Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Anticancer Res ; 34(12): 6981-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25503124

ABSTRACT

BACKGROUND: Positive transcription elongation factor-b (P-TEFb) is a complex containing CDK9 and a cyclin (T1, T2 or K). The effect of inhibition of P-TEFb by 5,6-dichloro-l-ß-D-ribofuranosyl benzimidazole (DRB) on cell radiosensitivity and the underlying mechanisms were investigated. MATERIALS AND METHODS: Six human cancer cell lines were subjected to (3)H-uridine incorporation, cell viability and clonogenic cell survival assays; cell-cycle redistribution and apoptosis assay; western blots and nuclear 53BP1 foci analysis after exposing the cells to DRB with/without γ-radiation. RESULTS: DRB suppressed colony formation and enhanced radiosensitivity of all cell lines. DRB caused a further increase in radiation-induced apoptosis and cell-cycle redistribution depending on p53 status. DRB prolonged the presence of radiation-induced nuclear p53 binding protein-1 (53BP1) foci and suppressed the expression of sirtuin-1 (SIRT1) and casein kinase 2-alpha (CK2α), suggesting an inhibition of DNA repair processes. CONCLUSION: Our findings indicate that DRB has the potential to increase the efficacy of radiotherapy and warrants further investigation using in vivo tumor models.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Dichlororibofuranosylbenzimidazole/pharmacology , Positive Transcriptional Elongation Factor B/antagonists & inhibitors , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Sirtuin 1/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/radiation effects , Casein Kinase II/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Repair/drug effects , Enzyme Inhibitors/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , M Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/radiation effects , Sirtuin 1/biosynthesis , Tumor Suppressor p53-Binding Protein 1
2.
Am J Clin Oncol ; 37(1): 90-100, 2014 Feb.
Article in English | MEDLINE | ID: mdl-22495455

ABSTRACT

Recent research on inhibitors of poly (ADP-ribose) polymerase (PARP) has demonstrated their potential for improving cancer therapy. They inhibit protein poly (ADP-ribosyl)ation and thus affect numerous molecular and cellular functions, including DNA repair and cell survival, that are critical for such physiological and patho-physiological states as carcinogenesis, inflammation, and resistance to cancer therapy. In this review, we describe the biological basis underlying the use of these agents in cancer therapy, providing data from preclinical studies that demonstrate the synergistic interaction of PARP inhibitors with radiation and chemotherapeutics. We also summarize initial clinical trials of PARP inhibitors for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Clinical Trials as Topic , DNA Repair , Drug Synergism , Humans , Inflammation , Necrosis , Neoplasms/radiotherapy , Neovascularization, Pathologic , Poly(ADP-ribose) Polymerases/metabolism , Radiotherapy, Adjuvant
3.
Int J Radiat Oncol Biol Phys ; 85(2): 472-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-22672748

ABSTRACT

PURPOSE: To test whether a cyclooxygenase-2 inhibitor (celecoxib) could reduce mortality resulting from radiation-induced pneumonitis. METHODS AND MATERIALS: Celecoxib was given to mice twice daily for 40 consecutive days starting on the day of local thoracic irradiation (LTI) or 40 or 80 days later. C3Hf/KamLaw mice were observed for morbidity, and time to death was determined. Results were analyzed using the Cox proportional hazards model. RESULTS: Timing of celecoxib relative to LTI determined efficacy. A significant reduction in time to death was achieved only when celecoxib was started 80 days after LTI, corresponding to the time when pneumonitis is expressed. For these mice the reduction in mortality was quantified as a hazard ratio for mortality of treated vs untreated of 0.36 (95% confidence interval [CI] 0.24-0.53), thus significantly less than 1.0. Correspondingly, the median lethal dose for treated mice (12.9 Gy; 95% CI 12.55-13.25 Gy) was significantly (P=.026) higher than for untreated mice (12.4 Gy; 95% CI 12.2-12.65 Gy). CONCLUSIONS: Celecoxib significantly reduced lung toxicity when administered months after LTI when the deleterious effects of radiation were expressed. The schedule-dependent reduction in fatal pneumonitis suggests that celecoxib could be clinically useful by reintroduction of treatment months after completion of radiation therapy. These findings may be important for designing clinical trials using cyclooxygenase-2 inhibitors to treat radiation-induced lung toxicity as a complement to concurrent radiation therapy of lung cancers.


Subject(s)
Cyclooxygenase 2 Inhibitors/administration & dosage , Pyrazoles/administration & dosage , Radiation Pneumonitis/prevention & control , Sulfonamides/administration & dosage , Animals , Celecoxib , Confidence Intervals , Dose-Response Relationship, Radiation , Drug Administration Schedule , Female , Lethal Dose 50 , Mice , Mice, Inbred C3H , Proportional Hazards Models , Radiation Pneumonitis/mortality , Time-to-Treatment
4.
Radiother Oncol ; 105(2): 241-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23010482

ABSTRACT

BACKGROUND AND PURPOSE: Although inhibition of epidermal growth factor receptor (EGFR) signaling during radiation led to improvement of tumor control and survival, novel strategies are needed to further improve the outcome of patients with locally advanced head and neck carcinoma. Because EGFR is known to interact with c-Src kinases, the present study investigated dasatinib (BMS-354825), an inhibitor of c-Src kinases, for its efficacy in enhancing radiosensitivity of human head and neck squamous cell carcinomas (HNSCC) in vitro and examined the underlying mechanisms for this effect. MATERIALS AND METHODS: Six HNSCC lines were exposed to dasatinib, radiation, or both, and assessed for c-Src and EGFR expression, cell survival and colony forming ability. Among these cell lines, HN-5 and FaDu lines were analyzed for induction of apoptosis, cell cycle re-distribution and for nuclear localization of EGFR, γ-H2AX and 53BP1 proteins. Immuno-precipitation and Western blots were performed to analyze the levels and binding of proteins involved in cell survival, apoptosis and DNA repair pathways. Suppression of c-Src by siRNA and subsequent clonogenic assay was performed in HN-5 cells. RESULTS: All six HNSCC lines that were examined expressed high levels of c-Src. Two (HN-5 and MDA-183) expressed higher levels of EGFR than other lines. Dasatinib suppressed cell survival of all cell lines tested independent of c-Src or EGFR levels but enhanced the radiosensitivity of HN-5 and MDA-183. HN-5 and FaDu were analyzed further. Dasatinib suppressed phosphorylation of c-Src in both cell lines, but decreased repair of radiation-induced DNA damage in HN-5 cells only as evidenced by suppression of c-Abl and Nbs-1 activity, inhibition of the association between c-Src and EGFR or Her-2, prolongation of nuclear γ-H2AX and 53BP1 foci and inhibition of EGFR nuclear localization and its association with DNA-PKcs. Finally, partial suppression of c-Src resulted in a small increase in HN-5 cell radiosensitivity. CONCLUSIONS: Our data demonstrate that dasatinib induces apoptosis and blocks DNA repair in EGFR-expressing HNSCC cells and improves radiotherapy outcome. These findings warrant further investigation using in vivo tumor models for potential translation into clinical testing.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Cell Nucleus/metabolism , DNA Repair/drug effects , ErbB Receptors/metabolism , Head and Neck Neoplasms/radiotherapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Radiation Tolerance/drug effects , Thiazoles/pharmacology , Active Transport, Cell Nucleus/drug effects , Apoptosis/radiation effects , CSK Tyrosine-Protein Kinase , Cell Cycle , Dasatinib , ErbB Receptors/analysis , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Squamous Cell Carcinoma of Head and Neck , src-Family Kinases/analysis , src-Family Kinases/metabolism
5.
Anticancer Res ; 32(8): 3029-35, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22843870

ABSTRACT

BACKGROUND: Targeting the epidermal growth factor receptor (EGFR) improved radiotherapy outcome by 10-15% in head and neck tumors (HNSCC). We tested the therapeutic benefits of co-targeting EGFR and insulin-like growth factor-1 receptor (IGF-1R) to further enhance tumor response to radiation. MATERIALS AND METHODS: Mice bearing FaDu tumor xenografts were treated with ganitumab (previously known as AMG479, an anti-IGF-1R antibody), panitumumab (an anti-EGFR antibody), or both in combination with fractionated doses of radiation. Tumor growth delay and tumor cure/recurrence served as end-points. RESULTS: The best tumor growth delay was achieved when ganitumab and panitumumab were given concurrently with radiation. Tumor cure/recurrence studies showed that combining ganitumab, panitumumab and radiation resulted in significantly higher radiocurability rates than use of either of the agents given with radiation. CONCLUSION: These findings provide the rationale for clinical testing of the combination of ganitumab and panitumumab for the treatment of HNSCC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/radiotherapy , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/radiotherapy , Radiation Tolerance , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Dose Fractionation, Radiation , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Mice , Panitumumab , Transplantation, Heterologous
6.
ACS Nano ; 6(3): 2497-505, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22316245

ABSTRACT

Current chemotherapeutics are characterized by efficient tumor cell-killing and severe side effects mostly derived from off-target toxicity. Hence targeted delivery of these drugs to tumor cells is actively sought. In an in vitro system, we previously demonstrated that targeted drug delivery to cancer cells overexpressing epidermal growth factor receptor (EGFR+) can be achieved by poly(ethylene glycol)-functionalized carbon nanovectors simply mixed with a drug, paclitaxel, and an antibody that binds to the epidermal growth factor receptor, cetuximab. This construct is unusual in that all three components are assembled through noncovalent interactions. Here we show that this same construct is effective in vivo, enhancing radiotherapy of EGFR+ tumors. This targeted nanovector system has the potential to be a new therapy for head and neck squamous cell carcinomas, deserving of further preclinical development.


Subject(s)
Carbon/chemistry , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Drug Carriers/chemistry , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Nanostructures/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Combined Modality Therapy , Humans , Male , Mice , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Polyethylene Glycols/chemistry , Tumor Burden/drug effects , Tumor Burden/radiation effects
7.
Invest New Drugs ; 30(6): 2113-20, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22127459

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/therapy , Indazoles/administration & dosage , Lung Neoplasms/therapy , Piperidines/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemoradiotherapy , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Poly(ADP-ribose) Polymerases/metabolism , Tumor Burden , Xenograft Model Antitumor Assays
8.
Anticancer Res ; 31(6): 2141-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21737634

ABSTRACT

AIM: To assess radiosensitzing potential of huachansu (HCS) and delineate the underlying mechanisms. MATERIALS AND METHODS: Lung cancer cell lines were exposed to HCS, radiation or both and subjected to survival assays, Western blots, apoptosis assay and immunocytochemical analysis. RESULTS: HCS suppressed the viability of all three lung lines tested and enhanced radiosensitivity of H460 and A549 (wild-type p53) only with no effect on H1299 (p53 null) cells. HCS prolonged the presence of radiation-induced γH2AX foci and increased radiation-induced apoptosis. Western blots showed that HCS increased cleaved caspase-3 and cleaved poly-(ADP-ribose) polymerase (PARP) levels, as well as reducing BCL-2 and p53 protein levels in H460 cells. CONCLUSION: HCS-enhanced radiosensitivity of human lung cancer lines appeared to be p53-dependent. Inhibition of DNA repair and increase in radiation-induced apoptosis may have served as underlying mechanisms. These data suggest that HCS may have potential to improve the efficacy of radiotherapy.


Subject(s)
Amphibian Venoms/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Amphibian Venoms/chemistry , Cardiac Glycosides/chemistry , Cardiac Glycosides/pharmacology , Cell Line, Tumor , Combined Modality Therapy , DNA Damage , DNA, Neoplasm/radiation effects , Dose-Response Relationship, Drug , Histones/biosynthesis , Histones/genetics , Humans , Lung Neoplasms/genetics
9.
Clin Cancer Res ; 17(7): 1815-27, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21350000

ABSTRACT

PURPOSE: We investigated whether vandetanib, an inhibitor of the tyrosine kinase activities of vascular endothelial growth factor receptor-2 (VEGFR-2), epidermal growth factor receptor (EGFR), and rearranged during transfection (RET), could augment the antitumor activity of radiation with or without cisplatin in preclinical in vitro and in vivo models of human head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN: OSC-19 and HN5 HNSCC cells that were cisplatin and radioresistant were treated with vandetanib, cisplatin, and radiation alone or in combination in vitro and in vivo using an orthotopic nude mouse model. Treatment effects were assessed using clonogenic survival assay, tumor volume, bioluminescence imaging, tumor growth delay, survival, microvessel density, tumor and endothelial cell apoptosis, and EGFR and Akt phosphorylation data. RESULTS: Vandetanib plus cisplatin radiosensitized HNSCC cells in vitro and in vivo. The combination treatment with vandetanib, cisplatin, and radiation was superior to the rest of treatments (including the double combinations) in antitumoral effects, prolonging survival, decreasing cervical lymph node metastases in vivo. It also increased both tumor and tumor-associated endothelial cell apoptosis and decreased microvessel density in vivo. An analysis of tumor growth delay data revealed that vandetanib plus cisplatin enhanced radioresponse in vivo. All vandetanib-containing treatments inhibited EGFR and Akt phosphorylation in vitro and in vivo. CONCLUSION: The addition of vandetanib to combination therapy with cisplatin and radiation was able to effectively overcome cisplatin and radioresistance in in vitro and in vivo models of HNSCC. Further study of this regimen in clinical trials may be warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/pathology , Piperidines/pharmacology , Quinazolines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Combined Modality Therapy , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Humans , Lymphatic Metastasis , Male , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Radiation Tolerance/drug effects , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
10.
Int J Radiat Oncol Biol Phys ; 79(4): 1179-87, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21129859

ABSTRACT

PURPOSE: The IGF1/IGF-1R signaling pathway has emerged as a potential determinant of radiation resistance in human cancer cell lines. Therefore we investigated the potency of monoclonal anti-IGF-1R antibody, A12, to enhance radiation response in upper respiratory tract cancers. METHODS AND MATERIALS: Cell lines were assessed for IGF-1R expression and IGF1-dependent response to A12 or radiation using viability and clonogenic cancer cell survival assays. In vivo response of tumor xenografts to 10 or 20 Gy and A12 (0.25-2 mg × 3) was assessed using growth delay assays. Combined treatment effects were also analyzed by immunohistochemical assays for tumor cell proliferation, apoptosis, necrosis, and vascular endothelial growth factor expression at Days 1 and 6 after start of treatment. RESULTS: A12 enhanced the radiosensitivity of HN5 and FaDu head-and-neck carcinomas in vitro (p < 0.05) and amplified the radioresponse of FaDu xenografts in a dose-dependent manner, with enhancement factors ranging from 1.2 to 1.8 (p < 0.01). Immunohistochemical analysis of FaDu xenografts demonstrated that A12 inhibited tumor cell proliferation (p < 0.05) and vascular endothelial growth factor expression. When A12 was combined with radiation, this resulted in apoptosis induction that persisted until 6 days from the start of treatment and in increased necrosis at Day 1 (p < 0.01, respectively). Combined treatment with A12 and radiation resulted in additive or subadditive growth delay in H460 or A549 xenografts, respectively. CONCLUSIONS: The results of this study strengthen the evidence for investigating how anti-IGF-1R strategies can be integrated into radiation and radiation-cetuximab regimen in the treatment of cancer of the upper aerodigestive tract cancers.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Head and Neck Neoplasms/radiotherapy , Lung Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dose-Response Relationship, Radiation , Female , Head and Neck Neoplasms/metabolism , Humans , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/metabolism , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , Necrosis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Radiation Tolerance , Receptor, IGF Type 1/metabolism , Time Factors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays/methods
11.
Invest New Drugs ; 29(4): 554-61, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20119866

ABSTRACT

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.


Subject(s)
Carbamates/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Triazines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Blotting, Western , Carbamates/therapeutic use , Carcinoma, Squamous Cell/pathology , Cell Count , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , ErbB Receptors/metabolism , Head and Neck Neoplasms/pathology , Histones/metabolism , Humans , Immunoprecipitation , Male , Mice , Mice, Nude , Radiation, Ionizing , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Triazines/therapeutic use , Xenograft Model Antitumor Assays
12.
Invest New Drugs ; 29(3): 514-22, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20024691

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Mitosis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Radiation Tolerance/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Histones/metabolism , Humans , Immunohistochemistry , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tubulin/metabolism
13.
Front Oncol ; 1: 52, 2011.
Article in English | MEDLINE | ID: mdl-22649768

ABSTRACT

PURPOSE: Preclinical findings suggest that adding targeted therapies to combination radiation-chemotherapy can enhance treatment efficacy; however, this approach may enhance normal tissue toxicity. We investigated the maximum tolerated dose, dose-limiting toxicities, and response rate when the selective cyclooxygenase-2 inhibitor celecoxib is added to concurrent irinotecan, cisplatin, and radiation therapy for patients with inoperable stage II-III non-small cell lung cancer (NSCLC). METHODS AND MATERIALS: Eighteen patients were analyzed in a phase I clinical dose-escalation trial. Celecoxib was given daily beginning 5 days before radiation followed by maintenance doses for 12 weeks. Toxicity was graded with the Common Terminology Criteria for Adverse Events V3.0 and response with the World Health Organization system. Primary endpoints were maximum tolerated dose of celecoxib and treatment toxicity; secondary endpoints were response and survival rates. RESULTS: The maximum tolerated dose of celecoxib was not reached, in part owing to discontinuation of the drug supply. At doses of 200 or 400 mg/day, no patients experienced any dose-limiting toxicity (acute grade ≥4 esophagitis or pneumonitis, neutropenic fever or thrombocytopenia requiring transfusion, or acute grade ≥3 diarrhea). Grade 3 toxicities were leukopenia (five patients), fatigue (3), pneumonitis (2), dyspnea (1), pain (1), and esophageal stricture (1). Interestingly, pulmonary fibrosis (a late toxicity) was no more severe in the higher-dose (400-mg) group and may have been less common than in the lower-dose group. The clinical response rate was 100% (8 complete, 10 partial). Two-year rates were: overall survival 65%; local-regional control 69%; distant metastasis-free survival 71%; and disease-free survival 64%. CONCLUSION: Although preliminary, our results suggest that adding celecoxib to concurrent chemoradiation for inoperable NSCLC is safe and can improve outcome without increasing normal tissue toxicity.

14.
Future Oncol ; 6(10): 1563-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21062156

ABSTRACT

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.


Subject(s)
Neoplastic Stem Cells/radiation effects , Radiation Tolerance/physiology , Animals , Humans
15.
Radiother Oncol ; 96(1): 108-15, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20451273

ABSTRACT

PURPOSE: Radiation therapy cures malignant tumors of the head and neck region more effectively when it is combined with application of the anti-EGFR monoclonal antibody cetuximab. Despite the successes achieved, we still do not know how to select patients who will respond to this combination of anti-EGFR monoclonal antibody and radiation. This study was conducted to elucidate possible mechanisms which cause the combined treatment with cetuximab and irradiation to fail in some cases of squamous cell carcinomas. METHODS AND MATERIALS: Mice bearing FaDu and A431 squamous cell carcinoma xenograft tumors were treated with cetuximab (total dose 3 mg, intraperitoneally), irradiation (10 Gy) or their combination at the same doses. Treatment was applied when tumors reached 8mm in size. To collect samples for further protein analysis (two-dimensional differential gel electrophoresis (2-D DIGE), mass spectrometry MALDI-TOF/TOF, Western blot analysis, and ELISA), mice from each group were sacrificed on the 8th day after the first injection of cetuximab. Other mice were subjected to tumor growth delay assay. RESULTS: In FaDu xenografts, treatment with cetuximab alone was nearly as effective as cetuximab combined with ionizing radiation, whereas A431 tumors responded to the combined treatment with significantly enhanced delay in tumor growth. Tumors extracted from the untreated FaDu and A431 xenografts were analysed for protein expression, and 34 proteins that were differently expressed in the two tumor types were identified. The majority of these proteins are closely related to intratumoral angiogenesis, cell adhesion, motility, differentiation, epithelial-to-mesenchymal transition (EMT), c-myc signaling and DNA repair. CONCLUSIONS: The failure of cetuximab to enhance radiation response in FaDu xenografts was associated with the initiation of the program of EMT and with c-myc up-regulation in the carcinoma cells. For this reason, c-myc and EMT-related proteins (E-cadherin, vimentin) may be considered as potential biomarkers to predict squamous cell carcinoma response after treatment with cetuximab in combination with radiation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Salivary alpha-Amylases/genetics , Animals , Antibodies, Monoclonal, Humanized , Blotting, Western , Cadherins/metabolism , Carcinoma, Squamous Cell/genetics , Cell Division/genetics , Cell Line, Tumor , Cetuximab , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Male , Mice , Mice, Nude , Radiation Tolerance/genetics , Radiation, Ionizing , Salivary alpha-Amylases/metabolism , Signal Transduction , Up-Regulation , Xenograft Model Antitumor Assays
16.
Anticancer Res ; 29(11): 4409-15, 2009 Nov.
Article in English | MEDLINE | ID: mdl-20032386

ABSTRACT

BACKGROUND: Imexon is an aziridine-containing small pro-oxidant molecule with promising antitumor activity in myeloma, lymphoma and lung and pancreatic cancer. Imexon is already in clinical trials in patients with advanced solid tumors. The present study examined the effects of imexon on H9 and Raji lymphoma cell lines in vitro when given in combination with ionizing radiation. MATERIALS AND METHODS: H9 and Raji lymphoma cells were grown in culture and exposed to imexon, radiation, or both. Cells were assessed for cell viability, glutathione content, induction of apoptosis, cell cycle distribution and also subject to Western blot analysis. RESULTS: Imexon inhibited cell proliferation in a dose-dependent manner. Imexon, given for 48 h prior to irradiation at a clinically achievable dose of 40 muM, potently enhanced the cell radiosensitivity. Imexon enhanced radiation-induced apoptosis and accumulated cells in G2/M phase of the cell cycle. Imexon induced caspase-3 activation and PARP cleavage. Alterations in glutathione levels were not observed at 40 microM of imexon. CONCLUSION: In conclusion, imexon efficiently augmented lymphoma cell radiosensitivity independently of glutathione and the underlying mechanisms include induction of apoptosis and cell cycle redistribution.


Subject(s)
Hexanones/pharmacology , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/radiotherapy , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Oxidation-Reduction
17.
Radiother Oncol ; 92(3): 383-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19695723

ABSTRACT

BACKGROUND AND PURPOSE: We recently demonstrated that C225 maintenance therapy after completion of radiotherapy further increased tumor radiocurability. The present study assessed mechanisms underlying the observed improvement in C225 efficacy in pre-irradiated tissue (tumor bed). MATERIALS AND METHODS: A431 xenografts growing in pre-irradiated and non-irradiated tissue were treated with C225. Tumors were assessed for growth delay, cell proliferation, hypoxia, EGFR and VEGF expressions. In vitro clonogenic survival of cells derived from these tumors was also assayed. RESULTS: Pre-irradiation of tumor bed induced growth retardation, reduction in Ki-67 labeling, and overexpression of HIF-1alpha, CA IX, EGFR and VEGF biomarkers. C225 treatment dramatically inhibited tumor growth in the irradiated tumor bed (P<0.0001), which was associated with further reduction in Ki-67 labeling, and reduced expression of HIF-1alpha, CA IX, EGFR and VEGF. Cells derived from tumors in the pre-irradiated bed showed increased sensitivity to C225. C225 was more cytotoxic against hypoxic than well-oxygenated A431 cells grown in vitro. CONCLUSION: A431 xenografts growing in pre-irradiated tumor bed exhibit enhanced sensitivity to C225. Pre-irradiated tissue microenvironment seems to render tumor cells more susceptible to C225 cytostatic and cytotoxic actions. If confirmed in other tumor models these findings support the use of C225 maintenance therapy after completion of radiotherapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , ErbB Receptors/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Cell Proliferation/radiation effects , Cetuximab , Disease Models, Animal , ErbB Receptors/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Probability , Radiation Tolerance/drug effects , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects
18.
Radiother Oncol ; 92(3): 316-22, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19615770

ABSTRACT

Radiotherapy plays a crucial role in the treatment of many malignancies; however, locoregional disease progression remains a critical problem. This has stimulated laboratory research into understanding the basis for tumor cell resistance to radiation and the development of strategies for overcoming such resistance. We know that some cell signaling pathways that respond to normal growth factors are abnormally activated in human cancer and that these pathways also invoke cell survival mechanisms that lead to resistance to radiation. For example, abnormal activation of the epidermal growth factor receptor (EGFR) promotes unregulated growth and is believed to contribute to clinical radiation resistance. Molecular blockade of EGFR signaling is an attractive strategy for enhancing the cytotoxic effects of radiotherapy and, as shown in numerous reports, the radiosensitizing effects of EGFR antagonists correlate with a suppression of the ability of the cells to repair radiation-induced DNA double strand breaks (DSBs). The molecular connection between the EGFR and its governance of DNA repair capacity appears to be mediated by one or more signaling pathways downstream of this receptor. The purpose of this review is to highlight what is currently known regarding EGFR signaling and the processes responsible for repairing radiation-induced DNA lesions that would explain the radiosensitizing effects of EGFR antagonists.


Subject(s)
DNA Repair/physiology , ErbB Receptors/radiation effects , Radiobiology , Signal Transduction/radiation effects , Animals , Cell Line, Tumor/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , ErbB Receptors/metabolism , Humans , Neoplasms/genetics , Neoplasms/radiotherapy , Radiation Injuries/prevention & control , Radiation Tolerance
19.
Int J Radiat Oncol Biol Phys ; 74(5): 1580-91, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19540073

ABSTRACT

At a meeting of the Translation Research Program of the Radiation Therapy Oncology Group held in early 2008, attendees focused on updating the current state of knowledge in cancer stem cell research and discussing ways in which this knowledge can be translated into clinical use across all disease sites. This report summarizes the major topics discussed and the future directions that research should take. Major conclusions of the symposium were that the flow cytometry of multiple markers in fresh tissue would remain the standard technique of evaluating cancer-initiating cells and that surrogates need to be developed for both experimental and clinical use.


Subject(s)
Biomedical Research , Clinical Trials as Topic , Neoplasms , Neoplastic Stem Cells , Radiation Oncology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/physiology , Cell Hypoxia , Drug Resistance, Neoplasm/physiology , Flow Cytometry/methods , Hematopoietic Stem Cells/pathology , Information Dissemination , Neoplasm Recurrence, Local/pathology , Neoplasms/chemistry , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Cells, Circulating/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/physiology , Neoplastic Stem Cells/radiation effects
20.
J Clin Oncol ; 27(20): 3370-8, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19380441

ABSTRACT

PURPOSE: In search of reliable biologic markers to predict the risk of normal tissue damage by radio(chemo)therapy before treatment, we investigated the association between single nucleotide polymorphisms (SNPs) in the transforming growth factor 1 (TGFbeta1) gene and risk of radiation pneumonitis (RP) in patients with non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: Using 164 available genomic DNA samples from patients with NSCLC treated with definitive radio(chemo)therapy, we genotyped three SNPs of the TGFbeta1 gene (rs1800469:C-509T, rs1800471:G915C, and rs1982073:T869C) by polymerase chain reaction restriction fragment length polymorphism method. We used Kaplan-Meier cumulative probability to assess the risk of grade > or = 3 RP and Cox proportional hazards analyses to evaluate the effect of TGFbeta1 genotypes on such risk. RESULTS: There were 90 men and 74 women in the study, with median age of 63 years. Radiation doses ranging from 60 to 70 Gy (median = 63 Gy) in 30 to 58 fractions were given to 158 patients (96.3%) and platinum-based chemotherapy to 147 (89.6%). Grade > or = 2 and grade > or = 3 RP were observed in 74 (45.1%) and 36 patients (22.0%), respectively. Multivariate analysis found CT/CC genotypes of TGFbeta1 rs1982073:T869C to be associated with a statistically significantly lower risk of RP grades > or = 2 (hazard ratio [HR] = 0.489; 95% CI, 0.227 to 0.861; P = .013) and grades > or = 3 (HR = 0.390; 95% CI, 0.197 to .774; P = 0.007), respectively, compared with the TT genotype, after adjustment for Karnofsky performance status, smoking status, pulmonary function, and dosimetric parameters. CONCLUSION: Our results showed that CT/CC genotypes of TGFbeta1 rs1982073:T869C gene were associated with lower risk of RP in patients with NSCLC treated with definitive radio(chemo)therapy and thus may serve as a reliable predictor of RP.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/radiotherapy , Polymorphism, Single Nucleotide , Radiation Pneumonitis/etiology , Transforming Growth Factor beta1/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , Dose-Response Relationship, Radiation , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Prognosis , Proportional Hazards Models , Radiation Pneumonitis/genetics , Radiotherapy/adverse effects , Radiotherapy/methods , Radiotherapy/statistics & numerical data , Risk Factors , Transforming Growth Factor beta1/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...