Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Int J Radiat Oncol Biol Phys ; 10(8): 1221-7, 1984 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6469742

RESUMEN

We have measured the rate of GSH resynthesis in plateau phase cultures of A549 human lung carcinoma cells subjected to a fresh medium change. Buthionine sulfoximine (BSO) blocks this resynthesis. Diethyl maleate (DEM) causes a decrease in accumulation of GSH. If DEM is added concurrently with BSO there is a rapid decline in GSH that is maximal in the presence of 0.5 mM DEM. GSH depletion rapidly occurs when BSO is added to log phase cultures which initially are higher in GSH content. Twenty-four hr treatment of A549 cells with BSO results in cells that are more radiosensitive in air and show a slight hypoxic radiation response. A 2 hr treatment with either 0.25 mM or 0.5 mM DEM results in some hypoxic sensitization and little increase in the aerobic radiation response. The 24 hr BSO + 2 hr DEM treatment sensitizes hypoxic cells to a greater degree than either agent alone but does not increase the aerobic response more than is seen with BSO alone. Cells treated simultaneously with BSO + DEM show little increase in the hypoxic radiation response, compared to DEM alone, but are more sensitive under aerobic conditions. Decreased cell survival for aerobically irradiated log phase A549 cells occurs within minutes after addition of a mixture of BSO + DEM. The decreased cell survival following aerobic irradiation, after prolonged treatment with BSO or acute exposure to BSO + DEM, may be in part due to inhibition of glutathione peroxidases. For example, glutathione-S-transferase, known to have glutathione peroxidase activity (non-selenium), is nearly completely inhibited by the BSO treatments. In addition, cellular capacity to react with peroxide (glutathione peroxidase, selenium containing) was also inhibited. We suggest that the enhanced aerobic radiation response is related to an inability of GSH depleted cells to inactivate either peroxy radicals or hydroperoxides that may be produced during irradiation of BSO treated cells. Furthermore, enhancement of the aerobic radiation response may be useful in vivo if normal tissue responses are not also increased.


Asunto(s)
Glutatión/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Neoplasias Pulmonares/patología , Maleatos/farmacología , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Factores de Tiempo
2.
Int J Radiat Oncol Biol Phys ; 16(5): 1219-23, 1989 May.
Artículo en Inglés | MEDLINE | ID: mdl-2715073

RESUMEN

From analytical expressions derived for the radical-repair (competition) model describing the relationship between cellular radiosensitivity and oxygen concentration, "K-curve" behavior has been quantified as a function of the concentration of the species S which restitutes the radiation-induced radicals to their original molecular configuration. If these species are identified with thiols, K-curves modified by fractionally depleting [S] through calculation can be compared with experimental data where cells have their thiols depleted using various means, for example, by chemical agents or by the use of cells with decreased thiols because of genetic deficiency. Families of curves have been calculated related both to the S-depleted and the non-S-depleted hypoxic control, the latter of which is used to calculate enhancement ratios. Comparison of the model with experimental data is made.


Asunto(s)
Reparación del ADN , Compuestos de Sulfhidrilo/metabolismo , Animales , Células/efectos de la radiación , Modelos Biológicos , Oxígeno/fisiología , Tolerancia a Radiación
3.
Int J Radiat Oncol Biol Phys ; 12(7): 1161-4, 1986 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2943713

RESUMEN

Chronic aerobic exposure of A549 human lung carcinoma cell cultures to 0.1 mM L-buthionine-S,R-sulfoximine and 1 mM misonidazole, or 1 mM SR-2508 results in inhibition of cell growth and decreased clonogenic survival. These patterns are not apparent with the individual drug treatments. Both parameters demonstrate maximum toxicity after 72 hr in culture, which parallels the time required to deplete A549 cells of glutathione with 0.1 mM L-BSO under these growth conditions. Toxicity appears to be related to hydrogen peroxide-produced during 1 electron reduction of the nitro compounds in the presence of oxygen. A549 cells have a lowered capacity to reduce peroxide due to the effect of thiol depletion on the enzymes GSH-peroxidase and GSH-S-transferase, which require the tripeptide as a substrate. The addition of catalase, another important enzyme involved in peroxide reduction, partially reverses the observed toxicity. 4-Hydroxypyrazole, which inhibits endogenous catalase activity, causes an increase in the observed cytotoxicity. Similar effects of L-BSO and 4-hydroxypyrazole are seen for toxicity due to hydrogen peroxide being added directly to cell cultures.


Asunto(s)
Metionina Sulfoximina/análogos & derivados , Misonidazol/toxicidad , Nitroimidazoles/toxicidad , Pirazoles/farmacología , Fármacos Sensibilizantes a Radiaciones/toxicidad , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Etanidazol , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/biosíntesis , Técnicas In Vitro , Metionina Sulfoximina/farmacología , Oxígeno/fisiología
4.
Int J Radiat Oncol Biol Phys ; 12(7): 1139-42, 1986 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3744931

RESUMEN

Our data show that A549 cells are increasingly radiosensitive with prolonged exposure to L-BSO. The resulting glutathione and protein thiol depleted cells show both loss of shoulder and slope modification. Furthermore, there is an increase in single strand DNA breaks and irrepairable cross-linking. The aerobic radiation damage in the thiol depleted state appears to be different from that obtained with hypoxic cells. Any postulated role for GSH in reducing or preventing peroxidative radiation damage must also include protection against single strand DNA breaks as well as involvement in repairing DNA-protein cross-links. The latter effect may be related to decreased protein thiol content as reflected in a decreased enzyme capacity to repair DNA damage.


Asunto(s)
Metionina Sulfoximina/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/farmacología , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Glutatión/metabolismo , Humanos , Técnicas In Vitro , Metionina Sulfoximina/farmacología , Oxígeno/fisiología , Factores de Tiempo
5.
Int J Radiat Oncol Biol Phys ; 16(5): 1311-4, 1989 May.
Artículo en Inglés | MEDLINE | ID: mdl-2715084

RESUMEN

We will review the relationships between glutathione (GSH), protein thiols, and cellular responses to radiation, peroxides, and peroxide-producing drugs. Our primary interest involves the behavior of sulfhydryls as electron and hydrogen carriers, and their capacity to protect various target molecules against radiation and peroxidative damage. We used reagents such as L-buthionine sulfoximine (LBSO), alone and in combination with N-ethyl maleimide (NEM), diamide, and dimethylfumarate, to decrease GSH so that it could no longer participate in the electron transfer reactions. Our results indicate that aerobic sensitization produced by GSH depletion can be further enhanced if electron-accepting agents, such as tertiary butyl hydroperoxide (t-BOOH), are present during irradiation. Hydroperoxide is a substrate for glutathione peroxidase and diverts electrons and hydrogen away from target molecules during its reduction. Sensitivity to radiation seems to be due to the inhibition of the mitochondria's capacity to reduce hydroperoxide. We will also report the mitochondria's ability to reduce the oxygen radicals produced by radiation and drugs. Data also indicate that t-BOOH oxidizes protein thiols which are enzymatically involved in repair of DNA damage.


Asunto(s)
Supervivencia Celular/efectos de la radiación , Glutatión/fisiología , Tolerancia a Radiación , Aerobiosis , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Peróxidos/farmacología , terc-Butilhidroperóxido
6.
Biochem Pharmacol ; 35(1): 77-90, 1986 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-2934068

RESUMEN

Misonidazole is a metabolically active drug. Its addition to cells causes an immediate alteration in cellular electron transfer pathways. Under aerobic conditions the metabolic alterations can result in futile cycling with electron transfer to oxygen and production of peroxide. Thiol levels are extremely important in protecting the cell against the peroxide formation and potentially hazardous conditions for hydroxyl radical production. Nevertheless such electron shunting out of cellular metabolism will result in alterations in pentose cycle, glycolysis and cellular capacity to reduce metabolites to essential intermediates needed in DNA metabolism (i.e. deoxyribonucleotides). Glutathione must be depleted to very low levels before toxic effects of misonidazole and other nitro compounds are manifested in cell death via peroxidative damage. Under hypoxic conditions misonidazole also diverts the pentose cycle via its own reduction; however, unlike the aerobic conditions, there are a number of reductive intermediates produced that react with non-protein thiols such as GSH as well as protein thiols. The reaction with protein thiols results in the inhibition of glycolysis and other as yet undetermined enzyme systems. The consequences of the hypoxic pretreatment of cells with nitro compounds are increased vulnerability to radiation and chemotherapeutic drugs such as L-PAM, cis-platinum and bleomycin. The role that altered enzyme activity has in the cellular response to misonidazole and chemotherapeutic agents remains to be determined. It is also clear that the GSH depleted state not only makes cells more vulnerable to oxidative stress but also to hypoxic intermediates produced by the reduction of misonidazole beyond the one electron stage. The relevancy of the present work to the proposed use of thiol depletion in vivo to enhance the radiation or chemotherapeutic response of tumor tissue lies with the following considerations. Apparently, spontaneous peroxidative damage to normal tissue such as liver can occur with GSH depletion to 10-20% of control and with other normal tissue when GSH reaches 50% of control. This situation can obviously become more critical if peroxide producing drugs are administered. The only advantage to such combined drug treatments would lie in the possibility that tumors vary in their catalase and peroxidase activity and consequently may be more vulnerable to oxidative stress (cf. review by Meister. Our tumor model, the A549 human lung carcinoma cell in vitro, appears to be an exception because it has catalase, peroxidase and a high content of GSH.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Nitroimidazoles/metabolismo , Animales , Butionina Sulfoximina , Catalasa , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Etanidazol , Radicales Libres , Glucosa/metabolismo , Glutatión/metabolismo , Glucólisis , Hexosafosfatos/metabolismo , Humanos , Hipoxia , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Microcuerpos/metabolismo , Microsomas/metabolismo , Misonidazol/metabolismo , Misonidazol/farmacología , Mitocondrias/metabolismo , Oxidación-Reducción , Pentosas/metabolismo , Peroxidasas/metabolismo , Compuestos de Sulfhidrilo/metabolismo
7.
Radiat Res ; 127(1): 75-80, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2068274

RESUMEN

Dimethyl fumarate (DMF) depletes intracellular glutathione (GSH) by covalent bond formation in a reaction mediated by GSH-S-transferase. Treatment of hypoxic Chinese hamster V79 cells with 5 mM DMF before irradiation radiosensitizes the cells, resulting in an enhancement ratio (ER) of about 2.7 with minimal toxicity, when the end point is clonogenic cell survival. Under the same conditions aerobic cells are sensitized, and ER of about 1.3 is found, and GSH is reduced to about 3% of control. Very similar results were obtained previously with Chinese hamster ovary (CHO) cells. In addition, new data presented here show that DMF treatment of V79 or CHO cells immediately after irradiation under hypoxic conditions sensitizes the cells, resulting in an ER of about 1.5, DMF treatment after irradiation under aerobic conditions results in an ER of 1.3, and this DMF treatment reduces protein thiols (PSH) to about 70% of control. When induction of DNA damage is measured using the neutral elution assay, treatment of V79 or CHO cells with DMF prior to irradiation under hypoxic conditions results in an ER of 1.9-2.0, but there is no enhancement of DNA damage when DMF is added after irradiation under hypoxic conditions or when cells are treated with DMF before or after irradiation under aerobic conditions. Based on these data we postulate that DMF radiosensitizes killing of hypoxic cells by two actions: depletion of GSH interferes with the chemical competition between damage fixation and repair, and depletion of PSH causes an inhibition of enzymatic repair processes. We also suggest that DMF sensitizes aerobic cells only by inhibition of enzymatic repair processes.


Asunto(s)
Supervivencia Celular/efectos de la radiación , Fumaratos/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Animales , Supervivencia Celular/efectos de los fármacos , Dimetilfumarato , Glutatión/metabolismo , Técnicas In Vitro , Oxígeno/fisiología , Factores de Tiempo
8.
Radiat Res ; 95(3): 620-36, 1983 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-6684311

RESUMEN

Experiments were conducted to investigate the effects of increasing or decreasing the yield of hydroxyl radicals (.OH) reacting with target molecules on the survival of CHO cells irradiated in a thin layer with single 3-nsec pulses of electrons. Dimethyl sulfoxide (DMSO), a known radioprotector, was used as an .OH scavenger. The gas nitrous oxide (N2O), which scavenges hydrated electrons and in the process generates an additional yield of .OH, was used in an attempt to produce sensitization by increasing the amount of .OH-induced cellular damage. It was found that DMSO at high concentration was an effective radioprotective agent in cells equilibrated with nitrogen, air, and N2O and irradiated at ultrahigh dose rates. Sensitization by N2O was observed, but only under certain conditions, specifically, when a high concentration of the .OH scavenger DMSO was also present. The enhancement ratio (ER) for oxygen sensitization was reduced in the presence of DMSO, as was the ER for sensitization by misonidazole. Interpretation of these results according to radiation chemistry considerations will be discussed.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Dimetilsulfóxido/farmacología , Radicales Libres , Tolerancia a Radiación , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Cricetinae , Relación Dosis-Respuesta en la Radiación , Electrones , Femenino , Misonidazol/farmacología , Óxido Nitroso/farmacología , Ovario , Oxígeno
9.
Radiat Res ; 115(3): 495-502, 1988 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-3174933

RESUMEN

Dimethylfumarate (DMF) depletes intracellular glutathione (GSH) by covalent bond formation in a reaction which may be mediated by GSH-S-transferase. In Chinese hamster ovary cells this depletion is rapid; e.g., 0.5 mM DMF depletes GSH to less than 10% of control in 5 min at room temperature. DMF is a very effective hypoxic cell radiosensitizer, with an enhancement ratio (ER) of about 3 obtained by a 5-min exposure of cells at room temperature to 5 mM DMF, without significant toxicity. At this same concentration of drug, there is a small enhancement of aerobic cells (ER = 1.3), but the 5 mM DMF in hypoxia results in nearly a complete collapse of the hypoxic dose-response curve to the same level as seen in air with DMF. It has been suggested previously that DMF sensitizes cells via electron affinic mechanisms. However, this appears not to be the case in this study, as shown by the fact that cells pretreated with DMF and then washed free of the drug remained equally radiosensitive as cells irradiated in the presence of the drug. This large enhancement of radiation sensitivity appears to be related to the drug's ability to deplete thiols; i.e., thiols appear to be a major factor responsible for radioresistance of hypoxic cells.


Asunto(s)
Fumaratos/farmacología , Glutatión/metabolismo , Fármacos Sensibilizantes a Radiaciones , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Cricetinae , Dimetilfumarato , Femenino , Cinética , Ovario , Oxígeno/farmacología
10.
Radiat Res ; 98(2): 370-80, 1984 May.
Artículo en Inglés | MEDLINE | ID: mdl-6539482

RESUMEN

Buthionine sulfoximine (BSO) inhibits the synthesis of glutathione (GSH), the major nonprotein sulfhydryl (NPSH) present in most mammalian cells. BSO concentrations from 1 microM to 0.1 mM reduced intracellular GSH at different rates, while BSO greater than or equal to 0.1 mM (i.e., 0.1 to 2.0 mM), resulting in inhibitor-enzyme saturation, depleted GSH to less than 10% of control within 10 hr at about equal rates. BSO exposures used in these experiments were not cytotoxic with the one exception that 2.0 mM BSO/24 hr reduced cell viability to approximately 50%. However, alterations in either the cell doubling time(s) or the cell age density distribution(s) were not observed with the BSO exposures used to determine its radiosensitizing effect. BSO significantly radiosensitized (ER = 1.41 with 0.1 mM BSO/24 hr) hypoxic, but not aerobic, CHO cells when the GSH and NPSH concentrations were reduced to less than 10 and 20% of control, respectively, and maximum radiosensitivity was even achieved with microM concentrations of BSO (ER = 1.38 with 10 microM BSO/24 hr). Furthermore, BSO exposure (0.1 mM BSO/24 hr) also enhanced the radiosensitizing effect of various concentrations of misonidazole on hypoxic CHO cells.


Asunto(s)
Supervivencia Celular/efectos de la radiación , Glutatión/fisiología , Metionina Sulfoximina/análogos & derivados , Misonidazol/farmacología , Nitroimidazoles/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Butionina Sulfoximina , Supervivencia Celular/efectos de los fármacos , Cricetinae , Cricetulus , Relación Dosis-Respuesta en la Radiación , Sinergismo Farmacológico , Femenino , Metionina Sulfoximina/farmacología , Ovario , Oxígeno , Factores de Tiempo
11.
Radiat Res ; 108(3): 238-50, 1986 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3797633

RESUMEN

The effect of changes in both the intracellular glutathione (GSH) concentration and the concentration of extracellular reducing equivalents on the aerobic radiosensitization was studied in three cell lines: CHO-10B4, V79, and A549. Intracellular GSH was metabolically depleted after the inhibition of GSH synthesis by buthionine sulfoximine (BSO), while the extracellular environment was controlled through the replacement of growth medium with a thiol-free salt solution and in some experiments by the exogenous addition of either GSH or GSSG. Each of the cell lines examined exhibited an enhanced aerobic radioresponse when the intracellular GSH was extensively depleted (GSH less than 1 nmol GSH/10(6) cells after 1.0 mM BSO/24 h treatment) and the complexity of the extracellular milieu decreased. Although the addition of oxidized glutathione (5 mM GSSG/30 min) to cells prior to irradiation was without effect, much or all of the induced radiosensitivity was overcome by the addition of reduced glutathione (5 mM GSH/15 min). However, the observation that the exogenous GSH addition restores the control radioresponse without increasing the intracellular GSH concentration was entirely unexpected. These results suggest that a number of factors exert an influence on the extent of GSH depletion and determine the extent of aerobic radiosensitization. Furthermore, the interaction of exogenous GSH with--but without penetrating--the cell membrane is sufficient to result in radiorecovery.


Asunto(s)
Glutatión/fisiología , Tolerancia a Radiación , Aerobiosis , Butionina Sulfoximina , Línea Celular , Membrana Celular/metabolismo , Medios de Cultivo , Glutatión/análisis , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología
12.
Radiat Res ; 93(3): 479-91, 1983 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-6856753

RESUMEN

The response of cultured CHO cells to ultrahigh-dose-rate radiation (approximately 10(9) Gy/sec) has been previously studied extensively using the thin-layer cell-handling technique developed in this laboratory. When the cells are equilibrated with a low concentration of oxygen, e.g., 0.44% O2, a breaking survival curve, due to radiolytic depletion of the oxygen, is observed. Hypoxic cells irradiated in the presence of the nitroimidazoles (e.g., misonidazole) are sensitized at ultrahigh dose rates in a dose-modifying manner, similar to that observed at conventional dose rates. These radiosensitizer compounds, if present in cells equilibrated with a low concentration of oxygen, prevent the breaking behavior of the survival curve, an observation believed to be due to the sensitizer interfering with the oxygen depletion process, leaving oxygen free to sensitize. Such experiments have recently been extended to studies with diamide, which, unlike the other sensitizers tested, acts primarily as a shoulder-modifying rather than a dose-modifying agent in hypoxic mammalian cells. These data indicate that diamide is active as a sensitizer at ultrahigh dose rates in a manner similar to that observed at conventional dose rates, and does modify the shape of the breaking survival curve observed with low concentrations of oxygen.


Asunto(s)
Compuestos Azo/farmacología , Supervivencia Celular/efectos de la radiación , Diamida/farmacología , Oxígeno , Animales , Línea Celular , Cricetinae , Cricetulus , Reparación del ADN/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Ovario
13.
Radiat Res ; 95(3): 437-55, 1983 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-6684310

RESUMEN

Cellular nonprotein thiols (NPSH) consist of glutathione (GSH) and other low molecular weight species such as cysteine, cysteamine, and coenzyme A. GSH is usually less than the total cellular NPSH, and with thiol reactive agents, such as diethyl maleate (DEM), its rate of depletion is in part dependent upon the cellular capacity for its resynthesis. If resynthesis is blocked by buthionine-S,R-sulfoximine(BSO), the NPSH, including GSH, is depleted more rapidly, Cellular thiol depletion by diamide, N-ethylmaleimide, and BSO may render oxygenated cells more sensitive to radiation. These cells may or may not show a reduction in the oxygen enhancement ratio (OER). Human A549 lung carcinoma cells depleted of their NPSH either by prolonged culture or by BSO treatment do not show a reduced OER but do show increased aerobic responses to radiation. Some nitroheterocyclic radiosensitizing drugs also deplete cellular thiols under aerobic conditions. Such reactivity may be the reason that they show anomalous radiation sensitization (i.e., better than predicted on the basis of electron affinity). Other nitrocompounds, such as misonidazole, are activated under hypoxic conditions to radical intermediates. When cellular thiols are depleted peroxide is formed. Under hypoxic conditions thiols are depleted because metabolically reduced intermediates react with GSH instead of oxygen. Thiol depletion, under hypoxic conditions, may be the reason that misonidazole and other nitrocompounds show an extra enhancement ratio with hypoxic cells. Thiol depletion by DEM or BSO alters the radiation response of hypoxic cells to misonidazole. In conclusion, we propose an altered thiol model which includes a mechanism for thiol involvement in the aerobic radiation response of cells. This mechanism involves both thiol-linked hydrogen donation to oxygen radical adducts to produce hydroperoxides followed by a GSH peroxidase-catalyzed reduction of the hydroperoxides to intermediates entering into metabolic pathways to produce the original molecule.


Asunto(s)
Compuestos Azo/farmacología , Diamida/farmacología , Glutatión/metabolismo , Maleatos/farmacología , Metionina Sulfoximina/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/farmacología , Compuestos de Sulfhidrilo/metabolismo , Animales , Butionina Sulfoximina , Supervivencia Celular , Células Cultivadas , Fenómenos Químicos , Química , Cricetinae , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Neoplasias Pulmonares , Metionina Sulfoximina/farmacología , Ovario , Oxígeno
14.
Radiat Res ; 116(3): 482-502, 1988 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3205910

RESUMEN

Therapeutic gain factors (TGFs) have been determined for three spontaneous tumors of the C3H mouse treated by photons + normobaric oxygen (O2(1) ATA), photons + hyperbaric oxygen (O2 3 ATA), photons + misonidazole, or fast neutrons. The tumors were early generation isotransplants of spontaneous tumors: MCaIV, a mammary carcinoma; FSaII, a fibrosarcoma; and SCCVII, a squamous cell carcinoma. The tumors, transplanted to the right leg, were 6 mm at start of treatment. Normal tissue responses studied were acute reaction of normal skin (all treatment modalities) and LD50 following irradiation of the upper abdomen (in test of photons + O2 at 1 or 3 ATA). Thus both the tumor and normal tissues would be classified as "acute responding." All subject tissues were at congruent to 34.5-35 degrees C at irradiation. Treatments were based on d(25)Be or p(43)Be fast neutron beams, 60Co and 137Cs photon beams. Treatments were given in 5 or 15 equal doses in 5 days. For photon treatments, TGFs (air/O2 3 ATA) were substantially and significantly larger than 1 for all three tumor systems treated at small or large doses per fraction when related to skin or abdominal tissue responses. The TGFs (air/O2 1 ATA) were greater than 1 at small doses per fraction for MCaIV and FSaII for skin as the normal tissue; the TGFs for all three tumors and at all doses per fraction would be greater than 1 when related to upper abdominal tissues. TGFs (O2 1 ATA/O2 3 ATA) for photon irradiation greater than 1 were found only for SCCVII and that obtained for both large and small doses per fraction. Misonidazole achieved impressive TGFs (air/air + miso or air/O2 1 ATA + miso); the drug was tested only at 10-12 Gy/fraction and relative to skin. RBEs(FN) for the three tumors were lower at 1.5-2 Gy(FN)/fraction than at 5-6 Gy(FN)/fraction, i.e. the opposite to that reported for normal tissue (RBE increases with decreasing dose per fraction). A TGF (relative to skin reaction) greater than 1 for fast neutron therapy was found only for SCCVII when treated at large doses/fraction; this was true for air or O2 1 ATA conditions.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Fibrosarcoma/radioterapia , Neoplasias Mamarias Experimentales/radioterapia , Animales , Terapia Combinada , Neutrones Rápidos , Femenino , Oxigenoterapia Hiperbárica , Masculino , Ratones , Ratones Endogámicos C3H , Misonidazol/uso terapéutico , Trasplante de Neoplasias , Oxígeno/uso terapéutico , Dosificación Radioterapéutica
15.
Med Phys ; 11(3): 231-41, 1984.
Artículo en Inglés | MEDLINE | ID: mdl-6429495

RESUMEN

The problem of neutrons produced by many of the high-energy x-ray therapy machines (10 MV and above) is reviewed, and the possible risk their presence poses to radiotherapy patients is estimated. A review of the regulatory background containing a summary of the recommendations of the U.S. Council of State Governments (USCSG), and of the International Electro-Technical Commission (IEC), as well as an indication that recommendations will be forthcoming from the National Council on Radiation Protection (NCRP) and the International Commission of Radiological Protection (ICRP) is presented. The neutrons in question are produced by high-energy photons (x rays) incident on the various materials of the target, flattening filter, collimators, and other essential components of the equipment. The neutron yield (per treatment dose) increases rapidly as the megavoltage is increased from 10 to 20 MV, but remains approximately constant above this. Measurements and calculations of the quantity, quality, and spatial distribution of these neutrons and their concomitant dose are summarized. Values of the neutron dose are presented as entrance dose, midline dose (10-cm depth), and integral dose, both within and outside of the treatment volume. These values are much less than the unavoidable photon doses which are largely responsible for treatment side effects. For typical equipment, the average neutron integral dose from accelerator-produced neutrons is about 4-7 g cGy (per treatment cGy), depending on the treatment plan. This translates into an average dose of neutrons [averaged over the body of a typical 70-kg (154 lb) patient] of 0.06-0.10 cGy for a treatment of 1000 cGy. Using these neutron doses and the best available neutron risk coefficients, it is estimated that 50 X 10(-6) fatal malignancies per year due to the neutrons may follow a typical treatment course of 5000 rads of 25-MV x rays. This is only about 1/60th of the average incidence of malignancies for the general population. Thus, the cancer risk to the radiotherapy patient from accelerator-produced neutrons poses an additional risk to the patient that is negligible in comparison.


Asunto(s)
Neutrones , Aceleradores de Partículas , Radioterapia de Alta Energía/instrumentación , Anomalías Inducidas por Radiación/etiología , Seguridad de Equipos , Humanos , Neoplasias Inducidas por Radiación/etiología , Dosis de Radiación , Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA