Elsevier

Chemico-Biological Interactions

Volume 224, 5 December 2014, Pages 128-135
Chemico-Biological Interactions

Resiniferatoxin induces death of bladder cancer cells associated with mitochondrial dysfunction and reduces tumor growth in a xenograft mouse model

https://doi.org/10.1016/j.cbi.2014.10.020Get rights and content

Highlights

  • RTX showed cytotoxic effects in bladder cancer cells.

  • RTX alters the redox homeostasis and induces necrosis in bladder cancer cells.

  • RTX reduces the tumor growth in a xenograft mouse model of bladder cancer.

  • Tumors from RTX-treated mice display reduced cell proliferation and increased necrosis.

  • RTX treatment does not induce inflammation in the tissues surrounding tumor.

Abstract

Bladder cancer (BC) is the fifth most common non-cutaneous malignancy and the most common form of BC in Western countries is transitional cell carcinoma. Resiniferatoxin (RTX) has found therapeutic usefulness for the treatment of bladder dysfunction but no data are available on its use as chemotherapeutic agent. The aim of this work is to evaluate the use of RTX as new anti-cancer drug in BC therapy. The effects of RTX on cell viability and cell death were evaluated on T24 and 5637 BC cell lines by MTT assay, cell cycle analysis, Annexin-V/PI staining and agarose gel electrophoresis of DNA. Mitochondrial depolarization and ROS production were assessed by flow cytometry. ADP/ATP ratio was measured by bioluminescence and caspase 3 cleavage by Western blot. For in vivo experiments, athymic nude mice, xenografted with T24 cells, received subcutaneous administrations of RTX. Tumor volumes were measured and immunohistochemistry was performed on tumor sections. Our data demonstrated that RTX influences cell cycle and induces necrotic cell death of BC cells by altering mitochondrial function, leading to depolarization, increase in ADP/ATP ratio and ROS production. Moreover, RTX is able to reduce tumor growth in a xenograft mouse model. Overall, we demonstrated that RTX induces necrotic cell death of BC cells and reduces tumor growth in a xenograft mouse model of BC, suggesting RTX as a new potential anti-cancer drug in BC chemotherapy.

Introduction

Bladder cancer (BC) is among the five most common malignancies worldwide. There are over 70,000 new cases of BC each year in the United States alone [1] and the most common form of BC in Western countries is transitional cell carcinoma (TCC) [2]. Approximately 30–40% of patients with high-risk non-muscle-invasive TCC of the bladder will progress to a more advanced disease within 5 years and up to 34% of them will ultimately die of bladder cancer [3]. Overall, only 20–40% of patients with advanced TCC have a 5-year survival rate, despite aggressive multimodal therapy and radical cystectomy remain the mainstay of treatment of muscle-invasive disease [4], [5]. To ameliorate patient life expectancy, improvement of current chemotherapeutic regimens and development of novel chemotherapeutic strategies are necessary. Recently, different therapeutic approaches based on targeting tumor mitochondria have been proposed [6], with the expectation that this novel class of agents could reduce tumor cells viability with an acceptable therapeutic index [7].

Resiniferatoxin (RTX) is a diterpene found in the latex of the cactus Euphorbia resinifera containing a homovanillic acid ester, a key structural motif of capsaicin, displaying analgesic activity and functioning as an ultrapotent capsaicin analog. RTX has found therapeutic usefulness in the urologic field in the treatment of bladder dysfunctions and painful bladder [8], [9]. Very few data are available on its use as chemotherapeutic agent. The anticancer activity of vanilloids such as capsaicin and dihydrocapsaicin can be mediated through both a direct pathway, independent of transient receptor potential vanilloid receptor 1 (TRPV1), the receptor for vanilloids, and an indirect pathway, through the interaction with TRPV1 and the subsequent intracellular calcium overload [10], [11], [12], [13], [14], [15], [16]. In regard to RTX, there are indications that mitochondria could be involved in the TRPV1-independent vanilloids-induced cell death. In pancreatic cancer tissue [13], squamous cell carcinoma and non-small lung cancer cell lines [14], [17] RTX causes non-vanilloid receptor mediated cell death.

This work is aimed to evaluate the potential use of RTX as new therapeutic strategy against BC through in vitro and in vivo pre-clinical experiments.

Section snippets

Cell lines

The p53 mutant T24 TCC and 5637 grade II BC cell lines, purchased from American Type Culture Collection (ATCC, Rockville, MD, USA), were maintained in RPMI-1640 medium (Lonza, Basel, Switzerland) supplemented with 10% heat-inactivated fetal bovine serum, 2.5 mM HEPES, 2 mM l-glutamine, 100 IU/ml of penicillin, 100 g/ml of streptomycin (Lonza) at 37 °C, 5% CO2 and 95% humidity. Normal human urothelial cells (NHUC) were purchased from ScienceCell Research laboratories (Carlsbad, CA, USA) and cultured

RTX induces necrotic cell death of BC cells independently from TRPV1

We initially evaluated the effects of different RTX doses (0.1–50 μM) on the viability of T24 and 5637 cell lines. As shown in Fig. 1a, RTX is able to reduce dose-dependently the growth of both cell lines at 24 h (IC50: 21.5 for T24 cells and 19.9 μM for 5637 cells). Since T24 but not 5637 cells [17], [18] express TRPV1 channel, we used RTX in combination with 5′-iodoresiniferatoxin (I-RTX), a strong competitive antagonist of TRPV1 receptor, to verify the involvement of TRPV1 in T24 cell growth

Discussion

We demonstrate for the first time that RTX is able to induce cell cycle arrest and necrotic cell death associated with mitochondrial dysfunction in different BC cell lines and, more importantly, it reduces in vivo the growth of human BC cells xenografted into nude mice. Thus, RTX can be considered as a new potential molecule for BC therapy. These results appear to be significant if we consider that, despite several efforts have been made for the development of new effective therapies, TCC of

Conclusion

Overall, we demonstrated that RTX treatment in vitro induces necrotic cell death of BC cells by affecting redox homeostasis and in vivo reduces tumor growth in a xenograft mouse model of BC. Therefore, since the high adaptability of RTX to a variety of pain problems and the relatively low toxicity when used locally (e.g. intravesical or peritumoral administration), it could represent a new promising strategy for the treatment of patients with BC.

Conflict of Interest

The authors declare that there are no conflicts of interest.

Transparency Document

.

Acknowledgements

This work was supported by FIRC national grant (number 11095) and by a grant from the Italian Ministry of University and Research, PRIN 2009–2011.

References (31)

  • H.B. Grossman et al.

    Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer

    N. Engl. J. Med.

    (2003)
  • L. Galluzzi et al.

    Mitochondria as therapeutic targets for cancer chemotherapy

    Oncogene

    (2006)
  • F. Cruz et al.

    Resiniferatoxin and botulinum toxin type A for treatment of lower urinary tract symptoms

    Neurourol. Urodyn.

    (2007)
  • P.K. Matsuoka et al.

    Intravesical treatment of painful bladder syndrome: a systematic review and meta-analysis

    Int. Urogynecol. J.

    (2012)
  • G. Santoni et al.

    TRPV channels in tumor growth and progression

    Adv. Exp. Med. Biol.

    (2011)
  • Cited by (12)

    • Natural products as a perspective for cancer pain management: A systematic review

      2019, Phytomedicine
      Citation Excerpt :

      Another fact that can contribute with the reduction of cancer pain is the antitumor activity of these compounds, once the tumor growth can compress nerves and nerve endings, causing pain. The antitumor potential of the 12 natural products involved in this review was analyzed and it was observed that many of them perform antitumor activity or cytotoxicity on tumor cells, such as carvacrol (Khan et al., 2018; Fan et al., 2015; Dai et al., 2016), triptolide (Zhang et al., 2018; Li et al., 2018; Huang 2018), resiniferatoxin (Farfariello et al., 2014), tanshinone IIA (Yen et al., 2018; Li et al., 2017; Zhang et al., 2017), euphol (Wang et al., 2013; Silva et al., 2018), quercetin (Calgarotto et al., 2018; Jia et al., 2018; Sun et al., 2018), morin (Jin et al., 2014; Gupta et al., 2013) and epigallocatechin-3-galate (Gan et al., 2017; Liu et al., 2017; Kwak et al., 2016). Besides, a literature review showed that cannabinoids inhibit tumor cell viability, invasion, metastasis and angiogenesis (McAllister et al., 2015) and may act as direct antitumor agents in a variety of aggressive cancers (Martín-Banderas et al., 2015).

    • Wittig derivatization of sesquiterpenoid polygodial leads to cytostatic agents with activity against drug resistant cancer cells and capable of pyrrolylation of primary amines

      2015, European Journal of Medicinal Chemistry
      Citation Excerpt :

      This undesirable mismatched interaction may well account for the loss of TRPV1 activity for 5. Although many reports investigating vanilloid agonists as potential anticancer agents have appeared in the literature, the involvement of TRPV1 in mediating their antiproliferative effects has been questioned on many occasions as vanilloid antagonists have consistently failed to prevent capsaicin or RTX-induced cell death [33–37]. In the related publication [55], we show that these observations also apply to α,β-unsaturated 1,4-dialdehyde terpenoids, such as 1 and 9-epipolygodial, and that cancer cell death induced by these compounds cannot be explained by TRPV1-targeting.

    • Anticancer Activity of Region B Capsaicin Analogs

      2023, Journal of Medicinal Chemistry
    • The impact of trpv1 on cancer pathogenesis and therapy: A systematic review

      2021, International Journal of Biological Sciences
    View all citing articles on Scopus
    1

    Valerio Farfariello and Sonia Liberati: equal contribution.

    View full text