Cancer Letters

Cancer Letters

Volume 356, Issue 2, Part B, 28 January 2015, Pages 656-668
Cancer Letters

Original Articles
Synergistic antitumor interactions between MK-1775 and panobinostat in preclinical models of pancreatic cancer

https://doi.org/10.1016/j.canlet.2014.10.015Get rights and content

Abstract

Pancreatic cancer remains a clinical challenge, thus new therapies are urgently needed. The selective Wee1 inhibitor MK-1775 has demonstrated promising results when combined with DNA damaging agents, and more recently with CHK1 inhibitors in various malignancies. We have previously demonstrated that treatment with the pan-histone deacetylase inhibitor panobinostat (LBH589) can cause down-regulation of CHK1. Accordingly, we investigated using panobinostat to down-regulate CHK1 in combination with MK-1775 to enhance cell death in preclinical pancreatic cancer models. We demonstrate that MK-1775 treatment results in increased H2AX phosphorylation, indicating increased DNA double-strand breaks, and activation of CHK1, which are both dependent on CDK activity. Combination of MK-1775 and panobinostat resulted in synergistic antitumor activity in six pancreatic cancer cell lines. Finally, our in vivo study using a pancreatic xenograft model reveals promising cooperative antitumor activity between MK-1775 and panobinostat. Our study provides compelling evidence that the combination of MK-1775 and panobinostat has antitumor activity in preclinical models of pancreatic cancer and supports the clinical development of panobinostat in combination with MK-1775 for the treatment of this deadly disease.

Introduction

Pancreatic cancer remains a very difficult disease to treat. The fourth most lethal cancer in the United States, pancreatic cancer has a grim prognosis, with a 5-year survival rate of 6% [1]. Standard chemotherapeutic care for pancreatic cancer involves treatment with gemcitabine, a nucleoside analogue, but offers only modest benefit [2]. Furthermore, combinations of other cytotoxic agents with gemcitabine have, in general, offered little improvement [3]. Thus, there is an obvious need to develop more effective therapy for this disease.

Cell cycle progression is a tightly regulated process. The cell cycle is driven by the activation of various cyclin dependent kinases (CDKs), which are regulated by a combination of cyclin expression and inhibitory phosphorylation. The Wee1 kinase is capable of phosphorylating CDK1 and CDK2 on Tyr-15 (Y15), preventing progression through G2/M and S phase, respectively [4], [5]. This phosphorylation can be removed by the CDC25 phosphatases, which are in turn inactivated when phosphorylated by CHK1 [6], [7]. Finally, CHK1 activity is controlled primarily by ATR kinase, which activates CHK1 by phosphorylation upon sensing replication stress or DNA damage [8], [9]. Thus, both the CHK1 and the Wee1 pathways contribute to maintaining inhibitory phosphorylation of CDKs.

MK-1775, the first potent and selective inhibitor of Wee1, has been investigated primarily as an agent that can target the G2/M checkpoint to exert toxicity specifically in p53-mutant cells [10]. It has been shown that, when combined with DNA damaging agents, MK-1775 is able to abrogate the G2/M checkpoint and enhance apoptosis, although recent studies have questioned the p53-dependence of these effects [10], [11], [12], [13], [14], [15], [16], [17], [18]. Importantly, it has been shown that Wee1 inhibition alone can induce DNA damage, likely through the induction of replication stress secondary to overactive CDKs and inhibition of DNA repair [19].

Histone deacetylase (HDAC) inhibitors (HDACIs) have been shown in vitro to have promising anti-cancer activity, but their single-agent effectiveness in the clinic has been only modest [20], [21], [22], [23], [24], [25], [26], [27]. However, there are many clinical trials investigating the role of HDACIs in combination therapies (NCT01242774, NCT01742793, NCT02061449, and NCT02145715, clinicaltrials.gov). Previous work from this lab has demonstrated the ability of HDACIs to synergize with standard chemotherapeutic agents, at least in part by enhancing DNA damage [28], [29], [30], [31]. Importantly, we recently demonstrated that treatment with the pan-HDACI panobinostat (LBH589) was able to down-regulate CHK1 [28], [29]. It has been reported that combined inhibition of Wee1 and CHK1 is effective at inducing cancer cell death [13], [32], [33], [34], leading us to consider the combination of MK-1775 and panobinostat for the treatment of pancreatic cancer.

In this work, we use pre-clinical pancreatic cancer models to investigate the effects of the combination of MK-1775 and panobinostat, and the mechanism by which panobinostat enhances MK-1775-induced apoptosis. We demonstrate that MK-1775 alone is able to induce DNA damage and activate CHK1 in a CDK-dependent fashion. Panobinostat treatment down-regulates CHK1 and synergizes with MK-1775 to enhance apoptosis and cell growth inhibition. Importantly, we demonstrate that in some cell lines, the CHK1 pathway is able to overcome single agent Wee1 inhibition and maintain phosphorylation of CDK1. This demonstrates a potential mechanism of resistance to treatment with MK-1775 and emphasizes the importance of combinations with agents such as panobinostat.

Section snippets

Drugs

MK-1775, panobinostat, LY2603618, and roscovitine were purchased from Selleck Chemicals (Houston, TX, USA).

Cell culture

The AsPC-1, BxPC-3, CFPAC-1, HPAC, MIAPaCa-2 and PANC-1 human pancreatic cancer cell lines were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA) and cultured as previously described [35]. The cell lines were authenticated by the University of Arizona Genetics Core Facility (Tucson, AZ, USA).

In vitro cytotoxicity assays

In vitro cytotoxicities of MK-1775, panobinostat, roscovitine, and

MK-1775 induces DNA damage and activates CHK1

To investigate the effects of MK-1775 treatment in pancreatic cancer cells, we first determined the expression levels of Wee1, PKMyt-1, p-CDK1, CDK1, p-CDK2, and CDK2 in 6 pancreatic cell lines by Western blot. The majority of cell lines expressed variable levels for all of the proteins (Fig. 1A). Then we determined MK-1775 sensitivity by MTT assays. MK-1775 IC50s varied, ranging from 470 nM (HPAC) to 13.2 µM (ASPC-1) (Fig. 1, Table 1).

Next, we treated the pancreatic cancer cell lines with

Discussion

Pancreatic cancer's poor prognosis highlights the need for new therapies. Recent studies have demonstrated that MK-1775 treatment results in increased phosphorylation of both H2AX and CHK1 [13], [33], [34], which we confirmed in six pancreatic cancer cell lines. We found that the increased phosphorylation of CHK1 and H2AX following MK-1775 treatment was dependent on CDK activity. Furthermore, we found that a CHK1 selective inhibitor (LY2603618) enhanced MK-1775 sensitivity in a synergistic

Conflict of interest

The authors declare no competing financial interests.

Acknowledgements

This study was supported by a Start-up Fund from Jilin University, Changchun, China, and a grant from the National Natural Science Foundation of China, NSFC 31271477. Mr. JTC is a predoctoral trainee supported by T32 CA009531 from the National Cancer Institute. The funders had no role in study design, data collection, analysis and interpretation of data, decision to publish, or preparation of the manuscript.

References (49)

  • H.A. Burris et al.

    Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial

    J. Clin. Oncol

    (1997)
  • T. Conroy et al.

    FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer

    N. Engl. J. Med

    (2011)
  • L.L. Parker et al.

    Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase

    Science

    (1992)
  • N. Watanabe et al.

    Regulation of the human WEE1Hu CDK tyrosine 15-kinase during the cell cycle

    EMBO J.

    (1995)
  • Y. Sanchez et al.

    Conservation of the Chk1 checkpoint pathway in mammals: linkage of DNA damage to Cdk regulation through Cdc25

    Science

    (1997)
  • C.Y. Peng et al.

    Mitotic and G2 checkpoint control: regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216

    Science

    (1997)
  • Q. Liu et al.

    Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint

    Genes Dev

    (2000)
  • H. Zhao et al.

    ATR-mediated checkpoint pathways regulate phosphorylation and activation of human Chk1

    Mol. Cell. Biol

    (2001)
  • H. Hirai et al.

    Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents

    Mol. Cancer Ther

    (2009)
  • M. Aarts et al.

    Forced mitotic entry of S-phase cells as a therapeutic strategy induced by inhibition of WEE1

    Cancer Discov

    (2012)
  • K.A. Bridges et al.

    MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells

    Clin. Cancer Res

    (2011)
  • L. Chaudhuri et al.

    CHK1 and WEE1 inhibition combine synergistically to enhance therapeutic efficacy in acute myeloid leukemia ex vivo

    Haematologica

    (2014)
  • A.D. Guertin et al.

    Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy

    Mol. Cancer Ther

    (2013)
  • H. Hirai et al.

    MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil

    Cancer Biol. Ther

    (2010)
  • Cited by (32)

    • Targeting Wee1 kinase as a therapeutic approach in Hematological Malignancies

      2021, DNA Repair
      Citation Excerpt :

      Furthermore, due to the effects of AZD1775 on the DDR pathway, which leads to increased DNA damage and cell death, it has synergistic effects in combination with DDR pathway targeting agents such as CHK1 and ATR inhibitors [82,83]. Moreover, synergistic effects in simultaneous inhibition of Wee1 kinase in combination with epigenetic modification targeting agents (Panobinostat, Vorinostat) have been reported [23,84]. There is evidence that AZD1775 increases DNA damages and sensitizes tumor cells to immunotherapeutic approaches, in which activated lymphocytes are inserting antitumor effects by recruiting co-stimulatory molecules [85].

    • Targeting replication stress in cancer therapy

      2023, Nature Reviews Drug Discovery
    View all citing articles on Scopus
    1

    G.W. and X.N. contributed equally to this study.

    View full text