Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Indian J Public Health ; 62(1): 21-26, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29512561

RESUMEN

BACKGROUND: Increasing antisocial and violent behaviors in adolescents and young adults present serious challenges for public health. Children with persistent high levels of aggressiveness are often associated with developing conduct disorders later in life. Early detection of highly aggressive children and sociodemographic risk-modifying factors are important for developing effective preventive strategies. OBJECTIVES: The present study was undertaken to assess levels of aggressiveness for detecting highly aggressive children in sample populations of primary school children in an urban setting and determine significant biosociocultural risk-modifying factors in this scenario. METHODS: The study was conducted during August-September, 2015 in 5 primary schools of South Delhi Municipal Corporation. Sociodemographic data on 2080 students were collected. Overall aggressiveness scores (OA-Scores) were estimated using a self-report questionnaire in Hindi. RESULTS: Categorizing students according to their OA-Scores, the data revealed that highly aggressive children constituted 4.3% of the study population. Analysis showed significant influence of (a) gender: boys displayed higher levels of aggressiveness compared to girls; (b) dietary pattern: omnivores showed higher aggressiveness than vegetarians; and (c) school environment: boys in mixed-sex (coeducational) schools displayed lower aggressiveness than from single-sex schools. Statistically significant influences of religion (Hindu/Muslim) and family type (joint/nuclear) on aggressiveness profiles were not noticeable. CONCLUSIONS: Vegetarian diets and mixed-sex education act as protective factors in the development of aggressiveness in children, especially among boys. Extending investigations to populations differing in geography and cultural backgrounds are warranted to verify present results.


Asunto(s)
Agresión , Dieta , Instituciones Académicas/estadística & datos numéricos , Población Urbana/estadística & datos numéricos , Violencia/estadística & datos numéricos , Adolescente , Factores de Edad , Niño , Ambiente , Femenino , Humanos , India , Masculino , Religión , Factores de Riesgo , Factores Sexuales , Factores Socioeconómicos
2.
J Cancer Res Ther ; 7(3): 314-24, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22044814

RESUMEN

BACKGROUND: Efficacy of photodynamic therapy can be enhanced by improving uptake, localization, and sub-cellular localization of sensitizers at the sensitive targets. MATERIALS AND METHODS: Uptake, localization, and photodynamic effects of hematoporphyrin derivative (HpD, Photosan-3; PS-3) and disulfonated aluminum phthalocyanine (AlPcS2) were studied either encapsulated in liposomes or conjugated to a monoclonal antibody to carcinoembryonic antigen (anti-CEA) in a brain glioma cell line, BMG-1. RESULTS: Although the total uptake with encapsulated or conjugated sensitizers was less than the free sensitizers, photodynamic efficiency was higher due to the localization of the sensitizer at the sensitive targets. Biodistribution of intravenously administered technetium (99m Tc)-labeled PS-3 analyzed by gamma camera imaging showed maximum accumulation in the liver followed by tumor. Tumor/muscle (T/N) ratio of free PS-3 was higher compared to encapsulated or conjugated PS-3 but the accumulation of PS-3 significantly reduced in brain and cutaneous tissue following modulated delivery. Pharmacokinetics suggested faster accumulation of encapsulated and conjugated PS-3 in the tumor. CONCLUSION: Localization of sensitizers at sensitive targets and reduced accumulation in normal tissues with liposome encapsulation and antibody conjugation suggest that these two delivery systems can potentially enhance the efficacy of photodynamic treatment.


Asunto(s)
Carcinoma de Ehrlich/tratamiento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Animales , Anticuerpos Monoclonales/inmunología , Antígeno Carcinoembrionario/inmunología , Carcinoma de Ehrlich/metabolismo , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Hematoporfirinas/administración & dosificación , Hematoporfirinas/farmacocinética , Hematoporfirinas/farmacología , Indoles/administración & dosificación , Indoles/farmacocinética , Indoles/farmacología , Liposomas , Hígado/metabolismo , Masculino , Ratones , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/farmacocinética , Compuestos Organometálicos/farmacología , Fármacos Fotosensibilizantes/farmacocinética , Distribución Tisular
3.
J Transl Med ; 8: 43, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20433757

RESUMEN

BACKGROUND: Photodynamic therapy (PDT) involves excitation of sensitizer molecules by visible light in the presence of molecular oxygen, thereby generating reactive oxygen species (ROS) through electron/energy transfer processes. The ROS, thus produced can cause damage to both the structure and the function of the cellular constituents resulting in cell death. Our preliminary investigations of dose-response relationships in a human glioma cell line (BMG-1) showed that disulphonated aluminum phthalocyanine (AlPcS2) photodynamically induced loss of cell survival in a concentration dependent manner up to 1 microM, further increases in AlPcS2concentration (>1 microM) were, however, observed to decrease the photodynamic toxicity. Considering the fact that for most photosensitizers only monotonic dose-response (survival) relationships have been reported, this result was unexpected. The present studies were, therefore, undertaken to further investigate the concentration dependent photodynamic effects of AlPcS2. METHODS: Concentration-dependent cellular uptake, sub-cellular localization, proliferation and photodynamic effects of AlPcS2 were investigated in BMG-1 cells by absorbance and fluorescence measurements, image analysis, cell counting and colony forming assays, flow cytometry and micronuclei formation respectively. RESULTS: The cellular uptake as a function of extra-cellular AlPcS2 concentrations was observed to be biphasic. AlPcS2 was distributed throughout the cytoplasm with intense fluorescence in the perinuclear regions at a concentration of 1 microM, while a weak diffuse fluorescence was observed at higher concentrations. A concentration-dependent decrease in cell proliferation with accumulation of cells in G2+M phase was observed after PDT. The response of clonogenic survival after AlPcS2-PDT was non-monotonic with respect to AlPcS2 concentration. CONCLUSIONS: Based on the results we conclude that concentration-dependent changes in physico-chemical properties of sensitizer such as aggregation may influence intracellular transport and localization of photosensitizer. Consequent modifications in the photodynamic induction of lesions and their repair leading to different modes of cell death may contribute to the observed non-linear effects.


Asunto(s)
Glioma/tratamiento farmacológico , Glioma/patología , Indoles/uso terapéutico , Compuestos Organometálicos/uso terapéutico , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Clonales , Daño del ADN , Humanos , Indoles/metabolismo , Indoles/farmacología , Compuestos Organometálicos/metabolismo , Compuestos Organometálicos/farmacología , Fármacos Fotosensibilizantes/farmacología , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Factores de Tiempo
4.
J Cancer Res Ther ; 5 Suppl 1: S21-6, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20009289

RESUMEN

Higher rates of glucose usage generally correlate with poor prognosis in several types of malignant tumours. Experimental studies (both in vitro and in vivo) have shown that 2-deoxy-D-glucose (2-DG), a glucose analog and glycolytic inhibitor, enhances radiation-induced damage selectively in tumor cells while protecting normal cells, thereby suggesting that 2-DG can be used as a differential radiomodifier to improve the efficacy of radiotherapy. Clinical trials undertaken to study the feasibility, safety, and validity of this suggested approach will be described. Based on 2-DG-induced radiosensitization observed in primary organ cultures of cerebral glioma tissues, clinical trials were designed taking into consideration the radiobiology of gliomas and pharmacokinetics of 2-DG. Phase I/II clinical trials have unequivocally demonstrated that a combination of 2-DG (200-300 mg 2-DG per kg body weight orally administered after overnight fasting, 20 min before irradiation) with large weekly fractions (5 Gy/fraction) of low-LET radiotherapy is well tolerated without any acute toxicity or late radiation damage to the normal brain tissue. Nonserious transient side effects similar to hypoglycemia induced disturbances like restlessness, nausea, and vomiting were observed at the 2-DG doses used. Data from these trials involving more than 100 patients have clearly indicated a moderate increase in the survival, with a significant improvement in the quality of life with clinicopathological evidence of protection of normal brain tissue. A phase III multicentric trial to evaluate the efficacy of the combined treatment is in progress. Directions for future studies are discussed.


Asunto(s)
Neoplasias Encefálicas/terapia , Ensayos Clínicos como Asunto , Desoxiglucosa/uso terapéutico , Glioblastoma/terapia , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Radioterapia , Terapia Combinada , Humanos
6.
Mol Imaging Biol ; 10(1): 1-18, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17957408

RESUMEN

Positron emission tomography (PET) using L-[methyl-(11)C]-methionine (MET) is the most popular amino acid imaging modality in oncology, although its use is restricted to PET centers with an in-house cyclotron facility. This review focuses on the role of MET-PET in imaging of cerebral gliomas. The biological background of tumor imaging with methionine is discussed with particular emphasis on cellular amino acid transport, amino acid utilization in brain, normal metabolism of methionine, and its alterations in cancer. The role of MET-PET in clinical management of cerebral gliomas in initial diagnosis, differentiation of tumor recurrence from radiation injury, grading, prognostication, tumor-extent delineation, biopsy planning, surgical resection and radiotherapy planning, and assessment of response to therapy is also reviewed in detail.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Metionina , Tomografía de Emisión de Positrones/métodos , Animales , Radioisótopos de Carbono , Humanos
7.
Cancer Biol Ther ; 5(7): 815-23, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16760658

RESUMEN

The glycolytic inhibitor 2-deoxy-D-glucose (2-DG) has been shown to enhance the cell death induced by radiation and other DNA damaging agents selectively in cells with high rates of glycolysis, like cancer cells. While energy linked modification of DNA and cellular repair processes have been suggested as possible mechanisms of sensitization, other effects such as global stress response cannot be excluded. In this pilot study, we have investigated the effect of 2-DG and radiation on the transcriptome in an attempt to elucidate how 2-DG impacts gene expression in undamaged verses irradiation (IR) damaged cells using a human malignant glioma cell line, U-87. Exponentially growing U-87 cells were exposed to various combinations of 2-DG and X-rays and total RNA was isolated four hours after exposure. Gene expression changes were elucidated using Affymetrix GeneChips. As expected, U-87 cells treated with 2-DG showed activation of several endoplasmic reticulum stress response genes. Selective RT-PCR and Western blotting confirmed these gene alterations. Given that glucose deprivation leads to p53 activation and 2-DG led to activation of p53 response genes in our present study (e.g., PMAIP1 and GADD45A), we examined the impact of transient p53 knockdown and observed that induction of PMAIP1 and GADD45A appear to be via p53-independent mechanisms. The majority of gene alterations seen with IR-treatment alone were consistent with previous reports. While most gene alterations seen with 2-DG and IR dual treatment were confirmed in the gene profiles seen with individual (2-DG or IR) treatments, several genes appeared differentially regulated between IR and 2-DG (e.g., DUSP8, IL8, GADD45B). Additionally, gene expression patterns suggested alterations in cell cycle regulation, apoptosis, and cytokine signaling pathways. Taken together, this study provides new insights into how the transcriptome of tumor cells are likely to be affected by a combined stress caused by IR and 2-DG.


Asunto(s)
Desoxiglucosa/farmacología , Perfilación de la Expresión Génica , Glioma/genética , Glioma/radioterapia , Tolerancia a Radiación/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Retículo Endoplásmico/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Glioma/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcripción Genética/efectos de la radiación , Proteína p53 Supresora de Tumor/agonistas , Rayos X
8.
Strahlenther Onkol ; 181(8): 507-14, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16044218

RESUMEN

BACKGROUND AND PURPOSE: Higher rates of glucose utilization and glycolysis generally correlate with poor prognosis in several types of malignant tumors. Own earlier studies on model systems demonstrated that the nonmetabolizable glucose analog 2-deoxy-D-glucose (2-DG) could enhance the efficacy of radiotherapy in a dose-dependent manner by selectively sensitizing cancer cells while protecting normal cells. Phase I/II clinical trials indicated that the combination of 2-DG, at an oral dose of 200 mg/kg body weight (BW), with large fractions of gamma-radiation was well tolerated in cerebral glioma patients. Since higher 2-DG doses are expected to improve the therapeutic gain, present studies were undertaken to examine the tolerance and safety of escalating 2-DG dose during combined treatment (2-DG + radiotherapy) in glioblastoma multiforme patients. PATIENTS AND METHODS: Untreated patients with histologically proven glioblastoma multiforme (WHO criteria) were included in the study. Seven weekly fractions of (60)Co gamma-rays (5 Gy/fraction) were delivered to the tumor volume (presurgical CT/MRI evaluation) plus 3 cm margin. Escalating 2-DG doses (200-250-300 mg/kg BW) were administered orally 30 min before irradiation after overnight fasting. Acute toxicity and tolerance were studied by monitoring the vital parameters and side effects. Late radiation damage and treatment responses were studied radiologically and clinically in surviving patients. RESULTS: Transient side effects similar to hypoglycemia were observed in most of the patients. Tolerance and patient compliance to the combined treatment were very good up to a 2-DG dose of 250 mg/kg BW. However, at the higher dose of 300 mg/kg BW, two out of six patients were very restless and could not complete treatment, though significant changes in the vital parameters were not observed even at this dose. No significant damage to the normal brain tissue was observed during follow-up in seven out of ten patients who received complete treatment and survived between 11 and 46 months after treatment. CONCLUSION: Oral administration of 2-DG combined with large fractions of radiation (5 Gy/fraction/week) is safe and could be tolerated in glioblastoma patients without any acute toxicity and late radiation damage to the normal brain. Further clinical studies to evaluate the efficacy of the combined treatment are warranted.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Desoxiglucosa/administración & dosificación , Glioblastoma/radioterapia , Radiofármacos/administración & dosificación , Administración Oral , Adulto , Anciano , Neoplasias Encefálicas/mortalidad , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Radioisótopos de Cobalto/uso terapéutico , Desoxiglucosa/efectos adversos , Fraccionamiento de la Dosis de Radiación , Femenino , Estudios de Seguimiento , Glioblastoma/mortalidad , Humanos , Estado de Ejecución de Karnofsky , Masculino , Persona de Mediana Edad , Radiofármacos/efectos adversos , Dosificación Radioterapéutica , Factores de Tiempo
9.
Technol Cancer Res Treat ; 3(3): 295-301, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15161322

RESUMEN

Biodistribution of technetium (99mTc) labeled hematoporphyrin derivative (HpD, Photosan-3) conjugated to a monoclonal antibody to carcinoembryonic antigen (anti-CEA) was compared following intravenous (i.v.) and intratumoral (i.t.) administration in solid Ehrlich ascites tumor bearing mice. Images of mice at different time intervals were acquired after injection of radiolabeled PS-3 in either conjugated or unconjugated forms. Quantitative estimation of the radiolabel in different tissues was performed by selecting the different region of interests (ROIs). Maximum accumulation of both free and antibody conjugated PS-3 following i.v. administration was observed in liver followed by tumor. Tumor/muscle (T/N) ratio was more with free PS-3 compared to conjugated PS-3. Pharmacokinetics of free and conjugated PS-3 was also different with faster accumulation of conjugated PS-3 in the tumor. With intratumoral administration of anti-CEA-PS-3-99mTc, specific accumulation and retention of the sensitizer was observed in the tumor tissue. Since, direct injection of antibody conjugated photosensitizer into the tumor resulted in longer retention of the dye in the tumor with no accumulation in the normal tissues, the present results imply that the toxicity to normal tissues could be reduced significantly with selective destruction of the tumor following photodynamic treatment with the use of i.t. administration of specific antibodies conjugated to photosensitizers.


Asunto(s)
Carcinoma de Ehrlich/terapia , Inmunoconjugados/farmacocinética , Fármacos Fotosensibilizantes/farmacocinética , Radioterapia/métodos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígeno Carcinoembrionario/inmunología , Carcinoma de Ehrlich/metabolismo , Hematoporfirinas , Inmunoconjugados/administración & dosificación , Inyecciones Intravenosas , Hígado/metabolismo , Ratones , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Tecnecio/administración & dosificación , Distribución Tisular
10.
Indian J Exp Biol ; 41(1): 33-40, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15267133

RESUMEN

Photodynamic therapy (PDT) using a photosensitizer, such as haematoporphyrin derivative (HpD), in conjunction with visible light is a promising new modality to treat localized cancer. Cell death caused by PDT (through the generation of reactive oxygen species) can occur either by apoptosis (interphase death or as a secondary event following mitosis) and/or necrosis depending on the cell type, concentration and intracellular localization of the sensitizer, and the light dose. Since, apoptosis induced by PDT treatment plays an important role in determining the photodynamic efficacy, in the present work we have investigated the role of apoptotic cell death in relation to the observed differences in sensitivity to HpD-PDT between a human glioma cell line (BMG-1) carrying wild-type tumour suppressor gene p53 and a human squamous carcinoma cell line (4451) with mutated p53. HpD (photosan-3; PS-3) -PDT induced apoptosis was studied by: [A] flow-cytometric analysis of DNA content (sub G0/G1 population); [B] phosphatidylserine externalization (Annexin-V +ve cells); [C] cell size and cytoskeleton reorganization (light-scatter analysis); and [D] fluorescence microscopy (morphological features). PS-3-PDT induced a significantly higher level of apoptosis in BMG-1 cells as compared to 4451 cells. This was dependent on the concentration of PS-3 as well as post-irradiation time in both the cell lines. At 2.5 microg/ml of PS-3 the fraction of BMG-1 cells undergoing apoptosis (60%) was nearly 6 folds higher than 4451 cells (10%). In BMG-1 cells the induction of apoptosis increased with PS-3 concentration up to 5 microg/ml (>80%). However, a decrease was observed at a concentration of 10 microg/ml, possibly due to a shift in the mode of cell death from apoptosis to necrosis. In 4451 cells, on the other hand, the increase in apoptosis could be observed even up to 10 microg/ml of PS-3 (60%). Present results show that the higher sensitivity to PS-3-PDT in glioma cells arise on account of a higher level of apoptosis and suggest that induction of apoptosis is an important determinant of photodynamic sensitivity in certain cell types.


Asunto(s)
Apoptosis , Fotoquimioterapia , Línea Celular Tumoral , Humanos , Fosfatidilserinas/metabolismo
11.
Indian J Physiol Pharmacol ; 46(1): 51-60, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12024957

RESUMEN

In an attempt to overcome resistance of hypoxic cells to radiotherapy, the combination of a hematoporphyrin derivative (Hpd) and 2-deoxy-D-glucose (2-DG), a promising radiomodifier, was evaluated by assessing the alterations in phosphorylated metabolites and bioenergetics induced in perfused Ehrlich ascites tumor (EAT) cells, using Phosphorus-31 Magnetic Resonance Spectroscopy (31P-MRS). By reducing flow rate of perfusion, a relatively hypoxic condition of tumor was simulated. Changes in bioenergetics levels induced by the combined treatment of Photosan, a Hpd, and 2-DG, under reduced perfusion conditions were more pronounced. Significantly higher uptake of 2-DG and irreversibility of the reduction in cellular bioenergetics induced by the combined treatment, observed under simulated hypoxic conditions, might have considerable implications in optimizing tumor radiotherapy using 2-DG as an adjuvant. These result also suggest the usefulness of this technique to easily simulate hypoxic conditions of tumors in vitro that could be used for rapid in vitro pharmacological evaluation of promising therapeutic strategies.


Asunto(s)
Carcinoma de Ehrlich/metabolismo , Metabolismo Energético/fisiología , Resonancia Magnética Nuclear Biomolecular , Tolerancia a Radiación/fisiología , Animales , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Metabolismo Energético/efectos de los fármacos , Resonancia Magnética Nuclear Biomolecular/métodos , Isótopos de Fósforo , Tolerancia a Radiación/efectos de los fármacos
12.
Radiat Res ; 157(5): 516-25, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11966317

RESUMEN

The hyper-radiosensitivity at low doses recently observed in vitro in a number of cell lines is thought to have important implications for improving tumor radiotherapy. However, cell-cell contact and the cellular environment influence cellular radiosensitivity at higher doses, and they may alter hyper-radiosensitivity in vivo. To confirm this supposition, we investigated the effects of cell density, multiplicity and nutritional deprivation on low-dose hypersensitivity in vitro. Cell survival in the low-dose range (3 cGy to 2 Gy) was studied in cells of two human glioma (BMG-1 and U-87) and two human oral squamous carcinoma (PECA-4451 and PECA-4197) lines using a conventional macrocolony assay. The effects of cell density, multiplicity and nutritional deprivation on hyper-radiosensitivity/induced radioresistance were studied in cells of the BMG-1 cell line, which showed prominent hypersensitivity and induced radioresistance. The induction of growth inhibition, cell cycle delay, micronuclei and apoptosis was also studied at the hyper-radiosensitivity-inducing low doses. Hyper-radiosensitivity/induced radioresistance was evident in the cells of all four cell lines to varying extents, with maximum sensitivity at 10-30 cGy, followed by an increase in survival up to 50 cGy-1 Gy. Both the glioma cell lines had more prominent hyper-radiosensitivity than the two squamous carcinoma cell lines. Low doses inducing maximum hyper-radiosensitivity did not cause significant growth inhibition, micronucleation or apoptosis in BMG-1 cells, but a transient G(1)/S-phase block was evident. Irradiating and incubating BMG-1 cells at high density for 0 or 4 h before plating, as well as irradiating cells as microcolonies, reduced hyper-radiosensitivity significantly, indicating the role of cell-cell contact-mediated processes. Liquid holding of BMG-1 cells in HBSS + 1% serum during and after irradiation for 4 h significantly reduced hyper-radiosensitivity, suggesting that hyper-radiosensitivity may be due partly to active damage fixation processes at low doses. Therefore, our findings suggest that the damage-induced signaling mechanisms influenced by (or mediated through) cell-cell contact or the cellular environment, as well as the lesion fixation processes, play an important role in hyper-radiosensitivity. Further studies are required to determine the exact nature of the damage that triggers these responses as well as for evaluating the potential of low-dose therapy.


Asunto(s)
Comunicación Celular/efectos de la radiación , Ciclo Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Apoptosis/efectos de la radiación , Neoplasias Encefálicas , Recuento de Células , División Celular/efectos de la radiación , Medios de Cultivo , Relación Dosis-Respuesta en la Radiación , Glioma , Humanos , Micronúcleos con Defecto Cromosómico/efectos de la radiación , Pruebas de Micronúcleos , Tolerancia a Radiación/efectos de la radiación , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...