included a significant portion of the stomach. These results suggest that when prescribing 50.4 Gy for esophageal cancer, there is no need to minimize the irradiated gastric volume or dose for the sake of preventing bleeding complications. Limiting stomach maximum doses to <58 Gy may also avoid bleeding, and particular caution should be taken in patients with other risk factors for bleeding, such as cirrhosis.

3.
Health Phys ; 119(3): 315-321, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32175929

RESUMEN

The Radiation and Nuclear Countermeasures Program at the National Institute of Allergy and Infectious Diseases (NIAID) mandated that medical countermeasures for treating Acute Radiation Syndrome (ARS) must have efficacy when administered at least 24 h after radiation exposure. At this time point, many cells within key target tissues, such as the hematopoietic system and the gastrointestinal (GI) tract, will already be dead. Therefore, drugs that promote the regeneration of surviving cells may improve outcomes. The serine/threonine kinase glycogen synthase kinase-3 (GSK-3) regulates stem and progenitor cell self-renewal and regeneration in the hematopoietic and GI compartments. We tested inhibition of GSK-3ß by SB216763 24 h after total body irradiation (TBI) and sub-total body irradiation (SBI). Here, we show that subcutaneous administration of SB216763 promotes the regeneration of surviving hematopoietic stem/progenitor cells (HSPCs), including myeloid progenitor cells, and improves survival of C57Bl/6 male mice when administered 24 h after TBI. However, these results were not recapitulated in female C57Bl/6 animals, suggesting a sex difference in GSK-3ß signaling in HSPCs. Subcutaneous administration of SB216763 in male mice stimulated activation of Sox2 transcription but failed to induce Sox2 transcription in female C57Bl/6 mice. Using TCF/lef-GFP reporter mice, we examined Wnt signaling in HSPCs of irradiated male and female mice treated with SB216763. GSK-3 inhibition elevated Wnt reporter activity in HSPCs isolated from male but not female mice. SB216763 did not mitigate hematopoietic ARS in males or females of a second strain of wild-type mice, C3H. In addition, administration of SB216763 did not mitigate hematopoietic ARS beyond the currently available standard approved therapy of ciprofloxacin and granulocyte-colony stimulating factor (G-CSF) in male C57Bl/6 mice. Further, SB216763 did not mitigate GI-ARS after SBI in C57Bl/6 male mice. The lack of efficacy in both sexes and multiple strains of mice indicate that SB216763 is not suitable for further drug development as a mitigator of ARS. Our studies demonstrate that activation of Wnt signaling in HSPCs promotes hematopoietic regeneration following radiation exposure, and targeting this pathway downstream of GSK-3ß may mitigate ARS in a sex- and strain-independent manner.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Hematopoyesis/efectos de la radiación , Indoles/uso terapéutico , Maleimidas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Protectores contra Radiación/uso terapéutico , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/enzimología , Médula Ósea/efectos de la radiación , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Hematopoyesis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores Sexuales , Especificidad de la Especie
4.
Cancer Res ; 79(4): 773-782, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30315114

RESUMEN

Stereotactic body radiotherapy is utilized to treat lung cancer. The mechanism of tumor response to high-dose radiotherapy (HDRT) is controversial, with competing hypotheses of increased direct tumor cell killing versus indirect effects on stroma including endothelial cells. Here we used dual recombinase technology in a primary murine lung cancer model to test whether tumor cells or endothelial cells are critical HDRT targets. Lenti-Cre deleted one or two copies of ataxia-telangiectasia mutated gene (Atm; KPAFL/+ or KPAFL/FL), whereas adeno-FlpO-infected mice expressed Cre in endothelial cells to delete one or both copies of Atm (KPVAFL/+ or KPVAFL/FL) to modify tumor cell or endothelial cell radiosensitivity, respectively. Deletion of Atm in either tumor cells or endothelial cells had no impact on tumor growth in the absence of radiation. Despite increased endothelial cell death in KPVAFL/FL mice following irradiation, tumor growth delay was not significantly increased. In contrast, a prolonged tumor growth delay was apparent in KPAFL/FL mice. Primary tumor cell lines lacking Atm expression also demonstrated enhanced radiosensitivity as determined via a clonogenic survival assay. These findings indicate that tumor cells, rather than endothelial cells, are critical targets of HDRT in primary murine lung cancer. SIGNIFICANCE: These findings establish radiosensitizing tumor cells rather than endothelial cells as the primary mechanism of tumor response to high-dose radiotherapy, supporting efforts to maximize local control by radiosensitizing tumors cells.See related commentary by Hallahan, p. 704.


Asunto(s)
Adenocarcinoma del Pulmón/radioterapia , Modelos Animales de Enfermedad , Células Endoteliales/efectos de la radiación , Neoplasias Pulmonares/radioterapia , Tolerancia a Radiación , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Proliferación Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Eliminación de Secuencia , Células Tumorales Cultivadas
5.
Int J Radiat Oncol Biol Phys ; 102(5): 1569-1576, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30056081

RESUMEN

PURPOSE: The delivery of radiation therapy to cure gastrointestinal (GI) cancers is often limited by normal tissue toxicity of the GI tract. Studies using genetically engineered mice have demonstrated an essential role of the cyclin-dependent kinase inhibitor p21 in protecting against GI acute radiation syndrome (GI-ARS). Here, we examined the impact of the Food and Drug Administration-approved, selective, cyclin-dependent kinase 4/6 inhibitor palbociclib (PD-0332991) on the development of GI-ARS induced by single-dose versus fractionated radiation in mice. METHODS AND MATERIALS: For the single-dose radiation study, C57BL/6J mice were treated with palbociclib or vehicle 28 and 4 hours before subtotal body irradiation (SBI). For the fractionated radiation study, C57BL/6J mice were exposed to fractionated SBI for 5 consecutive days. These mice were treated with palbociclib or vehicle either 28 and 4 hours before the first dose of irradiation or 4 hours before the first, third, and fifth doses of irradiation. RESULTS: Our data indicate that treatment with palbociclib before, but not after, a single fraction of SBI significantly ameliorated GI-ARS, improved the integrity of the GI barrier, and increased the number of surviving crypts in the small intestine. In addition, palbociclib did not protect tumor cell lines from radiation in vitro. In contrast to the results from the single-dose exposure, treatment with palbociclib before 5 daily fractions of SBI did not prevent GI-ARS. Moreover, we unexpectedly observed that GI-ARS was exacerbated in mice treated with palbociclib before and during 5 daily fractions of SBI. CONCLUSIONS: Our results demonstrate that treatment with palbociclib before a single dose of SBI protects mice from GI-ARS. In contrast, treatment with palbociclib before and during 5 daily fractions of SBI exacerbates GI-ARS in mice. These results emphasize the importance of conducting preclinical studies of radioprotectors with single-dose and fractionated radiation therapy.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Fraccionamiento de la Dosis de Radiación , Tracto Gastrointestinal/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Tracto Gastrointestinal/efectos de la radiación , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/uso terapéutico , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/patología
6.
Sci Transl Med ; 7(278): 278ra34, 2015 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-25761890

RESUMEN

Cancer clinics currently use high-dose stereotactic body radiation therapy as a curative treatment for several kinds of cancers. However, the contribution of vascular endothelial cells to tumor response to radiation remains controversial. Using dual recombinase technology, we generated primary sarcomas in mice with targeted genetic mutations specifically in tumor cells or endothelial cells. We selectively mutated the proapoptotic gene Bax or the DNA damage response gene Atm to genetically manipulate the radiosensitivity of endothelial cells in primary soft tissue sarcomas. Bax deletion from endothelial cells did not affect radiation-induced cell death in tumor endothelial cells or sarcoma response to radiation therapy. Although Atm deletion increased endothelial cell death after radiation therapy, deletion of Atm from endothelial cells failed to enhance sarcoma eradication. In contrast, deletion of Atm from tumor cells increased sarcoma eradication by radiation therapy. These results demonstrate that tumor cells, rather than endothelial cells, are critical targets that regulate sarcoma eradication by radiation therapy. Treatment with BEZ235, a small-molecule protein kinase inhibitor, radiosensitized primary sarcomas more than the heart. These results suggest that inhibiting ATM kinase during radiation therapy is a viable strategy for radiosensitization of some tumors.


Asunto(s)
Células Endoteliales/patología , Radiocirugia , Sarcoma/patología , Sarcoma/radioterapia , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Imidazoles/farmacología , Imidazoles/uso terapéutico , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Quinolinas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Sarcoma/tratamiento farmacológico , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
7.
J Clin Invest ; 124(8): 3325-38, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25036710

RESUMEN

Cells isolated from patients with ataxia telangiectasia are exquisitely sensitive to ionizing radiation. Kinase inhibitors of ATM, the gene mutated in ataxia telangiectasia, can sensitize tumor cells to radiation therapy, but concern that inhibiting ATM in normal tissues will also increase normal tissue toxicity from radiation has limited their clinical application. Endothelial cell damage can contribute to the development of long-term side effects after radiation therapy, but the role of endothelial cell death in tumor response to radiation therapy remains controversial. Here, we developed dual recombinase technology using both FlpO and Cre recombinases to generate primary sarcomas in mice with endothelial cell-specific deletion of Atm to determine whether loss of Atm in endothelial cells sensitizes tumors and normal tissues to radiation. Although deletion of Atm in proliferating tumor endothelial cells enhanced the response of sarcomas to radiation, Atm deletion in quiescent endothelial cells of the heart did not sensitize mice to radiation-induced myocardial necrosis. Blocking cell cycle progression reversed the effect of Atm loss on tumor endothelial cell radiosensitivity. These results indicate that endothelial cells must progress through the cell cycle in order to be radiosensitized by Atm deletion.


Asunto(s)
Eliminación de Gen , Tolerancia a Radiación/genética , Tolerancia a Radiación/fisiología , Sarcoma Experimental/genética , Sarcoma Experimental/radioterapia , Animales , Proteínas de la Ataxia Telangiectasia Mutada/deficiencia , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Endoteliales/patología , Células Endoteliales/efectos de la radiación , Corazón/efectos de la radiación , Humanos , Ratones , Ratones Transgénicos , Miocardio/patología , Recombinasas , Sarcoma Experimental/patología , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/radioterapia , Microambiente Tumoral/genética , Microambiente Tumoral/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA