Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Gene Ther ; 18(10): 751-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21869826

ABSTRACT

The herpes simplex virus thymidine kinase (HSV-TK) is the most widely used suicide gene in cancer gene therapy due to its superior anticancer activity with ganciclovir (GCV) compared with other HSV-TK substrates, such as 1-ß-D-arabinofuranosyl thymine (araT). We have evaluated the role of DNA damage as a mechanism for the superiority of GCV. Using γ-H2AX foci as an indicator of DNA damage, GCV induced ≥ sevenfold more foci than araT at similar cytotoxic concentrations. The number of foci decreased after removal of either drug, followed by an increase in Rad51 foci indicating that homologous recombination repair (HRR) was used to repair this damage. Notably, only GCV produced a late and persistent increase in γ-H2AX foci demonstrating the induction of unrepairable DNA damage. Both drugs induced the ATR damage response pathway, as evidenced by Chk1 activation. However, GCV resulted in greater activation of ATM, which coincided with the late induction of γ-H2AX foci, demonstrating the presence of DNA double-strand breaks (DSBs). The increase in DSBs after Rad51 induction suggested that they occurred as a result of a failed attempt at HRR. These data demonstrate that the late and unrepairable DSBs observed uniquely with GCV account for its superior cytotoxicity and further suggest that inhibition of HRR will enhance cytotoxicity with HSV-TK/GCV.


Subject(s)
DNA Breaks, Double-Stranded , Ganciclovir/toxicity , Simplexvirus/enzymology , Simplexvirus/genetics , Thymidine Kinase/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Checkpoint Kinase 1 , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Histones/metabolism , Humans , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Rad51 Recombinase/metabolism , Thymidine Kinase/metabolism , Tumor Suppressor Proteins/metabolism
2.
Gene Ther ; 9(15): 1023-30, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101433

ABSTRACT

We have previously demonstrated with several cell lines in vitro that hydroxyurea (HU) synergistically enhances ganciclovir (GCV)-mediated cytotoxicity in bystander cells. In this study, we evaluated the role of DNA synthesis inhibition on enhanced bystander killing and assessed whether addition of HU would improve the efficacy of the HSV-TK/GCV system in vivo. Compared with GCV treatment alone, addition of HU resulted in increased DNA synthesis inhibition and delayed progression through S phase following removal of drug. In a xenograft tumor model, 1:10 and 1:1 mixtures of HSVtk- and LacZ-expressing SW620 cells were injected s.c. in the flanks of nude mice and treated i.p. (100 mg/kg GCV, 1500 mg/kg HU) daily for 5 days. Tumors from mice treated with GCV alone grew rapidly and increased to 10 times their initial size in 15.7 +/- 1.8 and 16.0 +/- 0.9 days for 1:10 and 1:1 mixtures, respectively. However, when both GCV and HU were administered in combination, a single complete tumor regression was observed in both the 1:10 and 1:1 groups. In the remaining mice treated with GCV/HU, it took 23.2 +/- 2.1 (1:10) and 26.4 +/- 3.8 days (1:1) to obtain a similar 10-fold increase in tumor size.


Subject(s)
Antimetabolites/therapeutic use , Antiviral Agents/therapeutic use , Colonic Neoplasms/therapy , Ganciclovir/therapeutic use , Genetic Therapy/methods , Hydroxyurea/therapeutic use , Animals , Aphidicolin/therapeutic use , Bystander Effect , Cell Cycle/drug effects , Cell Line , Drug Synergism , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Simplexvirus/enzymology , Thymidine Kinase/genetics
3.
Cancer Res ; 60(21): 6080-8, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085531

ABSTRACT

Gemcitabine (2',2'-difluoro-2'-deoxycytidine; dFdCyd) has been shown to be a potent radiosensitizer in tumor cells both in vitro and in vivo. We evaluated the ability of dFdCyd to enhance the radiosensitivity of two human glioblastoma cell lines. The results demonstrated that U251 cells were more sensitive to the cytotoxicity of dFdCyd, and that dFdCyd was able to radiosensitize these cells. In contrast, D54 cells were more resistant to the cytotoxic effect of dFdCyd, and no radiosensitization occurred at any concentration of dFdCyd tested. Because radiosensitization by dFdCyd has been correlated with its ability to deplete dATP pools through inhibition of ribonucleotide reductase by dFdCyd diphosphate, we evaluated the metabolism of dFdCyd in both cell lines. At equitoxic concentrations of dFdCyd, both cell lines accumulated similar levels of the cytotoxic metabolite, dFdCyd triphosphate, as well as similar levels of dFdCyd monophosphate in DNA. In U251 cells, radiosensitizing concentrations of dFdCyd (10 or 25 nM; IC10 or IC50) depleted dATP by approximately 80% within 4 h. In contrast, 80 nM (IC50) was unable to deplete dATP by >30% within 4 h in D54 cells. Higher concentrations of dFdCyd or hydroxyurea, an inhibitor of ribonucleotide reductase that depleted dATP >90%, also did not produce radiosensitization in D54 cells. D54 cells were not resistant to radiosensitization because bromodeoxyuridine was able to induce radiosensitization. Because D54 cells express wild-type p53, whereas U251 cells express a mutant p53, the effect of dFdCyd and ionizing radiation on cell cycle progression was evaluated. Radiation alone produced a G1 block in D54 cells and a transient G2-M block in U251 cells. After a 24 h incubation with dFdCyd alone or in combination with ionizing radiation, U251 cells readily accumulated in S-phase, which remained elevated for at least 72 h, consistent with previous results in other mutant p53 cell lines. In addition, radiation enhanced the ability of dFdCyd to induce S-phase-specific cell death in U251 cells. In contrast, D54 cells showed a G1 block after dFdCyd and radiation exposure, with fewer cells in S-phase for at least 48 h after drug washout/irradiation. Furthermore, treatment with dFdCyd and/or radiation did not increase the amount of S-phase-specific cell death in D54 cells compared with control cells. These results suggest that the G1 block in D54 cells resulting from wild-type p53 induction prevented radiosensitization by dFdCyd.


Subject(s)
Cell Cycle/physiology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Nuclear Proteins , Radiation Tolerance/physiology , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Protein p53/physiology , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis/radiation effects , Bromodeoxyuridine/pharmacology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Deoxyadenine Nucleotides/metabolism , Deoxycytidine/metabolism , Deoxycytidine/toxicity , Glioblastoma/pathology , Humans , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-mdm2 , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/metabolism , Radiation-Sensitizing Agents/toxicity , Tumor Cells, Cultured/drug effects , Tumor Suppressor Protein p53/biosynthesis , Gemcitabine
4.
Cancer Res ; 60(6): 1631-6, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10749133

ABSTRACT

We have previously demonstrated (L. Z. Rubsam et al., Cancer Res., 59: 669-675, 1999) that low ganciclovir (GCV) triphosphate (TP) levels similar to cellular deoxynucleotide concentrations can induce multilog killing in cells stably expressing herpes simplex virus thymidine kinase (HSV-TK). In this study, we evaluated whether reducing the endogenous competitor of GCV-TP, dGTP, enhanced GCV-mediated cytotoxicity. In SW620 human colon carcinoma cells stably expressing HSV-TK, the addition of the ribonucleotide reductase inhibitor, hydroxyurea (HU), decreased cellular dGTP pools and simultaneously increased the accumulation of GCV-TP levels. The amount of GCV nucleotide transfer from HSV-TK-expressing to nonexpressing (bystander) cells was quantitated in physically separated pHook-expressing bystander cells. Elevation of the GCV-TP:dGTP ratio by HU resulted in increased levels of GCV nucleotides transferred from HSV-TK-expressing to bystander cells during a 24 h drug incubation and enhanced GCV monophosphate incorporation into DNA after drug removal. Isobologram analysis demonstrated that the combination of GCV and HU was additive in 100% HSV-TK cultures and synergistic in HSV-TK/bystander mixtures. IC50 values for GCV in 1:1 cocultures of HSV-TK-expressing and nonexpressing SW620 cells were reduced from 1.5 microM to 0.07 microM with 2 mM HU. A similar reduction was also observed with HT-29 cells and U251 cells. With 2 mM HU, IC50 values for GCV in 10:90, 5:95, and 1:99 SW620 HSV-TK-expressing and nonexpressing cocultures were reduced from 55 microM to 0.3 microM, 71 microM to 0.8 microM, and 118 microM to 7 microM, respectively. These results demonstrate the ability to pharmacologically enhance HSV-TK/GCV-mediated bystander killing and may have an important therapeutic impact.


Subject(s)
Antineoplastic Agents/pharmacology , Ganciclovir/pharmacology , Hydroxyurea/pharmacology , Thymidine Kinase/genetics , Cell Survival/drug effects , Coculture Techniques , DNA/drug effects , DNA/metabolism , Deoxyribonucleotides/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Ganciclovir/analogs & derivatives , Ganciclovir/metabolism , Humans , Inhibitory Concentration 50 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Simplexvirus/enzymology , Thymidine Kinase/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...