Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
BMC Cancer ; 14: 594, 2014 Aug 16.
Article in English | MEDLINE | ID: mdl-25128202

ABSTRACT

BACKGROUND: Regions within solid tumours often experience oxygen deprivation, which is associated with resistance to chemotherapy and irradiation. The aim of this study was to evaluate the radiosensitising effect of gemcitabine and its main metabolite dFdU under normoxia versus hypoxia and to determine whether hypoxia-inducible factor 1 (HIF-1) is involved in the radiosensitising mechanism. METHODS: Stable expression of dominant negative HIF-1α (dnHIF) in MDA-MB-231 breast cancer cells, that ablated endogenous HIF-1 transcriptional activity, was validated by western blot and functionality was assessed by HIF-1α activity assay. Cells were exposed to varying oxygen environments and treated with gemcitabine or dFdU for 24 h, followed by irradiation. Clonogenicity was then assessed. Using radiosensitising conditions, cells were collected for cell cycle analysis. RESULTS: HIF-1 activity was significantly inhibited in cells stably expressing dnHIF. A clear radiosensitising effect under normoxia and hypoxia was observed for both gemcitabine and dFdU. No significant difference in radiobiological parameters between HIF-1 proficient and HIF-1 deficient MDA-MB-231 cells was demonstrated. CONCLUSIONS: For the first time, radiosensitisation by dFdU, the main metabolite of gemcitabine, was demonstrated under low oxygen conditions. No major role for functional HIF-1 protein in radiosensitisation by gemcitabine or dFdU could be shown.


Subject(s)
Deoxycytidine/analogs & derivatives , Deoxyuridine/pharmacology , Hypoxia-Inducible Factor 1/metabolism , Radiation-Sensitizing Agents/pharmacology , Breast Neoplasms , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Deoxycytidine/pharmacology , Deoxyuridine/analogs & derivatives , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Gemcitabine
2.
BMC Cancer ; 10: 441, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20723210

ABSTRACT

BACKGROUND: Based on their different mechanisms of action, non-overlapping side effects and radiosensitising potential, combining the antimetabolites pemetrexed (multitargeted antifolate, MTA) and gemcitabine (2',2'-difluorodeoxycytidine, dFdC) with irradiation (RT) seems promising. This in vitro study, for the first time, presents the triple combination of MTA, dFdC and irradiation using various treatment schedules. METHODS: The cytotoxicity, radiosensitising potential and cell cycle effect of MTA were investigated in A549 (NSCLC) and CAL-27 (SCCHN) cells. Using simultaneous or sequential exposure schedules, the cytotoxicity and radiosensitising effect of 24 h MTA combined with 1 h or 24 h dFdC were analysed. RESULTS: Including a time interval between MTA exposure and irradiation seemed favourable to MTA immediately preceding or following radiotherapy. MTA induced a significant S phase accumulation that persisted for more than 8 h after drug removal. Among different MTA/dFdC combinations tested, the highest synergistic interaction was produced by 24 h MTA followed by 1 h dFdC. Combined with irradiation, this schedule showed a clear radiosensitising effect. CONCLUSIONS: Results from our in vitro model suggest that the sequence 24 h MTA --> 1 h dFdC --> RT is the most rational design and would, after confirmation in an in vivo setting, possibly provide the greatest benefit in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/therapy , Drug Interactions , Head and Neck Neoplasms/therapy , Lung Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Flow Cytometry , Glutamates/administration & dosage , Guanine/administration & dosage , Guanine/analogs & derivatives , Head and Neck Neoplasms/pathology , Humans , In Vitro Techniques , Lung Neoplasms/pathology , Pemetrexed , Radiotherapy Dosage , Tumor Cells, Cultured , Gemcitabine
3.
Future Oncol ; 6(9): 1485-96, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20919831

ABSTRACT

The combination of radiotherapy with chemotherapeutic agents that sensitize tumor cells to ionizing radiation has long been regarded as a promising strategy to enhance cancer therapy. Many chemotherapeutic agents interact with radiation and enhance the cytotoxic or anti-tumor effect of radiation through a number of mechanisms. These include an increase in initial radiation damage, inhibition of cellular repair, cell cycle redistribution, enhancement of apoptosis, counteracting hypoxia and overcoming accelerated repopulation. This article focuses on the role of cell cycle perturbations in the radiosensitivity of cancer cells.


Subject(s)
Cell Cycle/drug effects , Cell Cycle/radiation effects , Neoplasms/therapy , Radiation-Sensitizing Agents/pharmacology , Radiotherapy , Animals , Antineoplastic Agents/pharmacology , Combined Modality Therapy , Humans , Radiation Tolerance/drug effects
4.
BMC Cancer ; 8: 65, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18312675

ABSTRACT

BACKGROUND: Vinca alkaloids are an important class of anticancer agents and semisynthetic vinca alkaloids are developed to improve the therapeutic index of this class of drugs. In the present study, a direct comparison was made between vinflunine and vinorelbine regarding their radiosensitizing and cell cycle effects. METHODS: Four human tumour cell lines were tested under identical experimental conditions, using equitoxic concentrations of vinflunine and vinorelbine. RESULTS: Vinflunine and vinorelbine induced a comparable radiosensitizing effect (p-value never below 0.01) when cells were incubated for 24 h immediately prior to radiation. Regarding the cell cycle effects, a statistically significant concentration-dependent G2/M block was seen after 24 h incubation with vinorelbine in all tested cell lines. Similar results, with small cell line-related differences, were observed with vinflunine. CONCLUSION: The radiosensitizing effects of both semisynthetic vinca alkaloids were comparable (not statistically different) and nearly always cell line-specific and concentration-dependent. The cell cycle effects could be related to the observed radiosensitizing effects. Considering the more favourable toxicity profile of vinflunine, this agent might be more promising than vinorelbine for chemoradiation studies in the clinic.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Radiation-Sensitizing Agents/pharmacology , Vinblastine/analogs & derivatives , Antineoplastic Agents, Phytogenic/chemistry , Breast Neoplasms/pathology , Carcinoma/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Dose-Response Relationship, Drug , Female , Gamma Rays , Humans , Lung Neoplasms/pathology , Microtubules/drug effects , Molecular Structure , Structure-Activity Relationship , Tongue Neoplasms/pathology , Urinary Bladder Neoplasms/pathology , Vinblastine/chemistry , Vinblastine/pharmacology , Vinorelbine
5.
Cancer Chemother Pharmacol ; 62(2): 183-93, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17899084

ABSTRACT

PURPOSE: Vinflunine is an innovative microtubule inhibitor belonging to the vinca alkaloid class that possesses radiosensitising properties, which could lead to promising activity in chemoradiation studies in the clinic. METHOD: In the current study, different incubation times with vinflunine, immediately before radiation and different time intervals between vinflunine treatment and radiation were investigated, in vitro, using four different human tumour cell lines differing in cell type and p53 status. Results were correlated with the cell cycle distribution at the moment of radiation, in order to elucidate the role of cell cycle perturbations caused by vinflunine on its radiosensitising effect. RESULTS: Radiosensitisation was observed in all cell lines, and maximal radiosensitisation was both cell line- and schedule-dependent. The cell cycle distributions were cell line-dependent also, and when correlated with the observed radiosensitising effects could explain many (but not all) of the radiosensitising properties of vinflunine. CONCLUSION: The cell cycle perturbations caused by vinflunine may definitely have an impact on its radiosensitising potential, but other factors must play a role because of some unaccountable differences between cell cycle distribution and the radiosensitising potential.


Subject(s)
Cell Cycle , Gamma Rays , Radiation-Sensitizing Agents/pharmacology , Tubulin Modulators/pharmacology , Vinblastine/pharmacology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Flow Cytometry , Humans , Time Factors
6.
BMC Cancer ; 6: 142, 2006 May 30.
Article in English | MEDLINE | ID: mdl-16734894

ABSTRACT

BACKGROUND: Gemcitabine (dFdC) is an active antitumour agent with radiosensitising properties, shown both in preclinical and clinical studies. In the present study, the relation between deoxycytidine kinase (dCK) activity and the radiosensitising effect of gemcitabine was investigated in eight different human tumour cell lines. METHODS: Tumour cells were treated with dFdC (0-100 nM) for 24 h prior to radiotherapy (RT) (gamma-Co60, 0-6 Gy, room temperature). Cell survival was determined 7, 8, or 9 days after RT by the sulforhodamine B test. dCK activity of the cells was determined by an enzyme activity assay. RESULTS: A clear concentration-dependent radiosensitising effect of dFdC was observed in all cell lines. The degree of radiosensitisation was also cell line dependent and seemed to correlate with the sensitivity of the cell line to the cytotoxic effect of dFdC. The dCK activity of our cell lines varied considerably and differed up to three fold from 5 to 15 pmol/h/mg protein between the tested cell lines. In this range dCK activity was only weakly related to radiosensitisation (correlation coefficient 0.62, p = 0.11). CONCLUSION: Gemcitabine needs to be metabolised to the active nucleotide in order to radiosensitise the cells. Since dFdCTP accumulation and incorporation into DNA are concentration dependent, the degree of radiosensitisation seems to be related to the extent of dFdCTP incorporated into DNA required to inhibit DNA repair. The activity of dCK does not seem to be the most important factor, but is clearly a major factor. Other partners of the intracellular metabolism of gemcitabine in relation to the cell cycle effects and DNA repair could be more responsible for the radiosensitising effect than dCK activity.


Subject(s)
Deoxycytidine Kinase/metabolism , Deoxycytidine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Cell Survival , DNA Repair , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Deoxycytidine Kinase/drug effects , Drug Screening Assays, Antitumor , Humans , Tumor Cells, Cultured , Gemcitabine
7.
Cancer Chemother Pharmacol ; 58(2): 219-28, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16331496

ABSTRACT

PURPOSE: Gemcitabine is an active antitumour agent with radiosensitising properties. Gemcitabine is rapidly metabolised, intracellularly as well as extracellularly, by deoxycytidine deaminase to difluorodeoxyuridine (dFdU), a compound with little antitumour activity. However, plasma concentrations are maintained for a prolonged period (>24 h) at levels known to cause growth inhibition. This is the first study that investigates the radiosensitising potential of dFdU in vitro. METHODS: ECV304 and H292, human cancer cells, were treated with different concentrations dFdU (0-100 microM) during 24 h before radiation treatment (RT). The schedule dependency of the radiosensitising effect was studied by varying the interval between dFdU and radiation treatment. In addition, the cell cycle effect of dFdU was investigated with flow cytometry, and the induction of apoptosis under radiosensitising conditions was determined by Annexin V staining and caspase 3 cleavage. RESULTS: dFdU caused a clear concentration-dependent radiosensitising effect in both ECV304 and H292 cells. Dose enhancement factor (DEF) increased with an increasing concentration of dFdU: DEFs were 1.10, 1.60 and 2.17 after treatment with 10, 25 and 50 microM dFdU, respectively, in ECV304 cells and 1.08, 1.31 and 1.60 after treatment with 25, 50 and 100 microM, respectively, in H292 cells. DEFs decreased with an increasing interval of 0-24 h between dFdU treatment and radiation. Under radiosensitising conditions, the combination dFdU and radiation resulted in an increased induction of apoptosis. In addition, the cell cycle effect of dFdU, an arrest at the early S phase, is comparable with the cell cycle effect of gemcitabine. CONCLUSIONS: dFdU, the main metabolite of gemcitabine, causes a concentration- and schedule- dependent radiosensitising effect in vitro. Since the metabolite is present in plasma for a long period (>24 h) after treatment with gemcitabine, it might be partly responsible for the interaction between radiotherapy and gemcitabine. This observation might have important consequences for the optimal schedules of the combination gemcitabine and radiation therapy.


Subject(s)
Floxuridine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Floxuridine/pharmacology , Humans
8.
Cancer Chemother Pharmacol ; 58(2): 210-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16317558

ABSTRACT

PURPOSE: Vinflunine (VFL) is a novel third generation Vinca alkaloid with superior antitumour activity in preclinical models and an anticipated more favourable toxicity profile compared to the other Vinca alkaloids. METHOD: We investigate the radiosensitising properties of VFL and its cell cycle effects in four human tumour cell lines (ECV304, MCF-7, H292, and CAL-27). The sulforhodamine B test was used to determine cell survival, and cell cycle analysis was performed by flow cytometry. Radiosensitisation (RS) was represented by dose enhancement factors (DEFs). RESULTS: Twenty-four hours treatment with VFL before radiation caused dose-dependent RS in all cell lines. This was most pronounced in ECV304 cells with RS already at VFL concentrations that reduced cell survival by 10% (IC10). DEFs ranged from 1.57 to 2.29 in the different cell lines. A concentration-dependent G2/M block was observed (starting at 4 h of incubation). After maximal G2/M blockade cells started cycling again, mainly by mitosis, while a small portion of cells started a polyploid cell cycle. Also drug removal immediately caused recycling of cells and induction of a polyploid cell population. The polyploid cell population was most impressively noticeable after prolonged incubation with VFL (48 h), in particular in CAL-27 and ECV304. This was never observed in a tested normal fibroblast cell line (Fi 360). The fate of these cells is of particular interest, but yet uncertain. CONCLUSION: VFL has radiosensitising potential. The exact role of the cell cycle effects of VFL in its radiosensitising mechanism is still not fully elucidated and requires further study.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Radiation, Ionizing , Vinblastine/analogs & derivatives , Cell Line, Tumor , Humans , Vinblastine/pharmacology
9.
J Thorac Cardiovasc Surg ; 130(1): 125-30, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15999051

ABSTRACT

OBJECTIVE: Isolated lung perfusion is an experimental technique for the treatment of lung metastases. Single-agent isolated lung perfusion does not result in complete remission. We studied the in vivo and in vitro efficacy of combinations of gemcitabine, cisplatin, and melphalan. METHODS: In vitro, using the sulforhodamine B assay, CC531s cells were incubated with cisplatin, gemcitabine, or melphalan or with a combination of these drugs. One drug was added at concentrations causing 25% growth inhibition, whereas the second drug was added at variable concentrations. In vivo, left pulmonary metastases were induced in Wag/Rij rats by means of intravenous injection of CC531s adenocarcinoma cells. At day 7, rats underwent left isolated lung perfusion with gemcitabine (n = 7), cisplatin (n = 9), melphalan (n = 7), gemcitabine-cisplatin (n = 6), melphalan-gemcitabine (n = 6), and cisplatin-melphalan (n = 7). Death by means of metastatic disease was the end point. Survival and differences in survival were assessed by using Kaplan-Meier and log-rank testing. RESULTS: In vitro synergistic activity was observed for melphalan-gemcitabine, whereas other combinations showed additive or antagonistic activity. In vivo treated rats lived longer compared with control animals ( P < .0001). In isolated lung perfusion melphalan resulted in longer survival compared with gemcitabine ( P = .0016) and cisplatin ( P = .046). Isolated lung perfusion with melphalan-gemcitabine resulted in 67% survival of the rats after 90 days versus 0% in other groups. CONCLUSIONS: Isolated lung perfusion monotherapy or combination therapy with gemcitabine, cisplatin, or melphalan resulted in significantly longer survival compared with that seen in control animals. Isolated lung perfusion combination therapy with melphalan-gemcitabine resulted in the best survival either in vitro or in vivo.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Cancer, Regional Perfusion , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Lung Neoplasms/drug therapy , Adenocarcinoma/secondary , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Cisplatin/administration & dosage , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , In Vitro Techniques , Inhibitory Concentration 50 , Lung Neoplasms/secondary , Male , Melphalan/administration & dosage , Rats , Rats, Inbred Strains , Tumor Cells, Cultured , Gemcitabine
10.
Radiat Res ; 164(5): 642-50, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16238441

ABSTRACT

Gemcitabine has excellent radiosensitizing properties, as shown in both preclinical and clinical studies. Radiosensitization correlated with the early S-phase block of gemcitabine. In the present study, we investigated the role of TP53 in the radiosensitizing effect of gemcitabine. Isogenic A549 cells differing in TP53 status were treated with gemcitabine during the 24 h prior to irradiation. Cell survival was determined 7 days after irradiation by the sulforhodamine B test. In addition, cell cycle perturbation was determined by flow cytometry and TP53 expression by Western blot analysis. Gemcitabine caused a concentration-dependent radiosensitizing effect in all cell lines. Transformed A549 cells were less sensitive to the cytotoxic effect of gemcitabine. The cell cycle arrest early in the S phase was dependent on the drug dose but was comparable in the different cell lines and was not related to functional TP53. Using isogenic cell lines, we have shown that neither TP53 status nor the transfection procedure influenced the radiosensitizing effect of gemcitabine. Since both the radiosensitizing effect at equitoxic concentrations and the cell cycle effect of gemcitabine were independent of TP53 expression, it is likely that TP53 protein does not play a crucial role in the radiosensitizing mechanism of gemcitabine.


Subject(s)
Deoxycytidine/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Protein p53/physiology , Cell Cycle/drug effects , Cell Line, Tumor , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Humans , Tumor Suppressor Protein p53/analysis , Gemcitabine
11.
Int J Radiat Oncol Biol Phys ; 57(4): 1075-83, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14575839

ABSTRACT

PURPOSE: The mechanism of radiosensitization by gemcitabine is still unclear. It has been hypothesized that the accumulation of cells in early S phase may play a role in enhancing radiosensitivity. METHODS AND MATERIALS: The schedule dependency of the radiosensitizing effect was studied in ECV304, human bladder cancer cells, and H292, human lung cancer cells, by varying the incubation time and time interval between gemcitabine and radiation treatment. To determine the role of cell cycle perturbations in the radiosensitization, the influence of gemcitabine on the cell cycle at the moment of radiation was investigated by flow cytometry. RESULTS: The radiosensitizing effect increased with a longer incubation period: Dose enhancement factors varied from 1.30 to 2.82 in ECV304 and from 1.04 to 1.78 in H292 after treatment during 8-32 h, respectively. Radiosensitization decreased with an increasing interval: Dose enhancement factors varied from 2.26 to 1.49 in ECV304 and from 1.45 to 1.11 in H292 after an interval 0-24 h, respectively. Cells were blocked in the early S phase of the cell cycle by gemcitabine. The highest percentage S-phase cells was observed after treatment with the schedules that resulted in the highest radiosensitizing effect. CONCLUSIONS: We observed a clear schedule-dependent radiosensitization by gemcitabine. Our findings demonstrated a correlation between gemcitabine-induced early S-phase block and the radiosensitizing effect.


Subject(s)
Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Flow Cytometry , Humans , S Phase , Time Factors , Gemcitabine
12.
Ann Thorac Surg ; 76(4): 1036-40; discussion 1040, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14529980

ABSTRACT

BACKGROUND: Isolated lung perfusion (ILuP) proved to be superior for the treatment of lung metastases compared with intravenous (i.v.) injection. However its invasive character limits repetitive treatment. Blood flow occlusion (BFO) as a regional therapy with gemcitabine (GCB) was evaluated in a rat model. Lung levels of GCB were examined with different exposure times and flow rates and compared with ILuP and i.v.. Cell kill was studied in vitro. METHODS: In vitro survival of CC531 adenocarcinoma cells was determined after 10, 20, and 40 minutes of exposure to GCB. In vivo 48 Wag/Rij rats underwent BFO with GCB at a rate of 0.2 mL/min and 0.5 mL/min during 10, 20, 30, and 40 minutes. Statistical analysis was performed using Student's t test. RESULTS: In vitro, the dose of GCB resulting in 50% growth inhibition was 9.1 microg/mL, 7.2 microg/mL, and 2.2 microg/mL after 10, 20, and 40 minutes exposure respectively. In vivo, no significant difference in lung levels of GCB was observed between a flow rate of 0.2 mL/min compared with 0.5 mL/min at any exposure time point (p < 0.05). Lung tissue was saturated after 20 minutes. Blood flow occlusion resulted in a lower plasma levels and higher lung levels of GCB compared with i.v. injection of the maximal tolerated dose of 40 mg. CONCLUSIONS: Growth inhibition of CC531 cells in vitro increased with exposure time while lung tissue was saturated after 20 minutes of BFO. No difference in GCB lung levels were seen after BFO compared with ILuP. Systemic exposure after i.v. injection was higher compared with BFO but did not result in higher lung levels.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Chemotherapy, Cancer, Regional Perfusion , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Lung/metabolism , Adenocarcinoma/metabolism , Animals , Cell Survival/drug effects , In Vitro Techniques , Lung Neoplasms/metabolism , Male , Pulmonary Circulation , Rats , Rats, Inbred Strains , Tumor Cells, Cultured , Gemcitabine
13.
Cancer Chemother Pharmacol ; 51(3): 221-6, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12655440

ABSTRACT

PURPOSE: Since there is a growing interest in preclinical research on interactions between radiation and cytotoxic agents, this study focused on the development of an alternative to the very laborious clonogenic assay (CA). METHODS: The colorimetric sulforhodamine B (SRB) assay was compared to the clonogenic assay for radiosensitivity testing in two lung cancer cell lines (A549, H292), one colon cancer cell line (HT-29) and one breast cancer cell line (MCF-7). In addition, the combination of the radiosensitizing agent gemcitabine and radiation was investigated with both assays. RESULTS: The dose-response curves obtained with the SRB assay and the CA were very similar up to 6 Gy. The radiosensitivity parameters (SF(2), alpha, beta, MID and ID(50)) obtained from the SRB assay and the CA were not significantly different between H292, A549 and MCF-7 cells. The radiation dose-response curves for A549 and H292 cells pretreated with 4 n M gemcitabine for 24 h clearly showed a radiosensitizing effect with both assays. The dose-enhancement factors obtained with the SRB assay and the CA were 1.80 and 1.76, respectively, for A549 cells, and 1.52 and 1.41 for H292 cells. CONCLUSIONS: The SRB assay was shown to be as useful as the more traditional CA for research on chemotherapy/radiotherapy interactions in cell lines with moderate radiosensitivity. This assay will be used for more extensive in vitro research on radiosensitizing compounds in these cell lines.


Subject(s)
Colony-Forming Units Assay/methods , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Radiation-Sensitizing Agents/pharmacology , Radiotherapy , Breast Neoplasms/pathology , Colonic Neoplasms/pathology , Colorimetry , Combined Modality Therapy , Fluorescent Dyes , Humans , Lung Neoplasms/pathology , Rhodamines , Sensitivity and Specificity , Tumor Cells, Cultured , Gemcitabine
14.
PLoS One ; 7(10): e46536, 2012.
Article in English | MEDLINE | ID: mdl-23094027

ABSTRACT

BACKGROUND: Cell viability and motility comprise ubiquitous mechanisms involved in a variety of (patho)biological processes including cancer. We report a technical comparative analysis of the novel impedance-based xCELLigence Real-Time Cell Analysis detection platform, with conventional label-based endpoint methods, hereby indicating performance characteristics and correlating dynamic observations of cell proliferation, cytotoxicity, migration and invasion on cancer cells in highly standardized experimental conditions. METHODOLOGY/PRINCIPAL FINDINGS: Dynamic high-resolution assessments of proliferation, cytotoxicity and migration were performed using xCELLigence technology on the MDA-MB-231 (breast cancer) and A549 (lung cancer) cell lines. Proliferation kinetics were compared with the Sulforhodamine B (SRB) assay in a series of four cell concentrations, yielding fair to good correlations (Spearman's Rho 0.688 to 0.964). Cytotoxic action by paclitaxel (0-100 nM) correlated well with SRB (Rho>0.95) with similar IC(50) values. Reference cell migration experiments were performed using Transwell plates and correlated by pixel area calculation of crystal violet-stained membranes (Rho 0.90) and optical density (OD) measurement of extracted dye (Rho>0.95). Invasion was observed on MDA-MB-231 cells alone using Matrigel-coated Transwells as standard reference method and correlated by OD reading for two Matrigel densities (Rho>0.95). Variance component analysis revealed increased variances associated with impedance-based detection of migration and invasion, potentially caused by the sensitive nature of this method. CONCLUSIONS/SIGNIFICANCE: The xCELLigence RTCA technology provides an accurate platform for non-invasive detection of cell viability and motility. The strong correlations with conventional methods imply a similar observation of cell behavior and interchangeability with other systems, illustrated by the highly correlating kinetic invasion profiles on different platforms applying only adapted matrix surface densities. The increased sensitivity however implies standardized experimental conditions to minimize technical-induced variance.


Subject(s)
Biological Assay , Endpoint Determination , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen , Diffusion Chambers, Culture , Drug Combinations , Electric Impedance , Female , Gentian Violet , Humans , Laminin , Paclitaxel/pharmacology , Proteoglycans , Reproducibility of Results , Rhodamines , Sensitivity and Specificity
15.
Int J Radiat Oncol Biol Phys ; 80(2): 558-66, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21377279

ABSTRACT

PURPOSE: Whereas radiosensitization by gemcitabine is well studied under normal oxygen conditions, little is known about its radiosensitizing potential under reduced oxygen conditions. Therefore, the present study evaluated the impact of anoxia on gemcitabine-mediated radiosensitization. METHODS AND MATERIALS: The clonogenic assay was performed in three isogenic A549 cell lines differing in p53 status (24 h, 0-15 nM gemcitabine, 0-8 Gy irradiation, normoxia vs. anoxia). Using radiosensitizing conditions, cells were collected for cell cycle analysis and apoptosis detection. RESULTS: Whereas wild-type p53 A549-LXSN cells were more sensitive to radiation than p53-deficient A549-E6 cells, both cell lines showed similar radiosensitization by gemcitabine under normoxia and anoxia. Independent of p53 functionality, gemcitabine was able to overcome anoxia-induced G(0/1) arrest and established an (early) S phase block in normoxic and anoxic cells. The percentage early and late apoptotic/necrotic cells increased with the gemcitabine/radiation combination, with a significant difference between A549-LXSN and A549-E6. CONCLUSIONS: This study is the first to show that gemcitabine retains its radiosensitizing potential under low oxygen conditions. Although radiosensitization was observed in both p53 wild-type and p53-deficient cells, p53 status might influence induction of apoptosis after gemcitabine/radiation treatment, whereas no effect on cell cycle progression was noticed.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Hypoxia , Deoxycytidine/analogs & derivatives , Lung Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Protein p53 , Analysis of Variance , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Deoxycytidine/pharmacology , Humans , Lung Neoplasms/pathology , Necrosis , Resting Phase, Cell Cycle/drug effects , S Phase/drug effects , Tumor Suppressor Protein p53/deficiency , Gemcitabine
16.
Diagn Mol Pathol ; 20(4): 203-11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22089347

ABSTRACT

BACKGROUND: Formalin-fixed paraffin-embedded (FFPE) tissue is the most common tissue specimen widely available. Moreover, long clinical follow-up is on hand. Therefore, FFPE material is a precious source of material for identifying predictive and/or prognostic biomarkers in cancer research on the basis of gene expression. However, the main drawback of FFPE tissue is the significant reduction in quantity and quality of the extracted RNA. The aim of this study is the comparison of different commercially available kits for the RNA isolation in FFPE tissue material. METHODS: Five commercially available RNA isolation kits were tested and the concentration, purity, integrity, and raw cycle threshold values were determined. RESULTS: The mean total RNA concentrations were as follows: Qiagen 25957±19417 ng, Ambion 8249±2898 ng, SA Biosciences 8070±3700 ng, and Macherey-Nagel 622±394 ng. The mean A260/A280 ratios were as follows: Qiagen: 1.81, SA Biosciences: 0.66, Ambion: 1.03, and Macherey-Nagel: 1.04. The mean A260/A230 ratios were as follows: Qiagen: 1.88, SA Biosciences: 1.61, Ambion: 1.54, and Macherey-Nagel: 1.88. The RNA extractions from Epicentre could not be measured by the Nanodrop and, therefore, were excluded from further analysis. The mean RNA integrity number (range, 2.09 to 2.47) and the mean raw cycle threshold values (range, 33.43 to 35.37) were more or less the same for all the tested RNA isolation kits. CONCLUSIONS: Altogether, on the basis of the number of adequate isolations, the kit from Qiagen seems to be the most appropriate kit to be used in our further studies that require RNA isolation from FFPE material.


Subject(s)
RNA/analysis , RNA/isolation & purification , Reagent Kits, Diagnostic , Biomarkers, Tumor/analysis , Formaldehyde , Humans , Paraffin Embedding , RNA/genetics , Tissue Fixation
17.
Phys Med Biol ; 55(7): N167-78, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20208097

ABSTRACT

The clonogenic assay is the method of choice to determine cell reproductive death after in vitro irradiation treatment. Traditionally, colony quantification has been performed by manual counting, a very laborious, time-consuming and rather subjective task. In this study, we compared manual counting by two skilled investigators with automated counting using the freely available ClonoCounter program. Five human tumour cell lines were irradiated under normoxia (21% O(2)) or anoxia (<0.1% O(2)), after 24 h or 6 h anoxic preincubation. Colonies were quantified manually or using the ClonoCounter software. A positive correlation between the absolute number of colonies counted manually and automatically was shown. Though there was a general trend of underpredicting the absolute number of cell colonies when counting automatically, survival curves were very similar, and in none of the cell lines were significant differences in radiobiological parameters such as mean inactivation dose, surviving fraction at 2 Gy and oxygen enhancement ratio detected. Our results suggest that the ClonoCounter provides sufficient reliability to be implemented for counting human tumour colonies in in vitro irradiation experiments. In contrast to several previously reported computer-aided colony-counting methods, it is a freely available program, requiring only minimal instrument costs.


Subject(s)
Cell Count/methods , Image Interpretation, Computer-Assisted/methods , Oxidative Stress/radiation effects , Radiation Tolerance , Software Validation , Software , Tumor Stem Cell Assay/methods , Animals , Humans
18.
Cancer Lett ; 286(2): 180-8, 2009 Dec 28.
Article in English | MEDLINE | ID: mdl-19540039

ABSTRACT

Hypoxic tumour regions often contain viable cells that are more resistant to chemotherapy and/or radiotherapy, making it of key importance to analyse new combination treatments under both normoxic and hypoxic conditions. In this study, the impact of moderate hypoxia and anoxia on cellular characteristics was investigated in isogenic A549 cells differing in p53 status. VEGF expression, doubling time, cell cycle distribution, induction of apoptosis and p53 protein expression were evaluated. Radiation survival curves yielded an oxygen enhancement ratio of 1.16-1.67. In conclusion, an in vitro hypoxia model that will be highly useful to analyse chemoradiation interactions is presented.


Subject(s)
Apoptosis/drug effects , Oxygen/pharmacology , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism , Apoptosis/radiation effects , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Humans , Oxygen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Suppressor Protein p53/genetics , Vascular Endothelial Growth Factor A/genetics
19.
Oncologist ; 12(6): 690-712, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17602059

ABSTRACT

As it is now well established that human solid tumors frequently contain a substantial fraction of cells that are hypoxic, more and more in vitro research is focusing on the impact of hypoxia on the outcome of radiotherapy and chemotherapy. Indeed, the efficacy of irradiation and many cytotoxic drugs relies on an adequate oxygen supply. Consequently, hypoxic regions in solid tumors often contain viable cells that are intrinsically more resistant to treatment with radiotherapy or chemotherapy. Moreover, efforts have been made to exploit hypoxia as a potential difference between malignant and normal tissues.Nowadays, a body of evidence indicates that oxygen deficiency clearly influences some major intracellular pathways such as those involved in cell proliferation, cell cycle progression, apoptosis, cell adhesion, and others. Obviously, when investigating the effects of radiotherapy or chemotherapy or both combined under hypoxic conditions, it is essential to consider the influences of hypoxia itself on the cell. In this review, we first focus on the effects of hypoxia per se on some critical biological pathways. Next, we sketch an overview of preclinical and clinical research on radiotherapy, chemotherapy, and chemoradiation under hypoxic conditions.


Subject(s)
Hypoxia , Neoplasms/drug therapy , Neoplasms/radiotherapy , Apoptosis , Cell Hypoxia , Cell Proliferation , Humans , Medical Oncology/methods , Neoplasms/pathology
20.
Oncologist ; 10(1): 34-51, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15632251

ABSTRACT

The combination of gemcitabine and radiotherapy is a promising combined modality therapy. However, the clinical application of this combination has to be implemented carefully because of an increased toxicity to normal tissues. A body of experimental evidence shows that gemcitabine is a potent radiosensitizer in vitro and in vivo. The observations so far indicate that various mechanisms are responsible for the radiosensitizing effect. Although it is often difficult to transfer experimental data to the clinic, these studies offer the possibility to develop an improved schedule of administration for patient treatment, based on rational evidence in tumor biology. In the current review, the preclinical data that support the use of gemcitabine as a radiosensitizing agent and the clinical trials that have been conducted to date are summarized.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Cell Cycle , Clinical Trials as Topic , Combined Modality Therapy , DNA/biosynthesis , DNA Damage , DNA Repair , Deoxycytidine/pharmacology , Humans , Radiation-Sensitizing Agents/pharmacology , Rodentia , Tumor Cells, Cultured , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL