Cancer Letters

Cancer Letters

Volume 318, Issue 2, 28 May 2012, Pages 145-153
Cancer Letters

Targeting heat shock factor 1 with a triazole nucleoside analog to elicit potent anticancer activity on drug-resistant pancreatic cancer

https://doi.org/10.1016/j.canlet.2011.09.043Get rights and content

Abstract

Issued from a lead optimization process, we have identified a novel triazole nucleoside analog which elicits potent anticancer activity on drug-resistant pancreatic cancer. Most importantly, this compound targets heat shock response pathways by down-regulation of heat shock transcription factor 1 and consequential down-regulation of multiple heat shock proteins HSP27, HSP70 and HSP90. Down-regulation of these proteins caused the shut-down of several oncogenic pathways and caspase-dependent apoptosis resulting in a potent anticancer effect in vitro and in vivo. These results demonstrate the potential rewards gained in searching for anticancer candidates with multimodal actions on heat shock response pathways via HSF1 down-regulation.

Introduction

When subjected to insults such as heat shock or the exposure to heavy metal ions or cytotoxic agents, cells respond by over-expressing heat shock proteins (HSPs), which protect the cells and enable them to survive under otherwise stressful and lethal conditions [1], [2]. The whole process and related events is denoted the heat shock response (HSR). Regulation of the HSR is complicated and is linked to multiple cellular signaling pathways. While many questions remain concerning the HSR, increasing evidence suggests that heat shock factors (HSFs) mediate the inducible heat shock gene expression at the transcriptional level and hence regulate the HSR [3]. The most highly expressed HSFs in mammalian cells include HSF1, HSF2 and HSF4. Though all of them have been identified to modulate HSP expression, HSF1 is the most important regulator of the HSR in response to various stimuli [3], [4], [5]. Indeed HSF1 can regulate a multitude of signaling and metabolic pathways to promote the survival of cells. In addition, cancer cells are more dependent on HSF1 than normal cells for proliferation and survival due to the constant exposure of cancer cells to stresses both from internal oncogenic processes (such as mutated proteins and deregulated signal pathways) and external environmental factors (such as hypoxia and nutrient deprivation) [3]. Consequently, targeting HSF1 has attracted considerable attention as an appealing anticancer approach [3], [5], [6].

As the dominant upstream transcriptional regulator for stress inducible HSPs, HSF1 activates their transcription and regulates their expression in response to stress. Among the superfamily of HSPs, HSP90, HSP70 and HSP27 chaperones are particularly attractive as anticancer targets due to their function as master regulators of multiple oncoproteins and their tumorigenic properties [7], [8], [9], [10], [11], [12], [13], [14]. Different inhibitors of HSP90, mainly derived from the natural product geldanamycin, have led to encouraging results in clinical trials on multiple myeloma and gastrointestinal stromal tumors [15]. Antisense oligonucleotides developed to down-regulate HSP27 have also been demonstrated to show effective anticancer activity [16], [17], [18] and are at advanced stages in clinical trials for use in the treatment of several human cancers including lung, breast, prostate, bladder and ovarian [19]. As for HSP70, different types of inhibitors have been reported including small molecular chemical entities, antisenses and siRNAs, peptides, aptamers, etc. [14], although no candidate has yet entered into clinical trials for cancer treatment.

While current anticancer strategies focus mainly on the inhibition of individual HSPs, we believe that targeting multiple HSPs at once or their upstream controlling genes may be more effective. Given that HSF1 and HSPs form the HSR network and are up-regulated in cancer to promote cell survival and development, we postulated that targeting the heat shock response (HSR) pathways via the down-regulation of HSF1 might allow the simultaneous down-regulation of several HSPs, cause depletion of multiple oncoproteins and attack several pathways necessary for cancer development. This would ultimately lead to a more efficacious anticancer activity, while at the same time, reducing the likelihood of the tumor acquiring resistance to any single therapeutic pathway [14], [15], [20], [21].

In the present work, we disclose our finding of a triazole nucleoside analog which targets HSF1-mediated heat shock response pathways leading to the simultaneous down-regulation of the HSPs: HSP27, HSP70 and HSP90, and consequent depletion of their corresponding oncogenic client proteins. This triazole nucleoside analog yields effective caspase-dependent apoptosis with a potent anticancer effect in the drug-resistant pancreatic cancer model in vitro and in vivo. It is important to mention here that pancreatic cancer is one of the most lethal and devastating human cancers, constituting a major unsolved health problem worldwide [22], [23]. Conventional treatments have little impact on this disease due to its poor diagnosis, aggressive metastasis and rapid development of drug-resistance. The current standard chemotherapy for patients with advanced pancreatic cancer is the nucleoside drug gemcitabine [24], [25], though it is only moderately effective and results in a mere 12% response and 3% overall survival rate with a median survival of less than 6 months. Consequently, there is a pressing need to explore more efficacious drug candidates with novel modes of action to combat pancreatic cancer, particularly its drug-resistant form. Recently HSF1 was reported to play a pro-survival role in the pathogenesis of pancreatobiliary tumors [26]. Inhibition of HSF1 expression by the HSF1 siRNA sequences led to time-dependent death in pancreatic cancer cell lines in which a caspase-dependent apoptotic pathway was activated, whereas did not significantly decrease the viability of immortalized pancreatic ductal cells [26]. This hence suggested that down-regulation of HSF1 could emerge as cancer-specific therapy for pancreatic cancer. Targeting HSF1 using the triazole nucleoside disclosed here may therefore constitute a promising novel strategy.

Section snippets

Cell culture and reagents

Human pancreatic cancer MiaPaCa-2 and Panc-1 cells were purchased from American Type Culture Collection. The cells were grown in Dulbecco’s modified eagle’s medium (DMEM) (Gibco) supplemented with 10% fetal bovine serum (FBS). Gemcitabine (Gemzar) was purchased from Lilly France, SAS (Suresnes, France). (3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide, MTT) and Actinomycin D was purchased from Sigma–Aldrich. PE Annexin V and 7-aminoactinomycin D (7-AAD) was purchased from BD

Lead optimization results in more efficacious candidates

We have previously identified a triazole nucleoside analog 1 (Fig. 1A) which effectively down-regulated HSP27 and resulted in apoptosis-associated anticancer activity against drug-resistant pancreatic cancer [27]. During our optimization process based on 1, we found that by varying the alkyl chain length we could critically modulate the anticancer activity: the analogs 2 and 3 bearing shorter alkyl chains were almost inactive or showed little activity, whereas compounds 4 and 5 (Fig. 1A)

Discussion

The targeting of individual HSPs constitutes an emerging trend in the search for efficacious anticancer candidates with novel mechanisms of action, however these inhibitors sometimes cause counterproductive effects on anticancer efficiency via their induction of other HSPs. An example of this is shown with one of the earliest HSP90 inhibitors, 17-AAG, which was found to up-regulate the expression of HSP70, a HSP with strong cytoprotective properties [15]. As HSF1 is the master transcription

Conclusion

In conclusion, we identified a novel triazole nucleoside which can effectively target HSF1 leading to the simultaneous down-regulation of HSP27, HSP70 and HSP90. Following the suppression of these HSPs, their client oncoproteins such as STAT3 and AKT were also significantly depleted. The final global consequence was a caspase-dependent induction of apoptosis and a potent anticancer effect on drug-resistant pancreatic cancer in vitro and in vivo. To our knowledge, this is the first triazole

Acknowledgments

This project is supported by the National Mega Project on Major Drug Development (2009ZX09301-014), La Ligue Contre le Cancer (BL-8670), Wuhan University, CNRS, INSERM and INSERM Transfert. Y.X. is supported by la Fondation pour la Recherche Médicale, M.W. le program de bourses d’excellene d’Eiffel, and Y.F. the China Scholarship Council. We thank Joël Tardivel-Lacombe for his assistance with cell culture, and Sandrine Henri for the help with radioactivity experiments and Drs. Emily Witty and

References (45)

  • M. Wang et al.

    A novel arylethynyltriazole acyclonucleoside inhibits proliferation of drug-resistant pancreatic cancer cells

    Bioorg. Med. Chem. Lett.

    (2010)
  • Y. Xia et al.

    Targeting heat shock response pathways to treat pancreatic cancer

    Drug Discov. Today

    (2012)
  • D.A. Parsell et al.

    The function of heat-shock proteins in stress tolerance. Degradation and reactivation of damaged proteins

    Annu. Rev. Genet.

    (1993)
  • R.I. Morimoto et al.

    Stress-inducible responses and heat shock proteins: new pharmacologic targets for cytoprotection

    Nat. Biotechnol.

    (1998)
  • M. Åkerfelt et al.

    Heat shock factors: integrators of cell stress, development and lifespan

    Nat. Rev. Mol. Cell Biol.

    (2010)
  • L. Pirkkala et al.

    Roles of the heat shock transcription factors in regulation of the heat shock response and beyond

    FASEB J.

    (2001)
  • L. Whitesell et al.

    Inhibiting the transcription factor HSF1 as an anticancer strategy

    Expert Opin. Ther. Targets

    (2009)
  • E. de Billy et al.

    Drugging the heat shock factor 1 pathway: exploitation of the critical cancer cell dependence on the guardian of the proteome

    Cell Cycle

    (2009)
  • L. Whitesell et al.

    HSP90 and the chaperoning of cancer

    Nat. Rev. Cancer

    (2005)
  • C. Garrido et al.

    Heat shock proteins 27 and 70: anti-apoptotic proteins with tumorigenic properties

    Cell Cycle

    (2006)
  • D. Mahalingam et al.

    Targeting HSP90 for cancer therapy

    Br. J. Cancer

    (2009)
  • D.R. Ciocca et al.

    Heat shock proteins in cancer: diagnostic, prognostic, predictive, and treatment implications

    Cell Stress Chap.

    (2005)
  • Cited by (56)

    • Advance of structural modification of nucleosides scaffold

      2021, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Furthermore, bis-triazole-containing derivatives have similar activity to Ribavirin against tobacco mosaic virus (TMV) [121–123] (Fig. 18). The other compounds not only have good inhibitory effect on the proliferation of tumor cells, but also show different mechanisms of action from those of conventional nucleoside drugs, such as restriction of heat shock-related proteins, androgen receptors, transcription, and immune regulation [124–129]. The alkenyl compounds 165, 166, 168, 169 and the N aryl compounds 167, 170, 171, 172 exhibit potential viabilities without significant side effects on the drug-resistant pancreatic cancer MiaPaCa-2 cell line, of which 166, 167, 170, 171 and 172 are with a better potency than Gemcitabine (24).

    • Inhibiting Heat Shock Factor 1 in Cancer: A Unique Therapeutic Opportunity

      2019, Trends in Pharmacological Sciences
      Citation Excerpt :

      The MoA of dorsomorphin on the HSF1 pathway is unknown and there is no evidence to suggest a direct interaction between dorsomorphin and HSF1. CCT251236, PW3405, VM1, NZ28, and Emunin are small molecules identified through screening efforts, which inhibit the ‘HSF1 pathway’ at varying potency and with distinct phenotypic outcomes [101–105] (Table 1). Similar to the previously discussed molecules, there is no clear MoA for these compounds, raising concerns about specificity for HSF1.

    View all citing articles on Scopus
    View full text