Loratadine dysregulates cell cycle progression and enhances the effect of radiation in human tumor cell lines

Radiat Oncol. 2010 Feb 3:5:8. doi: 10.1186/1748-717X-5-8.

Abstract

Background: The histamine receptor-1 (H1)-antagonist, loratadine has been shown to inhibit growth of human colon cancer xenografts in part due to cell cycle arrest in G2/M. Since this is a radiation sensitive phase of the cell cycle, we sought to determine if loratadine modifies radiosensitivity in several human tumor cell lines with emphasis on human colon carcinoma (HT29).

Methods: Cells were treated with several doses of loratadine at several time points before and after exposure to radiation. Radiation dose modifying factors (DMF) were determined using full radiation dose response survival curves. Cell cycle phase was determined by flow cytometry and the expression of the cell cycle-associated proteins Chk1, pChk1(ser345), and Cyclin B was analyzed by western blot.

Results: Loratadine pre-treatment of exponentially growing cells (75 microM, 24 hours) increased radiation-induced cytotoxicity yielding a radiation DMF of 1.95. However, treatment of plateau phase cells also yielded a DMF of 1.3 suggesting that mechanisms other than cell cycle arrest also contribute to loratadine-mediated radiation modification. Like irradiation, loratadine initially induced G2/M arrest and activation of the cell-cycle associated protein Chk1 to pChk1(ser345), however a subsequent decrease in expression of total Chk1 and Cyclin B correlated with abrogation of the G2/M checkpoint. Analysis of DNA repair enzyme expression and DNA fragmentation revealed a distinct pattern of DNA damage in loratadine-treated cells in addition to enhanced radiation-induced damage. Taken together, these data suggest that the observed effects of loratadine are multifactorial in that loratadine 1) directly damages DNA, 2) activates Chk1 thereby promoting G2/M arrest making cells more susceptible to radiation-induced DNA damage and, 3) downregulates total Chk1 and Cyclin B abrogating the radiation-induced G2/M checkpoint and allowing cells to re-enter the cell cycle despite the persistence of damaged DNA.

Conclusions: Given this unique possible mechanism of action, loratadine has potential as a chemotherapeutic agent and as a modifier of radiation responsiveness in the treatment of cancer and, as such, may warrant further clinical evaluation.

Publication types

  • Research Support, N.I.H., Intramural

MeSH terms

  • Blotting, Western
  • Cell Cycle / drug effects*
  • Cell Cycle / radiation effects*
  • Cell Cycle Proteins / drug effects*
  • DNA Repair Enzymes / drug effects*
  • HT29 Cells
  • Humans
  • Loratadine / pharmacology*
  • Radiation Tolerance / drug effects
  • Radiation-Sensitizing Agents / pharmacology*

Substances

  • Cell Cycle Proteins
  • Radiation-Sensitizing Agents
  • Loratadine
  • DNA Repair Enzymes