Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Colloids Surf B Biointerfaces ; 105: 31-6, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23352945

ABSTRACT

This research is aimed to develop a nano-sized supramolecular micelle delivery system of cis-dichlorodiammine platinum (II) (CDDP) in order to achieve the passive tumor targeting. Firstly, star-shaped poly (γ-benzyl-L-glutamate) was synthesized by the ring-opening polymerization of γ-benzyl-L-glutamate-N-carboxyanhydride initiated with per-6-amino-ß-cyclodextrin. After removal of benzyl groups, ß-cyclodextrin based seven-armed poly (L-glutamic acid) (ß-CD-7PLGA) was obtained. ß-CD-7PLGA/CDDP complexes were prepared by the complex reaction between the carboxylic groups of ß-CD-7PLGA and CDDP. Further inclusion of ß-CD-7PLGA/CDDP complexes with adamantine terminated mPEG (mPEG-Ad) gave CDDP supramolecular micelles (mPEG-Ad@ß-CD-7PLGA/CDDP). The formation of mPEG-Ad@ß-CD-7PLGA/CDDP supramolecular micelles was confirmed by fluorescence spectrophotoscopy and particle size measurements. All the micelles showed spherical shape, and their sizes increased from 100 to 135 nm with the increase of PLGA arm molecular weight. mPEG-Ad@CD-7PLGA/CDDP micelles showed sustained drug release profiles over 50h in PBS. Compared with CDDP, mPEG-Ad@ß-CD-7PLGA/CDDP supramolecular micelles showed essential decreased cytotoxicity to KB cells, suggesting their great potential as the delivery carriers of CDDP.


Subject(s)
Amantadine/pharmacology , Cisplatin/pharmacology , Drug Delivery Systems , KB Cells/drug effects , Polyethylene Glycols/chemistry , Polyglutamic Acid/metabolism , beta-Cyclodextrins/chemistry , Antineoplastic Agents/pharmacology , Dopamine Agents/pharmacology , Humans , KB Cells/pathology , Micelles , Molecular Weight , Nanoparticles/administration & dosage , Particle Size , Polyethylene Glycols/metabolism , Polyglutamic Acid/chemistry , Spectrometry, Fluorescence , beta-Cyclodextrins/metabolism
3.
Eur Arch Otorhinolaryngol ; 267(4): 483-94, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20052589

ABSTRACT

This article gives an overview on different current strategies of assay-based response evaluation in head and neck squamous cell carcinomas (HNSCC) and critically summarizes their role and needs for future clinical evaluation. Due to a growing amount of data of phase III clinical trials of multimodality treatment options for HNSCC, treatment planning in regard to optimal outcome is becoming an interdisciplinary challenge. New concepts such as induction chemotherapy with bi- or ternary combinations of chemotherapeutics, integration of targeted therapies, concurrent and sequential chemoradiation concepts, and multimodality-based organ preservation strategies strongly compete with traditional definitive surgical procedures. Moreover, the outcome is difficult to predict due to heterogeneity of a tumor's response, impaired late functional outcome, and increased late toxicity if simultaneously applied to radiation. Retrospectively looking at non-responders with tumors classified as resectable, primary surgery is very likely to have achieved better results, since chemoradiation causes a high degree of early and late toxicities leading to extremely complicated terms and conditions in surgery following current multimodal therapeutic strategies. Unfortunately, predictive information on response characteristics of a given tumor before starting the therapy is not available in daily routine, although heterogeneity in response of a given tumor entity to treatments has been known for decades. Therefore, current therapy strategies for HNSCC still have to ignore this fact, creating an urgent need for the development of proper predictive assays. There are interesting clinical observations showing that response on induction chemotherapy may predict the outcome after radiotherapy. Some trials use this empiric phenomenon to pre-select non-responders for primary surgical treatment avoiding severe salvage complications after failure of complete chemoradiation treatment. Moving one step further, recent literature and our own investigations implicate that response evaluation of the individual patient's HNSCC in a suitable ex vivo assay just before starting the treatment is mature for clinical research. To this end, essential needs and hints are addressed and discussed.


Subject(s)
Carcinoma, Squamous Cell/therapy , Combined Modality Therapy/methods , Decision Making , Head and Neck Neoplasms/therapy , Antineoplastic Agents/pharmacology , Biopsy , Carcinoma, Squamous Cell/pathology , Feasibility Studies , Head and Neck Neoplasms/pathology , Humans , KB Cells/drug effects , KB Cells/pathology , Predictive Value of Tests , Reproducibility of Results , Salvage Therapy , Sensitivity and Specificity , Treatment Outcome
6.
Asia Pac J Clin Nutr ; 16 Suppl 1: 447-52, 2007.
Article in English | MEDLINE | ID: mdl-17392148

ABSTRACT

Lutein and zeaxanthin have various activities such as anti-age-related macular degeneration, anticataract, anticancer and cardiovascular diseases risk lowering, etc.; however, few studies have been reported on the role of free hydroxyl groups in the antitumor effects of lutein and zeaxanthin. The structure-activity relationship (SAR) of lutein and zeaxanthin di-acetylation derivatives has been investigated. The lutein and zeaxanthin were purified from corn protein residues and structurally modified by di-acetylation, which was characterized with UV/visible and HPLC/MS spectroscopy. The anti-proliferative effects of di-acetylated lutein or zeaxanthin on the human mouth epithelial cancer line KB cell were compared with controls of their original counterparts. Results showed that the anti-tumor activities of the di-acetylation of lutein and di-acetylation of zeaxanthin decreased significantly (p<0.05) at 50 micromol/L. Also the related IC50 dose was increased after di-acetylation. It was suggested that the hydroxyl groups contribute to the anti-tumor activity of lutein and zeaxanthin.


Subject(s)
Antineoplastic Agents/pharmacology , Lutein/pharmacology , Xanthophylls/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , KB Cells/pathology , KB Cells/ultrastructure , Lutein/chemistry , Structure-Activity Relationship , Xanthophylls/chemistry , Zeaxanthins
8.
Laryngoscope ; 112(5): 839-43, 2002 May.
Article in English | MEDLINE | ID: mdl-12150615

ABSTRACT

OBJECTIVES: Chemoprevention is a promising strategy to inhibit carcinoma before invasive tumors develop, but a new molecular target is desirable. Celecoxib is a newly developed cyclo-oxygenase (COX)-2 inhibitor with significantly less toxicity. The study was conducted to determine whether celecoxib is effective and safe in prevention of oral cancer. The antiangiogenic activity of celecoxib was studied to explore the potential mechanism involved. STUDY DESIGN: Randomized animal study. METHODS: The study consisted of two phases. In the phase 1,10 mice were used to determine the efficacy and safety of celecoxib with intradermal inoculation with oral carcinoma cells. The 10 mice were equally divided into two groups 5 mice (30 inoculated sites) in each group to receive 1,500 parts per million (ppm) celecoxib mixed in with the diet or to eat a normal diet, respectively, for 21 days. In phase 2, 10 more mice were inoculated to determine the effect of celecoxib on angiogenesis. Five mice received 3,000 ppm celecoxib in the diet, with the other five mice as control animals. The antiangiogenic activity was evaluated by comparing the density of newly growing microvessels after the inoculation. RESULTS: The results indicated that celecoxib significantly delayed cell growth and reduced tumor volume. There was statistical significance in the quantity of new vasculature in the tumor sites between the two groups. No toxic effect was found by means of measurement of body weight loss and microscopic dissection of organs. CONCLUSIONS: The study provided the first evidence to show the chemopreventive efficacy of celecoxib on oral cancer in a nude mouse model. Clinical trials are warranted to determine the efficacy in humans.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Squamous Cell/prevention & control , Cyclooxygenase Inhibitors/pharmacology , Mouth Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Sulfonamides/pharmacology , Administration, Oral , Animals , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Celecoxib , Cell Division/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Humans , KB Cells/drug effects , KB Cells/pathology , Mice , Mice, Nude , Mouth Neoplasms/blood supply , Mouth Neoplasms/pathology , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Pyrazoles
9.
Int Dent J ; 52 Suppl 3: 241-6, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12090461

ABSTRACT

AIM: Previous studies have demonstrated that methyl mercaptan (CH3SH), one of the main causes of oral malodour, might contribute to the initiation and progression of periodontal disease. These studies suggested that CH3SH may affect the epithelial cells of the gingival crevice, which form a barrier to the penetration of microbial substances. In this study, the effects of CH3SH on the epithelial cells and gingival fibroblasts were investigated. METHOD: Human oral epithelial carcinoma cell line (KB), human oral squamous cell carcinoma cell line (HSC-2), and human gingival fibroblasts (HGF) derived from healthy gingiva were used in this study. These cells were cultured in conditions of 5% CO2/95% air with or without CH3SH (10 ng/ml or 50 ng/ml) for 5 days. Cell numbers, proliferation and cytotoxicity were evaluated. RESULTS: CH3SH inhibited epithelial cell growth and proliferation at the concentration of 50 ng/ml, and a cytotoxic effect of CH3SH was also noted. On the other hand, HGF cells were not affected by 50 ng/ml CH3SH. CONCLUSION: High concentrations of CH3SH such as 50 ng/ml have an inhibitory effect on the growth and proliferation of epithelial cells, but not on those of fibroblasts.


Subject(s)
Gingiva/drug effects , Sulfhydryl Compounds/pharmacology , Analysis of Variance , Bromodeoxyuridine , Carcinoma, Squamous Cell/pathology , Cell Count , Cell Division/drug effects , Cell Survival , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Gingiva/pathology , Halitosis/metabolism , Humans , KB Cells/drug effects , KB Cells/pathology , L-Lactate Dehydrogenase/analysis , Statistics as Topic , Sulfhydryl Compounds/toxicity , Time Factors , Tumor Cells, Cultured
10.
Mol Med ; 8(11): 725-32, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12520089

ABSTRACT

BACKGROUND: Most tumors express death receptors and their activation represents a potential selective approach in cancer treatment. The most promising candidate for tumor selective death receptor-activation is tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/Apo2L, which activates the death receptors TRAIL-R1 and TRAIL-R2, and induces apoptosis preferentially in tumor cells but not in normal tissues. However, many cancer cells are not or only moderately sensitive towards TRAIL and require cotreatment with irradiation or chemotherapy to yield a therapeutically reasonable apoptotic response. Because chemotherapy can have a broad range of unwanted side effects, more specific means for sensitizing tumor cells for TRAIL are desirable. The expression of the cellular FLICE-like inhibitory protein (cFLIP) is regarded as a major cause of TRAIL resistance. We therefore analyzed the usefulness of targeting FLIP to sensitize tumor cells for TRAIL-induced apoptosis. MATERIALS AND METHODS: To selectively interfere with expression of cFLIP short double-stranded RNA oligonucleotides (small interfering RNAs [siRNAs]) were introduced in the human cell lines SV80 and KB by electroporation. Effects of siRNA on FLIP expression were analyzed by Western blotting and RNase protection assay and correlated with TRAIL sensitivity upon stimulation with recombinant soluble TRAIL and TRAIL-R1- and TRAIL-R2-specific agonistic antibodies. RESULTS: FLIP expression can be inhibited by RNA interference using siRNAs, evident from reduced levels of FLIP-mRNA and FLIP protein. Inhibition of cFLIP expression sensitizes cells for apoptosis induction by TRAIL and other death ligands. In accordance with the presumed function of FLIP as an inhibitor of death receptor-induced caspase-8 activation, down-regulation of FLIP by siRNAs enhanced TRAIL-induced caspase-8 activation. CONCLUSION: Inhibition of FLIP expression was sufficient to sensitize tumor cells for TRAIL-induced apoptosis. The combination of TRAIL and FLIP-targeting siRNA could therefore be a useful strategy to attack cancer cells, which are resistant to TRAIL alone.


Subject(s)
Apoptosis/drug effects , Carrier Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/pharmacology , RNA, Small Interfering/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Line, Transformed , Cell Survival , Drug Resistance, Neoplasm , Electroporation , Fibroblasts/drug effects , Flow Cytometry , Green Fluorescent Proteins , Humans , KB Cells/drug effects , KB Cells/pathology , Ligands , Luminescent Proteins/metabolism , Oligonucleotides/pharmacology , RNA, Messenger/metabolism , Ribonuclease, Pancreatic/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transfection
11.
Cancer Res ; 61(11): 4450-8, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389075

ABSTRACT

Vinblastine is an important antitumor agent that induces G(2)-M arrest and subsequent apoptosis in a wide variety of cell lines, but the molecular mechanisms that link mitotic arrest and apoptosis are poorly understood. The AP-1 transcription factor has been implicated in many critical cellular processes, including apoptosis, and is a major target of the c-Jun NH(2)-terminal kinase signaling pathway that is activated by vinblastine and other microtubule inhibitors. In this study we sought to determine the role of c-Jun NH(2)-terminal kinase/AP-1 in the response of KB3 carcinoma cells to vinblastine. For this purpose, we generated KB3 cell lines that stably expressed the c-Jun dominant-negative deletional mutant TAM67, which lacks the NH(2)-terminal transactivation domain. KB3-TAM67 cell lines displayed normal growth kinetics and essentially unaltered basal AP-1 activity, but vinblastine-induced phosphorylation of c-Jun and activating transcription factor-2, and AP-1 activation, were strongly inhibited. KB3-TAM67 cell lines arrested normally at G(2)-M in response to vinblastine, but were significantly more resistant to the drug, exhibiting markedly delayed apoptosis and increased overall survival, relative to control cells. To investigate the underlying mechanisms, differential expression of apoptotic regulatory genes was monitored by immunoblot and cDNA microarray analysis. We found that vinblastine treatment caused down-regulation of p53 and its target p21 and up-regulation of tumor necrosis factor alpha, Bak, and several other genes in control but not in KB3-TAM67 cells, identifying these genes as putative targets of vinblastine-inducible AP-1. These results demonstrate that vinblastine-inducible AP-1 plays a destructive, proapoptotic role and may do so by regulating the expression of a specific subset of target genes that promotes efficient apoptotic cell death following mitotic arrest.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Mitogen-Activated Protein Kinases/physiology , Transcription Factor AP-1/physiology , Vinblastine/pharmacology , Activating Transcription Factor 2 , Apoptosis/genetics , Apoptosis/physiology , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , JNK Mitogen-Activated Protein Kinases , KB Cells/drug effects , KB Cells/pathology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mitosis/drug effects , Phosphorylation/drug effects , Protein Structure, Tertiary , Signal Transduction/physiology , Transcription Factor AP-1/biosynthesis , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transfection , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
12.
Anticancer Drugs ; 11(5): 377-84, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10912954

ABSTRACT

We previously showed that conjugating doxorubicin to very large 70-500 kDa dextran decreased its removal rate from P-glycoprotein (P-gp) over-expressing, multidrug-resistant KB-V1 cells. Furthermore these conjugates could act synergistically with other cancer drugs. In the drug-sensitive 3-1 clone, but not in the V1 subclone which was 300-fold more resistant to free doxorubicin, conjugation led to a size-related decrease in toxicity. Here we identified the optimal size of dextran for avoiding P-gp-mediated efflux and yet preserving as much as possible doxorubicin toxicity. Chemically reduced, intracellularly stable 3.4-10 kDa conjugates were prepared. Confocal microscopy and fluorescence quenching experiments showed that these conjugates entered nuclei and interacted with DNA. In 3-1 cells, but not in V1 cells, cytotoxicity of conjugates decreased 14- to 45-fold linearly related to log size of the carrier (r=0.95). In V1 cells toxicity of the 10 kDa conjugate exceeded that of free doxorubicin. After conjugation the equilibrium binding constant of the DNA-drug complex (KA) decreased only by up to 3-fold. In 3-1 cells, but not in VI cells, DNA binding kinetics was an important factor and toxicity could be linearly correlated to 1/KA of conjugate (r=0.94). Drug accumulation decreased with an increase in dextran size but drug removal was decreased only in V1 cells. It appeared that drug uptake was also sensitive to dextran conjugation. In Vl cells drug removal was sensitive to the P-gp inhibitor verapamil or energy starvation. Ratios of V1/3-1 toxicity, drug accumulation and drug removal correlated linearly with log dextran size. When these ratios equaled 1, dextran sizes were estimated to be 32, 103 and 21 kDa, respectively.


Subject(s)
DNA, Neoplasm/metabolism , Dextrans/metabolism , Doxorubicin/metabolism , KB Cells/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Survival/drug effects , DNA, Neoplasm/drug effects , Dextrans/toxicity , Doxorubicin/analogs & derivatives , Doxorubicin/toxicity , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , KB Cells/metabolism , KB Cells/pathology , Regression Analysis , Rhodamines/metabolism , Spectrometry, Fluorescence , Time Factors
13.
Oral Oncol ; 36(2): 242-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10745179

ABSTRACT

Pingyangmycin (PYM; Bleomycin A(5)), an antitumour antibiotic is currently used during anticancer therapy. Previous experiments demonstrated that the therapeutic efficiency of PYM for treatment of malignant tumours is considered to be related to its ability to cause DNA strand breaks in vitro. However, very little is known about the interaction of PYM with the target cells, and it is still unclear how PYM enters the cells. In this study, cell death induced by PYM was studied in a human squamous cell carcinoma cell line (KB cells). In order to determine if cell death occurred by necrosis (reproductive cell death) or apoptosis (programmed cell death), KB cells were exposed to different concentrations of PYM and evaluated by biochemical and morphological criteria. Our results indicate that KB cells displayed an arrest in the G(2)-M phase of the cell cycle and became enlarged and polynucleated before dying at the low concentrations of PYM. In contrast, when cells were exposed to high concentrations of PYM, morphological changes identical to those usually associated with apoptosis were observed as well as internucleosomal digestion of genomic DNA. In conclusion, we demonstrate that PYM is able to induce two distinct modes of cell death depending on the doses of PYM.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Apoptosis , Bleomycin/analogs & derivatives , Carcinoma, Squamous Cell/drug therapy , Apoptosis/genetics , Bleomycin/therapeutic use , Carcinoma, Squamous Cell/pathology , DNA Fragmentation/drug effects , DNA, Neoplasm/drug effects , Dose-Response Relationship, Drug , Humans , KB Cells/drug effects , KB Cells/pathology , Necrosis
14.
Br J Cancer ; 81(3): 423-30, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10507766

ABSTRACT

This study has compared the preferential killing of three multidrug-resistant (MDR) KB cell lines, KB-C1, KB-A1 and KB-V1 by two inhibitors of glucosylceramide synthase, 1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP) and 1-phenyl-2-hexadecanoylamino-3-pyrrolidino-1-propanol (PPPP), to the killing produced by these compounds in the drug-sensitive cell line, KB-3-1. Both of the inhibitors caused much greater induction of apoptosis in each of the three MDR cell lines than in the drug-sensitive cell line, as judged by morphological assay and confirmed by poly-(ADP-ribose)-polymerase cleavage. The highest level of apoptosis was produced following 24-h exposure to 5 microM PPPP. This treatment produced 75.8 (+/- 7.1)%, 73.6 (+/- 9.8)% and 75.3 (+/- 6.4)% apoptotic cells in the three MDR cell lines respectively, compared to 19.0 (+/- 9.8)% in the drug-sensitive cell line. A reduction in glucosylceramide level following inhibitor treatment occurred in KB-3-1 cells as well as in the MDR cell lines, suggesting that the increased apoptotic response in the MDR cells reflected a different downstream response to changes in the levels of this lipid in these cells compared to that in the drug-sensitive cells. These results suggest that the manipulation of glucosylceramide levels may be a fruitful way of causing the preferential killing of MDR cells in vitro and possibly in vivo.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Glucosylceramides/metabolism , Glucosyltransferases/antagonists & inhibitors , KB Cells/drug effects , Morpholines/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Propanolamines/pharmacology , Pyrrolidines/pharmacology , Humans , KB Cells/pathology , Lipid Metabolism , Neoplasm Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism
15.
Bioelectromagnetics ; 20(1): 34-41, 1999.
Article in English | MEDLINE | ID: mdl-9915591

ABSTRACT

Electrochemical treatment (ECT) of cancer is a promising new method by which direct current is delivered into tumor tissue to induce tumor regression. The purpose of this study is to evaluate the effectiveness of ECT on human cancer cells and to investigate the factors that affect ECT. The biological mechanisms of ECT in cancer treatment were also explored. Using human KB cells, ECT was found to delay cell growth by using 0.3 coulombs (C)/ml (1.5 C in 5 ml of culture medium; 3 V, 400 microA for 62.5 min). From the results of a colony-forming assay, it was clearly demonstrated that increasing the ECT dose decreases tumor cell survival. A cytotoxicity study, in which a methylene blue assay was used, determined that, for 2.5 x 10(5) cells in culture, the 1D50 was 0.68 C/ml. For a fixed dose of 0.6 C/ml (3 C in 5 ml), using higher current and shorter treatment time resulted in better cell survival. Time, therefore, is an important factor. When cell concentration was altered, the survival was higher for increased cell concentrations. A thymidine incorporation assay indicated that the amount of [3H]thymidine incorporated into DNA decreased as the ECT dose increased. After treatment with 1 C/ml (5 C in 5 ml; 3 V, 400 microA for 208.4 min), pH at the anode decreased to 4.53 and at the cathode increased to 10.46. These results indicate that ECT is effective for killing human KB cells in vitro and that the toxicity effect is related to charge, current, and treatment time. The effect of pH alteration on cells is one of the mechanisms of ECT.


Subject(s)
Electric Stimulation Therapy , KB Cells/pathology , Cell Division , Cell Survival , Coloring Agents , DNA, Neoplasm/metabolism , Electrochemistry , Humans , Hydrogen-Ion Concentration , Methylene Blue , Neoplastic Stem Cells/pathology , Radiopharmaceuticals , Thymidine/metabolism , Time Factors , Tritium
16.
Oral Oncol ; 34(6): 524-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9930366

ABSTRACT

The radioresistant N10 and parental KB cell lines were examined for the expression of human DNA repair genes which were related to the repair of radiation-induced DNA damage by northern blot analysis using five kinds of DNA probes (XRCC1, XRCC3, XRCC5, RAD51, RAD52). In the unirradiated condition, N10 cells showed higher expression of XRCC1, XRCC3 and RAD51 mRNA than did KB cells. The X-irradiation induced a time-dependent increase in the mRNA levels of XRCC3 and RAD51 in both cell lines with a maximum at 2 h postirradiation. The XRCC1 mRNA in N10 was maintained at the same level even after irradiation, whereas that in KB was decreased after irradiation. There was no difference in the expression of XRCC5 and RAD52 mRNA between N10 and KB cells in both unirradiated and irradiated conditions. From these findings, it was suggested that XRCC1, XRCC3 and RAD51 contribute to the radioresistance in cell line N10.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/genetics , Blotting, Northern/methods , Cell Line , DNA, Complementary , Humans , KB Cells/pathology , KB Cells/radiation effects , RNA, Neoplasm/isolation & purification , Rad51 Recombinase , Radiation Tolerance , X-ray Repair Cross Complementing Protein 1
17.
Mutagenesis ; 12(1): 29-33, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9025094

ABSTRACT

The induction of chromosome-type aberrations is mediated by stabilization of the DNA-topoisomerase II (topo-II) complex, the cleavable complex (CC), induced by topo-II inhibitors. In the present study, in order to confirm the critical contribution of topo-II in producing chromosome-type aberrations, the inducibility of chromosome-type aberrations by topo-II inhibitors was compared between human epidermoid cancer KB cells and their mutant cells (KB/ VP-2 cells) which were resistant to etoposide (VP-16) and which have reduced levels of topo-II and its gene expression. KB/VP-2 cells were resistant to the cytotoxicity of topo-II inhibitors and most resistant to etoposide. In KB cells treated with etoposide which had accumulated CC, chromosome-type aberrations but not chromatid-type aberrations were efficiently induced, as has already been reported in Chinese hamster fibroblasts. In contrast, in KB/VP-2 cells with no accumulation of CC, etoposide induced mainly chromatid-type aberrations, with a few chromosome-type aberrations. Unlike etoposide, however, adriamycin, which was known to accumulate CC in Chinese hamster fibroblastic cells, neither induced chromosome-type aberrations nor accumulated CC in either KB or KB/VP-2 cells. No difference in cell incorporation of [3H]etoposide between the two cell lines was observed. These findings indicate that CC formation contributes to the induction of chromosome-type aberrations, although the reason why adriamycin could not accumulate CC in KB cells is not clear. This may suggest a mechanism for resistance to topo-II inhibitors in cancer chemotherapy.


Subject(s)
Chromosome Aberrations , DNA Topoisomerases, Type II/metabolism , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Topoisomerase II Inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , DNA/drug effects , DNA/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/metabolism , Etoposide/metabolism , Etoposide/pharmacokinetics , Humans , KB Cells/drug effects , KB Cells/pathology , Tritium
18.
Yao Xue Xue Bao ; 27(9): 656-61, 1992.
Article in Chinese | MEDLINE | ID: mdl-1293935

ABSTRACT

As a continuing part of our study on the chemistry and antitumor activity of podophyllotoxin, 11 new C-4 S-substituted podophyllotoxin derivatives were synthesised and screened in vitro against L1210 leukemia and KB cells. Thus, 4'-demethylepipodophyllotoxin was reacted with thiols in the presence of BF3. Et2O or trifluroacetic acid to give thioethers. In addition, 4-bromo-4-deoxy-4'-demethylepipodophyllotoxin, when reacted with thiols, also gave rise to corresponding thioether (4-alkylthio-4-deoxy-4'-demethylepipodophyllotoxins). Compounds 10 and 12 have the same activity as etoposide in the inhibition against L1210 leukemia, and compounds 9, 10, 12 and 15 also have comparable activity with etoposide against KB cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Podophyllotoxin/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Humans , KB Cells/pathology , Leukemia L1210/pathology , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Tumor Cells, Cultured/drug effects
19.
Auris Nasus Larynx ; 18(3): 235-69, 1991.
Article in German | MEDLINE | ID: mdl-1799331

ABSTRACT

In this study, we attempted to develop a technique, by which three-dimensional tumors were produced from two cultured head and neck tumor cell lines (Hep2, KB) and a colon adenocarcinoma cell line (HT29) using fibrinogen, thrombin, and double layered agar system. The three-dimensional tumor was large enough to perform the histologic study, which showed no significant histologic difference in comparison with the histologic findings of the xenografted tumor on nude mice. Furthermore, we applied this assay model to evaluate the antitumor effect of lymphokine activated killer (LAK) cells on the three-dimensional tumor produced by the technique. When tumor cells were cocultured with LAK cells, the damage of the three-dimensional structure due to the degeneration of tumor cells was observed. These findings suggest that the three-dimensional tumor may be useful to evaluate the antitumor effect of LAK cells in term of head and neck solid tumors.


Subject(s)
Culture Techniques/standards , KB Cells , Killer Cells, Lymphokine-Activated/physiology , Tumor Cells, Cultured , Adenocarcinoma , Antigens, Differentiation, T-Lymphocyte/analysis , Colonic Neoplasms , Culture Media/standards , Culture Techniques/methods , Evaluation Studies as Topic , HLA-DR Antigens/analysis , Head and Neck Neoplasms , Humans , Immunohistochemistry , KB Cells/immunology , KB Cells/pathology , Killer Cells, Lymphokine-Activated/immunology , T-Lymphocyte Subsets/chemistry , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/pathology
20.
J Gen Virol ; 70 ( Pt 8): 1975-86, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2527966

ABSTRACT

The cell line KB18 constitutively expresses adenovirus type 2 (Ad2) early region 1B (E1B) genes in the absence of E1A expression and thus is useful for understanding the function and properties of E1B gene products. We report here that KB18 cells complement the cyt and deg phenotypes of Ad2 cytocidal (cyt) mutants and an Ad12 cyt mutant. Thus the E1B 19K polypeptide in KB18 is functional. Expression of E1B mRNA and synthesis of the 19K polypeptide were studied in wild-type Ad2- and Ad2cyt15-infected KB cells at 15 h post-infection, and in KB18 cells. Although E1B mRNA synthesized in KB18 cells at 5 to 7% of the level in Ad2-infected KB cells, the amount of E1B 19K polypeptide formed was similar. In contrast, the amount of 19K protein in Ad2cyt15-infected KB cells was about one-tenth of that in Ad2-infected cells, although expression of E1B mRNA was comparable to that during Ad2 infection. The half-lives of the 19K polypeptide in wild-type-infected KB cells, in Ad2cyt15-infected KB cells and in KB18 were approximately 90 min, 25 min and 22 h, respectively. The cyt phenotype was expressed at 30 to 35 h post-infection, thus showing that the instability of the 19K polypeptide in Ad2cyt15-infected cells is not due to alteration of cell morphology or cell destruction.


Subject(s)
Adenoviruses, Human/physiology , Oncogene Proteins, Viral/physiology , Adenovirus Early Proteins , Adenoviruses, Human/genetics , Cell Transformation, Viral , DNA/metabolism , Half-Life , Humans , KB Cells/metabolism , KB Cells/microbiology , KB Cells/pathology , Molecular Weight , Mutation , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/genetics , Phenotype , RNA, Messenger/metabolism , RNA, Viral/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...