Radiation Oncology Radiosensitization by 2-benzoyl-3-phenyl-6,7-dichloroquinoxaline 1,4-dioxide under Oxia and Hypoxia in Human Colon Cancer Cells

BackgroundThe sensitizing effects of 2-benzoyl-3-phenyl-6,7-dichloroquinoxaline 1,4-dioxide (DCQ) and ionizing radiation (IR) were determined in four colon cancer cells and in FHs74Int normal intestinal cells.MethodsCell cycle modulation, TUNEL assay, clonogenic survival and DNA damage were examined under oxia or hypoxia. Effects on apoptotic molecules and on p-Akt and Cox-2 protein expression were investigated.ResultsThe four cell lines responded differently to DCQ+IR; HT-29 cells were most resistant. Combination treatment caused significant increases in preG1 (apoptosis) in HCT-116, while G2/M arrest occurred in DLD-1. DCQ potentiated IR effects more so under hypoxia than oxia. Pre-exposure of DLD-1 to hypoxia induced 30% apoptosis, and G2/M arrest in oxia. The survival rate was 50% lower in DCQ+IR than DCQ alone and this rate further decreased under hypoxia. FHs74Int normal intestinal cells were more resistant to DCQ+IR than cancer cells.Greater ssDNA damage occurred in DLD-1 exposed to DCQ+IR under hypoxia than oxia. In oxia, p-Akt protein expression increased upon IR exposure and drug pre-treatment inhibited this increase. In contrast, in hypoxia, exposure to IR reduced p-Akt protein and DCQ restored its expression to the untreated control. Apoptosis induced in hypoxic DLD-1 cells was independent of p53-p21 modulation but was associated with an increase in Bax/Bcl-2 ratio and the inhibition of the Cox-2 protein.ConclusionDCQ is a hypoxic cell radiosensitizer in DLD-1 human colon cancer cells.

[1]  A. López de Cerain,et al.  New quinoxalinecarbonitrile 1,4-di-N-oxide derivatives as hypoxic-cytotoxic agents. , 2000, European journal of medicinal chemistry.

[2]  H. Gali-Muhtasib,et al.  Quinoxaline 1,4-dioxides are novel angiogenesis inhibitors that potentiate antitumor effects of ionizing radiation. , 2004, International journal of oncology.

[3]  H. Gali-Muhtasib,et al.  Quinoxaline 1,4‐dioxides: Hypoxia‐selective therapeutic agents , 2002, Molecular carcinogenesis.

[4]  W. El-Deiry The role of p53 in chemosensitivity and radiosensitivity , 2003, Oncogene.

[5]  Stephen N. Jones,et al.  Regulation of p53 stability by Mdm2 , 1997, Nature.

[6]  K. Yoneda,et al.  Mn‐SOD antisense upregulates in vivo apoptosis of squamous cell carcinoma cells by anticancer drugs and γ‐rays regulating expression of the BCL‐2 family proteins, COX‐2 and p21 , 2001, International journal of cancer.

[7]  Giuseppe Schettino,et al.  New insights on cell death from radiation exposure. , 2005, The Lancet. Oncology.

[8]  P. Vaupel,et al.  Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects. , 2001, Journal of the National Cancer Institute.

[9]  S. Welz,et al.  Hypoxic radiosensitizers and hypoxic cytotoxins in radiation oncology. , 2003, Current medicinal chemistry. Anti-cancer agents.

[10]  K. Fukuchi,et al.  Phosphatidylinositol 3-kinase inhibitors, Wortmannin or LY294002, inhibited accumulation of p21 protein after gamma-irradiation by stabilization of the protein. , 2000, Biochimica et biophysica acta.

[11]  J. Masferrer,et al.  COX-2 Inhibitors as Radiosensitizing Agents for Cancer Therapy , 2003, American journal of clinical oncology.

[12]  Johan Bussink,et al.  Clinical studies of hypoxia modification in radiotherapy. , 2004, Seminars in radiation oncology.

[13]  Y. Matsui,et al.  Effects of p53 Mutations on Cellular Sensitivity to Ionizing Radiation , 2001, American journal of clinical oncology.

[14]  James B. Mitchell,et al.  Effects of hypoxia on radiation-responsive stress-activated protein kinase, p53, and caspase 3 signals in TK6 human lymphoblastoid cells. , 2005, Cancer research.

[15]  A. Levine,et al.  Cyclooxygenase-2 Suppresses Hypoxia-induced Apoptosis via a Combination of Direct and Indirect Inhibition of p53 Activity in a Human Prostate Cancer Cell Line* , 2005, Journal of Biological Chemistry.

[16]  G. Chowdhury,et al.  Redox-activated, hypoxia-selective DNA cleavage by quinoxaline 1,4-di-N-oxide. , 2001, Bioorganic & medicinal chemistry.

[17]  J. Brown,et al.  Exploiting the hypoxic cancer cell: mechanisms and therapeutic strategies. , 2000, Molecular medicine today.

[18]  I. Stratford,et al.  Pharmacological and biological evaluation of a series of substituted 1,4-naphthoquinone bioreductive drugs. , 2004, Biochemical pharmacology.

[19]  W. El-Deiry,et al.  Regulation of p53 downstream genes. , 1998, Seminars in cancer biology.

[20]  Z. Szewczuk,et al.  Direct solid-phase synthesis of quinoxaline-containing peptides , 2005 .

[21]  M. V. Heiden,et al.  Redox regulation of p53 during hypoxia , 2000, Oncogene.

[22]  Y Taya,et al.  Inhibition of cyclin-dependent kinase 2 by p21 is necessary for retinoblastoma protein-mediated G1 arrest after gamma-irradiation. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[23]  Peter Vaupel,et al.  Tumor microenvironmental physiology and its implications for radiation oncology. , 2004, Seminars in radiation oncology.

[24]  H. Choy,et al.  Enhancing radiotherapy with cyclooxygenase-2 enzyme inhibitors: a rational advance? , 2003, Journal of the National Cancer Institute.

[25]  E. Reyt,et al.  Chronic hypoxia protects against gamma-irradiation-induced apoptosis by inducing bcl-2 up-regulation and inhibiting mitochondrial translocation and conformational change of bax protein. , 2003, International journal of oncology.