32
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Sensitization of human cancer cells to gemcitabine by the Chk1 inhibitor MK-8776: cell cycle perturbation and impact of administration schedule in vitro and in vivo

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Chk1 inhibitors have emerged as promising anticancer therapeutic agents particularly when combined with antimetabolites such as gemcitabine, cytarabine or hydroxyurea. Here, we address the importance of appropriate drug scheduling when gemcitabine is combined with the Chk1 inhibitor MK-8776, and the mechanisms involved in the schedule dependence.

          Methods

          Growth inhibition induced by gemcitabine plus MK-8776 was assessed across multiple cancer cell lines. Experiments used clinically relevant “bolus” administration of both drugs rather than continuous drug exposures. We assessed the effect of different treatment schedules on cell cycle perturbation and tumor cell growth in vitro and in xenograft tumor models.

          Results

          MK-8776 induced an average 7-fold sensitization to gemcitabine in 16 cancer cell lines. The time of MK-8776 administration significantly affected the response of tumor cells to gemcitabine. Although gemcitabine induced rapid cell cycle arrest, the stalled replication forks were not initially dependent on Chk1 for stability. By 18 h, RAD51 was loaded onto DNA indicative of homologous recombination. Inhibition of Chk1 at 18 h rapidly dissociated RAD51 leading to the collapse of replication forks and cell death. Addition of MK-8776 from 18–24 h after a 6-h incubation with gemcitabine induced much greater sensitization than if the two drugs were incubated concurrently for 6 h. The ability of this short incubation with MK-8776 to sensitize cells is critical because of the short half-life of MK-8776 in patients’ plasma. Cell cycle perturbation was also assessed in human pancreas tumor xenografts in mice. There was a dramatic accumulation of cells in S/G 2 phase 18 h after gemcitabine administration, but cells had started to recover by 42 h. Administration of MK-8776 18 h after gemcitabine caused significantly delayed tumor growth compared to either drug alone, or when the two drugs were administered with only a 30 min interval.

          Conclusions

          There are two reasons why delayed addition of MK-8776 enhances sensitivity to gemcitabine: first, there is an increased number of cells arrested in S phase; and second, the arrested cells have adequate time to initiate recombination and thereby become Chk1 dependent. These results have important implications for the design of clinical trials using this drug combination.

          Related collections

          Most cited references26

          • Record: found
          • Abstract: found
          • Article: not found

          Targeting the replication checkpoint using SCH 900776, a potent and functionally selective CHK1 inhibitor identified via high content screening.

          Checkpoint kinase 1 (CHK1) is an essential serine/threonine kinase that responds to DNA damage and stalled DNA replication. CHK1 is essential for maintenance of replication fork viability during exposure to DNA antimetabolites. In human tumor cell lines, ablation of CHK1 function during antimetabolite exposure led to accumulation of double-strand DNA breaks and cell death. Here, we extend these observations and confirm ablation of CHK2 does not contribute to these phenotypes and may diminish them. Furthermore, concomitant suppression of cyclin-dependent kinase (CDK) activity is sufficient to completely antagonize the desired CHK1 ablation phenotypes. These mechanism-based observations prompted the development of a high-content, cell-based screen for γ-H2AX induction, a surrogate marker for double-strand DNA breaks. This mechanism-based functional approach was used to optimize small molecule inhibitors of CHK1. Specifically, the assay was used to mechanistically define the optimal in-cell profile with compounds exhibiting varying degrees of CHK1, CHK2, and CDK selectivity. Using this approach, SCH 900776 was identified as a highly potent and functionally optimal CHK1 inhibitor with minimal intrinsic antagonistic properties. SCH 900776 exposure phenocopies short interfering RNA-mediated CHK1 ablation and interacts synergistically with DNA antimetabolite agents in vitro and in vivo to selectively induce dsDNA breaks and cell death in tumor cell backgrounds.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Targeting the S and G2 checkpoint to treat cancer.

            Cell survival following DNA damage depends on activating checkpoints to arrest proliferation. Most cancer cells have dysregulated G1 checkpoints making them dependent on their S and G2 checkpoints, which are activated by ATR/Chk1 signalling. Thus, inhibiting ATR or Chk1 should selectively sensitise cancer cells to DNA damage. Genetic inactivation of ATR and Chk1 abrogates cell cycle arrest and enhances cytotoxicity following exposure to DNA-damaging agents. Similar effects were seen with small-molecule Chk1 inhibitors in preclinical studies, and clinical trial data are starting to emerge. Recently, potent ATR inhibitors have been identified that also sensitise cancer cells in vitro. ATR and Chk1 inhibitors might also cause 'synthetic lethality' in tumour cells defective in defined DNA repair pathways. Copyright © 2012 Elsevier Ltd. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The checkpoint kinases Chk1 and Chk2 regulate the functional associations between hBRCA2 and Rad51 in response to DNA damage.

              The cellular response to the introduction of double strand DNA breaks involves complexes of protein interactions that govern cell cycle checkpoint arrest and repair of the DNA lesions. The checkpoint kinases Chk1 and Chk2 phosphorylate the carboxy-terminal domain of hBRCA2, a protein involved in recombination-mediated DNA repair (HRR) and replication fork maintenance. Cells deficient in hBRCA2 are hypersensitive to DNA damaging agents. Phosphorylation of the residue in hBRCA2 targeted by the Chk1 and Chk2 kinases regulates its interaction with Rad51. Furthermore, the cell line lex1/lex2, which lacks the carboxy-terminal domain containing the phosphorylated residue, does not support localization of Rad51 to nuclear foci after exposure to UV or treatment with ionizing radiation (IR). The data show that either phosphorylation of Rad51 by Chk1 or phosphorylation of the carboxy-terminal domain of hBRCA2 by Chk1 or Chk2 plays a critical role in the binding of Rad51 to hBRCA2 and the subsequent recruitment of Rad51 to sites of DNA damage. While depletion of Chk1 from cells leads to loss of Rad51 localization to nuclear foci in response to replication arrest, cells lacking Chk2 also show a defect in Rad51 localization, but only in presence of double strand DNA breaks, indicating that each of these kinases may contribute somewhat differently to the formation of Rad51 nucleoprotein filaments depending on the type of DNA damage incurred by the cells.
                Bookmark

                Author and article information

                Journal
                BMC Cancer
                BMC Cancer
                BMC Cancer
                BioMed Central
                1471-2407
                2013
                21 December 2013
                : 13
                : 604
                Affiliations
                [1 ]Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Lebanon, NH USA
                [2 ]Duksung Women’s University, Seoul, Korea
                [3 ]Department of Radiology, Geisel School of Medicine at Dartmouth, Lebanon, NH USA
                [4 ]Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Rubin Building Level 6, Lebanon, NH USA
                Article
                1471-2407-13-604
                10.1186/1471-2407-13-604
                3878047
                24359526
                c9997999-bf94-4641-b62c-2aa09c5c146b
                Copyright © 2013 Montano et al.; licensee BioMed Central Ltd.

                This is an open access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 26 August 2013
                : 4 December 2013
                Categories
                Research Article

                Oncology & Radiotherapy
                chk1,gemcitabine,mk-8776,drug combinations,pancreas cancer xenografts,homologous recombination,cell cycle perturbation

                Comments

                Comment on this article