Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Invest New Drugs ; 30(6): 2113-20, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22127459

RESUMO

The poly-(ADP-ribose) polymerase (PARP) inhibitor, MK-4827, is a novel potent, orally bioavailable PARP-1 and PARP-2 inhibitor currently in phase I clinical trials for cancer treatment. No preclinical data currently exist on the combination of MK-4827 with radiotherapy. The current study examined combined treatment efficacy of MK-4827 and fractionated radiotherapy using a variety of human tumor xenografts of differing p53 status: Calu-6 (p53 null), A549 (p53 wild-type [wt]) and H-460 (p53 wt) lung cancers and triple negative MDA-MB-231 human breast carcinoma. To mimic clinical application of radiotherapy, fractionated radiation (2 Gy per fraction) schedules given once or twice daily for 1 to 2 weeks combined with MK-4827, 50 mg/kg once daily or 25 mg/kg twice daily, were used. MK-4827 was found to be highly and similarly effective in both radiation schedules but maximum radiation enhancement was observed when MK-4827 was given at a dose of 50 mg/kg once daily (EF = 2.2). MK-4827 radiosensitized all four tumors studied regardless of their p53 status. MK-4827 reduced PAR levels in tumors by 1 h after administration which persisted for up to 24 h. This long period of PARP inhibition potentially adds to the flexibility of design of future clinical trials. Thus, MK-4827 shows high potential to improve the efficacy of radiotherapy.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias da Mama/terapia , Indazóis/administração & dosagem , Neoplasias Pulmonares/terapia , Piperidinas/administração & dosagem , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Quimiorradioterapia , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Poli(ADP-Ribose) Polimerases/metabolismo , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Invest New Drugs ; 29(4): 554-61, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20119866

RESUMO

PURPOSE: The present study investigated the effect of AC480, a small molecule pan-HER tyrosine kinase inhibitor, on in vitro radiosensitivity and in vivo radioresponse of a human head and neck squamous cell carcinoma cell line. METHODS: HN-5 cells were exposed to γ-radiation with and without AC480 and assayed for proliferation, clonogenic survival, apoptosis, cell cycle distribution, and DNA damage. The cells were analyzed by immunoprecipitation and western blotting for proteins involved in apoptosis, cell cycle regulation, and the EGFR pathway. The effect of AC480 on tumor radioresponse was assessed by tumor growth delay assay using HN5 tumor xenografts generated in nude mice. RESULTS: At the molecular level, in HN-5 cells the agent inhibited the expression of pEGFR, pHER2, cyclins D and E, pRb, pAkt, pMAPK, pCDK1 and 2, CDK 6, and Ku70 proteins. The drug also induced accumulation of cells in the G1 cell cycle phase, inhibited cell growth, enhanced radiosensitivity, and prolonged the presence of γ-H2AX foci up to 24 h after radiation. AC480 did not increase the percentage of cells undergoing radiation-induced apoptosis. The drug given before and during irradiation improved the radioresponse of HN5 tumors in vivo. CONCLUSION: AC480 significantly enhanced the radiosensitivity of HN-5 cells, expressing both EGFR and Her2. The mechanisms involved in the enhancement included cell cycle redistribution and inhibition of DNA repair. Both in vitro and in vivo data from our study suggest that AC480 has potential to increase tumor response to radiotherapy.


Assuntos
Carbamatos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Receptor ErbB-2/antagonistas & inibidores , Triazinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Western Blotting , Carbamatos/uso terapêutico , Carcinoma de Células Escamosas/patologia , Contagem de Células , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Receptores ErbB/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Histonas/metabolismo , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Nus , Radiação Ionizante , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triazinas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Invest New Drugs ; 29(3): 514-22, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20024691

RESUMO

Check point kinases (Chk) play a major role in facilitating DNA repair upon radiation exposure. We tested the potency of a novel inhibitor of Chk1 and Chk2, XL-844 (provided by Exelixis Inc., CA, USA), to radiosensitize human cancer cells grown in culture and investigated the underlying mechanisms. HT-29 cells (a human colon cancer line) were exposed to XL-844, radiation, or both, and assessed for clonogenic cell survival. Treatment-dependent effects on phosphorylated forms of Chk proteins were assessed by Western blots. Further mechanistic investigations in HT-29 cells included cell cycle analysis by flowcytometry and assessment of DNA repair kinetics by immuno-cytochemistry (ICC) for nuclear appearance of the phosphorylated form of histone 2AX protein (γ-H2AX) staining. Cells undergoing mitotic catastrophe were identified by irregular pattern of mitotic spindle markers α and γ-tubulin staining by ICC. XL-844 enhanced radiosensitivity in a dose and schedule-dependent manner and the enhancement factor was 1.42 at 0.5 survival fraction. Mechanistically XL-844 abrogated radiation-induced Chk2 phosphorylation, induced pan-nuclear γ-H2AX, and prolonged the presence of radiation-induced γ-H2AX foci, and promoted mitotic catastrophe. In conclusion, our data showed that inhibition of Chk2 activity by XL-844 enhanced cancer cell radiosensitivity that was associated with inhibition of DNA repair and induction of mitotic catastrophe.


Assuntos
Antineoplásicos/farmacologia , Mitose/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Tolerância a Radiação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Histonas/metabolismo , Humanos , Imuno-Histoquímica , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
4.
Future Oncol ; 6(10): 1563-76, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21062156

RESUMO

Based on findings that cancer cell clonogens exhibit stem cell features, it has been suggested that cancer stem-like cells are relatively radioresistant owing to different intrinsic and extrinsic factors, including quiescence, activated radiation response mechanisms (e.g., enhanced DNA repair, upregulated cell cycle control mechanisms and increased free-radical scavengers) and a surrounding microenvironment that enhances cell survival mechanisms (e.g., hypoxia and interaction with stromal elements). However, these radiosensitivity features are probably dynamic in nature and come into play at different times during the course of chemo/radiotherapy. Therefore, different molecularly targeted radiosensitization strategies may be needed at different stages of therapy. This article describes potential sensitization approaches based on the dynamics and changing properties of cancer stem-like cells during therapy.


Assuntos
Células-Tronco Neoplásicas/efeitos da radiação , Tolerância a Radiação/fisiologia , Animais , Humanos
5.
Int J Radiat Biol ; 85(2): 107-15, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19280463

RESUMO

PURPOSE: Apoptosis, as a mode of cell death in irradiated cell populations, has been the subject of literarily hundreds if not thousands of published reports over the past few years. However, in spite of the large body of knowledge related to this subject, the role of apoptosis in determining tumor response to radiotherapy has been and remains poorly understood and controversial. Indeed, some previous reviews have suggested that apoptosis may not be important in this context. The purpose of the present review is to provide some examples of recently reported laboratory investigations that indicate that there is a reasonable expectation that the radiation-induced apoptosis observed has contributed to the tumor response. CONCLUSIONS: We review reports in four areas of research: Molecularly targeted agents, in vivo imaging, Bcl-2 and cancer stem cells. Examples are provided in each of these areas that we believe justify a reassessment of the role that apoptosis plays in radiation oncology.


Assuntos
Apoptose , Neoplasias/radioterapia , Animais , Anexina A5/análise , Dano ao DNA , Células Epiteliais/citologia , Receptores ErbB/antagonistas & inibidores , Humanos , Mesoderma/citologia , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
6.
Int J Radiat Oncol Biol Phys ; 70(1): 203-12, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17996386

RESUMO

PURPOSE: The purpose of the study was to elucidate the molecular mechanisms mediating radiosensitization of human tumor cells by the selective cyclooxygenase (COX)-2 inhibitor celecoxib. METHODS AND MATERIALS: Experiments were performed using bronchial carcinoma cells A549, transformed fibroblasts HH4dd, the FaDu head-and-neck tumor cells, the colon carcinoma cells HCT116, and normal fibroblasts HSF7. Effects of celecoxib treatment were assessed by clonogenic cell survival, Western analysis, and quantification of residual DNA damage by gammaH(2)AX foci assay. RESULTS: Celecoxib treatment resulted in a pronounced radiosensitization of A549, HCT116, and HSF7 cells, whereas FaDu and HH4dd cells were not radiosensitized. The observed radiosensitization could neither be correlated with basal COX-2 expression pattern nor with basal production of prostaglandin E2, but was depended on the ability of celecoxib to inhibit basal and radiation-induced nuclear transport of epidermal growth factor receptor (EGFR). The nuclear EGFR transport was strongly inhibited in A549-, HSF7-, and COX-2-deficient HCT116 cells, which were radiosensitized, but not in FaDu and HH4dd cells, which resisted celecoxib-induced radiosensitization. Celecoxib inhibited radiation-induced DNA-PK activation in A549, HSF7, and HCT116 cells, but not in FaDu and HH4dd cells. Consequentially, celecoxib increased residual gammaH2AX foci after irradiation, demonstrating that inhibition of DNA repair has occurred in responsive A549, HCT116, and HSF7 cells only. CONCLUSIONS: Celecoxib enhanced radiosensitivity by inhibition of EGFR-mediated mechanisms of radioresistance, a signaling that was independent of COX-2 activity. This novel observation may have therapeutic implications such that COX-2 inhibitors may improve therapeutic efficacy of radiation even in patients whose tumor radioresistance is not dependent on COX-2.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Reparo do DNA , Receptores ErbB/metabolismo , Pirazóis/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Sulfonamidas/farmacologia , Western Blotting , Celecoxib , Linhagem Celular Tumoral/efeitos da radiação , Sobrevivência Celular , Ciclo-Oxigenase 2/metabolismo , Proteína Quinase Ativada por DNA/metabolismo , Dinoprostona/metabolismo , Fibroblastos/efeitos da radiação , Células HCT116/efeitos da radiação , Humanos
7.
Clin Cancer Res ; 13(6): 1868-74, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363543

RESUMO

PURPOSE: 130-nm albumin-bound paclitaxel (nab-paclitaxel) is a novel solvent-free albumin-bound paclitaxel, designed to avoid solvent-related toxicity. Nab-paclitaxel has been successfully introduced into the clinic but its radiation-enhancing potential has not yet been evaluated. We conducted a preclinical evaluation of the radiation-modulating effects of nab-paclitaxel in tumor and normal tissues. EXPERIMENTAL DESIGN: Mice bearing syngeneic ovarian or mammary carcinomas were treated with nab-paclitaxel, radiation, or combination of both. Nab-paclitaxel was administered at 90 mg/kg, 1.5 times the maximum tolerated dose for solvent-based paclitaxel. End points were antitumor efficacy (growth delay, radiocurability, and cellular effects) and normal tissue toxicity (gut and skin). RESULTS: Nab-paclitaxel showed single-agent antitumor efficacy against both tumor types and acted as a radiosensitizer. Combined with radiation, nab-paclitaxel produced supra-additive effects when given before radiation. Nab-paclitaxel significantly increased radiocurability by reducing the dose yielding 50% tumor cure (TCD(50)) from 54.3 to 35.2 Gy. Tumor histology following nab-paclitaxel treatment was characterized by pronounced necrotic and apoptotic cell death and mitotic arrest. Nab-paclitaxel did not increase normal tissue radioresponse. CONCLUSIONS: Nab-paclitaxel exhibited strong antitumor efficacy against both tumors as a single agent and it improved radiotherapy in a supra-additive manner. These improved effects were achieved without increased normal tissue toxicity to either rapidly or slowly proliferating normal tissues although the drug dose was 1.5 times higher than the maximum tolerated dose of solvent-based paclitaxel. These preclinical findings show that combining nab-paclitaxel with radiotherapy would improve the outcome of taxane-based chemoradiotherapy. This novel taxane is thus a good candidate for testing in clinical chemoradiotherapy trials.


Assuntos
Adenocarcinoma/radioterapia , Albuminas/uso terapêutico , Neoplasias Mamárias Experimentais/radioterapia , Neoplasias Ovarianas/radioterapia , Paclitaxel/uso terapêutico , Radiossensibilizantes/uso terapêutico , Adenocarcinoma/patologia , Albuminas/química , Animais , Antineoplásicos/uso terapêutico , Terapia Combinada , Feminino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Neoplasias Ovarianas/patologia , Paclitaxel/química , Doses de Radiação , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação
8.
Int J Radiat Oncol Biol Phys ; 67(2): 568-72, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17236972

RESUMO

PURPOSE: C225 strongly enhances tumor radioresponse when given concurrently with radiotherapy. We investigated whether additional therapeutic benefit could be achieved by continuing maintenance treatment with C225 after the completion of fractionated radiotherapy. METHODS AND MATERIALS: A431 xenografts were treated with local irradiation or combined with C225 by two different schedules: (1) 6 h before the first dose of irradiation and at 3-day intervals for a total of 3 doses during the 7-day fractionated radiotherapy, or (2) 6 doses of C225 given both during radiotherapy and continuing for 3 additional doses after radiotherapy. Tumor cure was assessed by the radiation dose yielding local tumor control in 50% of animals (TCD50), and time to recurrence was also determined. RESULTS: Both treatment schedules increased radiocurability as evidenced by reductions in TCD50, but the effect was greater when C225 was given both during and after radiotherapy. C225 reduced the TCD50 of 83.1 (73.2-124.8) Gy by radiation only to 46.2 (39.1-57.5) Gy when given during radiotherapy and to 30.8 (22.2-38.0) Gy when given during and after radiotherapy. Dose modification factors were 1.8 when C225 was given during radiotherapy and 2.7 when given both during and after radiotherapy. C225 was also effective in delaying the onset of tumor recurrences, and was more effective when given as both concurrent and maintenance therapy. CONCLUSIONS: Data showed that C225 strongly enhanced the curative effect of fractionated radiation, and its effect was greater if administration was extended beyond the end of radiotherapy. This important finding may influence future designs of clinical trials combining anti-EGFR (anti-epidermal growth factor receptor) agents with radiotherapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Animais , Anticorpos Monoclonais Humanizados , Cetuximab , Fracionamento da Dose de Radiação , Esquema de Medicação , Feminino , Camundongos , Camundongos Nus , Recidiva Local de Neoplasia/prevenção & controle , Tolerância a Radiação/fisiologia
9.
Int J Radiat Oncol Biol Phys ; 67(3): 870-8, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17293237

RESUMO

PURPOSE: Clinical trials of antiangiogenic agents used alone for advanced malignancy have been disappointing but preclinical studies suggest that the addition of radiation therapy could improve antitumor efficacy. To test the hypothesis that antiangiogenic therapy combined with radiation therapy can overcome the limitations of antiangiogenic monotherapy, we studied the effects of endostatin combined with radiation on the growth and vascularization of A431 human epidermoid carcinomas growing intramuscularly in the legs of mice. METHODS AND MATERIALS: Mice with established A431 human epidermoid leg tumors were treated with radiation, endostatin, both radiation and endostatin, or vehicle control. The experiment was repeated and mice from each group were killed at 2, 7, and 10 days after irradiation so that tumor tissue could be obtained to further analyze the kinetics of the antitumor, antivascular, and antiangiogenic response to therapy. RESULTS: Endostatin enhanced the antitumor effects of radiation, and prolonged disease-free survival was observed in the combined treatment group. Endothelial cell proliferation was increased in tumors after irradiation but was blocked by the concurrent administration of endostatin, and the combination of endostatin with radiation enhanced endothelial cell apoptosis within 48 h after irradiation. Expression of vascular endothelial growth factor, interleukin-8, and matrix metalloproteinase-2 were increased in tumors after irradiation, and this increase was blocked by concurrent administration of endostatin. CONCLUSION: These data indicate that endostatin can block tumor revascularization after radiation therapy and thereby augment radioresponse.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/radioterapia , Endostatinas/uso terapêutico , Neovascularização Patológica/prevenção & controle , Proteínas Angiogênicas/metabolismo , Animais , Apoptose , Carcinoma de Células Escamosas/patologia , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Masculino , Camundongos , Camundongos Nus , Tolerância a Radiação , Transplante Heterólogo
10.
Int J Radiat Oncol Biol Phys ; 69(5): 1534-43, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17889445

RESUMO

PURPOSE: Conventional therapies for patients with lung cancer have reached a therapeutic plateau. We therefore evaluated the feasibility of combined vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) and epidermal growth factor (EGF) receptor (EGFR) targeting with radiation therapy in an orthotopic model that closely recapitulates the clinical presentation of human lung cancer. METHODS AND MATERIALS: Effects of irradiation and/or ZD6474, a small-molecule inhibitor of VEGFR2 and EGFR tyrosine kinases, were studied in vitro for human lung adenocarcinoma cells by using proliferation and clonogenic assays. The feasibility of combining ZD6474 with radiation therapy was then evaluated in an orthotopic model of human lung adenocarcinoma. Lung tumor burden and spread within the thorax were assessed, and tumor and adjacent tissues were analyzed by means of immunohistochemical staining for multiple parameters, including CD31, VEGF, VEGFR2, EGF, EGFR, matrix metalloproteinase-2 and -9, and basic fibroblast growth factor. RESULTS: ZD6474 enhanced the radioresponse of NCI-H441 human lung adenocarcinoma cells by a factor of 1.37 and markedly inhibited sublethal damage repair. In vivo, the combined blockade of VEGFR2 and EGFR by ZD6474 blocked pleural effusion formation and angiogenesis and enhanced the antivascular and antitumor effects of radiation therapy in the orthotopic human lung cancer model and was superior to chemoradiotherapy. CONCLUSIONS: When radiation therapy is combined with VEGFR2 and EGFR blockade, significant enhancement of antiangiogenic, antivascular, and antitumor effects are seen in an orthotopic model of lung cancer. These data provide support for clinical trials of biologically targeted and conventional therapies for human lung cancer.


Assuntos
Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/radioterapia , Neovascularização Patológica/prevenção & controle , Piperidinas/uso terapêutico , Quinazolinas/uso terapêutico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Proliferação de Células , Terapia Combinada/métodos , Reparo do DNA/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Estudos de Viabilidade , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Nus , Derrame Pleural/prevenção & controle , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/uso terapêutico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Drugs ; 67(6): 821-45, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17428102

RESUMO

Despite recent improvements in chemotherapy and radiation therapy in cancer management with the addition of biological agents, novel treatment approaches are needed to further benefit patients. Cyclo-oxygenase (COX)-2 inhibition represents one such possibility. COX-2 is an enzyme induced in pathological states such as inflammatory disorders and cancer, where it mediates production of prostanoids. The enzyme is commonly expressed in both premalignant lesions and malignant tumours of different types. A growing body of evidence suggests an association of COX-2 with tumour development, aggressive biological tumour behaviour, resistance to standard cancer treatment, and adverse patient outcome. COX-2 may be related to cancer development and propagation through multiple mechanisms, including stimulation of growth, migration, invasiveness, resistance to apoptosis, suppression of the immunosurveillance system, and enhancement of angiogenesis. Epidemiological data suggest that NSAIDs and selective COX-2 inhibitors might prevent the development of cancers, including colorectal, oesophageal and lung cancer. Preclinical investigations have demonstrated that inhibition of this enzyme with selective COX-2 inhibitors enhances tumour response to radiation and chemotherapeutic agents. These preclinical findings have been rapidly advanced to clinical oncology. Clinical trials of the combination of selective COX-2 inhibitors with radiotherapy, chemotherapy or both in patients with a number of cancers have been initiated, and preliminary results are encouraging. This review discusses the role of COX-2, its products (prostaglandins) and its inhibitors in tumour growth and treatment.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/fisiologia , Neoplasias/tratamento farmacológico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia Combinada , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias/fisiopatologia , Prostaglandinas/biossíntese , Prostaglandinas/fisiologia
12.
Oncol Rep ; 18(3): 645-51, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17671714

RESUMO

To determine whether a polymer-drug conjugate might improve tumor cell kill compared to the corresponding unconjugated agent when used in combination with radiation, we examined the antitumor activity of a water-soluble conjugate of N-(4-hydroxyphenyl) retinamide (4HPR) and poly(L-glutamic acid), PG-4HPR, in lung cancer cells and xenografts. The antiproliferative activity of 4HPR and PG-4HPR in human lung cancer A549 cells was evaluated and the response of the cells to radiation measured by clonogenic assay. Response to irradiation was evaluated by measuring tumor growth delay in nude mice bearing intramuscularly inoculated A549 tumors. Histologic responses were assessed by examination of apoptosis (TUNEL assay) and cell proliferation (Ki67 staining). In vitro, 4HPR and PG-4HPR inhibited the proliferation of A549 cells, with IC50 values of 25.8 microM and >50 microM, respectively, after 24 h of continuous exposure and 6.25 microM and 9.75 microM, respectively, after 120 h. Both agents increased radiosensitivity at an equivalent 4HPR concentration of 10 microM after 5 days of exposure, with enhancement factors of 1.40 and 1.43. In tumor xenografts, intravenous injection of 4HPR or PG-4HPR (30 mg eq. 4HPR/kg) enhanced radiosensitivity by 1.3 and 1.6, respectively, without apparent systemic toxicity. PG-4HPR augmented radiation-induced apoptosis and decreased cellular proliferation in vivo. The radiation response of A549 tumors was greater with PG-4HPR than with 4HPR, which may be attributed to increased delivery of 4HPR to the tumors and enhanced apoptotic response. These results suggest that a polymeric delivery system may be useful for modulating radiosensitivity.


Assuntos
Anticarcinógenos/uso terapêutico , Fenretinida/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/radioterapia , Apoptose , Linhagem Celular Tumoral , Humanos , Marcação In Situ das Extremidades Cortadas , Neoplasias Pulmonares/patologia , Ácido Poliglutâmico , Pró-Fármacos/uso terapêutico
13.
Clin Cancer Res ; 12(15): 4590-7, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16899606

RESUMO

PURPOSE: Early identification of esophageal cancer patients who are responding or resistant to combined chemoradiotherapy may lead to individualized therapeutic approaches and improved clinical outcomes. We assessed the ability of 3'-deoxy-3'-(18)F-fluorothymidine positron emission tomography (FLT-PET) to detect early changes in tumor proliferation after chemoradiotherapy in experimental models of esophageal carcinoma. EXPERIMENTAL DESIGN: The in vitro and ex vivo tumor uptake of [(3)H]FLT in SEG-1 human esophageal adenocarcinoma cells were studied at various early time points after docetaxel plus irradiation and validated with conventional assessments of cellular proliferation [thymidine (Thd) and Ki-67] and [(18)F]FLT micro-PET imaging. Imaging-histologic correlation was determined by comparing spatial Ki-67 and [(18)F]FLT distribution in autoradiographs. Comparison with fluorodeoxyglucose (FDG) was done in all experiments. RESULTS: In vitro [(3)H]FLT and [(3)H]Thd uptake rapidly decreased in SEG-1 cells 24 hours after docetaxel with a maximal reduction of over 5-fold (P = 0.005). The [(3)H]FLT tumor-to-muscle uptake ratio in xenografts declined by 75% compared with baseline (P < 0.005) by 2 days after chemoradiotherapy, despite the lack of change in tumor size. In contrast, the decline of [(3)H]FDG uptake was gradual and less pronounced. Tumor uptake of [(3)H]FLT was more closely correlated with Ki-67 expression (r = 0.89, P < 0.001) than was [(3)H]FDG (r = 0.39, P = 0.08). Micro-PET images depicted similar trends in reduction of [(18)F]FLT and [(18)F]FDG tumor uptake. Autoradiographs displayed spatial correlations between [(18)F]FLT uptake and histologic Ki-67 distribution in preliminary studies. CONCLUSIONS: FLT-PET is suitable and more specific than FDG-PET for depicting early reductions in tumor proliferation that precede tumor size changes after chemoradiotherapy.


Assuntos
Didesoxinucleosídeos , Neoplasias Esofágicas/diagnóstico por imagem , Compostos Radiofarmacêuticos , Animais , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Terapia Combinada , Didesoxinucleosídeos/farmacocinética , Neoplasias Esofágicas/patologia , Radioisótopos de Flúor , Humanos , Masculino , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Relação Estrutura-Atividade , Transplante Heterólogo , Trítio , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Int J Radiat Oncol Biol Phys ; 64(2): 551-61, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16414371

RESUMO

PURPOSE: The International Atomic Energy Agency (IAEA) held a Technical Meeting of Consultants to (1) discuss a selection of relatively new agents, not those well-established in clinical practice, that operated through a variety of mechanisms to sensitize tumors to radiation and (2) to compare and contrast their tumor efficacy, normal tissue toxicity, and status of development regarding clinical application. The aim was to advise the IAEA as to which developing agent or class of agents would be worth promoting further, by supporting additional laboratory research or clinical trials, with the eventual goal of improving cancer control rates using radiotherapy, in developing countries in particular. RESULTS: The agents under discussion included a wide, but not complete, range of different types of drugs, and antibodies that interfered with molecules in cell signaling pathways. These were contrasted with new molecular antisense and gene therapy strategies. All the drugs discussed have previously been shown to act as tumor cell radiosensitizers or to kill hypoxic cells present in tumors. CONCLUSION: Specific recommendations were made for more preclinical studies with certain of the agents and for clinical trials that would be suitable for industrialized countries, as well as trials that were considered more appropriate for developing countries.


Assuntos
Neoplasias/radioterapia , Radiossensibilizantes/uso terapêutico , Inibidores da Angiogênese/uso terapêutico , Apoptose/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Terapia Genética/métodos , Agências Internacionais , Oligorribonucleotídeos Antissenso/uso terapêutico
15.
Radiother Oncol ; 80(2): 185-91, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16905211

RESUMO

BACKGROUND AND PURPOSE: Concurrent chemo-radiotherapy before surgery is standard treatment protocol for esophageal cancer with a less than 30% complete response due to resistance to therapy. The aim of this study was to determine whether molecular targeting approach using an inhibitor of cyclin-dependent kinases, flavopiridol, can help overcome the resistance to radiotherapy. MATERIALS AND METHODS: SEG-1 cells (human esophageal adenocarcinoma) were exposed to gamma-rays with and without flavopiridol treatment and assayed for clonogenic survival, apoptosis, cell cycle distribution, and Western blot analysis. Efficacy of flavopiridol in enhancing tumor response to radiation was determined by tumor growth delay assay using SEG-1 tumor xenografts generated in nude mice. RESULTS: The clonogenic cell survival assay data showed that flavopiridol (300 nM, 24h), when given either before or after radiation, significantly enhanced the radiosensitivity of SEG-1 cells. The cells were accumulated at G1 phase of the cell cycle by flavopiridol that was associated with downregulation of p-cdk-1, p-cdk-2, cyclin D1 and p-Rb expression. Flavopiridol by itself induced apoptosis in SEG-1 cells and also enhanced the radiation-induced apoptosis, associated with an increase in cleaved poly ADP-ribose polymerase. Reduction in phosphorylation of RNA polymerase II by flavopiridol suggested that flavopiridol inhibited the transcriptional activity. In vivo studies with SEG-1 tumor xenografts showed that flavopiridol, either given before or after radiation, greatly enhanced the effect of tumor irradiation. CONCLUSIONS: Flavopiridol treatment significantly enhanced SEG-1 cell radiosensitivity as well as the radioresponse of SEG-1 tumor xenografts. The underlying mechanisms are multiple, including cell cycle redistribution, apoptosis, and transcriptional inhibition. These preclinical data suggest that flavopiridol has the potential to increase the radioresponse of esophageal adenocarcinomas.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/radioterapia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/radioterapia , Flavonoides/farmacologia , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta a Droga , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/patologia , Humanos , Camundongos , Camundongos Nus , Tolerância a Radiação/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Radiother Oncol ; 80(2): 192-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16905212

RESUMO

BACKGROUND AND PURPOSE: Synthetic oligodeoxynucleotides (ODNs) containing unmethylated cytosine-guanine (CpG) motifs bind to Toll-like receptor 9 (TLR9) and stimulate both innate and adaptive immune reactions and possess anti-tumor activity. We recently reported that CpG ODN 1826 strongly enhances radioresponse of both immunogenic [Milas L, Mason K, Ariga H, et al. CpG oligodeoxynucleotide enhances tumor response to radiation. Cancer Res 2004;64:5074-7] and non-immunogenic [Mason KA, Ariga H, Neal R, et al. Targeting toll-like receptor-9 with CpG oligodeoxynucleotides enhances tumor response to fractionated radiotherapy. Clin Cancer Res 2005;11:361-9] murine tumors. Using two immunogenic murine tumors, a fibrosarcoma (FSa) and a mammary carcinoma (MCa-K), the present study explored whether CpG ODN 1826 also improves the response of murine tumors to the chemotherapeutic agent docetaxel (DOC). MATERIALS AND METHODS: CpG ODN 1826 (100 microg) was given sc three times: when leg tumors were 6mm, when they grew to 8mm and again 1 week later. DOC (33 mg/kg iv) and local tumor radiation (10Gy) were given when tumors were 8mm. Effects of the treatments were assayed by tumor growth delay, defined as days for tumors to grow from 8 to 12 mm in diameter. RESULTS: Treatment with CpG ODN 1826 resulted in strongly enhanced response of FSa tumors to radiation and MCa-K tumors to the chemotherapeutic agent DOC. Enhancement of tumor treatment response was demonstrated by a strong prolongation in the primary tumor treatment endpoint, tumor growth delay. Coincidentally, this treatment also resulted in a higher rate of tumor cure than that observed after tumor radiotherapy or chemotherapy alone. When all three agents were combined the effect was comparable to that of the combination of CpG ODN 1826 with radiation in the case of FSa or of the combination of CpG ODN 1826 with DOC in the case of MCa-K. CONCLUSION: Overall results show that CpG ODN 1826 can markedly improve tumor response to radiation and chemotherapy (DOC), suggesting that CpG ODNs have potential to be beneficial when used singly or in combination with other standard treatment modalities such as taxane chemotherapy, radiotherapy or both.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , DNA/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/radioterapia , Taxoides/farmacologia , Animais , Terapia Combinada , DNA/administração & dosagem , Docetaxel , Sinergismo Farmacológico , Feminino , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos da radiação , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/radioterapia , Camundongos , Neoplasias Experimentais/genética , Oligodesoxirribonucleotídeos , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/genética , Sarcoma Experimental/patologia , Sarcoma Experimental/radioterapia , Taxoides/administração & dosagem
17.
Oncol Rep ; 16(3): 465-71, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16865244

RESUMO

S-1, a novel oral fluoropyrimidine, is increasingly used for the treatment of human cancer including gastrointestinal carcinomas. Using the 5-FU resistant DLD-1/FU human colon cancer cell xenografts, the present study investigated whether S-1 enhances the therapeutic efficacy of radiation and if so what are the underlying mechanisms. Nude mice bearing tumor xenografts were treated with radiation, S-1, or both. Tumor growth delay was the treatments' endpoint. To determine whether S-1 enhances intrinsic cell radiosensitivity, we performed clonogenic cell survival assay. Also we assessed the expression of thymidylate synthase (TS) using immunohistochemistry assay. While S-1 or 5 Gy were only slightly effective as single agents in delaying tumor growth, the combined treatment was highly effective. Clonogenic cell survival showed that S-1 strongly enhanced cell radiosensitivity. Immunohistochemistry showed that the expression of TS was down-regulated in tumors treated by S-1 plus radiation. Combined S-1 plus radiation treatment resulted in a synergistic effect in the therapy of 5-FU resistant human colon carcinoma xenografts (EF = 2.06). The effect could be attributed to the ability of S-1 to increase cell radiosensitivity (EF = 1.9) and to the down-regulation of TS involved in cellular processes leading to radio- and (or) chemo-resistance.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/radioterapia , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/uso terapêutico , Ácido Oxônico/uso terapêutico , Radiossensibilizantes/uso terapêutico , Tegafur/uso terapêutico , Administração Oral , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Combinação de Medicamentos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Clin Cancer Res ; 11(1): 361-9, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15671567

RESUMO

Synthetic oligodeoxynucleotides containing unmethylated CpG motifs detected by Toll-like receptor 9 of dendritic cells and B cells have potent immunomodulatory effects. CpG oligodeoxynucleotides induce cytokines, activate natural killer cells, and elicit T-cell responses leading to antitumor effects, including improved efficacy of chemotherapeutic agents and, as we reported recently, synergy between CpG oligodeoxynucleotide 1826 and single-dose radiotherapy of an immunogenic mouse fibrosarcoma. The present study extends this finding to the fractionated radiotherapy of the fibrosarcoma tumor and assesses the ability of CpG oligodeoxynucleotide 1826 to increase the radioresponse of a tumor (nonimmunogenic fibrosarcoma). The experiments used a murine immunogenic fibrosarcoma tumor, fibrosarcoma growing in the leg of mice, and response to radiotherapy was assessed by tumor growth delay and tumor cure rate (TCD50, radiation dose yielding 50% tumor cure). Multiple s.c. peritumoral or i.t. administrations of CpG oligodeoxynucleotide 1826 at a dose of 100 microg per mouse were given when established tumors were 6 mm in diameter. Local tumor irradiation was initiated when tumors grew to 8 mm in diameter; radiation was delivered in 1 to 9 Gy fractions given twice daily separated by 6 to 7 hours for 5 consecutive days to achieve a total dose of 10 to 90 Gy. CpG oligodeoxynucleotide 1826, given as a single agent, had only a small antitumor effect, but it dramatically enhanced fibrosarcoma response to radiotherapy. Although 83.1 (79.2-90.0) Gy total dose were needed to achieve tumor cures in 50% of mice treated with radiotherapy alone, only 23.0 (11.5-32.7) Gy total dose were needed in mice treated with both CpG oligodeoxynucleotide 1826 and radiotherapy. The magnitude of potentiation of tumor radioresponse at the TCD50 level was by a factor of 3.61, a much higher value than that (a factor of 1.93) that we reported for single-dose radiotherapy. Mice cured of their tumors by combined CpG oligodeoxynucleotide 1826 plus radiotherapy were highly resistant to s.c. tumor take or development of tumor nodules in the lung from i.v. injected tumor cells when rechallenged with fibrosarcoma cells 100 to 120 days after the treatment, suggesting the development of a memory response. CpG oligodeoxynucleotide 1826 also increased radioresponse of the nonimmunogenic fibrosarcoma tumor by a factor of 1.41 and 1.73 when CpG oligodeoxynucleotide 1826 was given s.c. and i.t., respectively. These findings show that CpG oligodeoxynucleotides are highly potent enhancers of tumor response to both single-dose and fractionated radiation and as such have potential to improve clinical radiotherapy.


Assuntos
Ilhas de CpG , Proteínas de Ligação a DNA/genética , Fracionamento da Dose de Radiação , Imunoterapia/métodos , Neoplasias/genética , Neoplasias/radioterapia , Receptores de Superfície Celular/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta à Radiação , Fibrossarcoma/química , Fibrossarcoma/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Oligonucleotídeos/química , Fatores de Tempo , Receptor Toll-Like 9
19.
Clin Cancer Res ; 11(9): 3342-8, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15867233

RESUMO

OBJECTIVES: Preclinical observations that selective cyclooxygenase-2 inhibitors enhance in vitro cell radiosensitivity and in vivo tumor radioresponse led to clinical trials testing therapeutic efficacy of these agents. Our study was designed to determine whether the COX-2 inhibitor celecoxib could be safely administered in doses within those approved by the Food and Drug Administration when used concurrently with thoracic radiotherapy in patients with poor prognosis non-small cell lung cancer (NSCLC). PATIENTS AND METHODS: The trial consisted of three cohorts of patients: (a) locally advanced NSCLC with obstructive pneumonia, hemoptysis, and/or minimal metastatic disease treated with 45 Gy in 15 fractions; (b) medically inoperable early-stage NSCLC treated with definitive radiation of 66 Gy in 33 fractions; and (c) patients who received induction chemotherapy but who were not eligible for concurrent chemoradiotherapy trials. These patients received 63 Gy in 35 fractions. Celecoxib was administered p.o. on a daily basis 5 days before and throughout the course of radiotherapy. Celecoxib doses were escalated from 200, 400, 600, to 800 mg/d given in two equally divided doses. Two to eight patients of each cohort were assigned to each dose level of celecoxib. RESULTS: Forty-seven patients were enrolled in this protocol (19 in cohort I, 22 in cohort II, and 6 in cohort III). The main toxicities were grades 1 and 2 nausea and esophagitis, and they were independent of the dose of celecoxib or radiotherapy schedule. Only two patients in group II developed grade 3 pneumonitis 1 month after treatment, one on 200 mg, and the other on 400 mg celecoxib. Celecoxib-related toxicity developed in 3 of 47 patients: an uncontrolled hypertension in one patient on 800 mg celecoxib and hemorrhagic episodes in 2 patients (shoulder hematoma in one and hemoptysis in the other) on 200 mg celecoxib who were on warfarin for other medical reasons. Of 37 patients evaluable for tumor response, 14 had complete response, 13 partial responses, and 10 stable or progressive disease. The actuarial local progression-free survival was 66.0% at 1 year and 42.2% at 2 years following initiation of radiotherapy. CONCLUSIONS: These results show that celecoxib can be safely administered concurrently with thoracic radiotherapy when given up to the highest Food and Drug Administration-approved dose of 800 mg/d, which we used. A maximal tolerated dose was not reached in this study. The treatment resulted in actuarial local progression-free survival of 66.0% at 1 year and 42.2% at 2 years, an encouraging outcome that warrants further assessment in a phase II/III trial.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/radioterapia , Pirazóis/uso terapêutico , Sulfonamidas/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Celecoxib , Estudos de Coortes , Terapia Combinada , Inibidores de Ciclo-Oxigenase/administração & dosagem , Inibidores de Ciclo-Oxigenase/efeitos adversos , Inibidores de Ciclo-Oxigenase/uso terapêutico , Fracionamento da Dose de Radiação , Relação Dose-Resposta a Droga , Esofagite/induzido quimicamente , Fadiga/induzido quimicamente , Feminino , Humanos , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Pneumonia/induzido quimicamente , Prognóstico , Pirazóis/administração & dosagem , Pirazóis/efeitos adversos , Dosagem Radioterapêutica , Dermatopatias/induzido quimicamente , Sulfonamidas/administração & dosagem , Sulfonamidas/efeitos adversos , Análise de Sobrevida , Taxa de Sobrevida , Fatores de Tempo , Resultado do Tratamento
20.
Cancer Res ; 63(12): 3263-7, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12810657

RESUMO

Flavopiridol, a cyclin-dependent kinase (cdk) inhibitor, can cause cell cycle arrest, induce apoptosis in cancer cells, and inhibit tumor cell growth in vivo. The present study investigated the in vitro radiosensitizing effect of flavopiridol and the underlying molecular mechanisms in a murine ovarian cancer cell line, OCA-I. Flavopiridol inhibited cell growth in a dose-dependent manner and enhanced cell radiosensitivity assessed by the clonogenic cell survival assay. A flavopiridol dose of 300 nM, given for 1 day, enhanced radiosensitivity by a factor of 2.1. Clonogenic cell survival after split-dose radiation showed that flavopiridol inhibited repair from radiation damage. In addition, flavopiridol treatment (300 nM, 1 day) resulted in decreased levels of Ku70 and Ku86 proteins that play a role in DNA repair processes, suggesting that DNA repair processes may have been disrupted by this agent. Flow cytometry analysis showed that flavopiridol (300 nM, 1 day) accumulated the cells in G(1) and G(2) phases, with a significant reduction in the S phase component. This cell cycle redistribution is likely another mechanism underlying flavopiridol-induced cell radiosensitivity. Flavopiridol down-regulated cyclin D1 and cyclin E protein levels and also inhibited phosphorylation of retinoblastoma protein, which is inconsistent with the observed cell cycle arrest. Among the cdks tested, cdk-9, the catalytic subunit of positive transcription elongation factor b, was significantly down-regulated by flavopiridol, suggesting that flavopiridol may modulate cellular transcription processes. Furthermore, flavopiridol on its own induced apoptosis in the OCA-I cells, whereas in combination with radiation, exerted no additional increase in apoptosis. Taken together, our data show that flavopiridol strongly augmented the response of ovarian carcinoma cells to radiation and that the underlying mechanisms included inhibition of sublethal DNA damage repair and cell cycle redistribution. At the molecular level, transcriptional regulation by flavopiridol may have been involved.


Assuntos
Antineoplásicos/farmacologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , DNA Helicases , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias Ovarianas/patologia , Piperidinas/farmacologia , Radiossensibilizantes/farmacologia , Animais , Antígenos Nucleares/análise , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspase 3 , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclina D1/biossíntese , Ciclina D1/genética , Ciclina E/biossíntese , Ciclina E/genética , Reparo do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/análise , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Feminino , Raios gama , Autoantígeno Ku , Camundongos , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Tolerância a Radiação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA